CN118141804A - Use of indole-3-propionic acid in preparing medicament for sensitization tumor immunotherapy - Google Patents

Use of indole-3-propionic acid in preparing medicament for sensitization tumor immunotherapy Download PDF

Info

Publication number
CN118141804A
CN118141804A CN202410254831.5A CN202410254831A CN118141804A CN 118141804 A CN118141804 A CN 118141804A CN 202410254831 A CN202410254831 A CN 202410254831A CN 118141804 A CN118141804 A CN 118141804A
Authority
CN
China
Prior art keywords
indole
propionic acid
tumor
pharmaceutical composition
mice
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202410254831.5A
Other languages
Chinese (zh)
Inventor
王良静
陈淑洁
贾丁佳成
王其温
戚亚东
王岚
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Zhejiang University ZJU
Original Assignee
Zhejiang University ZJU
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Zhejiang University ZJU filed Critical Zhejiang University ZJU
Priority to CN202410254831.5A priority Critical patent/CN118141804A/en
Publication of CN118141804A publication Critical patent/CN118141804A/en
Pending legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention belongs to the technical field of medicines, relates to application of indole-3-propionic acid in preparing a medicament for sensitization immunotherapy, and in particular relates to application of indole-3-propionic acid and a medicament containing the same in preparing a product for sensitization anti-PD-1 antibody. According to the research, the indole-3-propionic acid can obviously sensitize the mice for immunotherapy, the mice tumors are obviously reduced, the infiltrated CD8 + T cells are obviously increased, and the experimental mice do not show abnormal reaction, so that the indole-3-propionic acid can sensitize the mice for immunotherapy. The indole-3-propionic acid is a natural substance existing in organisms, has no heterogeneity and serious adverse reaction, has wide application potential, can be clinically combined with an immune checkpoint inhibitor, and provides a new strategy for preventing or treating tumors.

Description

Use of indole-3-propionic acid in preparing medicament for sensitization tumor immunotherapy
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to application of indole-3-propionic acid in preparation of a medicine for sensitized tumor immunotherapy.
Background
The tumor immunotherapy can restart the normal immune response of the organism, reduce the immune escape and the immune tolerance of the tumor, and effectively control and clear the tumor. Various immune checkpoint inhibitor drugs (ICIs), such as programmed cell death protein-1 (PD-1) and its ligand PD-L1 antibodies, have been used in clinical tumor immunotherapy. ICIs can enhance T cell activation, thereby increasing the toxic killing effect of immune cells on tumor cells. However, solid tumors such as colorectal cancer have low efficacy against PD-1 antibodies, which are closely related to the tumor immunosuppressive microenvironment and CD8 + T cells in tumor tissue. Multiple CD8 + T cell subsets have been found to affect anti-tumor responses. In particular, depletion of precursor T cells of TCF-1 +, characterized by high expression of the transcription factor T-cytokine 1 (TCF-1, encoded by TCF 7), is a major subset of responses to immunotherapy. The population of cells has an immune stem characteristic and is capable of self-renewal, proliferation and differentiation into effector CD8 + T cells, thereby limiting the occurrence of tumors. Since a clear correlation between Tcf7 deficiency and disruption of gut microbiota balance was observed in mice, a further study of whether gut microbiota and its metabolites could modulate the stem cell program of CD8 + T cells was needed.
Various tryptophan-indole metabolites of intestinal microbial origin, including indole-3-lactic acid (ILA), indole-3-acrylic acid (IA), indole-3-propionic acid (IPA), indole-3-acetic acid (IAA), and indole-3-acetamide (IAM), etc., have been demonstrated to have a broad range of biological effects. Specifically, IPA improves atherosclerosis, reduces radioactive toxicity and prolongs post-traumatic survival, but the effects of IPA on the management of cancer immunotherapy have not been reported.
Indole-3-propionic acid (Indolepropionic acid, IPA, CAS:830-96-6; molecular weight: 189.21; chemical formula: C 11H11NO2) is a ring of tryptophan indole metabolic pathway, and indole and its derivatives have been found to modulate immune microenvironment and are used in the prevention or treatment of rheumatoid arthritis and radiotherapy side-effects, but their effect on tumor immunotherapy is not known.
Various Immune Checkpoint Inhibitor (ICIs) drugs such as apoptosis protein-1 (PD-1) and its ligand PD-L1 mab have been used in clinical tumor immunotherapy. ICIs can enhance T cell activation, thereby increasing the toxic killing effect of immune cells on tumor cells. However, only about 15% of colorectal cancer (Colorectal Cancer, CRC) patients benefit from immune checkpoint blocking treatment, with a strong individual heterogeneity. Infiltration of T cells in colorectal cancer may better predict the patient's immunotherapeutic response compared to the mismatch repair deficiency (dMMR) or microsatellite instability (MSI) status of the tumor. Increasing evidence suggests that fecal transplantation or intestinal strain supplementation can affect tumor immunotherapy outcomes, such as fecal transplantation promoting therapeutic response of melanoma patients to PD-1 mab; bifidobacteria act synergistically to reduce tumor burden in mice; clostridium nucleatum enhances the therapeutic effect of PD-L1 mab in tumors, etc. Therefore, searching for immune checkpoint inhibitor sensitization pathway based on intestinal flora is expected to provide a new strategy for accurate immunotherapy of colorectal cancer.
In conclusion, the low effective rate of immune checkpoint inhibitors in solid tumors limits their wide clinical application. In view of this, there is a need to develop a product that is capable of sensitizing immune checkpoint inhibitors.
Disclosure of Invention
The first object of the present invention is to provide the use of indole-3-propionic acid in the manufacture of a medicament for use in the immunotherapy of sensitized tumors.
Further, the drug includes, but is not limited to, indole-3-propionic acid, and can also include pharmaceutically acceptable salts, or solvates, or prodrugs, or metabolites thereof;
further, the immunotherapy includes, but is not limited to, anti-PD-1 antibody therapy;
further, the tumors include, but are not limited to, colorectal cancer, breast cancer and melanoma.
In another aspect, the invention provides a medicament or pharmaceutical composition for use in the immunotherapy of a sensitized tumor, comprising indole-3-propionic acid and one or more pharmaceutically acceptable carriers, or comprising indole-3-propionic acid and one or more sensitizing immunotherapeutic agents.
Further, the medicament or pharmaceutical composition may also include, but is not limited to, a pharmaceutically acceptable salt of indole-3-propionic acid, or a solvate thereof, or a prodrug thereof, or a metabolite thereof;
further, the immunotherapy includes, but is not limited to, anti-PD-1 antibody therapy;
further, the tumors include, but are not limited to, colorectal cancer, breast cancer and melanoma;
Still further, the pharmaceutically acceptable carrier includes, but is not limited to, one or more of solvents, solubilizers, co-solvents, emulsifiers, flavoring agents, colorants, binders, disintegrants, fillers, lubricants, wetting agents, tonicity adjusting agents, pH adjusting agents, stabilizers, surfactants, preservatives, and the like;
further, the medicament or the pharmaceutical composition is a solid preparation, a semisolid preparation or a liquid preparation;
Still further, the solid formulations include, but are not limited to, tablets, capsules, granules, pills, and the like; the semi-solid formulations include, but are not limited to, gels, suppositories, ointments, and the like; such liquid formulations include, but are not limited to, emulsions, mixtures, suspensions, solutions, and the like.
Further, the drug or the drug composition is administered by intraperitoneal injection, oral lavage or intratumoral injection, wherein the dose of the indole-3-propionic acid is as follows: intraperitoneal injection or oral lavage is 60 mg/kg/day; intratumoral injection is 5-50 mu m, 50 mu L/day.
The invention has at least the following advantages and beneficial effects:
According to the invention, researches show that the indole-3-propionic acid has a remarkable sensitization effect on mouse immunotherapy, and can obviously reduce mouse tumors. The indole-3-propionic acid is a natural substance existing in organisms, has no heterogeneity and serious adverse reaction, and has wide application potential.
Drawings
FIG. 1 is an oral gavage indole-3-propionic acid sensitized colorectal carcinoma subcutaneous tumor model immunotherapy of example 1; graphically, a graph of tumor growth in mice is shown.
FIG. 2 is a model immunotherapy of example 2 against subcutaneous tumors of colorectal cancer sensitized by intratumoral injection of indole-3-propionic acid; in the figure, A is an intratumoral injection molding drawing; b is a graph of tumor growth in mice.
FIG. 3 is a comparison of the sensitization effect of the abdominal cavity injection of indole-3-propionic acid and indole-3-lactic acid in example 3 on colorectal carcinoma subcutaneous tumor model immunotherapy; graphically, a graph of tumor growth in mice is shown.
FIG. 4 is a graph showing that indole-3-propionic acid increases CD8 + T cell numbers in colorectal cancer immune microenvironment in example 4; in the figure, A is a representative graph of CD8 + T flow cytometry analysis in tumors; b is a statistical graph of the number of CD8 + T cells in the tumor.
FIG. 5 is an indole-3-propionic acid sensitized breast cancer subcutaneous tumor model immunotherapy of example 5; in the figure, A is a graph of tumor growth in mice; b is a statistical chart of the number of CD8 + T cells in the tumor; c is a statistical graph of the number of TCF-1 +CD8+ T cells in the tumor.
FIG. 6 is an indole-3-propionic acid sensitized melanoma subcutaneous tumor model immunotherapy of example 6; in the figure, A is a graph of tumor growth in mice; b is a statistical chart of the number of CD8 + T cells in the tumor; c is a statistical graph of the number of TCF-1 +CD8+ T cells in the tumor.
FIG. 7 is an in situ tumor model immunotherapy of indole-3-propionic acid sensitized breast cancer in example 7; in the figure, A is a graph of tumor growth in mice; b is a statistical chart of the number of CD8 + T cells in the tumor; c is a statistical graph of the number of TCF-1 +CD8+ T cells in the tumor.
FIG. 8 is an illustration of an indole-3-propionic acid sensitized breast cancer spontaneous tumor model immunotherapy of example 8; the graph is shown as a mouse tumor survival graph.
Detailed Description
The invention is described in further detail below with reference to the drawings and the specific embodiments.
Example 1: oral gavage indole-3-propionic acid sensitized colorectal cancer subcutaneous tumor model immunotherapy
Male C57BL/6 mice of 6-8 weeks are randomly divided into 4 groups, 7 mice in each group are freely drunk for 1 week before molding, drinking water containing 2mg/mL of metronidazole, 2mg/mL of penicillin, 2mg/mL of streptomycin and 1mg/mL of vancomycin, and the homogenized intestinal flora is respectively administrated with 0mg/kg of indole-3-propionic acid (control group), 6mg/kg (low dose group), 60mg/kg (medium dose group) and 300mg/kg (high dose group) of stomach according to the weight of the mice once per day. On the day of gavage, 1×10 6 of the mouse colon cancer cell line Mc38 cells were injected subcutaneously (100 μl per mouse). After 8 days of injection, 100 μg of anti-PD-1 antibody was given to each mouse intraperitoneally, one injection was given every three days, and tumor volumes were monitored every two days, as calculated as follows: volume = 0.5 xlxw 2, where L is the longest diameter and W is the shortest diameter. The experiment was terminated every other day after three PD-1 treatments, and tumor volumes were recorded and counted.
As shown in FIG. 1, compared with the control group, oral gavage of 6mg/kg indole-3-propionic acid cannot play a role in sensitization colorectal cancer immunotherapy, while oral gavage of 60mg/kg and 300mg/kg indole-3-propionic acid can remarkably increase the curative effect of colorectal cancer anti-PD-1 antibodies and reduce tumor load.
Example 2: intratumoral injection indole-3-propionic acid sensitized colorectal cancer subcutaneous tumor model immunotherapy
Male C57BL/6 mice of 6-8 weeks were randomly divided into 3 groups of 7 mice each, and 1 week of drinking water containing 2mg/mL metronidazole, 2mg/mL penicillin, 2mg/mL streptomycin and 1mg/mL vancomycin was freely drunk before molding, intestinal flora was homogenized, and 1X 10 6 of the Mc38 cells of the colon cancer cell line of the mice were injected subcutaneously (100. Mu.L per mouse). After 8 days of injection, 100. Mu.g of an anti-PD-1 antibody was given to each mouse by intraperitoneal injection, and 50. Mu.L of an anti-PD-1 antibody was injected by intratumoral injection into 0. Mu.M indole-3-propionic acid (control group), 5. Mu.M indole-3-propionic acid (low dose group) and 50. Mu.M indole-3-propionic acid (high dose group) at the same time as the injection. Tumor volumes were monitored every two days and the calculation formula was as follows: volume = 0.5 xlxw 2, where L is the longest diameter and W is the shortest diameter. The experiment was terminated every other day after three PD-1 treatments, and tumor volumes were recorded and counted.
As shown in fig. 2, compared with the control group, the intratumoral injection of 5 μm and 50 μm indole-3-propionic acid can significantly increase the curative effect of colorectal cancer anti-PD-1 antibody and reduce tumor load.
Example 3: intraperitoneal injection of indole-3-propionic acid sensitized colorectal cancer subcutaneous tumor model immunotherapy
Male C57BL/6 mice of 6-8 weeks are randomly divided into 2 groups, 6 mice in each group are freely drunk for 1 week before molding, drinking water containing 2mg/mL of metronidazole, 2mg/mL of penicillin, 2mg/mL of streptomycin and 1mg/mL of vancomycin, intestinal flora is homogenized, and 0mg/kg of indole-3-propionic acid (control group) and 60mg/kg of indole-3-propionic acid (experimental group) are respectively administrated according to the body weight of the mice, and the intraperitoneal injection amount is 200 mu L, and is once daily. On the day of injection, 1×10 6 of the mouse colon cancer cell line Mc38 cells were injected subcutaneously (100 μl per mouse). After 8 days of injection, 100 μg of anti-PD-1 antibody was given to each mouse intraperitoneally, one injection was given every three days, and tumor volumes were monitored every two days, as calculated as follows: volume = 0.5 xlxw 2, where L is the longest diameter and W is the shortest diameter. The experiment was terminated every other day after three PD-1 treatments, and tumor volumes were recorded and counted.
As shown in FIG. 3, compared with the control group, the experimental group (i.e. injecting 60mg/kg indole-3-propionic acid into the abdominal cavity) can remarkably increase the curative effect of the colorectal cancer anti-PD-1 antibody and reduce the tumor load.
The same experimental procedure was repeated and the results of intraperitoneal injection of indole-3-lactic acid (ILA) at 60mg/kg of mouse body weight showed no significant increase in the efficacy of the anti-PD-1 antibody against colorectal cancer as compared to the control group as shown in fig. 3.
Example 4: indole-3-propionic acid increases the number of CD8 + T cells in a colorectal carcinoma subcutaneous tumor model
The method used in example 1 was used to mold, after molding, subcutaneous tumors of mice were blunt-isolated, cut into small pieces of about 1mm 3, digested with collagenase type IV (Worthington, LS 004189) for 30 minutes and dissociated into single cells, and the number of cells in the collected single cell suspension was adjusted to 1X 10 6/mL with PBS. Each sample was first blocked for 10 minutes with 1 μg of anti-CD 16/32 antibody to Fc receptor (BioLegend, 101320). Then, the dead and live antibody FVS 510 (BD Biosciences, 564406) was added, the mixture was incubated at 4℃for 30min in the absence of light, the staining was stopped by adding an appropriate amount of FACS buffer, centrifugation was performed for 5min at 700g, and the supernatant was discarded. Antibody premix (Alexa Fluor 700-CD45(BioLegend,103128)、PECPCY5.5-CD3(BD Biosciences,551163)、BV605-CD4(BD Biosciences,563151) and APC-CY7-CD8 (BD Biosciences, 561967) were prepared, appropriate amounts of antibody were added to each sample, mixed well, incubated at 4 ℃ for 30min in the dark, stopped by adding PBS, centrifuged for 700g for 5min, the supernatant was discarded, washed 1x with PBS, resuspended in appropriate volumes and detected using flow cytometry. For TCF-1 staining, cells were incubated with the fixative working solution for 40 min at room temperature, washed with transcription factor staining buffer (Invitrogen TM, 00-5523-00), and stained with BV421-TCF-1 (BD Biosciences, 566692) for 40 min at room temperature. Appropriate volume was resuspended and then detected using a flow cytometer.
As shown in FIG. 4, oral gavage of 60mg/kg indole-3-propionic acid increased the number of CD8 + T cells in the colorectal cancer immune microenvironment.
Example 5: indole-3-propionic acid sensitized breast cancer subcutaneous tumor model immunotherapy
Female Balb/C mice of 6-8 weeks are randomly divided into 4 groups (A group, B group, C group and D group), 7 mice in each group are freely drunk for 1 week before molding, drinking water containing 2mg/mL metronidazole, 2mg/mL penicillin, 2mg/mL streptomycin and 1mg/mL vancomycin, and intestinal flora is homogenized, and 0mg/kg (A group), 60mg/kg (B group), 0mg/kg (C group) and 60mg/kg (D group) of indole-3-propionic acid are respectively administrated according to the weight of the mice, and the stomach is irrigated once per day. On the day of gastric lavage, 1×10 5 of 4T1 cells were injected subcutaneously into mice (100 μl per mouse). After 8 days of injection, each mouse was given 100 μg of intraperitoneal injection IgG antibody (group a), igG antibody (group B), anti-PD-1 antibody (group C), anti-PD-1 antibody (group D), and one injection was given every three days, and tumor volumes were monitored every two days, as follows: volume = 0.5 xlxw 2, where L is the longest diameter and W is the shortest diameter. The experiment was terminated every other day after three PD-1 treatments, and tumor volumes were recorded and counted.
As shown in fig. 5, the supplementation of indole-3-propionic acid can significantly increase the therapeutic effect of the breast cancer subcutaneous tumor model anti-PD-1 antibody, reduce tumor burden, and increase the numbers of CD8 + T cells and TCF-1 +CD8+ T cells in the breast cancer immune microenvironment.
Example 6: indole-3-propionic acid sensitized melanoma subcutaneous tumor model immunotherapy
Male C57BL/6 mice of 6-8 weeks are randomly divided into 4 groups (A group, B group, C group and D group), 7 mice of each group are freely drunk for 1 week before molding, drinking water containing 2mg/mL of metronidazole, 2mg/mL of penicillin, 2mg/mL of streptomycin and 1mg/mL of vancomycin, intestinal flora is homogenized, and 0mg/kg (A group), 60mg/kg (B group), 0mg/kg (C group) and 60mg/kg (D group) of indole-3-propionic acid are respectively administrated according to the weight of the mice, and the mice are irrigated with stomach once per day. On the day of gastric lavage, 1X 10 5 B16-F10 cells were injected subcutaneously into mice (100. Mu.L per mouse). After 8 days of injection, each mouse was given 100 μg of intraperitoneal injection IgG antibody (group a), igG antibody (group B), anti-PD-1 antibody (group C), anti-PD-1 antibody (group D), and one injection was given every three days, and tumor volumes were monitored every two days, as follows: volume = 0.5 xlxw 2, where L is the longest diameter and W is the shortest diameter. The experiment was terminated every other day after three PD-1 treatments, and tumor volumes were recorded and counted.
As shown in fig. 6, the supplementation of indole-3-propionic acid can significantly increase the efficacy of anti-PD-1 antibodies in the melanoma subcutaneous tumor model, reduce tumor burden, and increase the numbers of CD8 + T cells and TCF-1 +CD8+ T cells in the melanoma immune microenvironment.
Example 7: indole-3-propionic acid sensitized breast cancer in-situ tumor model immunotherapy
Female Balb/c mice of 6-8 weeks are randomly divided into 2 groups, 7 mice in each group are freely drunk for 1 week before molding, drinking water containing 2mg/mL of metronidazole, 2mg/mL of penicillin, 2mg/mL of streptomycin and 1mg/mL of vancomycin is homogenized, and indole-3-propionic acid is respectively administered for 0mg/kg, 60mg/kg of stomach irrigation according to the weight of the mice once daily. On the day of gastric lavage, 1×10 5 of 4T1 cells were injected onto a fourth group of mammary fat pads (100 μl per mouse). After 8 days of injection, 100 μg of anti-PD-1 antibody was given to each mouse intraperitoneally, one injection was given every three days, and tumor volumes were monitored every two days, as calculated as follows: volume = 0.5 xlxw 2, where L is the longest diameter and W is the shortest diameter. The experiment was terminated every other day after three PD-1 treatments, and tumor volumes were recorded and counted.
As shown in fig. 7, the supplementation of indole-3-propionic acid can significantly increase the curative effect of the anti-PD-1 antibody of the breast cancer in-situ tumor model, reduce tumor burden, and increase the numbers of CD8 + T cells and TCF-1 +CD8+ T cells in the breast cancer immune microenvironment.
Example 8: indole-3-propionic acid sensitized breast cancer spontaneous tumor model immunotherapy
Female MMTV-PyMT mice of 16 weeks are randomly divided into 2 groups, 10 mice in each group are freely drunk for 1 week before molding, drinking water containing 2mg/mL of metronidazole, 2mg/mL of penicillin, 2mg/mL of streptomycin and 1mg/mL of vancomycin is homogenized, and indole-3-propionic acid is respectively administrated for 0mg/kg, 60mg/kg of stomach irrigation once daily according to the weight of the mice. Each mouse was given 100 μg of anti-PD-1 antibody by intraperitoneal injection, one injection every three days, tumor volume was monitored every two days, and the calculation formula was as follows: volume = 0.5 xlxw 2, where L is the longest diameter and W is the shortest diameter. The experiment was terminated when individual tumors of the mice were grown to 1000mm 3, and survival curves were recorded and plotted.
As shown in the figure 8, the indole-3-propionic acid can be supplemented to remarkably increase the curative effect of the anti-PD-1 antibody of the spontaneous tumor model of the breast cancer, and prolong the survival time of mice.
Indole-3-propionic acid (IPA) used in examples 1-8 was purchased from Sigma company under the designation V900491; the PD-1 antibody used was InVivoMAb anti-mouse PD-1 (CD 279), purchased from BioXcell company, cat#BE0273; the reaction species: a Mouse; clone number: 29f.1a12; isotype: rat IgG2a; immunogens: recombinant PD-1-Ig fusion protein; the IgG antibody used was InVivoMAb rat IgG2a isotype control, purchased from BioXcell company, cat#be0089; clone number: 2A3, isotype: rat IgG2a, κ.
The above detailed description is intended to illustrate the present invention by way of example only and not to limit the invention to the particular embodiments disclosed, but to limit the invention to the precise embodiments disclosed, and any modifications, equivalents, improvements, etc. that fall within the spirit and scope of the invention as defined by the appended claims.

Claims (10)

1. The application of indole-3-propionic acid in preparing medicine for sensitization tumor immunotherapy.
2. The use according to claim 1, wherein said indole-3-propionic acid is capable of significantly increasing the number of CD8 + T cells in a tumor.
3. The use according to claim 1, wherein the tumor is colorectal cancer, breast cancer or melanoma.
4. The use according to claim 1, wherein the medicament comprises indole-3-propionic acid and/or a pharmaceutically acceptable salt thereof, a solvate thereof, a prodrug thereof or a metabolite thereof.
5. The use according to claim 1, wherein the medicament further comprises a pharmaceutical carrier and/or pharmaceutically acceptable excipients.
6. A pharmaceutical composition for the therapeutic effect of a sensitized anti-PD-1 antibody, comprising indole-3-propionic acid and one or more pharmaceutically acceptable carriers, or a pharmaceutical composition comprising indole-3-propionic acid and one or more therapeutic effects of a sensitized anti-PD-1 antibody.
7. The pharmaceutical composition of claim 6, wherein the pharmaceutically acceptable carrier comprises one or more of solvents, solubilizers, co-solvents, emulsifiers, flavoring agents, colorants, binders, disintegrants, fillers, lubricants, wetting agents, osmotic pressure regulators, pH regulators, stabilizers, surfactants, preservatives.
8. The pharmaceutical composition of claim 6, wherein the pharmaceutical composition is a solid formulation, a semi-solid formulation, or a liquid formulation.
9. The pharmaceutical composition of claim 6, wherein the solid formulation is a tablet, capsule, granule, or pill; the semisolid preparation is gel, suppository or ointment; the liquid preparation is emulsion, mixture, suspension or solution.
10. The use according to claim 1 and the pharmaceutical composition according to claim 6, wherein the medicament or pharmaceutical composition is administered by intraperitoneal injection, oral lavage or intratumoral injection, wherein the amount of indole-3-propionic acid administered is: intraperitoneal injection or oral lavage is 60 mg/kg/day; intratumoral injection is 5-50 mu m, 50 mu L/day.
CN202410254831.5A 2024-03-06 2024-03-06 Use of indole-3-propionic acid in preparing medicament for sensitization tumor immunotherapy Pending CN118141804A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202410254831.5A CN118141804A (en) 2024-03-06 2024-03-06 Use of indole-3-propionic acid in preparing medicament for sensitization tumor immunotherapy

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202410254831.5A CN118141804A (en) 2024-03-06 2024-03-06 Use of indole-3-propionic acid in preparing medicament for sensitization tumor immunotherapy

Publications (1)

Publication Number Publication Date
CN118141804A true CN118141804A (en) 2024-06-07

Family

ID=91289937

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202410254831.5A Pending CN118141804A (en) 2024-03-06 2024-03-06 Use of indole-3-propionic acid in preparing medicament for sensitization tumor immunotherapy

Country Status (1)

Country Link
CN (1) CN118141804A (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1292693A (en) * 1998-02-23 2001-04-25 南亚拉巴马州医学科学基金会 Indole-3-propionic acids, salts and esters thereof used as medicaments
WO2021222309A1 (en) * 2020-04-27 2021-11-04 Ixcela, Inc. Detection and modification of gut microbial population
CN114558007A (en) * 2021-12-06 2022-05-31 南京医科大学 Application of indole-3-lactic acid in preparation of anti-colorectal cancer drugs
KR20230111841A (en) * 2022-01-19 2023-07-26 한국과학기술원 Composition for preventing and treating cancer, containing tryptophan metabolite as an active ingredient

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1292693A (en) * 1998-02-23 2001-04-25 南亚拉巴马州医学科学基金会 Indole-3-propionic acids, salts and esters thereof used as medicaments
WO2021222309A1 (en) * 2020-04-27 2021-11-04 Ixcela, Inc. Detection and modification of gut microbial population
CN114558007A (en) * 2021-12-06 2022-05-31 南京医科大学 Application of indole-3-lactic acid in preparation of anti-colorectal cancer drugs
KR20230111841A (en) * 2022-01-19 2023-07-26 한국과학기술원 Composition for preventing and treating cancer, containing tryptophan metabolite as an active ingredient

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ETHAN SHELKEY等: "Immuno‑reactive cancer organoid model to assess effects of the microbiome on cancer immunotherapy", SCIENTIFIC REPORTS, vol. 12, 15 June 2022 (2022-06-15), pages 1 - 13 *

Similar Documents

Publication Publication Date Title
CN108601789A (en) Fourth Ring quinolone analogs combination treatment for treating cancer
JP5952890B2 (en) Pharmaceutical formulation for treating pancreatic cancer
CN110191876A (en) For applying the preparation of Eflornithine
CN108430472A (en) Combination for the hdac inhibitor and anti-PD-L1 antibody for the treatment of oophoroma
JPS61151123A (en) Immuno-regulator
CN114344325A (en) Application of bacteroides fragilis and zwitter-ion capsular polysaccharide thereof in preparation of medicines for preventing and treating genitourinary system tumors
CN114344339A (en) Application of bacteroides fragilis combined with immune checkpoint inhibitor in treatment of skin tumor
US20220362212A1 (en) Sublingual or buccal administration of dim for treatment of skin diseases
Song et al. A hydroxyethyl derivative of chrysin exhibits anti-inflammatory activity in dendritic cells and protective effects against dextran sodium salt-induced colitis in mice
CN114469986B (en) Application of bacteroides fragilis capsular polysaccharide A in combination with immune checkpoint inhibitor in preparation of medicines for treating digestive system tumors
CN115414390A (en) Probiotic compound preparation with functions of improving intestinal micro-ecological performance and enhancing treatment effect of tumor immune checkpoint inhibitor and application
CN114392356A (en) Application of bacteroides fragilis and immune checkpoint inhibitor in combination in treatment of digestive system tumor
CN118141804A (en) Use of indole-3-propionic acid in preparing medicament for sensitization tumor immunotherapy
KR20240063969A (en) Application of the association of C. gondii spores with pembrolizumab
CN110464845B (en) Combined medicine composition for breast cancer immunotherapy and application thereof
KR102316961B1 (en) Pharmaceutical composition of benzene derivatives as immunosuppressive agents
CN114404598A (en) Application of bacteroides fragilis capsular polysaccharide A combined with PD-1 inhibitor in preparation of medicine for treating skin tumor
Huang et al. Melatonin inhibits the formation of chemically induced experimental encapsulating peritoneal sclerosis through modulation of T cell differentiation by suppressing of NF-κB activation in dendritic cells
CN111514290B (en) Cucurbitacin composition and application thereof
JP2023513569A (en) Bacterial composition for the treatment of cancer
CN111569078A (en) Use of SIRT1 inhibitors for side effects caused by combination of VEGFR inhibitors and immune checkpoint inhibitors
CN109674788B (en) Application of carboxyamidotriazole and IDO1 inhibitor combination in resisting tumors
CN110446505A (en) For treating dermopathic conjoint therapy
CN116898964A (en) Application of rapamycin combined with PD-1 monoclonal antibody in preparation of colorectal cancer treatment drug
CN118185812A (en) Lactobacillus johnsonii and application thereof

Legal Events

Date Code Title Description
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination