WO2023048230A1 - Kit dispositif thérapeutique - Google Patents

Kit dispositif thérapeutique Download PDF

Info

Publication number
WO2023048230A1
WO2023048230A1 PCT/JP2022/035371 JP2022035371W WO2023048230A1 WO 2023048230 A1 WO2023048230 A1 WO 2023048230A1 JP 2022035371 W JP2022035371 W JP 2022035371W WO 2023048230 A1 WO2023048230 A1 WO 2023048230A1
Authority
WO
WIPO (PCT)
Prior art keywords
administration
cells
injection needle
tumor
cancer
Prior art date
Application number
PCT/JP2022/035371
Other languages
English (en)
Japanese (ja)
Inventor
大貴 有馬
滋典 野沢
真樹 平光
恵子 大津
善紀 米田
Original Assignee
テルモ株式会社
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 テルモ株式会社 filed Critical テルモ株式会社
Publication of WO2023048230A1 publication Critical patent/WO2023048230A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/282Platinum compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M5/00Devices for bringing media into the body in a subcutaneous, intra-vascular or intramuscular way; Accessories therefor, e.g. filling or cleaning devices, arm-rests
    • A61M5/14Infusion devices, e.g. infusing by gravity; Blood infusion; Accessories therefor
    • A61M5/158Needles for infusions; Accessories therefor, e.g. for inserting infusion needles, or for holding them on the body
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M5/00Devices for bringing media into the body in a subcutaneous, intra-vascular or intramuscular way; Accessories therefor, e.g. filling or cleaning devices, arm-rests
    • A61M5/14Infusion devices, e.g. infusing by gravity; Blood infusion; Accessories therefor
    • A61M5/168Means for controlling media flow to the body or for metering media to the body, e.g. drip meters, counters ; Monitoring media flow to the body
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M5/00Devices for bringing media into the body in a subcutaneous, intra-vascular or intramuscular way; Accessories therefor, e.g. filling or cleaning devices, arm-rests
    • A61M5/178Syringes

Definitions

  • the present invention relates to therapeutic device kits.
  • Intravenous administration is the standard administration method for anticancer drugs used in chemotherapy in cancer treatment.
  • intravenous administration of anticancer agents has the problem that only a small amount of the drug can reach the tumor, which may reduce the therapeutic effect and cause systemic side effects.
  • cancer immunotherapy which applies the immune system to treat diseases, is attracting attention.
  • the functions of the immune system are generally expressed through two mechanisms, humoral and cellular immunity. Since cell-mediated immunity has the ability to kill and eliminate cancer cells, it has been found to play an important role in cancer immunotherapy.
  • cytotoxic T cells hereinafter also simply referred to as CTLs
  • CTLs cytotoxic T cells
  • CTL induction is usually expressed as follows. That is, endogenous antigens such as proteins produced in virus-infected cells and cancer cells are ubiquitinated and then degraded into peptides by the proteasome. The cleaved peptides bind to major histocompatibility complex (MHC) class I molecules, and the resulting complexes are presented to CD8-positive T cells on the surface of antigen-presenting cells to activate CD8-positive T cells. be done. The activated CD8-positive T cells then differentiate into CTLs. Induction of CTLs has become a necessary factor for highly effective cancer immunotherapy.
  • MHC major histocompatibility complex
  • Japanese Patent Publication No. 2001-510806 discloses MHC class II ligands or MHC class II-like ligands as vaccine adjuvants for enhancing antigen-specific immune responses.
  • an immunotoxin comprising a single-chain variable region antibody fused to PE38 truncated Pseudomonas exotoxin and an immune checkpoint inhibitor are combined to treat tumors.
  • a medicament is disclosed for
  • the object of the present invention is to provide a method with a higher antitumor effect.
  • the present invention is a therapeutic device kit comprising an anticancer agent that induces immunogenic cell death and an administration device capable of directly administering the anticancer agent to tumor tissue, wherein the anticancer agent is an administration device is a therapeutic device kit that induces activation of anti-tumor immunity when administered using
  • FIG. 1 is a graph showing the tumor volume of each group in mouse drug efficacy evaluation.
  • FIG. 2 is a diagram showing survival curves of each group in mouse drug efficacy evaluation.
  • FIG. 4 shows a gating flow chart in flow cytometry; Total. of each group in CTL activation evaluation (mice) using flow cytometry. It is a graph which shows the number of CTLs.
  • Fig. 10 is a graph showing the number of AH-1 specific CTLs in each group in CTL activation evaluation (mice) using flow cytometry.
  • FIG. 10 is a graph showing the number of CD69+ CTLs in each group in CTL activation evaluation (mice) using flow cytometry.
  • FIG. 4 is a diagram showing the tip of an injection needle;
  • FIG. 4 is a diagram showing the tip of an injection needle;
  • FIG. 4 is a diagram showing the configuration of an outer cylinder that constitutes the administration device;
  • FIG. 4 is a diagram showing the configuration of an outer cylinder that constitutes the administration device;
  • FIG. 4 is a diagram showing an auxiliary tool that constitutes a therapeutic device kit;
  • FIG. 4 is a diagram showing an auxiliary tool that constitutes a therapeutic device kit;
  • FIG. 4 is a schematic diagram showing an endoscope, an administration device, etc.
  • FIG. 15 is a view showing the distal end portion of the endoscope shown in FIG. 14;
  • FIG. 15 is a diagram showing how an anticancer drug is administered to an affected area using the endoscope shown in FIG. 14;
  • the range "X to Y” includes X and Y and means "X or more and Y or less”.
  • operations and measurements of physical properties are performed under the conditions of room temperature (20° C. or higher and 25° C. or lower)/relative humidity of 40% RH or higher and 50% RH or lower.
  • One aspect of the present invention is a therapeutic device kit comprising an anticancer agent that induces immunogenic cell death and an administration device capable of directly administering the anticancer agent to tumor tissue, wherein the anticancer agent is , is a therapeutic device kit that induces activation of anti-tumor immunity when administered using an administration device. According to this aspect, it is possible to provide a therapeutic device kit with a higher antitumor effect.
  • Immunogenic cell death usually refers to cell death that is easily recognized as non-self by immune cells, accompanied by expression or release of DAMPs typified by calreticulin, ATP, and HMGB1.
  • Anticancer agents that cause immunogenic cell death are selected from the group consisting of doxorubicin, epirubicin, oxaliplatin, paclitaxel and pharmaceutically acceptable salts thereof It is preferably at least one, more preferably doxorubicin and/or a pharmaceutically acceptable salt thereof. That is, a preferred aspect of the present invention comprises doxorubicin and/or a pharmaceutically acceptable salt thereof, and an administration device capable of directly administering doxorubicin and/or a pharmaceutically acceptable salt thereof to tumor tissue.
  • a therapeutic device kit comprising: Doxorubicin salts include, for example, doxorubicin hydrochloride.
  • Examples of the pharmaceutically acceptable salts include salts at basic groups such as amino groups, and acidic groups such as hydroxyl groups and carboxyl groups.
  • Salts in basic groups include, for example, salts with inorganic acids such as hydrochloric, hydrobromic, phosphoric, boric, nitric and sulfuric acids; formic, acetic, lactic, citric, oxalic, fumaric, maleic, acids, salts with organic carboxylic acids such as succinic, malic, tartaric, aspartic, trichloroacetic and trifluoroacetic acids; and methanesulfonic, benzenesulfonic, p-toluenesulfonic, mesitylenesulfonic and naphthalenesulfonic acids. and salts with sulfonic acids such as
  • Salts in acidic groups include, for example, salts with alkali metals such as sodium and potassium; salts with alkaline earth metals such as calcium and magnesium; ammonium salts; and trimethylamine, triethylamine, tributylamine, pyridine, N,N- Nitrogen-containing organic bases such as dimethylaniline, N-methylpiperidine, N-methylmorpholine, diethylamine, dicyclohexylamine, procaine, dibenzylamine, N-benzyl- ⁇ -phenethylamine, 1-ephenamine and N,N'-dibenzylethylenediamine and salt with.
  • alkali metals such as sodium and potassium
  • alkaline earth metals such as calcium and magnesium
  • ammonium salts and trimethylamine, triethylamine, tributylamine, pyridine, N,N- Nitrogen-containing organic bases such as dimethylaniline, N-methylpiperidine, N-methylmorpholine, diethylamine
  • Anticancer agents induce activation of antitumor immunity, for example, by being administered using the administration device described later.
  • the activation of anti-tumor immunity is regulated by, for example, the dose of anti-cancer drugs.
  • activation of anti-tumor immunity specifically means that the number of cytotoxic T cells (CTL) is 150% or more, preferably 170% or more, compared to a control to which no anticancer agent is administered. , more preferably 200% or more.
  • CTL cytotoxic T cells
  • was measured using Total It can be measured by a method for measuring the number of CTLs.
  • the dosage of the anticancer drug is appropriately determined on a case-by-case basis, taking into consideration the patient's symptoms, age, sex, and the like.
  • an anticancer agent that induces immunogenic cell death can induce activation of antitumor immunity. Therefore, it is expected that the dosage can be reduced compared to the dosage of ordinary anticancer drugs.
  • the dose of the anticancer drug in humans is, for example, 0.1 mg/Kg/Day or less, may be 0.01 to 0.1 mg/Kg/Day, and may be 0.020 to 0.070 mg/Kg. /Day.
  • an anticancer drug such as doxorubicin hydrochloride usually requires administration at 0.20 mg/Kg or more.
  • the dose of the anticancer drug per tumor volume is, for example, 6 ⁇ g or more, may be 60 ⁇ g or less, or may be 20 ⁇ g or less per 100 mm 3 of tumor tissue.
  • the number of administrations of the anticancer drug per course is also not particularly limited, and it may be administered in a single dose or in multiple doses, for example, it can be administered once to three times per course.
  • the anticancer drug can be administered repeatedly at intervals of about 1 to 4 weeks.
  • the anticancer drug may be administered to multiple tumors.
  • the anticancer drug may constitute a composition together with pharmaceutically acceptable additives according to the desired product form.
  • “Pharmaceutically acceptable” means, within sound medical judgment, commensurate with a reasonable benefit/risk ratio, and without problems or complications such as excessive toxicity, irritation, or allergic reactions, in humans and animals. Used to refer to compounds, materials, compositions and/or dosage forms suitable for use in contact with tissue.
  • Pharmaceutically acceptable additives include solvents (e.g., physiological saline, water for injection, buffers, etc.), membrane stabilizers (e.g., cholesterol), tonicity agents (e.g., sodium chloride, glucose, glycerin, etc.), Oxidizing agents (eg, tocopherol, ascorbic acid, glutathione, etc.), preservatives (eg, chlorbutanol, parabens, etc.), and the like may be included.
  • Physiological saline means an inorganic salt solution adjusted to be isotonic with the human body, and may further have a buffering function. Saline solutions include saline containing 0.9 w/v % (weight/volume percent) sodium chloride, PBS and Tris-buffered saline, and the like.
  • administration device The specific configuration of the administration device is not particularly limited as long as the anticancer drug can be directly administered to the tumor tissue, but two administration devices will be exemplified below.
  • FIG. 7 is a schematic diagram showing a therapeutic device kit 1 including an administration device 100.
  • the treatment device kit 1 includes a medical device 200, an administration device 100, an instrument 300, and an auxiliary tool 400, as outlined with reference to FIG. Details will be described below.
  • the medical device 200 includes a tubular portion 210, a pressing portion 220, a sealing member 230, and a connecting member 240, as shown in FIG.
  • the cylinder part 210 has a semi-closed space for containing an anticancer drug.
  • the tubular portion 210 is configured in a tubular shape such as a cylinder, and has openings at both ends in the axial direction of the tubular shape.
  • a pressing portion 220 can be movably arranged in one of the openings (also referred to as a proximal opening).
  • a connecting member 240 can be attached to the other opening (also referred to as the distal opening).
  • the pressing part 220 includes a presser that can increase or decrease the size of the semi-closed space of the cylindrical part 210 .
  • the pusher of the pressing portion 220 is configured such that the distal end side is accommodated in the semi-closed space of the tubular portion 210 and the proximal end side is arranged outside the tubular portion 210 .
  • the pusher of the pressing portion 220 moves relative to the cylindrical portion 210 in the axial direction of the cylindrical portion 210 to form a semi-closed space in which a drug (anticancer drug, hereinafter also referred to as a drug) is contained. It is configured to change the size of a drug (anticancer drug, hereinafter also referred to as a drug) is contained. It is configured to change the size of
  • the pusher of the pressing part 220 As the size of the semi-closed space of the cylinder part 210 is reduced by the pusher of the pressing part 220, the amount of the drug contained in the semi-closed space is reduced, and flows through the lumen of the injection needle 120 to be administered to the patient.
  • the pusher of the pressing portion 220 may be manually operated by the operator by gripping it with fingers or the like, or may be operated by appropriately combining mechanical elements such as a motor and gears.
  • the sealing member 230 is configured to be attached to the distal end portion of the pusher of the pressing portion 220 in the axial direction.
  • the sealing member 230 prevents the medicine accommodated in the semi-closed space of the tubular portion 210 from flowing through other than the injection needle 120 by slidably fitting with the inner wall of the tubular portion 210 .
  • connection member 240 is attached to the opening on the distal end side of the tubular portion 210 .
  • the connecting member 240 is hollow so that the medicine can flow inside.
  • the connecting member 240 is attached to the distal end of the cylindrical portion 210 , and the inner cavity of the connecting member 240 is configured to communicate with the semi-closed space of the cylindrical portion 210 .
  • the administration device 100 includes an inner cylinder 110, an injection needle 120, an outer cylinder 130, a detection section 140, a notification section 150, and a control section (not shown), as shown in FIG. 7 and the like.
  • the inner cylinder 110 is connected to the medical device 200 and has a liquid feeding path for the anticancer drug flowing from the medical device 200 together with the injection needle 120 inside.
  • the inner cylinder 110 is provided with a connection portion 111 with the medical device 200 radially outward on the cylindrical side surface.
  • a detection unit 140 which will be described later, is provided in the liquid feeding path provided inside the inner cylinder 110. As shown in FIG.
  • the injection needle 120 is configured so that the anticancer drug can be directly administered to the tumor tissue of the living body.
  • the injection needle 120 includes a hollow member with a small diameter, and is configured to provide a liquid-feeding channel communicating with the liquid-feeding channel of the inner cylinder 110 inside the hollow member, and to provide a tip opening communicating with the liquid-feeding channel at the tip. are doing.
  • the dimensions of the injection needle 120 are appropriately selected depending on the administration target and the tumor position so that the injection needle 120 can be punctured from the body surface to the tumor existing in the body.
  • the longitudinal dimension can be configured to be 150 mm or more so that a tumor existing in the body can be punctured from the body surface.
  • the dimensions of the injection needle 120 may be, for example, 50-300 mm.
  • FIG. 8 is a view showing an opening for ejecting medicine in injection needle 120
  • FIG. 9 is a view showing an opening for ejecting medicine in injection needle 120a according to a modification of injection needle 120.
  • the injection needle 120 has an opening for discharging the anticancer drug in the present embodiment so as to face the distal end side in the longitudinal direction, as shown in FIG.
  • the opening of the injection needle is not limited to this, and in addition to the above, as shown in injection needle 120a in FIG.
  • An opening for ejecting medicine may be provided outward in the direction, and a plurality of such openings may be provided in the circumferential direction. In radially open configurations, the tip may be sharp.
  • the administration device has a drug solution discharge hole on the tip or side of the injection needle.
  • the injection needle 120 can perform an operation (priming) to circulate the anticancer drug to the tip of the liquid feeding channel of the injection needle 120 while using the auxiliary tool 400 described later.
  • the auxiliary tool 400 will be described later.
  • the administration device comprises a mechanism for safely loading the anticancer drug into the infusion needle.
  • the outer cylinder 130 includes a base portion 131 capable of storing at least part of the inner cylinder 110 and a distal end portion 132 capable of storing the distal end portion of the injection needle 120 .
  • 10 and 11 are diagrams showing the base portion 131 and the tip portion 132 of the outer cylinder 130.
  • FIG. The base portion 131 and the tip portion 132 are formed so as to be continuous in the longitudinal direction of the injection needle 120 in this embodiment.
  • the base portion 131 and the tip portion 132 are distinguished by a chain double-dashed line L1 shown in FIG. 10, and the base portion 131 and the tip portion 132 can each be called an "outer cylinder".
  • the detection unit 140 is configured to be provided in a liquid feed path provided inside the inner cylinder 110 .
  • the detection unit 140 includes a sensor capable of measuring the injection pressure when the injection needle 120 is used to administer the anticancer drug to the affected area.
  • the sensor of the detection unit 140 is not particularly limited as long as it can measure the pressure of the anticancer drug flowing through the liquid feeding channel, but a diaphragm type sensor can be given as an example.
  • the notification unit 150 is configured to be able to notify the pressure during injection of the anticancer drug measured by the sensor of the detection unit 140 .
  • the notification unit 150 is configured to be electrically connectable to the detection unit 140 by a wire or the like led from the sensor of the detection unit 140 to the outside of the inner cylinder 110 through the base end side of the inner cylinder 110 .
  • the specific configuration of the notification unit 150 is not particularly limited as long as it can notify the user of the pressure value obtained by the sensor associated with the detection unit 140.
  • the notification unit 150 may be a liquid crystal display or an organic EL display that displays the pressure value as an image using numerical values or graphs. etc.
  • the control unit is provided to control electrically connected components such as the detection unit 140 and the notification unit 150 in the administration device 100 .
  • the control unit is configured to include processors such as CPU and GPU, main memory such as RAM, auxiliary memory such as ROM, HDD and SSD.
  • the control unit can be housed inside the housing of the notification unit 150 or the like.
  • the device 300 is configured to make the first semi-closed space 413 of the tubular portion 410 of the auxiliary tool 400 negative pressure by the user's operation while connected to the auxiliary tool 400 (FIGS. 12 and 13).
  • the instrument 300 includes a tubular portion 310, a pressing member 320, a sealing member 330, and a connecting member 340, as shown in FIG.
  • the tubular portion 310 is the same as the tubular portion 210 of the medical device 200, the pressing member 320 is the same as the pressing portion 220, and the sealing member 330 is the same as the sealing member 230, so detailed description thereof will be omitted.
  • connection member 340 is configured to include a hollow member such as a tube through which fluid such as gas can flow.
  • the operator attaches one of the connection members 340 to the tube portion 310 and attaches the other to the connection portion 440 of the auxiliary tool 400, and moves the pressing member 320 so as to increase the semi-closed space of the tube portion 310. .
  • the pressure in the first semi-closed space 413 of the cylindrical portion 410 of the assisting device 400 can be made negative.
  • the instrument 300 is configured to include members similar to those of the medical device 200. is not limited to the same configuration as
  • auxiliary tool 12 and 13 are diagrams for explaining the auxiliary tool 400.
  • FIG. 1 the anticancer agent leaks out from the tip of the injection needle 120 during priming for circulating the anticancer agent in the liquid delivery path of the injection needle 120, so that the anticancer agent is released to the medical staff and others. Prevent splashing on the patient.
  • the assisting tool 400 includes a cylindrical portion 410 as shown in FIG. 12 and the like.
  • the tubular portion 410 of the auxiliary tool 400 includes a stopper 420 , a valve member 430 and a connecting portion 440 .
  • the cylindrical part 410 has a first semi-closed space 413 and a second semi-closed space 414 that form internal spaces so as to surround the tip of the hollow injection needle 120 through which the anticancer drug can flow.
  • the tubular portion 410 is configured in a tubular shape such as a cylinder provided with a first semi-closed space 413 and a second semi-closed space 414 .
  • the specific shape of the cylindrical portion 410 is not limited to a cylinder, and may be configured by a rectangular cylinder (polygonal column) other than a cylinder, as long as the medicine can be prevented from leaking to the outside during priming.
  • the cylindrical portion 410 has openings on both the distal end side and the proximal end side in the axial direction of the cylindrical shape to allow the first semi-closed space 413 or the second semi-closed space 414 to communicate with the outside.
  • the opening on the distal side that communicates the first semi-closed space 413 with the outside is the first opening 411
  • the opening on the proximal side that communicates the second semi-closed space 414 with the outside is the second opening 412. call.
  • the dimension of the first semi-closed space 413 in the direction of insertion of the injection needle 120 (dimension d1 in the vertical direction) in FIG.
  • the cylindrical portion 410 is made of a transparent material, it is possible to visually confirm that the anticancer drug is scattered from the injection needle 120 in the first semi-closed space 413 during priming, and to confirm the completion of priming. sell.
  • the stopper 420 is provided in the second semi-closed space 414, which is the internal space of the cylindrical portion 410 in this embodiment.
  • the stopper 420 is configured to allow the injection needle 120 of the administration device 100 to pass therethrough and to provide a third opening 421 that prevents the insertion of needle tubes other than the injection needle 120 .
  • the third opening 421 is configured such that the area of the cross section intersecting the axial direction becomes smaller as it advances in the axial direction from the direction of entering the first semi-closed space 413 of the tubular portion 410 . With such a configuration, the injection needle 120 can be easily inserted into the third opening 421 .
  • the third opening 421 is configured to have a stepped shape in which the radial dimension changes stepwise.
  • the third opening may be configured to have a tapered shape instead of or in addition to the stepped shape.
  • the valve member 430 is provided adjacent to the second opening 412 in the axial direction of the internal space of the tubular portion 410 .
  • the internal space of the cylindrical portion 410 can be divided by the valve member 430 into a first semi-closed space 413 on the first opening 411 side and a second semi-closed space 414 on the second opening 412 side.
  • the valve member 430 can be configured by a member similar to the elastically deformable elastic member that configures the sealing member 230 of the medical device 200 .
  • the valve member 430 has a substantially circular cross-sectional shape similar to the cross-sectional shape of the cylindrical portion 410 intersecting with the axial direction.
  • a valve member 430 is provided adjacent to the stopper 420 in this embodiment.
  • valve member 430 is configured such that a notch 431 is provided substantially in the central portion as shown in FIG. 12 and the like.
  • the distal end portion of the injection needle 120 is inserted through the valve member 430 from the notch 431 by elastically deforming the valve member 430 from the second opening 412 of the tubular portion 410 as shown in FIG. can enter (insert) into the first semi-closed space 413 of .
  • the valve member 430 holds the injection needle 120 while the tip of the injection needle 120 is inserted into the first semi-closed space 413 .
  • the valve member 430 prevents the anticancer drug from leaking from the second opening 412 side in the internal space in a state in which the injection needle 120 is inserted.
  • the notch 431 of the valve member 430 is formed in a cross shape in this embodiment.
  • the specific shape of the incision is not limited to a cross as long as the injection needle 120 can be inserted through the valve member and the anticancer drug does not flow out from the inserted portion in the inserted state.
  • the tip of the injection needle 120 can be inserted into the first semi-closed space 413 while the injection needle 120 is in close contact with the valve member, the valve member does not need to be provided with a notch.
  • the connecting portion 440 is provided on the side of the first opening 411 in the axial direction of the tubular portion 410 .
  • the connecting portion 440 is configured to be connectable to the device 300 that makes the first semi-closed space 413 (internal space) of the cylindrical portion 410 negative pressure, as will be described later.
  • connection part 440 is provided with a first opening 411 , and the first opening 411 is configured to communicate with the first semi-closed space 413 of the cylindrical part 410 . Accordingly, by operating the instrument 300 with the connecting member 340 and the like of the instrument 300 attached to the connecting portion 440, the pressure in the first semi-closed space 413 of the cylindrical portion 410 can be made negative.
  • connection part 440 has a hollow cylindrical shape with an axis parallel to the insertion direction of the injection needle 120 in the same manner as the cylinder part 410 in this embodiment.
  • the specific shape of the connection portion is not limited to the above. It may be configured with other tubular shapes such as prisms.
  • the assisting tool 400 is not limited to the method of making the semi-closed space 413 negative pressure. A method in which the drug is pushed out into the
  • materials constituting the treatment device kit 1 are not particularly limited.
  • the inner cylinder 110, the outer cylinder 130, the cylinder part 210, the cylinder part 410, the pressing part 220, the pressing member 320, the connecting member 240, the connecting member 340, etc. can be made of plastic such as polypropylene or polyethylene.
  • Seal member 230 and seal member 330 can be made of butyl rubber, silicon rubber, elastomer, or the like.
  • the injection needle 120 can be made of stainless steel or the like.
  • connection member 240 of the medical device 200 connects the connection member 240 of the medical device 200 to the connection portion 111 of the administration device 100 and connects the connection member 340 of the instrument 300 to the connection portion 440 of the auxiliary tool 400 .
  • the operator inserts the tip of the injection needle 120 of the administration device 100 into the notch 431 of the valve member 430 of the auxiliary tool 400 with the injection needle 120 exposed from the tip 132 of the outer cylinder 130, The tip of the injection needle 120 is placed in the first semi-closed space 413 of the cylindrical portion 410 .
  • the operator performs an operation to axially move the pressing member 320 so that the internal space of the cylindrical portion 310 of the instrument 300 is widened. As a result, the pressure in the first semi-closed space 413 of the assisting device 400 becomes negative.
  • the anticancer drug contained in the inner space of the tubular portion 210 of the medical device 200 moves from the inner space of the tubular portion 210 to the lumen of the injection needle 120 .
  • the operator moves the anticancer drug placed in the tube part 210 into the lumen of the injection needle 120 while the tip of the injection needle 120 is placed in the first semi-closed space 413 of the tube part 410 . , it is possible to visually confirm that the anticancer drug has come out from the tip of the injection needle 120 into the first semi-closed space 413 (priming).
  • the operator withdraws the tip of the injection needle 120 from the first semi-closed space 413 of the tubular portion 410 and accommodates the injection needle 120 in the tip 132 of the outer tube 130 .
  • injection needle 120 is passed through valve member 430 while coming out of first semi-closed space 413 of cylindrical portion 410 .
  • the anticancer drug attached to the outer surface of the injection needle 120 can be attached to the valve member 430 and retained in the first semi-closed space 413 of the cylindrical portion 410 .
  • the operator forms a small incision around the patient's abdomen. Then, the operator percutaneously punctures the injection needle 120 and the distal end portion 132 of the outer tube 130 accommodating the injection needle 120 under echo to gain access to the front of the tumor. Then, the operator pierces only the injection needle 120 into the inside of the tumor, moves the pressing portion 220 relative to the cylinder portion 210 in the axial direction so that the internal space of the cylinder portion 210 is reduced, and injects the anticancer agent. is administered to the affected area (see Figure 10). After completing the administration of the anticancer drug, the operator accommodates the tip of the injection needle 120 in the tip 132 of the outer cylinder 130 (see FIG. 11), and removes the administration device 100 from the body.
  • the treatment device kit 1 has been described above as including the instrument 300 and the auxiliary tool 400 .
  • the anticancer agent described in this specification can exhibit a high antitumor effect
  • the treatment device kit that does not include the instrument 300 and the auxiliary tool 400 is also included in one embodiment of the present invention.
  • the longitudinal dimension of the injection needle 120 of the administration device 100 is set to 50 mm or more. Therefore, the injection needle 120 approached by puncturing from the body surface can puncture the tumor and administer the anticancer drug to the affected area. That is, in one aspect of the present invention, the administration device includes an injection needle having a length that allows it to penetrate from outside the body to the tumor.
  • the administration device 100 is configured such that the distal end portion 132 of the outer cylinder 130 can store the distal end portion of the injection needle 120 .
  • the administration needle 120 that has come into contact with the tumor after administration of the anticancer drug to the affected area can be prevented from coming into contact with the living body on the route from the affected area to the body surface. Therefore, tumor dissemination and anticancer drug leakage can be prevented.
  • the administration device further comprises a barrel housing the injection needle.
  • the administration device 100 has a sensor related to the detection unit 140 arranged in the liquid feed path of the injection needle 120, and the detection unit 140 is electrically connected to the notification unit 150 that notifies the pressure measured by the detection unit 140. It consists of As a result, the administration device 100 can administer the anticancer drug to the affected area and at the same time inform the user of the injection pressure during the injection of the anticancer drug. That is, in one aspect of the present invention, the administration device measures and reports the injection pressure at the same time as administering the anticancer drug.
  • One aspect of the present invention is a therapeutic device kit comprising an anticancer agent that induces immunogenic cell death and an administration device capable of directly administering the anticancer agent to tumor tissue, wherein the anticancer agent is , using an administration device having an injection needle with a length that can be pierced from outside the body to the tumor.
  • One aspect of the present invention is a therapeutic device kit comprising an anticancer agent that induces immunogenic cell death and an administration device capable of directly administering the anticancer agent to tumor tissue, wherein the administration device is an anticancer agent.
  • the injection pressure is measured and reported at the same time as the drug is administered.
  • One aspect of the present invention is a therapeutic device kit comprising an anticancer agent that induces immunogenic cell death and an administration device capable of directly administering the anticancer agent to tumor tissue, wherein the administration device is an injection needle has a chemical discharge hole on the tip or side of the
  • One aspect of the present invention is a therapeutic device kit comprising an anticancer agent that induces immunogenic cell death and an administration device capable of directly administering the anticancer agent to tumor tissue, wherein the administration device is an injection needle Equipped with a mechanism for safely filling anticancer drugs inside.
  • FIGS. 14 to 16 are diagrams explaining another form of anticancer drug administration using the administration device 100 described above.
  • the administration method of the anticancer drug can be configured as follows other than the first embodiment.
  • the anticancer drug is administered to the affected area by puncturing the affected area using the above-described treatment device kit 1 and an endoscope.
  • the endoscope includes a video system body 10 and a videoscope 20 as shown in FIG. Since a known endoscope can be used in this embodiment, a portion of the endoscope related to injection needle 120 of administration device 100 will be mainly described below.
  • the video system main body 10 performs processing for recording information captured by the video scope 20 and displaying it on a monitor (display).
  • the videoscope 20 includes an operation section 30 and an insertion section 40 .
  • the insertion portion 40 includes an elongated member and is configured to provide a plurality of lumens therein.
  • the insertion section 40 includes an imaging lumen 41, an irradiation lumen 42, a fluid circulation lumen 43, and a treatment instrument insertion lumen 44, as shown in FIG.
  • the imaging lumen 41 is provided as a lumen for installing a device for acquiring images of the inside of the body, such as a CCD camera.
  • the irradiation lumen 42 is provided as a lumen in which a lighting device such as a light is installed so as to enable a clear image to be taken of the imaging location inside the body.
  • the fluid circulation lumen 43 is provided as a lumen for circulating fluid such as water and air as needed.
  • the treatment instrument insertion lumen 44 is configured as a lumen in which a treatment instrument such as forceps is inserted from the base end side and exposed from the distal end side of the elongated member to install the treatment instrument for various treatments.
  • the injection needle 120 of the administration device 100 constituting the treatment device kit 1 can be passed through the treatment instrument insertion lumen 44 .
  • the operator When administering an anticancer drug using an endoscope, the operator removes the tip of the injection needle 120 from the auxiliary tool 400 after priming as in the first embodiment. Then, the operator carries the distal end of the insertion section 40 of the endoscope to the affected area using the operation section 30 . After that, the operator inserts the injection needle 120 into the treatment instrument insertion lumen 44, and administers the anticancer agent from the distal end of the injection needle 120 to the affected area as shown in FIG.
  • the anticancer drug is administered to the affected area by the injection needle 120 inserted through the treatment instrument insertion lumen 44 of the endoscope as shown in FIG. be able to.
  • Tumor tissue to which an anticancer agent is directly administered includes cancer tissue. That is, the therapeutic device kit of this embodiment can be a therapeutic device kit for cancer therapy or prevention.
  • the tumor is a solid tumor because the administration device can easily access the tumor directly and administer the anticancer drug.
  • Solid tumors include hepatocellular carcinoma, colorectal cancer, rectal cancer, colon cancer, breast cancer, esophageal cancer, gastric cancer, bile duct cancer, pancreatic cancer, malignant melanoma, non-small cell lung cancer, and small cell cancer.
  • Lung cancer, head and neck cancer eg oral cavity cancer, nasopharyngeal cancer, oropharyngeal cancer, hypopharyngeal cancer, laryngeal cancer, salivary gland cancer and tongue cancer
  • renal cell carcinoma eg clear renal cell carcinoma
  • ovarian cancer e.g., serous ovarian cancer and ovarian clear cell adenocarcinoma
  • nasopharyngeal cancer uterine cancer (e.g., cervical cancer and endometrial cancer)
  • Anal cancer eg, anal canal cancer
  • urothelial cancer eg, bladder cancer, upper urinary tract cancer, ureter cancer, renal pelvic cancer, and urethral cancer
  • prostate cancer fallopian tube cancer
  • primary peritoneal cancer malignant pleural mesothelioma
  • gallbladder cancer biliary tract cancer
  • biliary tract cancer eg, uveal melanoma and Merkel cell carcinoma
  • testi
  • a preferred embodiment of the present invention is used such that an anticancer drug is administered in combination with an immune checkpoint inhibitor.
  • an anticancer drug is administered in combination with an immune checkpoint inhibitor.
  • Immune checkpoint receptors exist on T cells and interact with ligands expressed on antigen-presenting cells. T cells recognize and activate antigens presented on MHC molecules and initiate immune responses, but T cell activation is regulated by immune checkpoint receptor-ligand interactions occurring in parallel. Immune checkpoint receptors are co-stimulatory and inhibitory, and the balance between the two regulates T cell activation and immune responses.
  • Cancer cells express ligands for inhibitory immune checkpoint receptors and use these receptors to escape destruction by cytotoxic T cells.
  • Immune checkpoint inhibitors inhibit the function of immune checkpoints of receptors or ligands, and include, for example, inhibitory receptor antagonists and co-stimulatory immune checkpoint receptor agonists.
  • antagonist includes various substances that interfere with the activation of receptors by the binding of receptors and ligands. Examples include substances that bind to receptors and interfere with receptor-ligand binding, and substances that bind to ligands and interfere with receptor-ligand binding.
  • Antagonists against inhibitory immune checkpoints include antagonistic antibodies that bind to inhibitory immune checkpoint molecules (inhibitory receptors or ligands for the receptors), inhibitory immune checkpoint ligands designed based on Also included are soluble polypeptides that do not activate receptors, vectors capable of expressing the polypeptides, and the like.
  • antagonists against inhibitory immune checkpoint receptors include anti-PD-1 antibodies, anti-CTLA-4 antibodies, anti-LAG-3 antibodies, anti-TIM-3 antibodies, anti-BTLA antibodies, and the like.
  • Antagonists against ligands for inhibitory immune checkpoint receptors include anti-PD-L1 antibodies, anti-PD-L2 antibodies, anti-CD80 antibodies, anti-CD86 antibodies, anti-GAL9 antibodies, and anti-HVEM antibodies.
  • immune checkpoint inhibitors consist of anti-PD-1 antibody, anti-PD-L1 antibody, anti-PD-L2 antibody and anti-CTLA-4 antibody because of their high anti-tumor effect when used in combination with anti-cancer agents. It is preferably at least one selected from the group, more preferably an anti-PD-1 antibody and/or an anti-PD-L1 antibody, and even more preferably an anti-PD-1 antibody.
  • antibodies such as anti-PD-1 antibody, anti-PD-L1 antibody, and anti-PD-L2 include monoclonal antibodies, polyclonal antibodies, single-chain antibodies, and modified antibodies (for example, "humanized antibodies” in which only the antigen recognition site is humanized). ), chimeric antibodies, bifunctional antibodies capable of recognizing two epitopes simultaneously, fragment antibodies (eg, F(ab′) 2 , Fab′, Fab or Fv fragments), and the like.
  • Antibodies can be of any class, such as IgA, IgD, IgE, IgG, IgM. From the viewpoint of specific binding to antigens, it is more preferable to use monoclonal antibodies.
  • Monoclonal antibodies and polyclonal antibodies can be produced in consideration of conventionally known methods.
  • the administration method of the immune checkpoint inhibitor may be the same route as the anticancer drug or a different route.
  • the administration method of the immune checkpoint inhibitor is not particularly limited, and oral administration, intravenous injection, intraarterial injection, subcutaneous injection, intradermal injection, intraperitoneal injection, intramuscular injection, intrathecal injection, transdermal administration. Alternatively, parenteral administration such as percutaneous absorption may be used.
  • the administration method of the immune checkpoint inhibitor is preferably intraperitoneal injection or intravenous injection, and systemic injection. More preferably, it is an intravenous injection that works.
  • the order of administration of the anticancer drug and immune checkpoint inhibitor is not particularly limited, and the anticancer drug and immune checkpoint inhibitor may be administered simultaneously or at different times. Moreover, when administering with a time lag, the immune checkpoint inhibitor may be administered after administration of the anticancer agent, or the anticancer agent may be administered after administration of the immune checkpoint inhibitor.
  • Immune checkpoint inhibitors act on immune checkpoint receptors-ligands on T cells to obtain their effects. By administering an immune checkpoint inhibitor after inducing T cells, it is believed that an increase in the antitumor effect is exhibited more, so an immune checkpoint inhibitor is administered after administering an anticancer drug. is preferred. Therefore, the anticancer drug is preferably administered before the immune checkpoint inhibitor is administered. In addition, in the mode of administration with a time lag, administration routes may be the same or different as long as they are administered with a time lag.
  • compositions containing anticancer agents and immune checkpoint inhibitors are exemplified.
  • a preferred form is a combination of separately formulated anticancer agents and immune checkpoint inhibitors. That is, in this aspect, it is preferable that the therapeutic device kit is a combination of an anticancer drug, an administration device, and an immune checkpoint inhibitor.
  • the order of administration of the anticancer drug and the immune checkpoint inhibitor is not particularly limited, and the anticancer drug and the immune checkpoint inhibitor may be administered at the same time, or may be administered at different times. may be administered at any time.
  • the immune checkpoint inhibitor when administering with a time lag, may be administered after administration of the anticancer agent, or the anticancer agent may be administered after administration of the immune checkpoint inhibitor. It is preferable to administer the anticancer drug and then administer the immune checkpoint inhibitor, since the antitumor effect is more enhanced. A remarkable antitumor effect can be obtained by such an embodiment.
  • the administration routes In the mode of administration with a time lag, the administration routes may be the same or different as long as they are administered with a time lag.
  • the therapeutic device kit is preferably a therapeutic device kit for cancer therapy or prevention.
  • the dosage form when an anticancer agent and an immune checkpoint inhibitor are administered in combination is not particularly limited as long as an administration route, administration frequency and dosage suitable for each are adopted.
  • a composition containing a cancer drug and an immune checkpoint inhibitor that is, administration as a single formulation, (2) two formulations obtained by separately formulating an anticancer drug and an immune checkpoint inhibitor Simultaneous administration by the same administration route, (3) administration of two formulations obtained by separately formulating an anticancer agent and an immune checkpoint inhibitor with a time lag by the same administration route, (4) anti-cancer Simultaneous administration of two formulations obtained by separately formulating a cancer drug and an immune checkpoint inhibitor through different administration routes; Examples include administration of two formulations with a time lag through different administration routes.
  • the drugs to be administered are not limited to these two drugs, and other drugs may be added.
  • a preferred mode of administration for combined administration is to administer the immune checkpoint inhibitor after administering the anticancer drug.
  • Another aspect of the present invention includes administering an effective amount of an anticancer agent that causes immunogenic cell death to a subject in need of treatment or prevention directly into tumor tissue using an administration device, It is a method of treating or preventing disease.
  • a subject in need of treatment or prevention is administered an effective amount of an anticancer drug that causes immunogenic cell death using an administration device directly to tumor tissue, and an immune check is performed.
  • a method of treating or preventing a disease comprising administering an effective amount of a point inhibitor. It is particularly preferred that the disease is cancer.
  • the above subjects are preferably mammals, particularly preferably humans.
  • the anticancer drug and the immune checkpoint inhibitor are administered with a time lag, it is necessary to administer the drug at intervals sufficient to enhance the antitumor effect.
  • Specific dosing intervals are appropriately determined on a case-by-case basis, taking into consideration the patient's symptoms, age, sex, and the like.
  • anticancer drugs and immune checkpoint inhibitors may be administered in a fixed cycle.
  • the administration cycle it is preferable to adjust the administration cycle appropriately so as to suit the combined use.
  • Specific administration frequency, dosage, drip administration time, administration cycle, and the like are appropriately determined according to individual cases, taking into account the patient's symptoms, age, sex, and the like.
  • immune checkpoint inhibitors are conventionally known, and is administered, for example, once to several times a day in the range of 2-3 mg/kg/day.
  • the dosage is the same as when administered alone or lower than the usual administration route (e.g., when administered alone 0.10 to 0.99 times the maximum dose of ).
  • the dosage weight (mg/kg/Day) ratio of immune checkpoint inhibitors and anticancer drugs is also determined appropriately on an individual basis, taking into account the patient's symptoms, age, gender, etc.
  • ⁇ Preparation of test substance administration solution > ⁇ Anti-mouse PD-1 antibody ( ⁇ PD-1) administration solution preparation (100 ⁇ g/200 ⁇ L/head) 0.88 mL of ⁇ PD-1 (7.24 mg/mL) solution was taken and mixed with 11.8 mL of PBS to give 0.5 mg/mL.
  • Doxorubicin hereinafter also simply referred to as Dox
  • Dox 10 mg / mL solution Adriacin Injection 10 (contains 10 mg (potency) of doxorubicin hydrochloride of the Japanese Pharmacopoeia in 1 vial) 1 physiological saline (Saline) 0 mL was injected to dissolve the powder.
  • FBS Fluorescence-Activated fetal bovine serum
  • RPMI-1640 model number; A10491-01
  • Trypsin-EDTA model number; 25200-056
  • Penicillin-Streptomycin model number; 15140-122
  • PBS model number; 14190-144
  • isoflurane inhalation anesthetic was purchased from Mylan Pharmaceutical Co., Ltd. and used.
  • Cell culture Frozen cells (CT26.WT, ATCC, CRL-2638) were thawed in a hot bath at about 37°C and added to the culture medium (10% FBS, RPMI-1640 with Penicillin-Streptomycin added) heated to 37°C. I put it in. Then, it was centrifuged (1000 rpm, 3 min, room temperature) using a KUBOTA table top cooling centrifuge 2810 (SN Sharp J90124-A000). The supernatant was discarded, the culture medium was added, and the cells were well suspended by pipetting. Transferred to a culture flask and cultured under environmental conditions of 37° C. and 5% CO 2 concentration. Passaging was performed when the cells reached 70-90% confluence.
  • the cells After removing the culture medium from the culture flask and washing with PBS, the cells were detached with a 0.25% Trypsin-EDTA solution. A culture medium was added, the supernatant was discarded by centrifugation, and the cells were appropriately diluted with the culture medium and cultured. The cells were adjusted to reach the required number of cells, subcultured, and used for transplantation after reaching the required number of cells.
  • a WT cell suspension (5 ⁇ 10 5 cells/50 ⁇ L/head) was injected with a 1 mL syringe for tuberculin (Terumo Syringe, SS-01T) equipped with a 23G ⁇ 1′′ injection needle (Terumo injection needle, NN-2325R). It was implanted subcutaneously on the right back.
  • Grouping The day of transplantation is Day 0, animals with a tumor volume of about 41 mm 3 to about 145 mm 3 on Day 8 are selected, and the average tumor volume of each group is uniform according to the group composition described in "B.1.”
  • the groups were divided as follows. At the time of grouping, animals with tumors that were too small or too large, those with tumors that were divided into two, or those with flattened tumors were excluded from grouping.
  • Tumor volume ( mm3 ) 1/2 x L x W x W L: long diameter of tumor (mm), W: short diameter of tumor (mm).
  • PBS or ⁇ PD-1 was intraperitoneally administered at 200 ⁇ L/head, and the remaining solution after administration was discarded.
  • a 1 mL syringe for tuberculin (Terumo Syringe, SS-01T) fitted with a 26G ⁇ 1/2′′ injection needle (Terumo injection needle, NN-2613S) was used.
  • Saline or Dox was administered via tail vein or intratumor. In the tail vein administration, administration was performed at 5 mL/kg based on the most recent body weight value, and in the intratumoral administration, administration was performed at 20 ⁇ L/site assuming a body weight of 20 g.
  • a needle-implanted syringe (27G ⁇ 1/2′′, FN syringe, SS-010F2713) was used for tail vein administration, and a needle-implanted syringe (29G ⁇ 1/2′′, FN syringe) was used for intratumoral administration. , SS-010F2913) was used.
  • Administration was carried out three times on Days 8, 11 and 14 with any vehicle or test substance. In Groups 3, 5, 8, 10 and 12, ⁇ PD-1 was administered after an interval of 1 hour or more after Dox administration.
  • the group in which doxorubicin was directly administered to the tumor tissue has a high antitumor effect.
  • a higher antitumor effect can be obtained by using an immune checkpoint inhibitor in combination.
  • the antitumor effect is particularly high, and the immune checkpoint inhibitor administration at a dose at which the antitumor effect is not seen alone, the anticancer drug Since the antitumor effect was enhanced compared to intratumoral administration alone, it is a synergistic effect of the anticancer drug and the immune checkpoint inhibitor alone.
  • the antitumor effect is not necessarily enhanced. Considering these circumstances, it can be said that the synergistic effect of the combination is a surprising result.
  • Rat PK test A. Experimental method 1 . Animal Species Used: Rat, Strain: Crl: CD (SD), Gender: Female, Age: 5 Weeks, Microbial Control: SPF 2. Reagent used
  • Group composition The group composition consisted of two groups, an intratumoral administration group and an intravenous administration group.
  • the cells were collected from the T75 flask together with the medium into a 50 mL tube, and about 10 mL of DMEM medium was used to collect the remaining cells into a 50 mL tube. After centrifugation using the aforementioned centrifuge (1000 rpm, 5 minutes, room temperature), the supernatant was removed. The cells were resuspended by adding 10 mL of DMEM medium and counted using a hemocytometer, resulting in a cell suspension of about 2.3 ⁇ 10 6 cells/mL. The cell count was obtained by adding 1 mL to one T75 flask containing 10 mL DMEM medium and placing it in a CO 2 incubator. The following day, the T75 flask was harvested along with the medium. Of the total volume of about 35 mL, 2 mL was taken and transferred to one T75 flask containing 10 mL of DMEM medium.
  • the cells were harvested together with the medium from one T75 flask. Further, about 5 mL of DMEM medium was added to the T75 flask to collect the remaining cells (twice). After centrifugation using the aforementioned centrifuge (1000 rpm, 5 minutes, room temperature), the supernatant was removed and 10 mL of DMEM medium was added for resuspension to obtain a cell suspension of about 3.7 ⁇ 10 6 cells/mL. Approximately 1 mL of this was added to two T75 flasks (containing 10 mL of DMEM medium) and placed in a CO 2 incubator.
  • the cells were harvested together with the medium from two T75 flasks. Additional approximately 5 mL of DMEM medium was added to each T75 flask to collect the remaining cells (twice). After centrifugation using the aforementioned centrifuge (1000 rpm, 5 minutes, room temperature), the supernatant was removed and 20 mL of DMEM medium was added for resuspension to obtain a cell suspension of approximately 3.3 ⁇ 10 6 cells/mL. Approximately 0.6 mL of this was added to 15 T75 flasks (containing 10 mL of DMEM medium) and placed in a CO 2 incubator.
  • the cells were collected together with the medium from 15 T75 flasks. Additional approximately 5 mL of DMEM medium was added to each T75 flask to collect the remaining cells (twice). After centrifugation using the aforementioned centrifuge (1000 rpm, 5 minutes, room temperature), the supernatant was removed and 10 mL of DMEM medium was added for resuspension to obtain a cell suspension of about 3.1 ⁇ 10 7 cells/mL. 8 mL of this cell suspension was taken and centrifuged using the aforementioned centrifuge (1000 rpm, 5 minutes, room temperature). got
  • flank skin was pinched and 13762-MAT-BIII cells were implanted subcutaneously at one site per animal, at about 4 ⁇ 10 6 cells/100 ⁇ L per site.
  • Tumor diameter measurement and grouping On the 4th, 7th, and 9th days after cell transplantation, under the same anesthesia as during cell transplantation, the size of tumors formed in the flanks was measured using a vernier caliper, and the major and minor diameters were measured once per animal. measured times. The tumor size was calculated by the formula of major axis ⁇ (shorter axis) 2 ⁇ 2. Based on the tumor size on the 9th day, 2 mice and 3 mice were selected for the intravenous administration group and the intratumor administration group, respectively, in descending order of size.
  • Drug administration Drug preparation 1 mL of physiological saline was added to each of 2 vials of Adriacin Injection 10 and dissolved together to prepare about 2 mL of doxorubicin hydrochloride 10 mg/mL solution. To 1.6 mL of the 10 mg/mL solution of doxorubicin hydrochloride was added 6.4 mL of physiological saline to prepare a 2 mg/mL solution of doxorubicin hydrochloride. Assuming a body weight of 200 g, 0.2 mg of doxorubicin hydrochloride (drug volume: 0.1 mL) was administered to all individuals so that the dose would be 1 mg/kg.
  • Drug administration Administration was performed under the same anesthesia as during cell transplantation.
  • ⁇ Intratumor administration> (1) A syringe filled with doxorubicin hydrochloride (2 mg/mL) was set on a syringe pump (TE-361, Terumo Corporation) and an extension tube was connected. A 3-way stopcock was connected to the distal end and a manometer and an extension tube were connected. (2) A syringe needle for administration was connected to the distal end of the extension tube. The line was filled with the drug solution before administration. (3) After confirming the position of the tumor by applying an ultrasonic probe to the periphery of the tumor, an injection needle (22G) was inserted to confirm that the tip of the needle was near the center of the tumor.
  • a syringe filled with doxorubicin hydrochloride (2 mg/mL) was set on a syringe pump (TE-361, Terumo Corporation) and an extension tube was connected. A 3-way stopcock was connected to the distal end and a manometer and an extension tube were connected.
  • Plasma collection, euthanasia, and sample processing were performed in the same manner as (5), (6), and (7) during intratumoral administration.
  • Doxorubicin hydrochloride concentration measurement ⁇ Reagent> Doxorubicin hydrochloride (product number: D1515, Sigma-Aldrich Corp.) was used as a standard substance. Daunorubicin hydrochloride (product number: 30450, Sigma-Aldrich Corp.) was used as an internal standard.
  • the standard stock solution is Doxorubicin hydrochloride 1.07 mg (1.00 mg as doxorubicin, correction factor: 1.07) is accurately weighed and dissolved in methanol to 100 ⁇ g/mL, and the internal standard stock solution is Daunorubicin hydrochloride 1.0 mg. Weighed and dissolved in methanol to 100 ⁇ g/mL.
  • the standard solution was prepared by appropriately diluting the standard stock solution with methanol to 10.0 to 10000 ng/mL before use.
  • the internal standard solution was appropriately diluted with methanol and prepared to 200 ng/mL just before use.
  • ⁇ Pretreatment> Measurement of Doxorubicin Concentration in Plasma The measurement sample was thawed at room temperature, stirred, centrifuged (2000 ⁇ g, 4° C., 5 minutes), and 20 ⁇ L of the supernatant was collected in a microtest tube and used for the experiment. Deproteinization was performed with methanol, and after centrifugation, the supernatant was transferred to a glass test tube and concentrated to dryness under a nitrogen stream. The residue was redissolved by adding 0.1% formic acid-acetonitrile (70:30, v/v) to prepare an actual measurement sample.
  • a homogenate was prepared by adding 5 times the amount of methanol to the total weight of the rat tissue and crushing it. At the time of use, the tissue homogenate was centrifuged (2000 ⁇ g, 4° C., 5 minutes), and 20 ⁇ L of the supernatant was collected in a microtest tube and used for pretreatment. Deproteinization was performed with methanol, and after centrifugation, the supernatant was transferred to a glass test tube and concentrated to dryness under a nitrogen stream. The residue was redissolved by adding 0.1% formic acid-acetonitrile (70:30, v/v) to prepare an actual measurement sample.
  • 0.1% formic acid-acetonitrile 70:30, v/v
  • the plasma doxorubicin concentration is low due to the direct administration of the device into the tumor, despite the high intratumoral doxorubicin concentration. Therefore, direct administration into the tumor using the device is expected to reduce systemic side effects as well as a high antitumor effect.
  • a cell suspension was prepared so that Ninety animals were under isoflurane anesthesia (induction anesthesia: 2.0-3.0%, maintenance anesthesia: 2.0%), the graft site (lower right abdomen) was shaved, and prepared CT26. WT cells were implanted subcutaneously in the right flank (between the forelimb and hindlimb) using a 29G FN syringe (5 ⁇ 10 5 cells/100 ⁇ L/mouse). The day of transplantation was defined as Day0.
  • Tumor minor diameter and major diameter were measured on the day of grouping (Day 8), and tumor volume was obtained from the following formula.
  • Tumor volume ( mm3 ) 1/2 x L x W x W L: long diameter of tumor (mm), W: short diameter of tumor (mm) 1.4.
  • Grouping Grouping was performed based on the tumor volume on Day 8. At this time, 42 animals with the smallest tumor volume were excluded after excluding "individuals with distorted tumors". Forty-eight of the 90 cancer-bearing mice were divided into the following groups so that the average tumor volume was uniform.
  • Preparation of test substance administration solution ⁇ Preparation of Dox administration solution> Dox 10 mg/mL: Inject 1.0 mL of Saline into a 10 vial for adriacin injection to dissolve the powder and obtain a stock solution.
  • ⁇ i. v. (Intra-tail vein administration) administration solution The above 10 mg/mL stock solution was diluted 100-fold according to Table 14 below, and Dox 1 mg/kg i.v. v. It was used as an administration solution.
  • ⁇ PD-1 antibody Anti-mouse PD-1 antibody
  • 0.50 mL of ⁇ PD-1 stock solution (7.24 mg/mL) was added to 6.74 mL of PBS and mixed to give a concentration of 100 ⁇ g/200 ⁇ L/head, and ⁇ PD-1 antibody administration solution (0.5 mg/mL) and bottom.
  • Groups 1, 2, 3, and 6 were administered Saline or Dox under isoflurane anesthesia (induction anesthesia: 2.0-3.0%, maintenance anesthesia: 2.0%). was administered intratumorally (it) at 20 ⁇ L/head using a 29G FN syringe.
  • Dox was administered in the tail vein (i.v.) with a 27G injection needle and a 1 mL thermosyringe after calculating the dosage of Dox at 10 mL/kg based on the body weight of each individual under wakefulness.
  • PBS or ⁇ PD-1 antibody was intraperitoneally administered at 200 ⁇ L/head using a two-stage top needle and a 1 mL thermosyringe under awake conditions.
  • a hemolytic agent (ACK Lysing Buffer) was added to samples in which contamination with blood was observed, and after suspension, the samples were allowed to stand at room temperature for 1 to 2 minutes. rice field).
  • RPMI1640+10% FBS (9 mL) was added, and after centrifugation at 400 ⁇ g, 4° C., 5 min, the supernatant was decanted and the cells were loosened by tapping. After confirming that there was no residual hemolysis, 15 mL of PBE buffer (Rinsing Solution: BSA Solution 20:1) was added and mixed.
  • Lymph node isolated cell staining (dead cell staining, blocking, tetramer staining, antibody staining) ⁇ Dead cell staining> 1 mL of PEB buffer was added to the round tube containing the cells and mixed. After centrifugation at 400 ⁇ g, 4° C., 5 min, the supernatant was decanted, the cells were gently loosened by vortexing, and 1 mL of PBS was added.
  • ⁇ Tetramer staining> A cocktail for AH-1 tetramer staining (APC-AH-1 tetramer; 5 ⁇ L, Stain Buffer; 20 ⁇ L per tube) was prepared, and 25 ⁇ L was added to the round tube after the blocking reaction.
  • APC-AH-1 tetramer staining cocktail APC- ⁇ -galactosidase tetramer; 5 ⁇ L, Stain Buffer; 20 ⁇ L per bottle
  • 25 ⁇ L was added to a round tube. After addition of the tetramer, the mixture was lightly vortexed and allowed to stand at 4°C, shielded from light, for 40 minutes.
  • Antibody staining cocktail per CP-CD45 antibody; 2.5 ⁇ L, PE-Cy7-CD3 antibody; 2.5 ⁇ L, APC-Cy7-CD4 antibody; 2.5 ⁇ L, FITC-CD8 antibody; 10 ⁇ L, PE-CD69 antibody ; 2.5 ⁇ L, Stain Buffer; 5 ⁇ L) were prepared, and 25 ⁇ L were added to the round tube after tetramer staining.
  • Isotype Control contains Isotype tetramer staining cocktail (per bottle PerCP-Isotype antibody; 2.5 ⁇ L, PE-Cy7-Isotype antibody; 2.5 ⁇ L, APC-Cy7-Isotype antibody; 2.5 ⁇ L, FITC -Isotype antibody; 10 ⁇ L, PE-Isotype antibody; 2.5 ⁇ L, Stain Buffer; 5 ⁇ L) is added, and Isotype Control (AH-1 tetramer, CD69) is an Isotype tetramer staining cocktail (PerCP-CD45 antibody per bottle; 2.5 ⁇ L, PE-Cy7-CD3 antibody; 2.5 ⁇ L, APC-Cy7-CD4 antibody; 2.5 ⁇ L, FITC-CD8 antibody; 10 ⁇ L, PE-Isotype antibody; 2.5 ⁇ L, Stain Buffer; 5 ⁇ L) was added . After addition of the antibody staining cocktail, the mixture was lightly vortetramer sta
  • Fig. 3 The FSC-A/SSC-A plot was developed from All Events to select cell populations.
  • a Zombie Aqua/FSC-A plot was developed from the cell population, and living cells from which the dead cell population (Zombie Aqua-positive cells) had been removed were selected.
  • CD45/SSC-A was expanded from living cells and the CD45+ population was selected.
  • CD3/SSC-A was expanded from the CD45+ population to select the CD3+ population.
  • FSC-H/FSC-A was expanded from the CD3+ cell population, doublets were removed, and the Single cell 1 population was selected.
  • SSC-H/SSC-A was developed from the single cell 1 population, doublets were removed, and the single cell 2 population was selected.
  • Expand CD8/CD4 from the Single cell 2 population, CD8+ cells to Total. CTL and CD4+ cells were used as helper T cells.
  • Total. CTLs were expanded with CD8/CD69, and CD69+ cells were defined as CD69+ CTLs (activated CTLs).
  • Total. CTL was expanded with CD8/AH-1 Tetramer, and AH-1 Tetramer+ cells were designated as AH-1 specific CTL (cancer antigen-specific CTL).
  • Results Figures 4 to 6 show the average value and standard error of each group for the number of cells in each group.
  • Figure 4 shows Total.
  • Figure 5 shows the AH-1 specific CTL number and
  • Figure 6 shows the CD69+ CTL number.
  • the number of CTLs means the total number of CTLs
  • the number of AH-1 specific CTLs means CTLs that specifically recognize tumors
  • the number of CD69+ CTLs means CTLs in an activated state.
  • Table 15 shows the rate of increase in CTL, AH-1 specific CTL, and CD69+ CTL relative to controls.
  • each administration device described in the administration device section can be used as the administration device.
  • 1 therapeutic device kit 100 dosing device, 120, 120a injection needle, 130 outer cylinder, 140 detection unit, 150 annunciation unit;

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Vascular Medicine (AREA)
  • Hematology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Biomedical Technology (AREA)
  • Anesthesiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Infusion, Injection, And Reservoir Apparatuses (AREA)

Abstract

Le but de la présente invention est de fournir une méthode présentant un excellent effet antitumoral. La solution selon l'invention concerne un kit dispositif thérapeutique comportant un médicament anticancéreux destiné à induire une mort cellulaire immunogène, et un dispositif d'administration pouvant administrer le médicament anticancéreux en direction d'un tissu tumoral, le médicament anticancéreux induisant l'activation d'une immunité antitumorale du fait de son administration à l'aide du dispositif d'administration.
PCT/JP2022/035371 2021-09-27 2022-09-22 Kit dispositif thérapeutique WO2023048230A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2021-156771 2021-09-27
JP2021156771 2021-09-27

Publications (1)

Publication Number Publication Date
WO2023048230A1 true WO2023048230A1 (fr) 2023-03-30

Family

ID=85720798

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2022/035371 WO2023048230A1 (fr) 2021-09-27 2022-09-22 Kit dispositif thérapeutique

Country Status (1)

Country Link
WO (1) WO2023048230A1 (fr)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012128129A1 (fr) * 2011-03-22 2012-09-27 オリンパス株式会社 Aiguille de perforation, appareil d'administration de médicament et pompe à seringue
US9399101B1 (en) * 2012-02-03 2016-07-26 Patrick Jean Le Pivert Needle system for tissue perfusion
WO2022138740A1 (fr) * 2020-12-23 2022-06-30 テルモ株式会社 Dispositif médical et trousse de dispositif médical
WO2022168862A1 (fr) * 2021-02-08 2022-08-11 テルモ株式会社 Instrument médical

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012128129A1 (fr) * 2011-03-22 2012-09-27 オリンパス株式会社 Aiguille de perforation, appareil d'administration de médicament et pompe à seringue
US9399101B1 (en) * 2012-02-03 2016-07-26 Patrick Jean Le Pivert Needle system for tissue perfusion
WO2022138740A1 (fr) * 2020-12-23 2022-06-30 テルモ株式会社 Dispositif médical et trousse de dispositif médical
WO2022168862A1 (fr) * 2021-02-08 2022-08-11 テルモ株式会社 Instrument médical

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SHI YINGGE, LI DONG, HE CHAOLIANG, CHEN XUESI: "Design of an Injectable Polypeptide Hydrogel Depot Containing the Immune Checkpoint Blocker Anti‐PD‐L1 and Doxorubicin to Enhance Antitumor Combination Therapy", MACROMOLECULAR BIOSCIENCE, WILEY-VCH VERLAG GMBH, DE, vol. 21, no. 6, 1 June 2021 (2021-06-01), DE , pages 2100049, XP093051273, ISSN: 1616-5187, DOI: 10.1002/mabi.202100049 *

Similar Documents

Publication Publication Date Title
ES2938321T3 (es) Métodos para tratar el cáncer de piel mediante la administración de un inhibidor de PD-1
RU2708374C2 (ru) Комбинированная терапия для лечения рака
JP7384949B2 (ja) 肺癌の処置のための抗pd-1抗体
JP2014132009A5 (fr)
WO2014142220A1 (fr) Agent antitumoral
US20170106067A1 (en) Combinatorial immunotherapy for pancreatic cancer treatment
US20130006034A1 (en) Methods of enhancing the response to radiation in tumor therapy using anti-dll4 antibodies
CN110831629B (zh) 用于治疗癌症的包括abx196的组合
US20160237159A1 (en) Methods and compositions for regulatory t-cell ablation
EP3925973A1 (fr) Compositions vaccinales et méthodes pour rétablir la fonction de la voie nkg2d contre les cancers
WO2023048230A1 (fr) Kit dispositif thérapeutique
AU2020308283A1 (en) Small molecule inhibitors for treating cancer in a subject having tumors with high interstitial pressure
Sun et al. Tumor interstitial fluid and gastric cancer metastasis: an experimental study to verify the hypothesis of “tumor-phlegm microenvironment”
CN117043193A (zh) 通过施用新辅助pd-1抑制剂治疗癌症的方法
Song et al. Blocking transforming growth factor-beta reduces the migration and invasion of the residual tumour after TAE
Coventry et al. Immuno-chemotherapy using repeated vaccine treatment can produce successful clinical responses in advanced metastatic melanoma
WO2020150567A1 (fr) Fibroblastes et microvésicules correspondantes pour la réduction de la toxicité associée à l'immunothérapie anticancéreuse
Kang et al. Effectiveness of antibody-drug conjugate (ADC): Results of in vitro and in vivo studies
RU2771759C2 (ru) Антитела против pd-1 для лечения рака легких
US20190170756A1 (en) Tirc7 based diagnostic and therapy of solid cancer
CN108472366A (zh) 用于免疫介导的癌症疗法的组合物和方法
WO2024059833A1 (fr) Utilisation d'anticorps anti-ctla-4 pour traiter le carcinome adénoïde kystique
Patel et al. The New Era Drug: Dostarlimab Having Effective Clinical Trial & Combination Clinical Therapies
Jørgensen et al. Peptide vaccination against PD-L1 in multiple myeloma: A phase I trial
EA045900B1 (ru) Комбинация антител против pd-1 и облучения для лечения злокачественной опухоли

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22872981

Country of ref document: EP

Kind code of ref document: A1