WO2024012464A1 - Utilisation d'un composé d'imidazotétrazine - Google Patents

Utilisation d'un composé d'imidazotétrazine Download PDF

Info

Publication number
WO2024012464A1
WO2024012464A1 PCT/CN2023/106847 CN2023106847W WO2024012464A1 WO 2024012464 A1 WO2024012464 A1 WO 2024012464A1 CN 2023106847 W CN2023106847 W CN 2023106847W WO 2024012464 A1 WO2024012464 A1 WO 2024012464A1
Authority
WO
WIPO (PCT)
Prior art keywords
dlbcl
cell
lymphoma
cell lymphoma
tumor
Prior art date
Application number
PCT/CN2023/106847
Other languages
English (en)
Chinese (zh)
Inventor
尚宝虎
马静静
侯晓磊
Original Assignee
北京紫萌医药科技有限公司
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 北京紫萌医药科技有限公司 filed Critical 北京紫萌医药科技有限公司
Publication of WO2024012464A1 publication Critical patent/WO2024012464A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41881,3-Diazoles condensed with other heterocyclic ring systems, e.g. biotin, sorbinil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention belongs to the field of medicine, and more specifically, the present invention relates to new anti-tumor uses of imidazotetrazine compounds, and the use of pharmaceutical preparations at least including them as active ingredients in the prevention and treatment of diffuse lymphoma.
  • lymphoma According to the "World Health Organization Pathological Classification Criteria for Lymphatic System Tumors", there are currently nearly 70 known pathological types of lymphoma, which can be roughly divided into two categories: Hodgkin's lymphoma and non-Hodgkin's lymphoma. Among them, Hodgkin's lymphoma Lymphoma accounts for about 10% of lymphomas, and non-Hodgkin lymphoma accounts for about 90% of all lymphoma cases, and the incidence rate is increasing year by year. Non-Hodgkin's lymphoma is divided into two categories: B cell type and T/NK cell type, of which B cell type lymphoma accounts for about 70% and T/NK cell type lymphoma accounts for about 30%. With the basic and clinical research With the continuous development, the classification of lymphoma is still further refined and improved.
  • DLBCL diffuse large B lymphoma
  • NHL non-Hodgkin lymphoma
  • DLBCL may be related to congenital inheritance, out-of-control apoptosis mechanism, and DNA repair defects.
  • the specific cause of DLBCL is still unclear.
  • the clinical efficacy of DLBCL treatment has improved significantly, mainly due to the traditional combination of rituximab and chemotherapy as initial treatment and the refinement of high-dose chemotherapy and autologous stem cell transplantation strategies to treat DLBCL.
  • both treatment options have the problem of drug resistance in DLBCL treatment, which affects the clinical application of related drugs. Therefore, new treatment strategies are urgently needed.
  • TMZ The imidazotetrazine compound 3-methyl-4-oxo-8-imidazo[5,1-d][1,2,3,5]tetrazinecarboxamide
  • Temodar A chemotherapeutic agent approved for the treatment of brain tumors (marketed by Schering under the trademark Temodar in the United States and in Europe under the trademark Temodal).
  • Temodal a chemotherapeutic agent approved for the treatment of brain tumors
  • TMZ degrades via pH-dependent hydrolysis to its active metabolite MTIC (3-methyl-(triazen-1-yl)imidazole-4-carboxamide).
  • Temodar capsules are currently indicated in the United States for the treatment of adult patients with newly diagnosed glioblastoma multiforme and refractory anaplastic astrocytoma, and Temodar capsules are currently approved in Europe for the treatment of patients with malignant glial tumors. tumor.
  • TMZ has been reported to be used in the treatment of a variety of lymphomas, such as the treatment of T/NK cell type (Ma Xuejun, Fudan Study on the Treatment Patterns, Radiotherapy Plans and Prognostic Related Factors of Early Extranodal NK/T Cell Lymphoma Nasal Type University doctoral thesis), treatment of primary central nervous system lymphoma (O Asif and M Nehal, Temozolomide for Relapsed Primary CNS Lymphoma Journal of the College of Physicians and Surgeons Pakistan 2012,Vol.22(9):594-595 ), treatment of leukemia and lymphoma (Wang Chunmei, study on the synthesis and biological activity of temozolomide derivatives, Guizhou University master's thesis).
  • the object of the present invention is to provide the application of imidazotetrazine compounds, especially TMZ, in the preparation of medicines for preventing or treating diffuse lymphoma.
  • the present invention surprisingly found that imidazotetrazine compounds selectively treat tumors other than the primary site. Or diffuse lymphoma whose metastasis site is the central nervous system has excellent antagonistic effects.
  • the first aspect of the present invention provides imidazotetrazine compounds, or their solvates, hydrates, polymorphs, prodrugs or pharmaceutically acceptable salts for the preparation of medicaments for the treatment and/or prevention of diffuse lymphoma.
  • imidazotetrazine compound is represented by formula I:
  • R 1 is hydrogen or C1-C3 alkyl
  • R 2 is amino or amino substituted by C1-C3 alkyl
  • R 3 is hydrogen or C1-C3 alkyl
  • the DLBCL is selected from diffuse large B-cell lymphoma whose primary site and/or metastatic site is not the central nervous system (except for DLBCL whose primary site and/or metastatic site is the central nervous system). ).
  • the DLBCL is selected from the group consisting of unspecified DLBCL, other large B-cell lymphoma, high-grade B-cell lymphoma, and unclassifiable B-cell lymphoma.
  • the DLBCL is selected from the group consisting of centroblast DLBCL, immunoblastic variant DLBCL, anaplastic DLBCL, spindle cell variant DLBCL, signet ring cell-like variant DLBCL, T cell/histiocyte enriched DLBCL, primary cutaneous DLBCL, EBV-positive DLBCL, chronic inflammation-associated large B-cell lymphoma, lymphomatoid granuloma, large B-cell lymphoma with IRF4 rearrangement, large B-cell lymphoma with IRF4/MUM1 rearrangement cellular lymphoma, primary mediastinal large B-cell lymphoma, intravascular large B-cell lymphoma, ALK-positive large B-cell lymphoma, plasmablastic lymphoma, HHV8-positive DLBCL, primary effusion lymphoma, high Grade B-cell lymphoma with MYC, BCL2 and/or BCL6 rearrangements, high-grade B-cell lymphom
  • the DLBCL is primary DLBCL, or transformed DLBCL, or advanced and/or relapsed, and/or refractory DLBCL, or DLBCL that has previously received systemic therapy, or DLBCL that has failed chemotherapy drugs and/or targeted drugs and/or immunotherapy.
  • the cells of the DLBCL are selected from the group consisting of human diffuse histological lymphoma cell SU-DHL-2, human diffuse lymphoma cell line TMD-8, and human diffuse large cell lymphoma cell SU-DHL-6 one or more of them.
  • a second aspect of the present invention provides the use of a pharmaceutical composition in the preparation of medicaments for the treatment and/or prevention of DLBCL, the pharmaceutical composition comprising an imidazotetrazine compound represented by Formula I or a solvate thereof , hydrates, polymorphs, prodrugs or pharmaceutically acceptable salts thereof, and pharmaceutically acceptable excipients,
  • R 1 is hydrogen or C1-C3 alkyl
  • R 2 is amino or amino substituted by C1-C3 alkyl
  • R 3 is hydrogen or C1-C3 alkyl
  • R1 is methyl
  • R2 is amino
  • R3 is hydrogen
  • R 1 is methyl
  • R 2 is amino
  • R 3 is hydrogen
  • the DLBCL is selected from diffuse large B-cell lymphoma whose primary site and/or metastatic site is not the central nervous system.
  • the DLBCL is selected from the group consisting of unspecified DLBCL, other large B-cell lymphoma, high-grade B-cell lymphoma, and unclassifiable B-cell lymphoma.
  • the DLBCL is selected from the group consisting of centroblast DLBCL, immunoblastic variant DLBCL, anaplastic DLBCL, spindle cell variant DLBCL, signet ring cell-like variant DLBCL, T cell/histiocyte enriched DLBCL, primary cutaneous DLBCL, EBV-positive DLBCL, chronic inflammation-associated large B-cell lymphoma, lymphomatoid granuloma, large B-cell lymphoma with IRF4 rearrangement, large B-cell lymphoma with IRF4/MUM1 rearrangement cellular lymphoma, primary mediastinal large B-cell lymphoma, intravascular large B-cell lymphoma, ALK-positive large B-cell lymphoma, plasmablastic lymphoma, HHV8-positive DLBCL, primary effusion lymphoma, high Grade B-cell lymphoma with MYC, BCL2 and/or BCL6 rearrangements, high-grade B-cell lymphom
  • the DLBCL is primary DLBCL, or transformed DLBCL, or advanced and/or relapsed, and/or refractory DLBCL, or DLBCL that has previously received systemic therapy, or DLBCL that has failed chemotherapy drugs and/or targeted drugs and/or immunotherapy.
  • the cells of the DLBCL are selected from the group consisting of human diffuse histological lymphoma cell SU-DHL-2, human diffuse lymphoma cell line TMD-8, and human diffuse large cell lymphoma cell SU-DHL-6 one or more of them.
  • a third aspect of the present invention also provides a pharmaceutical combination, including an imidazotetrazine compound and a second therapeutic agent that can be used to treat DLBCL.
  • the imidazotetrazine compound is represented by Formula I:
  • R 1 is hydrogen or C1-C3 alkyl
  • R 2 is amino or C1-C3 alkyl substituted amino
  • R 3 is hydrogen or C1-C3 alkyl
  • R1 is methyl
  • R2 is amino
  • R3 is hydrogen
  • R 1 is methyl
  • R 2 is amino
  • R 3 is hydrogen, i.e., the imidazotetrazine is temozolomide (3-methyl-4-oxo-8-imidazo[5,1 -d][1,2,3,5]tetrazinecarboxamide).
  • the second therapeutic agent is one or more of chemotherapy drugs, small molecule targeted anti-tumor drugs, immunotherapy drugs, and antibody drugs.
  • the second therapeutic agent is cyclophosphamide.
  • the fourth aspect of the present invention relates to the use of the pharmaceutical combination described in the third aspect in the preparation of medicaments for the treatment and/or prevention of DLBCL.
  • the DLBCL is selected from DLBCL other than DLBCL whose primary site and/or metastatic site is the central nervous system.
  • the DLBCL is selected from the group consisting of unspecified DLBCL, other large B-cell lymphoma, high-grade B-cell lymphoma, and unclassifiable B-cell lymphoma.
  • the DLBCL is selected from the group consisting of centroblast DLBCL, immunoblastic variant DLBCL, anaplastic DLBCL, spindle cell variant DLBCL, signet ring cell-like variant DLBCL, T cell/histiocyte enriched DLBCL, primary cutaneous DLBCL, EBV-positive DLBCL, chronic inflammation-associated large B-cell lymphoma, lymphomatoid granuloma, large B-cell lymphoma with IRF4 rearrangement, large B-cell lymphoma with IRF4/MUM1 rearrangement cellular lymphoma, primary mediastinal large B-cell lymphoma, intravascular large B-cell lymphoma, ALK-positive large B-cell lymphoma, plasmablastic lymphoma, HHV8-positive DLBCL, primary effusion lymphoma, high Grade B-cell lymphoma with MYC, BCL2 and/or BCL6 rearrangements, high-grade B-cell lymphom
  • the DLBCL is primary DLBCL, or transformed DLBCL, or advanced and/or relapsed, and/or refractory DLBCL, or DLBCL that has previously received systemic therapy, or DLBCL that has failed chemotherapy drugs and/or targeted drugs and/or immunotherapy.
  • the cells of the DLBCL are selected from the group consisting of human diffuse histological lymphoma cell SU-DHL-2, human diffuse lymphoma cell line TMD-8, and human diffuse large cell lymphoma cell SU-DHL-6 one or more of them.
  • Another aspect of the present invention relates to a method for treating and/or preventing DLBCL in a subject, the method comprising administering to the subject an effective amount of an imidazotetrazine compound or a solvate, hydrate thereof Materials, polymorphs, prodrugs or pharmaceutically acceptable salts or pharmaceutical combinations thereof, wherein the imidazotetrazine compound is represented by formula I:
  • R 1 is hydrogen or C1-C3 alkyl
  • R 2 is amino or amino substituted by C1-C3 alkyl
  • R 3 is hydrogen or C1-C3 alkyl
  • R1 is methyl
  • R2 is amino
  • R3 is hydrogen
  • R 1 is methyl, R 2 is amino; R 3 is hydrogen.
  • the imidazotetrazine is 3-methyl-4-oxo-8-imidazo[5,1-d][1,2,3,5]tetrazinecarboxamide.
  • the pharmaceutical combination further includes a second therapeutic agent capable of treating DLBCL.
  • the second therapeutic agent is one or more of chemotherapy drugs, small molecule targeted anti-tumor drugs, immunotherapy drugs, and antibody drugs.
  • the second therapeutic agent is cyclophosphamide.
  • the subject is a mammal, preferably a human.
  • the DLBCL is selected from DLBCL other than DLBCL whose primary site and/or metastatic site is the central nervous system.
  • the DLBCL is selected from the group consisting of unspecified DLBCL, other large B-cell lymphoma, high-grade B-cell lymphoma, and unclassifiable B-cell lymphoma.
  • the DLBCL can be selected from the group consisting of centroblast DLBCL, immunoblastic variant DLBCL, anaplastic DLBCL, spindle cell variant DLBCL, signet ring cell-like variant DLBCL, T cell/histiocyte enrichment DLBCL, primary cutaneous DLBCL, EBV-positive DLBCL, chronic inflammation-associated large B-cell lymphoma, lymphomatoid granuloma, large B-cell lymphoma with IRF4 rearrangement, large B-cell lymphoma with IRF4/MUM1 rearrangement B-cell lymphoma, primary mediastinal large B-cell lymphoma, intravascular large B-cell lymphoma, ALK-positive large B-cell lymphoma, plasmablastic lymphoma, HHV8-positive DLBCL, primary effusion lymphoma, High-grade B-cell lymphoma with MYC, BCL2 and/or BCL6 rearrangements, high-grade B-cell
  • the DLBCL can be primary DLBCL, or transformed DLBCL, or late-stage and/or relapsed, and/or refractory DLBCL, or DLBCL that has previously received systemic therapy, Or DLBCL that has failed chemotherapy drugs and/or targeted drugs and/or immunotherapy.
  • the cells of the DLBCL are selected from the group consisting of human diffuse histological lymphoma cell SU-DHL-2, human diffuse lymphoma cell line TMD-8, and human diffuse large cell lymphoma cell SU-DHL-6 one or more of them.
  • Subjects for administration include, but are not limited to: humans (i.e., males or females of any age group, e.g., pediatric subjects (e.g., infants, children, adolescents) or adult subjects (e.g., young Adults, middle-aged adults or older adults)) and/or non-human animals, for example, mammals, such as primates (e.g., cynomolgus monkeys, rhesus monkeys), cattle, pigs, horses, sheep, Goats, rodents, cats and/or dogs.
  • the subject is human.
  • the subject is a non-human animal.
  • the terms "person,” “patient,” and “subject” are used interchangeably herein.
  • treatment includes an action in a subject suffering from a particular disease, disorder, or condition that reduces the severity of the disease, disorder, or condition, or delays or slows down the disease, disorder, or condition.
  • therapeutic treatment the development of a disorder or condition
  • preventive treatment the effects that occur before a subject begins to suffer from a specific disease, disorder or condition.
  • an "effective amount" of a compound is an amount sufficient to elicit a target biological response.
  • the effective amount of a compound of the present invention may vary depending on factors such as, for example, the biological target, the pharmacokinetics of the compound, the disease being treated, the mode of administration, and the condition of the subject. Age health conditions and symptoms.
  • the effective amount includes a therapeutically effective amount and a preventive effective amount.
  • a "therapeutically effective amount" of a compound as used herein is an amount sufficient to provide a therapeutic benefit in treating a disease, disorder, or condition, or to cause one or more symptoms associated with the disease, disorder, or condition The amount to delay or minimize.
  • a therapeutically effective amount of a compound is that amount of therapeutic agent that, when used alone or in combination with other therapies, provides a therapeutic benefit in the treatment of a disease, disorder, or condition.
  • the term "therapeutically effective amount” may include an amount that improves overall treatment, reduces or avoids symptoms or causes of a disease or disorder, or enhances the therapeutic effect of other therapeutic agents.
  • a prophylactically effective amount of a compound as used herein is an amount sufficient to prevent a disease, disorder or condition, or to prevent one or more symptoms associated with a disease, disorder or condition, or to prevent a disease , the amount of recurrence of a disorder or condition.
  • a prophylactically effective amount of a compound is that amount of therapeutic agent that, when used alone or in combination with other agents, provides a prophylactic benefit in preventing a disease, disorder, or condition.
  • the term “prophylactically effective amount” may include an amount that improves overall prophylaxis, or an amount that enhances the prophylactic effect of other prophylactic agents.
  • solvate refers to a form of a compound or a salt thereof that is combined with a solvent, usually formed by a solvolysis reaction. Physical association may include hydrogen bonding.
  • solvents include water, methanol, ethanol, acetic acid, DMSO, THF, ether, etc.
  • Suitable solvates include pharmaceutically acceptable solvates and further include stoichiometric and non-stoichiometric solvates. In some cases, the solvate will be capable of isolating, for example, when one or more solvents separate When incorporated into the crystal lattice of a crystalline solid.
  • “Solvate” includes both solution solvates and isolable solvates. Representative solvates include hydrates, ethanolates, and methoxides.
  • hydrate refers to a compound combined with water. Typically, the ratio of the number of water molecules contained in a hydrate of a compound to the number of molecules of the compound in the hydrate is determined.
  • a hydrate of a compound may be represented, for example, by the general formula R.xH2O , where R is the compound and x is a number greater than zero.
  • a given compound may form more than one hydrate type, including, for example, monohydrate (x is 1), lower hydrate (x is a number greater than 0 and less than 1), for example, hemihydrate (R ⁇ 0.5H 2 O)) and polyhydrates (x is a number greater than 1, for example, dihydrate (R ⁇ 2H 2 O) and hexahydrate (R ⁇ 6H 2 O)).
  • monohydrate x is 1
  • lower hydrate x is a number greater than 0 and less than 1
  • hemihydrate R ⁇ 0.5H 2 O
  • polyhydrates x is a number greater than 1, for example, dihydrate (R ⁇ 2H 2 O) and hexahydrate (R ⁇ 6H 2 O)
  • the compounds of the invention may be in amorphous or crystalline forms (polymorphs). Furthermore, the compounds of the present invention may exist in one or more crystalline forms. Accordingly, the present invention includes within its scope all amorphous or crystalline forms of the compounds of the invention.
  • polymorph refers to a crystalline form of a compound (or a salt, hydrate or solvate thereof) in a specific crystal packing arrangement. All polymorphs have the same elemental composition. Different crystalline forms often have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, optoelectronic properties, stability and solubility. Recrystallization solvent, crystallization rate, storage temperature, and other factors can lead to the dominance of one crystalline form. Various polymorphs of the compounds can be prepared by crystallization under different conditions.
  • prodrugs are also included within the context of the present invention.
  • the term "prodrug” as used herein refers to a compound that is converted in the body to its active form having a medical effect, for example, by hydrolysis in the blood.
  • Pharmaceutically acceptable prodrugs are described in T. Higuchi and V. Stella, Prodrugs as Novel Delivery Systems, Vol. 14 of A.C.S. Symposium Series, Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, and D. Fleisher, S. Ramon and H. Barbra "Improved oral drug delivery: solubility limitations overcome by the use of prodrugs", Advanced Drug Delivery Reviews (1996) 19 (2) 115-130, which are incorporated into this article as a reference .
  • a prodrug is any covalently bonded compound of the invention that releases the parent compound in the body when administered to a patient.
  • Prodrugs are typically prepared by modifying functional groups in a manner such that the modification can be cleaved by conventional manipulations or in vivo to yield the parent compound.
  • Prodrugs include, for example, compounds of the invention in which a hydroxyl, amino or thiol group is bonded to any group that can be cleaved to form a hydroxyl, amino or thiol group when administered to a patient.
  • representative examples of prodrugs include, but are not limited to, amide derivatives of amino functionality that may be present in the compound.
  • esters such as methyl ester, ethyl ester, etc. can be used.
  • the ester itself may be reactive and/or hydrolyzable under human body conditions.
  • Suitable pharmaceutically acceptable in vivo hydrolyzable ester groups include those that readily break down in the human body to release the parent acid or salt thereof.
  • “Pharmaceutically acceptable” means a pharmaceutical composition that is generally safe, non-toxic and neither biologically or otherwise undesirable for use in the preparation of a pharmaceutical composition, and includes that is acceptable for human pharmaceutical use. accepted.
  • “Pharmaceutically acceptable salts” include, but are not limited to, acid addition salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, etc.; or with organic acids such as acetic acid, trifluoroacetic acid, propionic acid, Caproic acid, heptanoic acid, cyclopentane propionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid Acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid
  • the imidazotetrazine compound described in the present invention is represented by Formula I:
  • R 1 is hydrogen or C1-C3 alkyl
  • R 2 is amino or amino substituted by C1-C3 alkyl
  • R 3 is hydrogen or C1-C3 alkyl
  • R1 is methyl
  • R2 is amino
  • R3 is hydrogen
  • R 1 is methyl, R 2 is amino; R 3 is hydrogen, that is, the imidazotetrazine compound is temozolomide.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the imidazotetrazine compound represented by Formula I of the present invention (also referred to as "active component") and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition contains an effective amount of the imidazotetrazine compound represented by Formula I of the present invention.
  • the pharmaceutical composition contains a therapeutically effective amount of the imidazotetrazine compound represented by Formula I of the present invention.
  • the pharmaceutical composition contains a prophylactically effective amount of the imidazotetrazine compound represented by Formula I of the present invention.
  • compositions of the present invention refer to non-toxic carriers, adjuvants or vehicles that do not destroy the pharmacological activity of the compounds with which they are formulated.
  • Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of the present invention include, but are not limited to, ion exchangers, aluminum oxide, aluminum stearate, lecithin, serum proteins (such as human serum albumin) protein), buffer substances (such as phosphate), glycine, sorbic acid, potassium sorbate, partial glyceride mixture of saturated vegetable fatty acids, water, salt or electrolyte (such as protamine sulfate), disodium hydrogen phosphate, potassium hydrogen phosphate , sodium chloride, zinc salt, silica gel, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylate, wax, polyethylene-polyoxypropylene- Block polymers, polyethylene glycols, and
  • kits eg, pharmaceutical packaging.
  • Kits provided may include a compound of the invention, other therapeutic agents, and first and second containers (e.g., vials, ampoules, bottles, syringes, and/or dispersible packaging or other) containing the compounds of the invention, other therapeutic agents. suitable container).
  • provided kits may also optionally include a third container containing pharmaceutical excipients for diluting or suspending the compounds of the invention and/or other therapeutic agents.
  • the compound of the invention and the other therapeutic agent provided in the first container and the second container are combined to form a unit dosage form.
  • the present invention also provides a pharmaceutical combination, including the above-mentioned imidazotetrazine compound and a second therapeutic agent that can be used to treat DLBCL.
  • a pharmaceutical combination including the above-mentioned imidazotetrazine compound and a second therapeutic agent that can be used to treat DLBCL.
  • the imidazotetrazine compound and the second therapeutic agent may be administered to the subject separately or sequentially.
  • the second therapeutic agent may be one or more of chemotherapy drugs, small molecule targeted anti-tumor drugs, immunotherapy drugs, and antibody drugs.
  • parenteral administration as used herein includes subcutaneous administration, intradermal administration, intravenous administration, intramuscular administration, intraarticular administration, intraarterial administration, intrasynovial administration, and intrasternal administration.
  • intracerebrospinal membrane drug administration intralesional drug administration, and intracranial injection or infusion techniques.
  • an effective amount of a compound provided herein is administered.
  • the amount of compound actually administered can be determined by the physician depending on the circumstances, including the condition being treated, the route of administration chosen, the compound actually administered, the age, weight and response of the individual patient, the severity of the patient's symptoms, etc. .
  • a compound provided herein is administered to a subject at risk of developing the condition, typically on the advice of and under the supervision of a physician, at dosage levels as described above.
  • Subjects at risk of developing a particular condition generally include subjects with a family history of the condition or those who have been determined by genetic testing or screening to be particularly susceptible to developing the condition.
  • compositions provided herein can also be administered over a long period of time ("chronic administration").
  • Long-term administration refers to the administration of a compound or pharmaceutical composition thereof over a long period of time, for example, 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or administration may be continued indefinitely, For example, the remainder of the subject's life.
  • chronic administration is intended to provide a constant level of the compound in the blood over an extended period of time, eg, within a therapeutic window.
  • a pharmaceutical composition may be administered as a bolus injection, eg, in order to increase the concentration of the compound in the blood to an effective level.
  • the bolus dose depends on the target systemic levels of the active ingredient through the body, e.g., an intramuscular or subcutaneous bolus dose provides a slow release of the active ingredient, whereas a bolus dose delivered directly into the vein (e.g., via an IV drip) ) can be delivered more quickly, allowing the concentration of active ingredients in the blood to quickly increase to effective levels.
  • the pharmaceutical composition may be administered as a continuous infusion, for example, by IV infusion, thereby providing a steady-state concentration of the active ingredient in the subject's body. Additionally, in other embodiments, a bolus dose of the pharmaceutical composition may be administered first, followed by a continuous infusion.
  • Oral compositions may take the form of bulk liquid solutions or suspensions, or bulk powders. More typically, however, the compositions are provided in unit dosage form to facilitate precise dosing.
  • dosage unit form refers to physically discrete units suitable as unitary dosages for human patients and other mammals, each unit containing a predetermined quantity of active material suitable to produce the desired therapeutic effect in association with a suitable pharmaceutical excipient.
  • Typical unit dosage forms include prefilled, premeasured ampoules or syringes for liquid compositions, or pills, tablets, capsules, and the like in the case of solid compositions.
  • the compound will generally be a minor component (from about 0.1 to about 50% by weight, or preferably from about 1 to about 40% by weight), with the remainder being various components useful in forming the desired administration form. carriers or excipients and processing aids.
  • Liquid forms suitable for oral administration may include suitable aqueous or non-aqueous carriers as well as buffering agents, suspending and dispersing agents, coloring agents, flavoring agents, and the like.
  • Solid forms may include, for example, any of the following components, or compounds of similar nature: binders, such as microcrystalline cellulose, tragacanth, or gelatin; excipients, such as starch or lactose; disintegrants, For example, alginic acid, sodium carboxymethyl starch or corn starch; lubricant, for example, magnesium stearate; glidant, for example, colloidal silicon dioxide; sweetener, for example, sucrose or saccharin; or flavoring agent, for example, Mint, methyl salicylate or orange flavoring; stabilizers such as tartaric acid, aspartic acid, glutamic acid, etc.
  • binders such as microcrystalline cellulose, tragacanth, or gelatin
  • excipients such as starch or lactose
  • Injectable compositions are typically based on injectable sterile saline or phosphate buffered saline, or other injectable excipients known in the art. As stated previously, in such compositions the active compound is typically a minor component, often about 0.05 to 10% by weight, with the remainder being injectable excipients and the like.
  • Transdermal compositions are typically formulated as topical ointments or creams containing the active ingredients.
  • the active ingredients When formulated as an ointment, the active ingredients are typically combined with a paraffin or water-miscible ointment base.
  • the active ingredient may be formulated as a cream with, for example, an oil-in-water cream base.
  • Such transdermal formulations are well known in the art and often include other components for promoting stable skin penetration of the active ingredient or formulation. All such known transdermal formulations and components are included within the scope provided by this invention.
  • transdermal administration may be achieved using reservoir or porous membrane types, or a variety of solid matrix patches.
  • compositions for oral administration, injection or topical administration are merely representative.
  • Other materials and processing techniques are described in Part 8 of Remington's Pharmaceutical Sciences, 17th edition, 1985, Mack Publishing Company, Easton, Pennsylvania, which article is incorporated by reference.
  • the compounds of the present invention may also be administered in sustained release form or from a sustained release drug delivery system.
  • sustained release materials can be found in Remington's Pharmaceutical Sciences.
  • the imidazotetrazine compounds represented by formula I have value as anti-tumor agents.
  • the compounds of the invention have value as anti-proliferative, apoptotic and/or anti-invasive agents in the containment and/or treatment of solid and/or liquid tumor diseases.
  • the compounds of the present invention are expected to be useful in preventing or treating diffuse lymphoma and the like.
  • Anti-cancer effects useful for treating cancer in patients include, but are not limited to, anti-tumor effects, response rates, time to disease progression, and survival rates.
  • the anti-tumor effects of the treatment method of the present invention include, but are not limited to, inhibition of tumor growth, delay of tumor growth, tumor regression, tumor shrinkage, prolonged tumor regeneration after treatment is stopped, and slowing of disease progression.
  • Anticancer effects include preventive treatment as well as treatment of existing disease.
  • the effective amount of the compound of the present invention is usually at an average daily dose of 0.01 mg to 100 mg compound/kg patient body weight, preferably 0.1 mg to 30 mg compound/kg patient body weight, in single or multiple administrations.
  • the compounds of the present invention may be administered to the patient in need of such treatment at a daily dosage ranging from about 1 mg to about 3500 mg per patient, preferably 10 mg to 1000 mg.
  • the daily dose per patient may be 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 120, 150, 160, 180, 200, 240, 250, 300, 350, 360, 400, 500, 600, 700, 800, 900 or 1000mg.
  • Administration can be one or more times daily, weekly (or several days apart), or on an intermittent schedule.
  • the compound may be administered on a weekly basis (eg, every Monday) one or more times per day, occasionally or for several weeks, eg, 4-10 weeks.
  • the compound may be administered daily for several days (e.g., 2-10 days), followed by several days (e.g., 1-30 days) without administration of the compound, and the cycle may be repeated indefinitely or a given number of times, e.g., 4-10 cycle.
  • a compound of the invention may be administered daily for 5 days, then 9 days off, then again daily for 5 days, then 9 days off, and so on, repeating the cycle indefinitely or a total of 4-10 times.
  • the compounds of the present invention can be prepared into any dosage form and administered to cancer patients in an appropriate administration method. Those skilled in the art can appropriately select the dosage of the product of the present invention according to the patient's symptoms and physical health.
  • the daily dose can be given as a single dose or divided into multiple doses. Dosage varies based on symptoms, age, etc.
  • the preferred dosage is, in the case of oral administration to adults, a dosage of 0.1 mg/kg (preferably 0.1 mg/kg) as the lower limit and a dosage of 20 mg/kg (preferably 10 mg/kg) as the upper limit.
  • the dose is 0.02 mg/kg (preferably 0.01 mg/kg) as the lower limit and 10 mg/kg (preferably 2 mg/kg) as the upper limit, and is administered 1 to 6 times a day depending on the symptoms. .
  • TTZ tumor necrosis factor-producing glioma U87 and U251 cultured in vitro
  • human lymphocytoma Mino human Burkitt's tumor Raji
  • human B lymphocyte tumor Ramos human multiple myeloma peripheral blood B lymphocyte tumor RPMI8226
  • human diffuse tissue lymphoma SU-DHL-2 human diffuse tissue lymphoma SU-DHL-2
  • (ABC type) diffuse lymphoma TMD-8 human diffuse large cell lymphoma cell SU-DHL-6 and other human tumor cells inhibit the proliferation.
  • TMZ Dissolve TMZ API (hereinafter referred to as TMZ) with DMSO and dilute it to 364.5 ⁇ M, 121.5 ⁇ M, 40.5 ⁇ M, 13.5 ⁇ M, 4.5 ⁇ M, 1.5 ⁇ M, 0.5 ⁇ M, 0.167 ⁇ M, 0.056 ⁇ M as drug stock solution, and store in the refrigerator Refrigerate at 4°C until ready to use.
  • TMZ Dissolve TMZ API
  • SU-DHL-2 Mino, Raji, Ramos, RPMI 8226, TMD-8, SU-DHL-6: Add an appropriate volume of DMSO and vortex to completely dissolve TMZ to prepare a stock solution with a concentration of 243mM, and dilute it to 486 ⁇ M and 162 ⁇ M. , 54.0 ⁇ M, 18.0 ⁇ M, 6.00 ⁇ M, 2.00 ⁇ M, 0.667 ⁇ M, 0.222 ⁇ M, 0 ⁇ M, stored in -90°C ⁇ -60°C refrigerator.
  • IC50 is calculated by the following formula
  • Min, Max and Slope represent the minimum value, maximum value and slope respectively.
  • Example 2 Inhibitory effect of TMZ on tumor growth in vivo of human diffuse tissue lymphoma SU-DHL-2 xenograft tumor model
  • Reagents RPMI-1640 powder (Gibco; Cat. No.: 31800-022), fetal bovine serum (FBS, Gibco; Cat. No.: 10099-141C), cyan-streptobis antibody (HyClone; Cat. No.: SV30010; Lot No.: J220018), PBS ( 0.01M, pH 7.4, Sangon Biotech; Cat. No.: B040100-0005), N-N-dimethylacetamide (aladdin; Cat. No.: D108095), sterile water for injection (Zhejiang Dubang Pharmaceutical).
  • mice Female NOD/SCID mice (number: 62; age: 6-7 weeks) were purchased from Viton Lever and raised in the SPF animal room of Suzhou Shengsu New Drug Development Co., Ltd. at a temperature of 20-25°C and a relative humidity of 40 % ⁇ 70%, with light and dark lighting for 12 hours each; animals can drink water and eat freely. After about 5 days of normal feeding, mice with good physical condition after veterinary examination can be selected for this experiment. Before grouping, use a marker to mark the base of the animal's tail. After grouping, each animal will be marked with an ear cutout.
  • Human diffuse tissue lymphoma cells SU-DHL-2 were obtained from Kebai Biotech (liquid nitrogen cryopreservation in our laboratory).
  • SU-DHL-2 cells were routinely cultured in RPMI-1640 culture medium containing 10% fetal bovine serum; they were passaged according to the cell growth conditions, with a passage ratio of 1:2 to 1:5.
  • Collect SU-DHL-2 cells in the logarithmic growth phase count the cells and resuspend them in serum-free RPMI-1640 culture medium. Adjust the cell concentration to 1.5 ⁇ 10 8 cells/mL; pipette the cells with a pipette to disperse them evenly. Put it into a 50-mL centrifuge tube, and place the centrifuge tube in an ice box; use a 1-mL syringe to draw the cell suspension, and inject it into the subcutaneous skin of the axilla of the front right limb of the mouse. Each animal is inoculated with 100 ⁇ L (1.5 ⁇ 10 7 cells/ only) to establish a SU-DHL-2 mouse xenograft tumor model.
  • the tumor diameter was measured using an electronic vernier caliper, and the data was input into an Excel spreadsheet to calculate the tumor volume.
  • the tumor volume reached 100-300mm 3
  • the day of grouping was regarded as the first day of the experiment (D1).
  • the tumor diameter was measured twice a week, the tumor volume was calculated, and the animal body weight was weighed and recorded.
  • TMZ Before each administration, weigh an appropriate amount of TMZ, add an appropriate volume of DMA to it, vortex to completely dissolve the compound, add an appropriate amount of sterile water for injection, vortex to mix the liquid evenly, and then add it to the dosage preparation.
  • the proportions of DMA and sterile water for injection were 10% and 90%, and the final concentrations of the dosage preparations were 1.0, 2.0 and 3.0 mg ⁇ mL -1 respectively.
  • the animal grouping and dosing schedule are shown in Table 5.
  • the test samples were administered starting on the day of grouping (D1), and the dosing period lasted for 3 weeks (the specific dosing time can be adjusted according to the progress of the experiment).
  • the dosage, route and time of administration of the above compounds can be adjusted in a timely manner according to the progress of the experiment.
  • Dosage regimen for drug efficacy experiments in mouse transplanted tumor model Note: “ig” means intragastric administration, “QD” means once a day, and “QW” means once a week.
  • TV initial is the tumor volume measured during group administration
  • TV t is the tumor volume measured at each time during the administration period.
  • RTV T represents the RTV of the treatment group
  • RTV C represents the RTV of the solvent control group.
  • TGI (%) 100% ⁇ [1–(TV t(T) –TV initial(T) )/(TV t(C) –TV initial(C) )]
  • TV t (T) represents the tumor volume measured each time in the treatment group
  • TV initial (T) represents the tumor volume in the treatment group when administered in groups
  • TV t (C) represents the tumor volume measured each time in the solvent control group
  • TV initial(C) represents the tumor volume of the solvent control group during group administration.
  • BW initial is the body weight of the animals during group administration
  • BW t is the body weight of the animals measured each time during the administration period.
  • Animal weight loss rate 100% ⁇ (BW initial - BW t )/BW initial
  • BW t represents the animal body weight measured each time during the administration period
  • BW initial represents the animal body weight during group administration.
  • IR (%) 100% ⁇ (W C - W T )/W C
  • WC represents the tumor weight in the solvent control group
  • WT represents the tumor weight in the treatment group
  • experimenters and veterinarians need to continuously observe the physical signs and health status of the experimental animals. Any abnormal performance of the animal, such as pain, depression, reduced activity, etc., must be recorded in the original experimental record. If the abnormal performance of experimental animals exceeds the provisions of IACUC-related animal welfare documents, the veterinarian can determine whether to terminate the experiment.
  • Table 6 Average tumor volume ⁇ SE (mm 3 ) and tumor growth inhibition rate (TGI) of animals in each group during the administration period Note: “*” indicates that there is a significant difference between the tumor volume and the solvent control group (P ⁇ 0.05), and “**” indicates that there is a highly significant difference between the tumor volume and the solvent control group (P ⁇ 0.01).
  • Group A was the solvent control group.
  • the animals were given blank solvent by gavage.
  • the tumors of the animals grew well and no spontaneous regression was found.
  • the average tumor volume reached 1163 ⁇ 133mm 3 .
  • Group B is the positive control drug doxorubicin group. Severe toxicity occurred at the dose of 7.5 mg ⁇ kg -1 QW. The animals' weight initially dropped severely and then all died. The tumor growth inhibition rate on D11 was 109% (P ⁇ 0.01), which has a significant inhibitory effect on tumor growth.
  • Group C is the TMZ low-dose group, with a dose of 10 mg ⁇ kg -1 QD.
  • the tumor growth inhibition rate on D22 was 114% (P ⁇ 0.05), showing an extremely excellent anti-tumor effect.
  • D11 the anti-tumor effect was already better than The safety of the positive control drug is significantly better than that of the positive control drug group.
  • Group D is the TMZ medium-dose group, with a dose of 20 mg ⁇ kg -1 QD. Although the tumor growth inhibition rate on D22 is the same as that of the low-dose group, both 114% (P ⁇ 0.05), it has a faster onset of action than the low-dose group. It showed an extremely excellent anti-tumor effect. By D8, the anti-tumor effect had reached that of the positive control drug. By D11, the anti-tumor effect had been better than that of the positive control drug, and its safety was significantly better than that of the positive control drug group.
  • Group E is a high-dose TMZ group, with a dose of 30 mg ⁇ kg -1 QD. Although the tumor growth inhibition rate was 116% on D22 (P ⁇ 0.05), the tumors completely disappeared, and the effect was faster than that of the low- and medium-dose groups. It showed extremely excellent anti-tumor effect, especially by D4, the anti-tumor effect was better than that of the positive control drug, and its safety was significantly better than that of the positive control drug group.
  • Example 3 Inhibitory effect of TMZ on tumor growth in vivo of human diffuse tissue lymphoma TMD-8 xenograft tumor model
  • cyclophosphamide was used as a control and the human diffuse large B lymphoma TMD-8 mouse transplant tumor model was used to evaluate the anti-tumor effect of TMZ.
  • Reagents RPMI-1640 powder (Gibco; Cat. No.: 31800-022); Fetal bovine serum FBS (Gibco; Cat. No.: 10099-141C); HyClone; Cat. No.: SV30010); PBS (Beyotime; Cat. No.: ST447 ); Sterile water for injection (Zhejiang Dubang Pharmaceutical); Normal saline (Guizhou Tiandi Pharmaceutical); N-N-dimethylacetamide (aladdin; Cat. No.: D108095).
  • mice Female CB17/SCID mice (number: 75; age: 6-8 weeks) were purchased from Viton Lever and raised in the SPF animal room of Suzhou Shengsu New Drug Development Co., Ltd. at a temperature of 20-25°C and a relative humidity of 40 % ⁇ 70%, with light and dark lighting for 12 hours each; animals can drink water and eat freely. After 7 days of normal feeding, mice with good physical condition after veterinary examination can be selected for this experiment. Before grouping, use a marker to mark the base of the animal's tail. After grouping, each animal will be marked with an ear cutout.
  • Human diffuse large B lymphoma TMD-8 cells were purchased from Shanghai Huzhen Industrial Co., Ltd. and were frozen in liquid nitrogen in our laboratory.
  • TMD-8 cells Under the conditions of 5% CO 2 , 37°C, and saturated humidity, TMD-8 cells were routinely cultured in RPMI-1640 culture medium containing 10% fetal bovine serum. According to the cell growth conditions, they were digested and passaged with 0.25% trypsin. Passage 1 to 2 times per week, with a passage ratio of 1:3 to 1:4.
  • TMD-8 cells in the logarithmic growth phase, count the cells, and resuspend them in a culture medium containing 30% serum-free RPMI-1640 and 70% Matrigel. Adjust the cell concentration to 5.0 ⁇ 10 7 cells/mL; place the cells on ice. In the box, use a 1-mL syringe to draw the cell suspension and inject it into the subcutaneous tissue of the axilla of the front right limb of the mouse. Each animal is inoculated with 200 ⁇ L (1.0 ⁇ 10 7 cells/mouse) to establish a TMD-8 transplanted tumor model. Observe the status of the animals regularly, use electronic vernier calipers to measure the tumor diameter, input the data into an Excel spreadsheet, calculate the tumor volume, and monitor tumor growth.
  • the day of grouping was regarded as the first day of the experiment (D1). After the start of the experiment, the tumor diameter was measured twice a week, the tumor volume was calculated, and the animal body weight was weighed and recorded.
  • DMA N-N-dimethylacetamide
  • sterile water for injection Take appropriate volumes of DMA (N-N-dimethylacetamide) and sterile water for injection respectively, mix them evenly, use them as blank solvents, and store them at room temperature for later use.
  • the proportions of DMA and sterile water for injection in the mixed solvent are 10% and 90% respectively.
  • the animal grouping and dosing schedule are shown in Table 8.
  • the first dose was started on the day of grouping (D1), and the dosing volume was 10 mL/kg.
  • Table 9 Average tumor volume ⁇ SE (mm 3 ) and tumor growth inhibition rate (TGI) of animals in each group during the administration period Note: “*” indicates that there is a significant difference between the tumor volume and the solvent control group (P ⁇ 0.05), and “**” indicates that there is a highly significant difference between the tumor volume and the solvent control group (P ⁇ 0.01).
  • mice The effects on the body weight of TMD-8 tumor-bearing mice are shown in Table 10.
  • TMZ at doses of 1.25, 2.5 and 5.0 mg/kg (ig, QD), the tumor volume inhibition rate TGI was greater than 60%, and p ⁇ 0.01, showing significant inhibition on the growth of transplanted tumors.
  • the effective dose is lower than 1.25 mg/kg (ig, QD); TMZ has a significant inhibitory effect on the growth of human diffuse large B lymphoma TMD-8 transplanted tumor at a dose of 1.25 mg ⁇ kg -1 (ig, QD). Better than CTX (25mg ⁇ kg-1,iv,QW) dose.
  • TMZ had no significant effect on the body weight of human diffuse large B lymphoma TMD-8 tumor-bearing mice at the dose set in this experiment, and no obvious drug-related abnormal reactions were observed during the administration;
  • TMZ (1.25mg/kg, i.g., QD) combined with CTX (25mg/kg, i.v., QW) has a weight loss rate of less than 5%, which also shows good safety.
  • Example 4 Inhibitory effect of TMZ and its preparations on tumor growth in vivo of human diffuse tissue lymphoma SU-DHL-6 xenograft tumor model
  • This experiment used cyclophosphamide as a control and used the human diffuse large B lymphoma SU-DHL-6 mouse transplant tumor model to evaluate the anti-tumor effects of TMZ and its preparations alone and in combination with cyclophosphamide.
  • Reagents RPMI-1640 powder (Gibco; Cat. No.: 31800-022); Fetal bovine serum FBS (Gibco; Cat. No.: 10099-141C); HyClone; Cat. No.: SV30010; Lot No.: J220018); PBS (Sangon) Biotech; Cat. No.: B040100-0005); Sterile water for injection (Zhejiang Dubang Pharmaceutical); Normal saline (Guizhou Tiandi Pharmaceutical); N-N-dimethylacetamide (aladdin).
  • mice Female NOD/SCID mice (number: 85; age: 6-8 weeks) were purchased from Viton Lever and kept in the SPF animal room of Suzhou Shengsu New Drug Development Co., Ltd. at a temperature of 20-25°C and a relative humidity of 40 % ⁇ 70%, with light and dark lighting for 12 hours each; animals can drink water and eat freely. After 7 days of normal feeding, mice with good physical condition after veterinary examination can be selected for this experiment. Before grouping, use a marker to mark the base of the animal's tail. After grouping, each animal will be marked with an ear cutout.
  • Human large cell lymphoma cells SU-DHL-6 were obtained from the Kunming Cell Bank of the Chinese Academy of Sciences (CAS(KM), cryopreserved in liquid nitrogen in our laboratory).
  • SU-DHL-6 cells Under the conditions of 5% CO 2 , 37°C, and saturated humidity, SU-DHL-6 cells were routinely cultured in RPMI-1640 culture medium containing 10% fetal bovine serum; according to the cell growth conditions, they were digested and passaged with 0.25% trypsin. , passaged 1 to 2 times a week, with a passage ratio of 1:3 to 1:4.
  • SU-DHL-6 cells Under the conditions of 5% CO 2 , 37°C, and saturated humidity, SU-DHL-6 cells were routinely cultured in RPMI-1640 culture medium containing 10% fetal bovine serum; according to the cell growth conditions, they were digested and passaged with 0.25% trypsin. , passaged 1 to 2 times a week, with a passage ratio of 1:3 to 1:4.
  • Collect SU-DHL-6 cells in the logarithmic growth phase count the cells and resuspend them in culture medium containing 50% serum-free RPMI-1640 and 50% Matrigel. Adjust the cell concentration to 3.0 ⁇ 10 7 cells/mL; place the cells In an ice box, use a 1-mL syringe to draw the cell suspension, and inject it into the mouse's front right armpit subcutaneously. Each animal is inoculated with 200 ⁇ L (6.0 ⁇ 10 6 cells/mouse) to establish a SU-DHL-6 transplanted tumor model. Observe the status of the animals regularly, use electronic vernier calipers to measure the tumor diameter, input the data into an Excel spreadsheet, calculate the tumor volume, and monitor tumor growth.
  • the day of grouping was regarded as the first day of the experiment (D1). After the start of the experiment, the tumor diameter was measured twice a week, the tumor volume was calculated, and the animal body weight was weighed and recorded.
  • TMZ capsule i.e., the aforementioned TMZ preparation, the same below
  • TMZ preparation the same below
  • take out the contents into a glass bottle add 75 ml of sterile water for injection, vortex, and ultrasonic for 20 to 30 minutes to prepare a mixture containing 0.5 mg/ml (API) dosage preparation; then add an appropriate volume of sterile water for injection, and dilute to obtain dosage preparations with final concentrations of 0.125, 0.25 and 0.5 mg/mL respectively, which are ready for use.
  • API 0.5 mg/ml
  • TMZ raw material drug delivery preparation cyclophosphamide (CTX) delivery preparation and blank vehicle are the same as in Example 3.
  • the animal grouping and dosing schedule are shown in Table 11.
  • the first dose was started on the day of grouping (D1), and the dosing volume was 10 mL/kg.
  • i.v. means intravenous injection
  • i.g. means intragastric administration
  • QD means once a day
  • QW means once a week.
  • Table 12 Average tumor volume ⁇ SE (mm 3 ) and tumor growth inhibition rate (TGI) of animals in each group during the administration period Note: “*” indicates that there is a significant difference between the tumor volume and the solvent control group (P ⁇ 0.05), and “**” indicates that there is a highly significant difference between the tumor volume and the solvent control group (P ⁇ 0.01).
  • Cyclophosphamide (CTX) at a dose of 25 mg/kg (iv, QW) has no obvious inhibitory effect on the growth of transplanted tumors, and the tumor volume inhibition rate TGI is less than 60%, while TMZ preparations at 1.25, 2.5 and 5.0 mg/kg ( ig, QD) dose, has a significant inhibitory effect on the growth of transplanted tumors, and the tumor volume inhibition rate TGI is greater than 60%, and p ⁇ 0.01;
  • TMZ preparation (1.25mg/kg, ig, QD) and CTX (25mg/ kg, iv, QW) combination therapy has a significant inhibitory effect on the growth of transplanted tumors, and the tumor inhibition rate in the combination group is significantly higher than that of TMZ preparation (1.25 mg/kg, ig, QD) and CTX (25 mg/kg, iv, QW) Used alone, it shows obvious synergistic and/or additive effects; TMZ has a significant inhibitory effect on the
  • TMZ raw material drug has a significant inhibitory effect on the growth of transplanted tumors at a dose of 2.5 mg/kg (ig, QD), which is equivalent to the TMZ preparation (2.5 mg/kg, ig, QD) alone group.
  • Example 5 Inhibitory effect of TMZ on tumor growth in human brain glioma cell U87 transplanted tumor model in vivo
  • Test purpose To test the inhibitory effect of TMZ on the in vivo tumor growth of human glioma cell U87 nude mouse transplanted tumor model.
  • Test instruments centrifuge, American Thermo Company, model ThermoSorvall ST 40; Hitachi 7600 fully automatic biochemical analyzer equipped with corresponding reagents; electronic balance Sartorius Company, model: BSA3202S-CW; electronic balance, METTLER-TOLEDO, model: LE204; small animals Weight balance, Shanghai Yueping Scientific Instrument Co., Ltd., model: YP1002; pipette: eppendorf; IVC mouse cage, product of Suzhou Teaching Cage Factory.
  • Test site and U87 cells Beijing Yongxin Kangtai Technology Development Co., Ltd.
  • U87 cells are human malignant glioma cells.
  • the normal cell morphology is epithelial and grows adherently.
  • the tumors were inoculated into nude mice and passaged twice. After the second passage, the tumors were removed and used for formal experiments.
  • Reagents RPMI-1640 culture medium, fetal bovine serum (FBS), cyan-streptomycin, PBS (0.01M, pH 7.4), Matrigel, sterile water for injection.
  • mice Take nude mice with a body weight of 16 ⁇ 2g and adaptively raise them in the experimental animal center for one week.
  • Select tumor-bearing mice with vigorous tumor growth and no ulceration, and sacrifice them by cervical dislocation.
  • the animals are disinfected with alcohol under sterile conditions on a clean bench. Skin, cut the skin, peel off the tumor. Cut the tumor tissue into about 1.5mm3 and inoculate it subcutaneously in the right armpit of the animal with a trocar.
  • Test drug It is a white powder. It is provided by Beijing Zimeng Pharmaceutical Co., Ltd. and the dosage is calculated. The dosage is 40 mg/kg, which is equivalent to the dosage of clinical patients. Store it dry and protected from light.
  • Dosing method Oral administration, once a day; Dosing cycle: 19 days; Solvent: Animal drinking pure water
  • the relative tumor volume (RTV) is calculated based on the measurement results.
  • the animal grouping and dosing schedule are shown in Table 14. The first dose will be started on the day of grouping (D1).
  • Table 15 Average tumor volume ⁇ SE (mm 3 ) and tumor growth inhibition rate (TGI) of animals in each group during the administration period Note: “*” indicates that there is a significant difference between the tumor volume and the solvent control group (P ⁇ 0.05), and “**” indicates that there is a highly significant difference between the tumor volume and the solvent control group (P ⁇ 0.01).
  • the compounds of the present invention have good anti-proliferative effects on glioma.
  • the body weight of the animals in the TMZ group continued to be low, once reduced by 7.9%, and compared with the blank control group, the weight of the animals once decreased by approximately 15%.
  • TMZ intracranial inoculation
  • the glioma animal model is not intracranial inoculation
  • TMZ has a significant advantage in its ability to penetrate the blood-brain barrier among anti-tumor drugs, its exposure in brain tissue is less than half of that in the circulation system.
  • TMZ is the only globally approved Indications, but the effect and safety of TMZ on DLBCL far exceed that of glioma.
  • primary central nervous system lymphoma also known as microglioma cells
  • TMZ's better blood-brain barrier penetration only use TMZ's better blood-brain barrier penetration.
  • the use of TMZ has not produced significant effects (M Kazuhiko, N Ryo et al. Randomized phase III study of high-dose methotrexate and whole-brain radiotherapy with/without temozolomide for newly diagnosed primary CNS lymphoma: JCOG1114C, Neuro-Oncology, 1– 12, 2022). Therefore, the present invention mainly provides the use of TMZ in the treatment and/or prevention of DLBCL other than diffuse large B-cell lymphoma (DLBCL) whose primary site or metastatic site is the central nervous system.
  • DLBCL diffuse large B-cell lymphoma
  • the imidazotetrazine compounds proposed by the present invention have excellent antagonistic effects and excellent safety on diffuse lymphoma whose primary site or metastasis site is not the central nervous system.

Landscapes

  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne une nouvelle utilisation d'un composé d'imidazotétrazine, ou un solvate de celui-ci, un hydrate de celui-ci, un polymorphe de celui-ci, un promédicament de celui-ci ou un sel pharmaceutiquement acceptable de celui-ci, et en particulier l'utilisation dans la préparation d'un médicament pour le traitement et/ou la prévention d'un lymphome diffus à grandes cellules B chez un sujet.
PCT/CN2023/106847 2022-07-11 2023-07-11 Utilisation d'un composé d'imidazotétrazine WO2024012464A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210809678 2022-07-11
CN202210809678.9 2022-07-11

Publications (1)

Publication Number Publication Date
WO2024012464A1 true WO2024012464A1 (fr) 2024-01-18

Family

ID=89535469

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/106847 WO2024012464A1 (fr) 2022-07-11 2023-07-11 Utilisation d'un composé d'imidazotétrazine

Country Status (1)

Country Link
WO (1) WO2024012464A1 (fr)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114469949A (zh) * 2020-10-27 2022-05-13 正大天晴药业集团股份有限公司 用于治疗弥漫大b细胞淋巴瘤的喹啉衍生物

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114469949A (zh) * 2020-10-27 2022-05-13 正大天晴药业集团股份有限公司 用于治疗弥漫大b细胞淋巴瘤的喹啉衍生物

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
HUANG, YAN ET AL.: "Advances in the Treatment of Relapsed/Refractory Primary Central Nervous System Lymphoma", WORLD LATEST MEDICINE INFORMATION, CHINA, vol. 18, no. 66, 31 December 2018 (2018-12-31), China, pages 29 - 30, XP009553172, ISSN: 1671-3141, DOI: 10.19613/j.cnki.1671-3141.2018.66.015 *
RENI, M. MASON, W. ZAJA, F. PERRY, J. FRANCESCHI, E. BERNARDI, D. DELL'ORO, S. STELITANO, C. CANDELA, M. ABBADES: "Salvage chemotherapy with temozolomide in primary CNS lymphomas: preliminary results of a phase II trial", EUROPEAN JOURNAL OF CANCER, ELSEVIER, 34TH EORTC-NCI-AACR SYMPOSIUM ON MOLECULAR TARGETS AND CANCER THERAPEUTICS, BARCELONA, SPAIN, vol. 40, no. 11, 1 July 2004 (2004-07-01), pages 1682 - 1688, XP004519919, ISSN: 0959-8049, DOI: 10.1016/j.ejca.2004.03.008 *
VIOLETTA V. LESHCHENKO ET AL.: "Integrative Genomic Analysis of Temozolomide Resistance in Diffuse Large B-Cell Lymphoma.", CLINICAL CANCER RESEARCH, vol. 20, no. 2, 15 January 2014 (2014-01-15), XP055197747, DOI: 10.1158/1078-0432.CCR-13-0669 *
WANG, SHUAI: "Clinical Effects of Temozolomide in the Treatment of Recurrent Primary Central Nervous System Lymphoma", JOURNAL OF NANJING MEDICAL UNIVERSITY (NATURAL SCIENCES), vol. 39, no. 4, 30 April 2019 (2019-04-30), CN , pages 563 - 565, XP009552637, ISSN: 1007-4368, DOI: 10.7655/NYDXBNS20190418 *

Similar Documents

Publication Publication Date Title
EP3600270B1 (fr) Composés et compositions pour le traitement de troubles hématologiques
CN111225911B (zh) 用于治疗血液病的化合物和组合物
US20230159559A1 (en) Compound for inhibiting and inducing degradation of egfr kinase
US10080756B2 (en) Combination methods for treating cancers
RU2762893C2 (ru) Применение ингибитора ezh2 в комбинации с ингибитором btk в получении лекарственного средства для лечения опухоли
JP2023502742A (ja) Alk2阻害剤及びjak2阻害剤を含む併用療法
CA3030967C (fr) Combinaison d'un inhibiteur de bcl-2 et d'un inhibiteur de mcl-1, utilisations et compositions pharmaceutiques associees
JP6462582B2 (ja) がんの治療のための方法および組成物
WO2024012464A1 (fr) Utilisation d'un composé d'imidazotétrazine
JP7156287B2 (ja) Axl阻害剤を有効成分として含むがん治療剤
WO2022228363A1 (fr) Utilisation médicale d'une composition
US20220331330A1 (en) Combination therapies for the treatment of cancer
BR112020024481A2 (pt) formulações/composições compreendendo ibrutinib
KR20200055117A (ko) Pi3 키나아제 억제제 및 세포독성 항암제를 포함하는 암의 예방 또는 치료용 약학적 조성물
WO2024088392A1 (fr) Combinaison pharmaceutique pour le traitement d'une tumeur, composition pharmaceutique et son utilisation
WO2021197334A1 (fr) Association pharmaceutique et son utilisation
CN115300511A (zh) 一种组合物的医药用途
TW202241415A (zh) 通路調節劑、含其的醫藥組成物、其用途和採用其的治療方法
JP2022532186A (ja) 非晶質のpi3k阻害剤及びこれを含有する医薬組成物
CN117835974A (zh) 使用bcl-2抑制剂治疗b细胞恶性肿瘤的方法

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: 23838955

Country of ref document: EP

Kind code of ref document: A1