CN113116895A - Quinoline derivatives for the treatment of neuroblastoma - Google Patents

Quinoline derivatives for the treatment of neuroblastoma Download PDF

Info

Publication number
CN113116895A
CN113116895A CN202011629017.5A CN202011629017A CN113116895A CN 113116895 A CN113116895 A CN 113116895A CN 202011629017 A CN202011629017 A CN 202011629017A CN 113116895 A CN113116895 A CN 113116895A
Authority
CN
China
Prior art keywords
neuroblastoma
pharmaceutically acceptable
formula
compound
temozolomide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN202011629017.5A
Other languages
Chinese (zh)
Other versions
CN113116895B (en
Inventor
苏雨栋
赵强
李龙
闫杰
李敏
陈鹏
王阳阳
尹洁
郑鉴
刘邓
徐萍
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Chia Tai Tianqing Pharmaceutical Group Co Ltd
Tianjin Medical University Cancer Institute and Hospital
Original Assignee
Chia Tai Tianqing Pharmaceutical Group Co Ltd
Tianjin Medical University Cancer Institute and Hospital
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 Chia Tai Tianqing Pharmaceutical Group Co Ltd, Tianjin Medical University Cancer Institute and Hospital filed Critical Chia Tai Tianqing Pharmaceutical Group Co Ltd
Priority to CN202011629017.5A priority Critical patent/CN113116895B/en
Publication of CN113116895A publication Critical patent/CN113116895A/en
Application granted granted Critical
Publication of CN113116895B publication Critical patent/CN113116895B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • 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
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis

Abstract

The invention relates to application of a quinoline derivative in treating neuroblastoma, belonging to the field of medicines. In particular, the present invention relates to quinoline derivatives for use in the treatment of neuroblastoma. Its advantages are: the nilotinib can obviously inhibit the growth, migration and invasion of an NB cell line, induce the cell cycle arrest of the NB cell line and promote the apoptosis of the NB cell line.

Description

Quinoline derivatives for the treatment of neuroblastoma
Technical Field
The invention belongs to the field of medicines, and particularly relates to application of a quinoline derivative in treating neuroblastoma.
Background
Neuroblastoma (neuroblastoma) is an embryonic tumor that originates from primitive neural crest cells and is the most common extracranial solid tumor in children. Accounts for 8-10% of all children malignant tumors, and the annual incidence rate is 0.3-5.5/10 ten thousands. Neuroblastoma is derived from undifferentiated sympathetic ganglion cells, so that tumors can occur in any area where embryonic sympathetic ganglion cells are present. The suprarenal gland is the most common primary site, followed by the abdominal sympathetic ganglion, thoracic sympathetic ganglion, cervical sympathetic ganglion, and pelvic sympathetic ganglion, with about 1% of patients failing to find primary tumors. Neuroblastoma can metastasize to lymph nodes, bone marrow, bone, dura mater, orbit, liver and skin, and in rare cases also to the lung and cranium.
The prognosis of neuroblastoma is closely related to the age of diagnosis, clinical stage, type of tumor pathology, copy number of N-myc gene and DNA index. The American child tumor tissue (COG) divides neuroblastoma into low, medium and high risk groups according to the five indexes. The treatment of the neuroblastoma of children is difficult, the single treatment prognosis is poor, and the treatment of the neuroblastoma at present is a multidisciplinary combined treatment mainly comprising operations, chemoradiotherapy and autologous stem cell transplantation. With the gradual improvement of diagnosis and treatment technologies, the survival rate of the neuroblastoma of children in China also improves year by year, but OS is still lower than that of Europe and America, and a novel and efficient treatment method is urgently needed clinically.
Summary of The Invention
In a first aspect, the present application provides the use of a pharmaceutical composition comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier for the manufacture of a medicament for the treatment of neuroblastoma. The present application also provides a method of treating neuroblastoma comprising administering to a patient in need thereof a pharmaceutical composition comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier. The present application also provides the use of a pharmaceutical composition comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier for the treatment of neuroblastoma.
In a second part, the present application provides a combination comprising (I) a compound of formula I, or a pharmaceutically acceptable salt thereof; and (ii) at least one second therapeutic agent.
In a third aspect, the present application also provides the use of a combination for the manufacture of a medicament for the treatment of neuroblastoma. The application also provides the use of the combination for treating neuroblastoma.
In a fourth aspect, the present application also provides a method of treating neuroblastoma comprising administering to a patient in need thereof the combination of the present application. The combination includes (I) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) at least one second therapeutic agent.
Disclosure of Invention
In a first aspect, the present application provides the use of a pharmaceutical composition comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier for the manufacture of a medicament for the treatment of neuroblastoma.
Figure BDA0002878117640000021
In the present application, the neuroblastoma includes, but is not limited to, low-risk, medium-risk, and high-risk neuroblastoma. In some embodiments, the neuroblastoma is a high risk neuroblastoma.
In some embodiments, the neuroblastoma includes, but is not limited to, an undifferentiated, differentiated, poorly differentiated form of neuroblastoma. In some embodiments, the neuroblastoma includes, but is not limited to, an extracranial neuroblastoma.
In some embodiments, the neuroblastoma includes, but is not limited to, the following histopathological types: NB type (schwannian stroma-poor type), GNB promiscuous type (intermixed), GN maturity type (mating), GNB nodal type (nodularia).
In some embodiments, the neuroblastoma is a locally advanced, advanced and/or metastatic neuroblastoma. In some embodiments, the neuroblastoma is a metastatic neuroblastoma. In some embodiments, the neuroblastoma is a relapsed and/or refractory neuroblastoma. In some embodiments, the neuroblastoma is a recurrent neuroblastoma; in some embodiments, the neuroblastoma is a refractory neuroblastoma. In some embodiments, the neuroblastoma includes, but is not limited to, an ALK gene mutation, a PHOX2B gene mutation, a deletion at position 1p36 or 11q 14-23, a TP53-R337H mutation, a CDKN1C mutation, an HRAS mutation, and/or a MYCN gene amplified neuroblastoma. In some embodiments, the neuroblastoma is a 11q normal and differentiated neuroblastoma, or a 11q abnormal or undifferentiated neuroblastoma.
In the present application, the patient suffering from neuroblastoma is a patient under the age of 21 years; in some embodiments, the neuroblastoma patient is a patient under the age of 18. In some embodiments, the neuroblastoma patient has previously received a hematopoietic stem cell transplant. In some embodiments, the subject having neuroblastoma has previously received an autologous hematopoietic stem cell transplant.
In one embodiment of the present application, the neuroblastoma patient has previously received first line therapy. In a specific embodiment of the present application, the neuroblastoma is a relapsed and/or refractory neuroblastoma that fails first line therapy. In some embodiments, the neuroblastoma patient has previously undergone surgical resection, chemotherapy, radiation therapy, targeted therapy, immunotherapy, and/or hematopoietic stem cell transplantation. In some embodiments, the neuroblastoma is a neuroblastoma that fails a chemotherapeutic drug, a targeted drug, and/or an immunotherapy. In a specific embodiment of the present application, the neuroblastoma patient has previously received chemotherapy. In a specific embodiment of the present application, the neuroblastoma is a neuroblastoma that fails chemotherapy.
In some specific embodiments, the neuroblastoma is a neuroblastoma that fails treatment with an anthracycline; in some specific embodiments, the neuroblastoma is a neuroblastoma that has failed a chemotherapeutic regimen comprising an anthracycline. In some specific embodiments, the neuroblastoma is a neuroblastoma that has failed treatment with one or more of a platinum-based drug, etoposide, vincristine, doxorubicin, cyclophosphamide, ifosfamide, topotecan, temozolomide, irinotecan, doxorubicin, mitoxantrone, methotrexate, cytarabine, procarbazine, gemcitabine, lenalidomide, dituximab (dinutuximab), dinutuximab beta, 13-cis-retinoic acid, granulocyte-macrophage colony stimulating factor GM-CSF, interleukin 2, an ALK inhibitor, 131I-MIBG.
In some embodiments, the pharmaceutical composition comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier further comprises at least one second therapeutic agent.
The second therapeutic drug comprises one or more of chemotherapy drugs, targeted drugs, immunotherapy, 13-cis-retinoic acid, granulocyte-macrophage colony stimulating factor GM-CSF, interleukin 2 and 131I-MIBG.
In some embodiments, the second therapeutic agent is selected from chemotherapy and immunotherapy. In some embodiments, the second therapeutic agent is selected from an anti-tumor antibody agent and an immunotherapeutic agent. In some embodiments, the second therapeutic agent comprises one or more of a topoisomerase inhibitor, a plant alkaloid, an antibiotic antineoplastic agent, an alkylating agent, an ALK inhibitor, an antineoplastic antibody drug, an immunotherapeutic drug.
In some embodiments, the second therapeutic agent is selected from one or more of a platinum-based agent, etoposide, an anthracycline, a vinblastine, granulocyte-macrophage colony stimulating factor GM-CSF, interleukin 2, 131I-MIBG, 13-cis-retinoic acid, mesna, cyclophosphamide, ifosfamide, dituximab, dinutuximab beta, busulfan, melphalan, and topotecan.
In some embodiments, the second therapeutic agent is selected from one or more of the group consisting of:
one or more of cyclophosphamide, doxorubicin, dexaximab, dinutuximab beta, vincristine, busulfan, melphalan, carboplatin, cisplatin, etoposide, 13-cis-retinoic acid, interleukin 2, GM-CSF, and temozolomide;
platinum drugs (e.g., carboplatin, cisplatin) and etoposide;
platinum drugs (e.g., carboplatin, cisplatin), etoposide, and melphalan;
vincristine + doxorubicin + cyclophosphamide + mesna;
vincristine + cyclophosphamide + mesna;
topotecan;
topotecan + etoposide;
topotecan + vincristine + doxorubicin;
topotecan + temozolomide;
topotecan + cyclophosphamide;
temozolomide;
temozolomide + irinotecan;
irinotecan;
carboplatin + irinotecan and/or temozolomide;
busulfan + melphalan;
temozolomide + irinotecan + dexuximab;
an ALK inhibitor;
131I-MIBG;
anti-GD-2 (anti-ganglioside 2) antibody; and
anti-GD-2 antibody + isotretinoin;
anti-GD-2 antibody + interleukin-2;
anti-tumor antibody drugs + isotretinoin;
anti-tumor antibody drugs + interleukin 2.
In a preferred embodiment, the second therapeutic agent is selected from the group consisting of irinotecan and temozolomide.
In a specific embodiment, the second therapeutic agent is selected from irinotecan and temozolomide for a treatment cycle every 3 weeks.
In some embodiments, wherein the compound of formula I or a pharmaceutically acceptable salt thereof is present at 4-10mg/m2The daily dosage of (a); in some embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is present at 5-8mg/m2The daily dosage of (a); in some embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is present at 5-7mg/m2The daily dosage of (a);
in some specific embodiments, the second therapeutic agent is irinotecan and temozolomide, wherein the compound of formula I or a pharmaceutically acceptable salt thereof is present at 7mg/m2The daily dose of (a), administered on days 1-14; irinotecan with the concentration of 50mg/m2The dose of (1), administered on days 1-5; temozolomide 100mg/m2The dose of (4), administered on days 1-5.
In a second aspect, the present application provides a combination comprising (I) a compound of formula I, or a pharmaceutically acceptable salt thereof; and (ii) at least one second therapeutic agent.
In some embodiments, there is provided a combination for treating neuroblastoma, comprising: (i) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) one or more of a chemotherapeutic agent, a targeted agent, an immunotherapy, 13-cis-retinoic acid, granulocyte-macrophage colony stimulating factor GM-CSF, interleukin 2, 131I-MIBG, and optionally in combination with radiation therapy.
In some embodiments, there is provided a combination for treating neuroblastoma, comprising: (i) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) one or more chemotherapeutic agents, optionally in combination with radiation therapy. In some embodiments, it further comprises (iii) one or more of 13-cis-retinoic acid, granulocyte-macrophage colony-stimulating factor GM-CSF, interleukin 2, 131I-MIBG; in some embodiments, the chemotherapeutic agent is selected from one or more of the group consisting of platinum-based agents, temozolomide, etoposide, anthracyclines, vinblastine, camptothecin and analogues thereof, cyclophosphamide, ifosfamide, doxorubicin, topotecan, busulfan, melphalan. In some embodiments, the chemotherapeutic agent is selected from irinotecan and temozolomide; in some embodiments, every 3 weeks is a treatment cycle wherein the compound of formula I or a pharmaceutically acceptable salt thereof is at 4-10mg/m2The daily dosage of (a); in some embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is present at 5-8mg/m2The daily dosage of (a); in some embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is present at 5-7mg/m2The daily dosage of (a); in some particular embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is present at 7mg/m2The daily dose of (a), administered on days 1-14; irinotecan with the concentration of 50mg/m2The dose of (1), administered on days 1-5; temozolomide 100mg/m2The dose of (4), administered on days 1-5.
In some embodiments, there is provided a combination for treating neuroblastoma, comprising: (i) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) at least one small molecule targeted antineoplastic agent, and optionally in combination with radiation therapy; in some embodiments, it further comprises (iii) one or more of 13-cis-retinoic acid, granulocyte-macrophage colony-stimulating factor GM-CSF, interleukin 2, 131I-MIBG; in some embodiments, the small molecule targeted anti-tumor drug is an ALK inhibitor.
In some embodiments, there is provided a combination for treating neuroblastoma, comprising: (i) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) at least one anti-tumor antibody drug, optionally in combination with radiation therapy. In some embodiments, it further comprises (iii) one or more of 13-cis-retinoic acid, granulocyte-macrophage colony-stimulating factor GM-CSF, interleukin 2, and 131I-MIBG. In some embodiments, the anti-tumor antibody drug is selected from the group consisting of an anti-GD-2 antibody; in some embodiments, the anti-tumor antibody drug is selected from the group consisting of dexuximab, dinutuximab beta.
In some embodiments, there is provided a combination for treating neuroblastoma, comprising: (i) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) at least one immunotherapeutic drug, optionally in combination with radiation therapy. In some embodiments, it further comprises (iii) one or more of 13-cis-retinoic acid, granulocyte-macrophage colony-stimulating factor GM-CSF, interleukin 2, and 131I-MIBG.
In some embodiments, there is provided a combination for treating neuroblastoma, comprising: (i) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) an anti-tumor antibody drug and at least one chemotherapeutic drug, optionally in combination with radiation therapy. In some embodiments, it further comprises (iii) one or more of 13-cis-retinoic acid, granulocyte-macrophage colony stimulating factor GM-CSF, interleukin 2, and 131I-MIBG.
In some embodiments, there is provided a combination for treating neuroblastoma, comprising: (i) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) camptothecin and analogues thereof and/or temozolomide, optionally in combination with radiotherapy; in some embodiments, it further comprises (iii) one or more of 13-cis-retinoic acid, granulocyte-macrophage colony-stimulating factor GM-CSF, interleukin 2, and 131I-MIBG.
In some embodiments, there is provided a combination for treating neuroblastoma, comprising: (i) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) vinblastine and/or temozolomide, optionally in combination with radiotherapy; in some embodiments, it further comprises (iii) one or more of 13-cis-retinoic acid, granulocyte-macrophage colony-stimulating factor GM-CSF, interleukin 2, and 131I-MIBG.
In some embodiments, there is provided a combination for treating neuroblastoma, comprising: (i) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) a platinum and/or etoposide, optionally in combination with radiation therapy. In some embodiments, it further comprises (iii) one or more of 13-cis-retinoic acid, granulocyte-macrophage colony-stimulating factor GM-CSF, interleukin 2, and 131I-MIBG.
In some embodiments, there is provided a combination for treating neuroblastoma, comprising: (i) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) one or more of cyclophosphamide, vincristine, doxorubicine, etoposide, and optionally in combination with radiation therapy. In some embodiments, it further comprises (iii) one or more of 13-cis-retinoic acid, granulocyte-macrophage colony stimulating factor GM-CSF, interleukin 2, and 131I-MIBG.
In some embodiments, there is provided a combination for treating neuroblastoma, comprising: (i) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) lenalidomide, optionally in combination with radiation therapy. In some embodiments, there is provided a combination for treating neuroblastoma, comprising: (i) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) one or both of gemcitabine and vinorelbine, optionally in combination with radiation therapy.
In some embodiments, the second therapeutic agent is selected from one or more of the group consisting of:
one or more of cyclophosphamide, doxorubicin, dexaximab, dinutuximab beta, vincristine, busulfan, melphalan, carboplatin, cisplatin, etoposide, 13-cis-retinoic acid, interleukin 2, GM-CSF, and temozolomide;
platinum drugs (e.g., carboplatin, cisplatin) and etoposide;
platinum drugs (e.g., carboplatin, cisplatin), etoposide, and melphalan;
vincristine + doxorubicin + cyclophosphamide + mesna;
vincristine + cyclophosphamide + mesna;
topotecan;
topotecan + etoposide;
topotecan + vincristine + doxorubicin;
topotecan + temozolomide;
topotecan + cyclophosphamide;
temozolomide;
temozolomide + irinotecan;
irinotecan;
carboplatin + irinotecan and/or temozolomide;
busulfan + melphalan;
temozolomide + irinotecan + dexuximab;
an ALK inhibitor;
131I-MIBG;
anti-GD-2 (anti-ganglioside 2) antibody; and
anti-GD-2 antibody + isotretinoin;
anti-GD-2 antibody + interleukin-2;
anti-tumor antibody drugs + isotretinoin;
anti-tumor antibody drugs + interleukin 2.
In a third aspect, the present application also provides the use of a combination for the manufacture of a medicament for the treatment of neuroblastoma.
In a fourth aspect, the present application also provides a method of treating neuroblastoma comprising administering to a patient in need thereof the combination of the present application. The combination comprises (I) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) at least one second therapeutic agent.
The present application provides a method for treating a patient having neuroblastoma. In some embodiments of the present application, the patient has previously received chemotherapy and/or targeted drugs and/or immunotherapeutic drugs and/or radiation therapy. In some embodiments, the patient is treated with chemotherapy and/or targeted drugs and/or immunotherapeutic drugs and/or radiation before complete remission of disease progression occurs. In some embodiments, the patient has failed to complete remission or partial remission following chemotherapy and/or targeted and/or immunotherapeutic drugs and/or radiation therapy.
The present application provides a method of treating neuroblastoma comprising administering to a patient in need thereof a compound of formula I or a pharmaceutically acceptable salt thereof and at least one second therapeutic agent. In some embodiments, the present application provides a method of treating neuroblastoma in a patient in need of treatment, comprising administering to the patient a compound of formula I, or a pharmaceutically acceptable salt thereof, and at least one second therapeutic agent. In some embodiments, the present application provides a method of treating neuroblastoma in a patient in need thereof, comprising administering to the patient a compound of formula I, or a pharmaceutically acceptable salt thereof, and at least one second therapeutic agent. In some embodiments, the present application provides a method of treating neuroblastoma in a first line therapy failure, comprising administering to a patient in need thereof a compound of formula I, or a pharmaceutically acceptable salt thereof, and at least one second therapeutic agent. In one embodiment, the present application provides a method of treating refractory relapsed neuroblastoma, comprising administering to a patient in need thereof a compound of formula I, or a pharmaceutically acceptable salt thereof, and at least one second therapeutic agent.
In some versions of the present application, the patient has not previously received systemic treatment.
The method of administration can be determined comprehensively on the basis of the activity, toxicity of the drug, tolerance of the patient, and the like. In some embodiments of the present application, the use or method of treatment, including but not limited to, the second therapeutic agent may be administered once, twice, three times, or four times daily (qd), every other day (qod), every 3 days (q3d), every 4 days (q4d), every 5 days (q5d), weekly (q1w), every 2 weeks (q2w), every 3 weeks (q3w), or every 4 weeks (q4w), e.g., twice daily (bid), twice weekly (biw), three times daily (tid), four times daily (qid), etc. In some embodiments of the present application, the second therapeutic agent may also be administered in an intermittent manner for use or method of treatment. The intermittent administration includes a dosing period and a rest period, for example, a second therapeutic agent is administered daily during the dosing period, then the administration is stopped for a period of time during the rest period, followed by the dosing period, then the rest period, and so on, which may be repeated multiple times.
In some embodiments of the present application, the use or method of treatment, including but not limited to the compound of formula I or a pharmaceutically acceptable salt thereof, may be administered at a dose of 8-12mg for 2 weeks on a dosing schedule of 1 week off; and/or, in a dosing regimen of 2 weeks on continuous dosing, and 2 weeks off.
In some embodiments, a compound of formula I, or a pharmaceutically acceptable salt thereof, is administered on days 1-14 of each cycle, with one cycle of administration every 21 days.
In some embodiments, the second therapeutic agent and the compound of formula I or a pharmaceutically acceptable salt thereof each have the same or different treatment cycles. In some specific embodiments, the second therapeutic agent and the compound of formula I or a pharmaceutically acceptable salt thereof have the same treatment cycle, e.g., one treatment cycle every 1 week, every 2 weeks, every 3 weeks, or every 4 weeks. In some specific embodiments, the second therapeutic agent and the compound of formula I or a pharmaceutically acceptable salt thereof are each one treatment cycle every 3 weeks.
Optionally, the second therapeutic agent can be used in combination with chemotherapeutic adjuvants including, but not limited to, leucovorin (CF), aldehydo, mesna, bisphosphonates, amifostine, hematopoietic Colony Stimulating Factors (CSFs), cyclosporine.
In addition, the present application provides a kit for treating neuroblastoma, comprising a compound of formula I or a pharmaceutically acceptable salt thereof and at least one second therapeutic agent, each packaged separately, together with instructions for treating neuroblastoma.
Neuroblastoma
In some embodiments of the present application, the neuroblastoma, its clinical stage, includes, but is not limited to, a locally advanced, and/or advanced (e.g., stage III/IV) and/or metastatic neuroblastoma. In some embodiments, the neuroblastoma is a relapsed and/or refractory neuroblastoma. In some embodiments, the neuroblastoma is recurrent; in certain embodiments, the neuroblastoma is refractory.
In the present application, the neuroblastoma includes, but is not limited to, low-risk, medium-risk, and high-risk neuroblastoma. In some embodiments, the neuroblastoma is a high risk neuroblastoma.
In some embodiments, the neuroblastoma includes, but is not limited to, an undifferentiated, differentiated, poorly differentiated form of neuroblastoma. In some embodiments, the neuroblastoma includes, but is not limited to, an extracranial neuroblastoma.
In some embodiments, the neuroblastoma includes, but is not limited to, the following histopathological types: NB type (schwannian stroma-poor type), GNB promiscuous type (intermixed), GN maturity type (mating), GNB nodal type (nodularia).
In some embodiments, the neuroblastoma includes, but is not limited to, an ALK gene mutation, a PHOX2B gene mutation, a deletion at position 1p36 or 11q 14-23, a TP53-R337H mutation, a CDKN1C mutation, an HRAS mutation, and/or a MYCN gene amplified neuroblastoma. In some embodiments, the neuroblastoma is a 11q normal and differentiated neuroblastoma, or a 11q abnormal or undifferentiated neuroblastoma.
In the present application, the patient suffering from neuroblastoma is a patient under the age of 21 years; in some embodiments, the neuroblastoma patient is a patient under the age of 18.
In some embodiments, the neuroblastoma is a neuroblastoma that was previously surgically excised. In some embodiments, the neuroblastoma patient has previously received a hematopoietic stem cell transplant. In some embodiments, the neuroblastoma is a neuroblastoma that was previously transplanted with autologous hematopoietic stem cells.
In some embodiments, the neuroblastoma is a neuroblastoma that fails radiation therapy. In some embodiments, the neuroblastoma is a neuroblastoma that fails a first line therapy. In some embodiments, the neuroblastoma is a neuroblastoma that fails treatment with a chemotherapeutic and/or targeted drug. In some embodiments, the neuroblastoma is a neuroblastoma that had previously received at least two chemotherapy regimens. In some embodiments, the neuroblastoma is a neuroblastoma that has failed treatment with surgery and/or radiation and/or chemotherapy drugs. In some specific embodiments, the neuroblastoma is a neuroblastoma that has failed treatment with one or more of a platinum-based drug (e.g., carboplatin, cisplatin), an anthracycline antineoplastic, etoposide, vincristine, doxorubicin, cyclophosphamide, ifosfamide, topotecan, temozolomide, irinotecan, dobestatin, mitoxantrone, methotrexate, cytarabine, procarbazine, gemcitabine, lenalidomide, dituximab, 13-cis-retinoic acid, granulocyte-macrophage colony stimulating factor GM-CSF, interleukin 2, an ALK inhibitor, 131I-MIBG, and ibrutinib.
A second therapeutic agent
In the present application, the second therapeutic agent includes, but is not limited to, one or more of chemotherapeutic drugs, targeted drugs, immunotherapy, 13-cis-retinoic acid, granulocyte-macrophage colony stimulating factor GM-CSF, interleukin 2, and 131I-MIBG. The targeted drugs comprise small molecule targeted antitumor drugs and antitumor antibody drugs.
The second therapeutic agent described herein includes, but is not limited to, one or more of a chemotherapeutic agent, a small molecule targeted antineoplastic agent, an immunotherapeutic agent, an anti-tumor antibody agent.
In some embodiments, the second therapeutic agent is selected from chemotherapy and immunotherapy. In some embodiments, the second therapeutic agent is selected from an anti-tumor antibody agent and an immunotherapeutic agent. In some embodiments, the second therapeutic agent comprises one or more of a topoisomerase inhibitor, an ALK inhibitor, a plant alkaloid, an antibiotic antineoplastic agent, an alkylating agent, an antineoplastic antibody drug, and an immunotherapeutic drug.
In some embodiments, the second therapeutic agent is selected from one or more of the group consisting of platinum-based drugs, etoposide, anthracyclines, vinblastine, camptothecin and analogs thereof, granulocyte-macrophage colony stimulating factor GM-CSF, interleukin 2, 131I-MIBG, 13-cis-retinoic acid, mesna, cyclophosphamide, ifosfamide, dituximab, dinutuximab beta, busulfan, melphalan, topotecan.
In some embodiments, the second therapeutic agent is an anthracycline including, but not limited to, one or more of epirubicin, doxorubicin, daunorubicin, pirarubicin, amrubicin, idarubicin, mitoxantrone, adriamycin, valrubicin, zorubicin, pixantrone, and doxorubicin pyrans.
In some embodiments, the second therapeutic agent is a platinum-based agent including, but not limited to, one or more of cisplatin, carboplatin, nedaplatin, oxaliplatin, miriplatin, triplatin tetranitrate, phenanthroline, picoplatin, satraplatin, and leplatin.
In some embodiments, the second therapeutic agent is a fluoropyrimidine derivative including, but not limited to, one or more of gemcitabine, capecitabine, ancitabine, fluorouracil, bifuran fluorouracil, doxifluridine, tegafur, carmofur, trifluridine.
In some embodiments, the second therapeutic agent is a camptothecin and analogs thereof; in some embodiments, the second therapeutic agent is a camptothecin and analogs thereof and an anti-tumor antibody drug; the camptothecin and the analogues thereof include but are not limited to one or more of camptothecin, hydroxycamptothecin, 9-aminocamptothecin, 7-ethylcamptothecin, irinotecan and topotecan.
In some embodiments, the second therapeutic agent is selected from one or more of the group consisting of:
one or more of cyclophosphamide, doxorubicin, dexaximab, dinutuximab beta, vincristine, busulfan, melphalan, carboplatin, cisplatin, etoposide, 13-cis-retinoic acid, interleukin 2, GM-CSF, and temozolomide;
platinum drugs (e.g., carboplatin, cisplatin) and etoposide;
platinum drugs (e.g., carboplatin, cisplatin), etoposide, and melphalan;
vincristine + doxorubicin + cyclophosphamide + mesna;
vincristine + cyclophosphamide + mesna;
topotecan;
topotecan + etoposide;
topotecan + vincristine + doxorubicin;
topotecan + temozolomide;
topotecan + cyclophosphamide;
temozolomide;
temozolomide + irinotecan;
irinotecan;
carboplatin + irinotecan and/or temozolomide;
busulfan + melphalan;
temozolomide + irinotecan + dinotefuran;
an ALK inhibitor;
131I-MIBG;
anti-GD-2 (anti-ganglioside 2) antibody; and
anti-GD-2 antibody + isotretinoin;
anti-GD-2 antibody + interleukin-2;
anti-tumor antibody drugs + isotretinoin;
anti-tumor antibody drugs + interleukin 2.
Chemotherapy medicine
In the present application, the chemotherapeutic agents include, but are not limited to, one or more of alkylating agents (e.g., platinum-based drugs, busulfan, melphalan, cyclophosphamide, ifosfamide, procarbazine, dacarbazine, temozolomide, etc.), plant alkaloids (e.g., vinblastine, taxanes, podophyllotoxin (e.g., etoposide, teniposide), camptothecin and analogs thereof), antibiotic antineoplastics (e.g., anthracyclines, mitomycin, bleomycin, etc.), antimetabolites (e.g., methotrexate, 5-fluorouracil, cytarabine, capecitabine, gemcitabine, fludarabine, nelarabine, etc.), topoisomerase inhibitors (e.g., irinotecan, topotecan, etoposide, etc.).
Examples of the chemotherapeutic drugs described in the present application include, but are not limited to, platinum drugs (e.g., oxaliplatin, miriplatin, cisplatin, carboplatin, nedaplatin, dicycloplatin, ledaplatin, Lobaplatin, carboplatin, picoplatin), fluoropyrimidine derivatives (e.g., gemcitabine, capecitabine, antacitabine, fluorouracil, difurofluorouracil, doxifluridine, tegafur, carmofur, trifluridine), taxanes (e.g., paclitaxel, albumin-bound paclitaxel, and docetaxel), camptothecins and analogs thereof (e.g., camptothecin, hydroxycamptothecin, 9-aminocamptothecin, 7-ethylcamptothecin, irinotecan, topotecan), vinblastines (vinorelbine, vincristine, vindesine, vinflunine, and the like, Anthracyclines (epirubicin, doxorubicin (adriamycin), daunorubicin, pirarubicin, amrubicin, idarubicin, mitoxantrone, aclarubicin, valrubicin, zorubicin, pixantrone, doxorubicin pyrans), cytarabine, thioguanine, pemetrexed, carmustine, melphalan, etoposide, teniposide, mitomycin, ifosfamide, cyclophosphamide, azacitidine, methotrexate, bendamustine, pentalimutin, liposomal doxorubicin, actinomycin D (dactinomycin), bleomycin, pingyangmycin, temozolomide, dacarbazine, pellomycin, eribulin, plinabulin (plinabulin), Sapacitabine, troosulfan (treosulfan), 153Sm-EDTMP, tijiao, levoasparaginase, pemetrexed, cephalotaxine, procarbazine, ARC-100, and encequidar.
In certain embodiments, the chemotherapeutic agent is one or more of the platinum group including, but not limited to, cisplatin, carboplatin, nedaplatin, oxaliplatin, miriplatin, triplatin tetranitrate, phenanthroline, picoplatin, satraplatin, leplatin, dicycloplatin, and the like.
Immunotherapy medicine
In the present application, the immunotherapeutic drugs include, but are not limited to, one or more of interferons (interferon α, interferon α -1b, interferon α -2b), interleukins, sirolimus (temsirolimus), everolimus (everolimus), ridaforolimus (ridaforolimus), temsirolimus, and lenalidomide.
Small molecule targeted antitumor drug
In the application, the small molecule targeted antitumor drug includes but is not limited to a protein kinase regulator. The acting target of the small molecule targeted antitumor drug comprises but is not limited to Fascin-1 protein, HDAC (histone deacetylase), Proteasome, CD38, SLAMF7(CS1/CD319/CRACC), Proteasome, RANKL, EGFR (epidermal growth factor receptor), Anaplastic Lymphoma (ALK), MET gene, ROS1 gene, HER2 gene, RET gene, BRAF gene, PI3K signal path, DDR2 (discoid death receptor 2) gene, FGFR1 (fibroblast growth factor receptor 1), NTRK1 (neurotrophic tyrosine kinase type 1 receptor) gene and KRAS gene; the target of the small molecule targeted antitumor drug also comprises COX-2 (cyclooxygenase-2) and APE1 (apurinic/apyrimidinic endonuclease)Enzymes), VEGFR (vascular endothelial growth factor receptor), CXCR-4 (chemokine receptor-4), MMP (matrix metalloproteinase), IGF-1R (insulin-like growth factor receptor), Ezrin, PEDF (pigment epithelium derived factor), AS, ES, OPG (osteoprotegerin), Src, IFN, ALCAM (leukocyte activation adhesion factor), HSP, JIP1, GSK-3 (glycogen synthesis kinase 3 sugar), CyclinD1 (cell cycle regulatory protein), CDK4/6(cyclin-dependent kinase), TIMP1 (tissue metalloproteinase inhibitor), THBS3, PTHR1 (parathyroid hormone-related protein receptor 1), TEM7 (human tumor vascular endothelial marker 7), COPS3, cathepsin K, T cell surface antigen (CD4), Aurora a kinase, fusion protein, PNP, cyclic depsipeptide, DHFR.
In some embodiments, the small molecule targeted anti-tumor drug is a protein kinase inhibitor. Wherein, the protein kinase inhibitor includes but is not limited to tyrosine kinase inhibitor, serine and/or threonine kinase inhibitor, Poly ADP Ribose Polymerase (PARP) inhibitor.
In some embodiments, the small molecule targeted antineoplastic agents include, but are not limited to, imatinib, sunitinib, Nilotinib (Nilotinib), bosutinib (bosutinib), saratinib (Saracatinib), pazopanib, trabectedidin (trabecteddin), regorafenib, Cediranib (Cediranib), bortezomib, Panobinostat (Panobinostat), Carfilzomib (Carfilzomib), ixazomi (ixazoib), apatinib, erlotinib, afatinib, crizotinib, Ceritinib (ceinib), Vemurafenib (veinaftifib), delafenib (dabrafafenib), cabozatinib (cabozatinib), gefitinib (gefitinib), darutinib (Dactinib), erlotinib (Dasatinib), albertinib (albertinib), erlotinib (Nilotinib), erlotinib (Osteinib), erlotinib (Nilotinib), erlotinib (lanotinib), erlotinib (valtinib), erlotinib (valcanitinib), erlotinib (valtinib), erlotinib (Lorentinib (Loniceb), erlotinib (Lorentinib (Lorentib), erlotinib (Lapatitinib), and Evotini, Sermititinib (Selumetinib), Sorafenib (Sorafenib), tematinib (Olmutinib), Vofolitinib (Savoltinib), Fujitinib (Fruquintinib), Entretinib (Entretinib), Dasatinib (Dasatinib), Ensartinib (Ensartinib), Lenvatinib (Lenvatinib), itacetitinib, pyrrolitinib (Pyropyib), Bimetinib (Binimetinib), Erdamitinib (Erdasatinib), Acxitinib (Axitinib), Neratinib (Neratinib), Cobimitinib (Cobimetinib), Alcalitinib (Acalbutinib), Famitinib (Famitatinib), Maratinib (Masitinib), Initubib (Initinib), Lancinib (Cobimetinib), Doxilitinib (XO-803, Blafenib-1205, BLemnib-PC, L-1205, Lvcitabine, Lb-PC, Lpiniboborinib, Lvi-PC-D, Lpiniboborinib, Lb, Lvi-IIb, Lb, Lvi-IIb, Lb, Ln-Lb, Ln-Lb, L, seliciclib, OSE-2101, APL-101, berzosertib, idelalisib, lerociclib, ceralasertib, PLB-1003, tomivosertib, AST-2818, SKLB-1028, D-0316, LY-3023414, allitinib, MRTX-849, AP-32788, AZD-4205, lifrafenib, vactor, mivebresib, napabustin, stravatinib, TAS-114, molibresib, CC-223, ricerandib, CK-101, LXTH-254, simotinib, GSK-3368715, TAS-0728, magatinib, tepotiib, HS-10296, D-4547, meresteinib, pterostimulil, SAF-4927, ASK-3627, AZS-369, AZ-53, SANTIANtictinib-0735, AST-077, AST-3, AST-3727, AST-3, AST-3, SALTAIC-369, SANTIATIB, SAID, SANTRITIIB-1, SANTRITIIB, SAID-1, SANTI, SAID-3695, SAID-377, SAID-379, SAID-379, SAID, epitinib succinate, tesevatinib, SPH-1188-11, BPI-15000, copanlisib, niraparib, olaparib, veliparib, talazoparib tosylate, DV-281, Siremaddin, Telaglenastat, MP-0250, GLG-801, ABTL-0812, bortezomib, tucidinostat, vorinostat, reminiostat, epadostat, tazemetostat, entinostat, mocetinostat, quisinostat, LCL-161, KML-001, Bellistat, Pralatrexate, Ropidexin (Roepsin), Sedamide (Chidamide), Alweisib, Piletisindin, Piletisindine, or a plurality of them.
In some embodiments, the small molecule targeted anti-tumor drug is an ALK kinase inhibitor, examples of which include, but are not limited to, one or more of crizotinib, ceritinib, bugatinib, loratinib, aletinib (Alectinib), buitinib, and enzatinib.
In some embodiments, the small molecule targeted anti-tumor drug is one or more of Bomaniciclib, vennoclad, fenretinide, ABTL-0812, OTS-167, SF-1126, nifurtimox, eflornithine, ABTL-0812, CBL-0137, ONC-201, fadraccib, LY-3295668erbumine, rose bengal sodium salt (rose bengal sodium), vennoclad, copanlisib, tamibatene, Lauretonib, ensantinib, and entrecitinib.
Antitumor antibody medicine
In the present application, the target of the anti-tumor antibody drug includes, but is not limited to, any one or more of PD-1, PD-L1, cytotoxic T lymphocyte antigen 4 (cytoxic T-lymphocyte antigen 4, CTLA-4), platelet-derived growth factor receptor alpha (PDGFR- α), Vascular Endothelial Growth Factor (VEGF), human epidermal growth factor receptor-2 (HER2), Epidermal Growth Factor Receptor (EGFR), ganglioside GD2, B cell surface protein CD20, B cell surface protein CD52, B cell surface protein CD38, B cell surface protein CD319, B cell surface protein CD30, B cell surface protein CD19, CD3, CD 276.
In some embodiments, the anti-tumor antibody drugs described herein are inhibitors of the CD20(Cluster of differentiation 20) antigen; in some embodiments, the anti-tumor antibody drug described herein is an MDM2 inhibitor; in some embodiments, the anti-tumor antibody drug described herein is a GD2 inhibitor; in some embodiments, the antibody drug is an inhibitor of the interaction between the PD-1 receptor and its ligand PD-L1; in some embodiments, the antibody agent is a cytotoxic T-lymphocyte antigen 4 (CTLA-4) inhibitor. In some embodiments, the antibody drug is a platelet-derived growth factor receptor alpha (PDGFR-alpha) inhibitor. In some embodiments, the anti-PD-1 or PD-L1 antibody is an anti-PD-1 or PD-L1 monoclonal antibody.
In some embodiments, the anti-PD-1 antibody can be selected from any one or more of Nivolumab, palboceprizumab (Pembrolizumab), debarvilleuzumab (Durvalumab), teriepril mab (torelizumab, JS-001), Cendilizumab (IBI308), Carrelizumab (Camrelizumab), tirelin mab (BGB-A317), Jennuzumab (GB226), Lilizumab (LZM009), HLX-10, BAT-1306, AK103(HX008), AK104 (Combo organism), CS1003, SCT-I10A, F520, SG001, GLS-010.
In some embodiments, the anti-PD-L1 antibody may be selected from any one or more of Atezolizumab, Avelumab, KL-A167, SHR-1316, BGB-333, JS003, STI-A1014(ZKAB0011), KN035, MSB2311, HLX-20, CS-1001, SGN-35, Polatuzumab, Tafasitamab, Brentuximab.
In some embodiments, the anti-CTLA-4 antibody may be selected from any one or more of Ipilimumab (Iplimumab), Tremelimumab (Tremelimumab), AGEN-1884, BMS-986249, BMS-986218, AK-104, IBI 310.
In some embodiments, the platelet-derived growth factor receptor alpha (PDGFR-alpha) inhibitor is an anti-PDGFR alpha antibody. In some embodiments, the anti-PDGFR α antibody is an anti-PDGFR α monoclonal antibody. In some specific embodiments, the anti-PDGFR α antibody is Olaratumab (olarataumab).
In some specific embodiments, the anti-tumor antibody drug includes, but is not limited to, any one or more of nafitamab, omburtamab, OGD-201, APN-301, racotumomab, dextuximab, dinutuximab beta, enoblituzumab, the CD3 antagonist, nivatoramab.
A compound of formula I or a pharmaceutically acceptable salt thereof
The compound of formula I has the chemical name 1- [ [ [4- (4-fluoro-2-methyl-1H-indol-5-yl) oxy-6-methoxyquinolin-7-yl ] oxy ] methyl ] cyclopropylamine, having the structural formula:
Figure BDA0002878117640000151
in the application, all related to the nilotinib refer to the compound of the formula I.
The compound of formula I may be administered in its free base form, or in the form of its salts, hydrates, and prodrugs, which convert in vivo to the free base form of the compound of formula I. For example, pharmaceutically acceptable salts of compounds of formula I are within the scope of the present application and can be generated from various organic and inorganic acids according to methods well known in the art.
In some embodiments, the compound of formula I is administered as the hydrochloride salt. In some embodiments, the compound of formula I is administered as the monohydrochloride salt of the compound of formula I. In some embodiments, the compound of formula I is administered as the dihydrochloride salt. In some embodiments, the compound of formula I is administered as a crystalline form of the hydrochloride salt. In a particular embodiment, the compound of formula I is administered in the form of a crystal of the dihydrochloride salt.
The compound of formula I, or a pharmaceutically acceptable salt thereof, the second therapeutic agent, may be administered by a variety of routes including, but not limited to, a route selected from: oral, parenteral, intraperitoneal, intravenous, intraarterial, transdermal, sublingual, intramuscular, rectal, transbuccal, intranasal, inhalation, vaginal, intraocular, topical, subcutaneous, intralipid, intraarticular, intraperitoneal, and intrathecal. In a particular embodiment, administration is by oral administration.
The amount of the compound of formula I or a pharmaceutically acceptable salt thereof, and the second therapeutic agent administered can be determined based on the severity of the disease, the response to the disease, any treatment-related toxicities, the age and health of the patient.
In some embodiments, the compound of formula I, or a pharmaceutically acceptable salt thereof, is administered in a daily dose of 3 mg to 30 mg. In some embodiments, the compound of formula I, or a pharmaceutically acceptable salt thereof, is administered in a daily dose of 5 mg to 20 mg. In some embodiments, the daily dose of a compound of formula I, or a pharmaceutically acceptable salt thereof, administered is from 8mg to 16 mg. In some embodiments, the compound of formula I, or a pharmaceutically acceptable salt thereof, is administered in a daily dose of 10mg to 14 mg. In a particular embodiment, the compound of formula I or a pharmaceutically acceptable salt thereof is administered in a daily dose of 8 mg. In a particular embodiment, a compound of formula I or a pharmaceutically acceptable salt thereof is administered in a daily dose of 10 mg. In a particular embodiment, a compound of formula I or a pharmaceutically acceptable salt thereof is administered in a daily dose of 12 mg. In this application, for example, with respect to tablets or capsules, "12 mg of a compound of formula I on a unit dose basis" means that each tablet or capsule ultimately produced contains 12mg of a compound of formula I.
In some embodiments, the amount of a compound of formula I, or a pharmaceutically acceptable salt thereof, administered may also be calculated based on the body surface area of the patient. In some embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is present at 4-10mg/m2The daily dosage of (a); it will be understood by those skilled in the art that if the patient has a body surface area of 1m2If so, the amount of the compound of formula I or a pharmaceutically acceptable salt thereof administered is 4-10 mg. In some embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is present at 5-8mg/m2The daily dosage of (a); in some embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is present at 5-7mg/m2The daily dosage of (a); in some embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is present at 4, 5, 6, 7, 8mg/m2The daily dose of (a).
The compound of formula I or a pharmaceutically acceptable salt thereof, the second therapeutic agent may be administered one or more times per day. In some embodiments, the compound of formula I, or a pharmaceutically acceptable salt thereof, is administered once daily. In one embodiment, the administration is once daily in an oral solid formulation.
In the above-mentioned therapeutic methods, the administration method can be comprehensively determined depending on the activity and toxicity of the drug, the tolerance of the patient, and the like. Preferably, the compound of formula I or a pharmaceutically acceptable salt thereof is administered in an intermittent manner. The intermittent administration includes a dosing period during which the compound of formula I, or a pharmaceutically acceptable salt thereof, may be administered one or more times per day and a rest period. For example, a compound of formula I or a pharmaceutically acceptable salt thereof is administered daily for a dosing period, then the administration is stopped for a period of time during a rest period, followed by a dosing period, then a rest period, and so on, which may be repeated multiple times. Wherein the ratio of the administration period to the withdrawal period in days is 2: 0.5-5, preferably 2: 0.5-3, more preferably 2: 0.5-2, and still more preferably 2: 0.5-1.
In some embodiments, the administration is discontinued for 2 weeks. In some embodiments, administration is 1 time per day for 14 days followed by 14 days off; followed by 1 administration per day for 14 days and then 14 days, so that the administration may be repeated several times at 2-week intervals.
In some embodiments, the administration is discontinued for 1 week for 2 weeks. In some embodiments, administration is 1 time per day for 14 days followed by 7 days of discontinuation; the administration is followed 1 time per day for 14 days and then discontinued for 7 days, so that the administration is repeated multiple times at 1 week intervals for 2 weeks of continuous administration.
In some embodiments, the administration is continued for 5 days and discontinued for 2 days. In some embodiments, administration is 1 time per day for 5 days, followed by 2 days off; the administration is followed 1 time per day for 5 days and then discontinued for 2 days, such that the administration is repeated multiple times at intervals of 5 consecutive days with 2 days discontinued.
In certain particular embodiments, the administration is oral at a dose of 12mg once daily for 2 weeks with 1 week rest.
Combined medicine
Each component of the combination described herein may optionally be used in combination with one or more pharmaceutically acceptable carriers, wherein the components may each independently, or some or all of them together contain a pharmaceutically acceptable carrier and/or excipient. The combination agents described herein may be formulated separately from each other or some or all of them may be co-formulated. Preferably, the components of the combination are formulated separately or each in a suitable pharmaceutical composition. In some embodiments, the combination of the present application may be formulated as a pharmaceutical composition suitable for single or multiple administration. In some particular embodiments, the pharmaceutical composition containing a compound of formula I or a pharmaceutically acceptable salt thereof may be selected from solid pharmaceutical compositions including, but not limited to, tablets or capsules.
The components of the combination of the present application may be administered separately from each other or some or all of them may be co-administered. The components of the combination as claimed may be administered substantially simultaneously, or some or all of them may be administered substantially simultaneously.
The components of the combination of the present application may be administered independently of each other, or some or all of them together in a suitable route, including, but not limited to, orally or parenterally (by intravenous, intramuscular, topical or subcutaneous routes). In some embodiments, the components of the combination of the present application may be administered orally or by injection, such as intravenously or intraperitoneally, independently of each other, or in part or all together.
The components of the combination of the present application may each independently, or some or all of them, be prepared in suitable dosage forms, including, but not limited to, tablets, troches, pills, capsules (e.g. hard capsules, soft capsules, enteric capsules, microcapsules), elixirs, granules, syrups, injections (intramuscular, intravenous, intraperitoneal), granules, emulsions, suspensions, solutions, dispersions and dosage forms for sustained release formulations for oral or non-oral administration.
In some embodiments of the present application, the combination is a fixed combination. In some embodiments, the fixed combination is in the form of a solid pharmaceutical composition or a liquid pharmaceutical composition.
The present invention provides a kit comprising (a) at least one unit dose of a pharmaceutical composition of a compound of formula I, or a pharmaceutically acceptable salt thereof, and (b) instructions for treating neuroblastoma.
The invention also provides a kit comprising (a) at least one unit dose of a formulation suitable for oral administration of a compound of formula I or a pharmaceutically acceptable salt thereof and (b) instructions for use in the treatment of neuroblastoma in a spaced-apart administration. In certain embodiments, a kit is provided comprising (a) at least one unit dose of a tablet or capsule of a compound of formula I, or a pharmaceutically acceptable salt thereof, and (b) instructions for use in the treatment of neuroblastoma in a spaced-apart administration. In some more typical embodiments, the neuroblastoma is a neuroblastoma that fails treatment with a chemotherapeutic and/or targeted drug. The term "unit dose" refers to a pharmaceutical composition packaged in a single package for ease of administration. Such as tablets or capsules.
In some embodiments of the present application, the combination is a non-fixed combination. In some embodiments, the second therapeutic agent and the compound of formula I, or a pharmaceutically acceptable salt thereof, in the non-fixed combination are each in the form of a pharmaceutical composition.
In some embodiments of the present application, the compound of formula I, or a pharmaceutically acceptable salt thereof, is administered simultaneously or sequentially with one or more second therapeutic agents. In certain embodiments, the one or more second therapeutic agents are administered to the patient prior to administration of the compound of formula I or a pharmaceutically acceptable salt thereof. In certain embodiments, the one or more second therapeutic agents are administered to the patient after administration of the compound of formula I or a pharmaceutically acceptable salt thereof. In some embodiments, the compound of formula I, or a pharmaceutically acceptable salt thereof, and one or more second therapeutic agents are administered to the patient sequentially.
In some embodiments, there is also provided a kit of combination for treating neuroblastoma, comprising: (a) a pharmaceutical composition containing a compound of formula I or a pharmaceutically acceptable salt thereof as an active ingredient; and (b) a pharmaceutical composition containing a chemotherapeutic drug as an active ingredient. In some embodiments, there is also provided a kit of combination for treating neuroblastoma, comprising: (a) a pharmaceutical composition containing a compound of formula I or a pharmaceutically acceptable salt thereof as an active ingredient; (ii) a And (b) a pharmaceutical composition containing a small molecule targeted antitumor drug as an active ingredient. In some embodiments, there is also provided a kit of pharmaceutical compositions for treating neuroblastoma, comprising: (a) a pharmaceutical composition containing a compound of formula I or a pharmaceutically acceptable salt thereof as an active ingredient; and (b) a pharmaceutical composition containing an immunotherapeutic agent as an active ingredient. In some embodiments, there is also provided a kit of parts for a pharmaceutical combination for treating neuroblastoma, comprising (a) a pharmaceutical composition comprising a compound of formula I or a pharmaceutically acceptable salt thereof as an active ingredient; and (b) a pharmaceutical composition containing the antibody drug as an active ingredient.
In some embodiments, there is also provided a kit of combination for treating neuroblastoma, comprising: (a) a first pharmaceutical composition comprising as an active ingredient a compound of formula I or a pharmaceutically acceptable salt thereof; and (b) a second pharmaceutical composition comprising an alkylating agent and/or camptothecin and analogues thereof as active ingredients; in some embodiments, a kit of pharmaceutical compositions for treating neuroblastoma is also provided, comprising (a) a first pharmaceutical composition comprising as an active ingredient a compound of formula I or a pharmaceutically acceptable salt thereof; and (b) a second pharmaceutical composition comprising temozolomide and/or camptothecin and analogues thereof as active ingredients; in some embodiments, a kit of pharmaceutical compositions for treating neuroblastoma is also provided, comprising (a) a first pharmaceutical composition comprising as an active ingredient a compound of formula I or a pharmaceutically acceptable salt thereof; and (b) a second pharmaceutical composition containing temozolomide and/or irinotecan as an active ingredient.
Herein, unless otherwise indicated, the dosages and ranges provided herein are based on the molecular weight of the compound of formula I in its free base form.
Unless otherwise indicated, the following terms used in the specification and claims shall have the following meanings for the purposes of this application.
In the application, all related to the nilotinib refer to the compound of the formula I.
By "patient" is meant a mammal, preferably a human.
By "pharmaceutically acceptable" is meant that it is used to prepare pharmaceutical compositions that are generally safe, non-toxic, and neither biologically nor otherwise undesirable, and include that they are acceptable for human pharmaceutical use.
"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, and the like; or with organic acids such as acetic, trifluoroacetic, propionic, hexanoic, heptanoic, cyclopentanepropionic, glycolic, pyruvic, lactic, malonic, succinic, malic, maleic, fumaric, tartaric, citric, benzoic, cinnamic, mandelic, methanesulfonic, ethanesulfonic, 1, 2-ethanedisulfonic, 2-hydroxyethanesulfonic, benzenesulfonic, p-chlorobenzenesulfonic, p-toluenesulfonic, 3-phenylpropionic, trimethylacetic, t-butylacetic, dodecylsulfuric, gluconic, glutamic, hydroxynaphthoic, salicylic, stearic acid and the like.
By "treatment" is meant any administration of a therapeutically effective amount of a compound and includes:
(1) inhibiting the disease (i.e., arresting the further development of the pathology and/or symptomatology) in a human experiencing or exhibiting the pathology or symptomatology of the disease, or
(2) Ameliorating the disease (i.e., reversing the pathology and/or symptomatology) in a human experiencing or exhibiting the pathology or symptomatology of the disease.
"treatment failure" refers to the condition where toxic side effects are not tolerated after dose adjustment and disease progression occurs for a short period of time during treatment. Wherein intolerance includes, but is not limited to, hematologic toxicity that remains at grade IV (platelet lowering grade III and above) and non-hematologic toxicity that remains at grade III or above after dose adjustment.
"CR" means complete remission. "PR" refers to partial remission. "PD" refers to disease progression. "SD" refers to stable disease.
"about" in this application means within + -5% of the specified numerical range given, preferably within + -2% and more preferably within + -1%.
In this document, unless otherwise indicated, the terms "comprises, comprising and including" or equivalents thereof, are open-ended and mean that elements, components and steps other than those listed are included.
As used herein, "First-line treatment" or "First-line therapy" are used interchangeably to refer to the First, or First, treatment recommended for a disease; for cancer, there are generally a number of alternative treatment options for first line therapy.
In the application, the nilotinib can obviously inhibit the growth, migration and invasion of neuroblastoma cell lines 9464D, 975A2, IMR-32 and SK-N-BE (2), induce the cell cycle block of the neuroblastoma cell lines, and promote the neuroblastoma cell lines to die. Obviously inhibit the growth of the mouse model tumor of neuroblastoma, and has the advantages of dose dependence of curative effect, high dose and good safety. Can also obviously activate the immune microenvironment of the neuroblastoma, improve the infiltration percentage of CD4 and CD 8T cells in the tumor and spleen, reduce the number of MDSC, enhance the INF-gamma and TNF-alpha secretion capacity of CD4 and CD 8T cells and improve the MHC-I expression of neuroblastoma tumor cells, namely the killing capacity of CD4 and CD 8T cells and the antigen presentation function of the tumor cells are enhanced. The erlotinib remarkably up-regulates immune activation related genes such as T cell activation and antigen presentation, and remarkably down-regulates angiogenesis related genes.
All patents, patent applications, and other established publications are herein expressly incorporated by reference for the purpose of description and disclosure. These publications are provided solely for their disclosure prior to the filing date of the present application. All statements as to the date of these documents or representation as to the contents of these documents is based on the information available to the applicant and does not constitute any admission as to the correctness of the dates of these documents or the contents of these documents. Moreover, any reference to such publications in this specification does not constitute an admission that the publications form part of the common general knowledge in the art in any country.
Drawings
FIG. 1 inhibition of neuroblastoma cell line proliferation by Arotinib
FIG. 2 inhibitory Effect of Arotinib on clonal formation of neuroblastoma cell line
FIG. 3 inhibition of neuroblastoma cell cycle by Arotinib
FIG. 4 promotion of neuroblastoma cell line apoptosis by Arotinib
FIG. 5 inhibition of neuroblastoma cell line migration by Arotinib
FIG. 6 inhibition of neuroblastoma cell line invasion by Arotinib
FIG. 7A inhibitory Effect of Arotinib on mouse neuroblastoma
FIG. 7B inhibitory Effect of Arotinib on mouse neuroblastoma
FIG. 8A activation of the mouse neuroblastoma immune microenvironment by Arotinib
FIG. 8B activation of the neuroblastoma immune microenvironment in mice by Arotinib
Detailed Description
The present application is further illustrated with reference to specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present application.
Example 1
1.1 Material cell lines 9464D and 975A2 were given by Dr.Rimas Orientas (Seattle Children's Research Institute), both cell lines were derived from N-myc expanded C57BL/6 mice, neuroblastoma spontaneously growing in the adrenal gland, the mouse tumor model mimicking the pathogenesis of neuroblastoma, and the mice were normal in immune function, largely mimicking the true pathogenic background of the patient. Human neuroblastoma cell lines IMR-32 and SK-N-BE (2) were purchased from American Type Culture Collection (ATCC). C57BL/6 mice were purchased from Beijing Sibefu Biotechnology, Inc.
1.2 reagents MEM (Medium), DMEM (Medium) and R1640 medium were purchased from Gibco, USA. 0.25%, 0.05% trypsin was purchased from Gibco, USA. Fetal bovine serum was purchased from israel BI. The CCK8 kit, the cell cycle kit and the apoptosis kit are all purchased from Beijing Solebao Biotechnology GmbH. Transwell cells (24-well cell, 8 μm pore size) and Mtrigel matrigel were purchased from Corning, USA. Fluorescently labeled antibodies were purchased from BioLegend, usa, primary fluorescent test antibodies from Abcam, uk, and secondary antibodies from CST, usa.
1.3 Instrument incubator (Thermo corporation, USA), flow cytometer (BD corporation, USA), inverted fluorescence microscope (Olympus corporation, Japan), upright fluorescence microscope (Nikon corporation, Japan), enzyme-linked plate reader (Thermo corporation, USA), high-speed refrigerated centrifuge (Eppendorf corporation, Germany), cryomicrotome (Leica corporation, Switzerland), refrigerator (Haier corporation, China), 80 ℃ cryorefrigerator (Haier corporation, China), liquid nitrogen tank (Thermo corporation, USA), clean bench (ESCO corporation, New Gangpo).
1.4 Experimental methods
1.4.1 cell culture cell lines 9464D and 975A2 were cultured in DMEM medium containing 10% fetal bovine serum, 100u/ml penicillin and 100. mu.g/ml streptomycin. The cell lines IMR-32 and SK-N-BE (2) were cultured in DMEM medium and R1640 medium containing 10% fetal bovine serum, 100u/ml penicillin and 100. mu.g/ml streptomycin, respectively. All cultured in a constant temperature incubator at 37 ℃ and 5% CO 2. When the cell fusion degree is about 80-90%, 9464D is digested with 0.25% pancreatin for 2 minutes, 975A2 is digested with 0.05% pancreatin for 2 minutes, IMR-32 and SK-N-BE (2) are digested with 0.05% pancreatin for 30 seconds, then the cells are collected in a 15ml centrifuge tube, centrifuged at 4 ℃ and 1000r/min for 5 minutes, and subcultured.
1.4.2 cell proliferation assay (CCK8 assay) 9464D, 975A2, IMR-32 and SK-N-BE (2) cells were collected in the logarithmic growth phase, counted, added to 96-well plates at 3500/100. mu.L/well, cultured for 24 hours to allow the cells to stick to the walls, then treated with different concentrations of anrotinib (0.25, 0.5, 1,2, 4, 8, 16, 32, 64. mu. mol/L) for 24 hours, after which 10. mu.L of CCK8 solution was added to each well, the plates were incubated in an incubator for 1-4 hours, the absorbance value at 450nm was measured with an enzyme marker, and the cell inhibition rate was calculated.
1.4.3 plate cloning experiments the harvested 9464D, 975A2, IMR-32 and SK-N-BE (2) cell pellets were resuspended and counted in the corresponding medium, and each cell was seeded in 6-well plates with a 50, 100, 200 cell density gradient per well. Cells were treated with different concentrations of apratinib (0, 1,2 μmol/L) and incubated in a constant temperature incubator for about 2 weeks. The medium was discarded, washed twice with PBS phosphate buffered saline, and 1ml of 4% paraformaldehyde was added to each well to fix the cells for 15 minutes. The paraformaldehyde is discarded, the membrane is washed twice with PBS, and 0.1% crystal violet staining solution is added into each hole to stain for 15 minutes. The staining solution was discarded, washed twice with PBS, air dried naturally and photographed. The number of colony formation was visually counted, and the colony formation rate was calculated.
1.4.4 cell cycle assay 9464D, 975A2, IMR-32 and SK-N-BE (2) cells were treated with different concentrations of apratinib (0, 1,2, 4. mu. mol/L) for 24 hours, respectively, harvested, centrifuged at 1000r/min for 5min and washed twice with PBS. Cell fixation was performed with 70% ethanol for more than 2 hours or overnight. After this time, the sample was centrifuged at 1400r/min for 5min, ethanol was discarded, PBS was washed twice, and 10. mu.L of 2% PBS-Triron X100 and 0.4. mu.L of ki67-APC antibody were added to each tube and incubated at 4 ℃ for 30 min. After a single PBS wash, 250. mu.L of propidium iodide (RNase-containing) was added and a water bath was kept at 37 ℃ in the dark for 30 minutes. The cells can then be stored at 4 ℃ in the dark. Detection was performed using a flow cytometer within 24 hours. Flowjo software was used for analysis to calculate the cell cycle phase ratio. The experiment was repeated three times.
1.4.5 apoptosis assay 9464D, 975A2, IMR-32 and SK-N-BE (2) cells were treated with different concentrations of apratinib (0, 1,2, 4. mu. mol/L) for 24 hours, respectively, harvested, centrifuged at 1000r/min for 5min and washed twice with PBS. Discarding the supernatant, adding 250. mu.L of 1XAnnexinV Binding Buffer working solution to resuspend the cells, adding 2.5. mu.L of Annexin V-APC and 2.5. mu.L of PI staining solution to the cell suspension, and incubating for 15-20 min at room temperature in the dark. Immediately after the reaction was completed, detection was performed using a flow cytometer. And analyzing by using Flowjo software, and calculating the proportion of the apoptotic cells. The experiment was repeated three times.
1.4.6 cell Scoring assay the harvested 9464D, 975A2, IMR-32 and SK-N-BE (2) cell pellets were resuspended in the appropriate medium and counted, 6X 10 cells per well in 6-well plates5And culturing the cells in an incubator. When the cell fusion rate reached 100%, the medium was discarded, scratched with a 200 μ L sterile pipette tip perpendicular to the 6-well plate, and then washed twice with PBS. The PBS was discarded, serum-free medium was added, the cells were treated with 1. mu. mol/L of apratinib, and the cells were placed in an incubator for further culture. Photographs were taken at fixed positions at 0, 24, and 48 hours after scratching, respectively. The distance of the scratch was measured using OLYMPUS cellSens Entry software to calculate the cell mobility. The experiment was repeated three times.
1.4.7Transwell cell invasion assay Mtrigel matrigel was diluted (ice-on-ice) using the manufacturer's recommended dilution method, 100. mu.L of the dilution was added to the upper chamber of the Transwell chamber, placed in an incubator for 1-4 hours, and the upper liquid medium was discarded. The collected 9464D, 975A2, IMR-32 and SK-N-BE (2) cell pellets were resuspended and counted in 1X 10 serum-free medium5Each 100. mu.L/well was added to the upper chamber, and 600. mu.L of the corresponding 20% fetal bovine serum medium was added to the lower chamber. Both the Transwell cells and the 24-well plates contained the same concentration of antrotinib (4. mu. mol/L). The cultivation was continued in the incubator for 24 hours. The medium in the upper and lower chambers was discarded, and 600. mu.L of 4% paraformaldehyde-fixed cells were added to the lower chamber for 15 minutes. The paraformaldehyde was discarded and the PBS was washed twice. Add 600. mu.L of 0.1% crystal violet stain to the lower chamber for 1 min, wash twice with PBS, wipe off the cells in the upper chamber with a cotton swab, and air dry the chamber naturally. And taking 5 fields at random for each chamber under the microscope to take a picture, counting cells by adopting ImageJ software, and calculating the number of the invaded cells. The experiment was repeated three times.
1.4.8 subcutaneous tumor-forming animal experiments 5-6 weeks old C57BL/6 mice were selected for feeding and two murine N-myc expansion neuroblastoma cell lines of 9464D and 975A2 were used for subcutaneous tumor formation. Firstly, preparing tumor cell suspension according to the proportion of 1 × 106Cells/mouse, diluted to 100. mu.L, were inoculated subcutaneously in the right dorsal aspect of the mice. About 3-4 weeks after tumor implantation, subcutaneous injectionAfter the tumor volume is measured, dividing the tumor volume into 4 groups according to the volume, namely a control group, an Arotinib treatment group, a Bomaciclib treatment group and an Arotinib combined Bomaciclib treatment group, wherein each group comprises 6 mice, then performing intragastric lavage treatment for 21 days, measuring the tumor volume and the weight of the mice once every 3 days, and drawing a tumor growth curve. After the treatment was stopped, the mice were sacrificed by cervical dislocation, subcutaneous tumors and spleens were removed with surgical instruments, weighed, photographed, recorded for tumors, and total splenic cell counts were performed. Statistics and plots were performed using GraphpadPrism8 software.
1.4.9 flow cytometry analysis of tumor immune microenvironment after 21 days of anitinib treatment, mice were sacrificed by cervical dislocation, subcutaneous tumor tissues and spleens were removed with sterile surgical instruments and processed into single cell suspensions, and finally stained with fluorescently labeled antibodies (CD3, CD4, CD8, CD11b, CD11c, CD19, CD25, CD40, CD45, Gr1, F4/80, NK1.1, NKp46, FoxP3, PD1, Ki67, TNF α, IFN γ, GzmB, H2, β 2m, MULT1, PDL 1). After the reaction was completed, detection was performed using a flow cytometer. And analyzing by using Flowjo software, and calculating the ratio of various immune cells.
2. Results of the experiment
2.1 Arotinib significantly inhibits neuroblastoma cell proliferation
The CCK8 experiment result shows that the cell inhibition rate is higher with the increase of the concentration of the angutinib. And the inhibition rate of the same concentration of the nilotinib on the cells is increased along with the time. The above results indicate that the inhibition of 9464D, 975A2, IMR-32 and SK-N-BE (2) cell lines by Arotinib is concentration and time dependent. The plate cloning experiment result shows that the nilotinib can obviously inhibit the cloning formation of the neuroblastoma cell line (see fig. 1 and fig. 2).
IC50A cell line 975A2 9464D IMR-32 SK-N-BE(2)
24h (micromole) 23.49 15.72 24.71 23.27
48h (micromole) 6.683 4.091 6.452 6.396
72h (micromolar) 2.770 2.051 2.514 2.320
2.2 Arotinib significantly blocks neuroblastoma cell cycle
Experimental results of the effect of anitinib on cell cycle of 9464D, 975A2, IMR-32 and SK-N-BE (2) cell lines show that anitinib can significantly induce G2\ M cycle arrest in neuroblastoma cell lines, and that cell cycle arrest is stronger with increasing anitinib concentration (FIG. 3).
2.3 Arotinib significantly promotes neuroblastoma cell apoptosis
The effect of Arotinib on apoptosis of 9464D, 975A2, IMR-32 and SK-N-BE (2) cell lines was examined by flow cytometry. The results show that the neuroblastoma cell line can be remarkably promoted to undergo apoptosis, and the apoptosis rate is higher along with the increase of the concentration of the neuroblastoma (figure 4).
2.4 Arotinib significantly inhibits neuroblastoma cell migration
The results of the scratch experiment show that the cell lines 9464D, 975A2, IMR-32 and SKN-BE (2) have significantly reduced migration ability after the anitinib treatment, and the cell migration rate is more significantly different from that of the control group with the time (FIG. 5).
2.5 Arotinib significantly inhibits neuroblastoma cell invasion
The results of the Transwell cell invasion experiments show that the invasion capacity of 9464D, 975A2, IMR-32 and SK-N-BE (2) cell lines is remarkably reduced after the treatment of the anitinib (FIG. 6).
2.6 Arotinib significantly inhibited growth of subcutaneous neuroblastoma in mice
The experimental result of a mouse subcutaneous tumor-forming animal shows that the anitinib obviously inhibits the growth of mouse subcutaneous neuroblastoma and reduces the tumor load. The targeted drug Bomaciclib was able to enhance the therapeutic effect of erlotinib on neuroblastoma (FIG. 7A, FIG. 7B), wherein NS is control, Abe is Bomaciclib, and Anlo is Arotinib.
2.7 Arotinib significantly activates the neuroblastoma immune microenvironment
Flow cytometry analysis results showed that antratinib significantly increased the percentage of CD4 and CD 8T cell infiltration in mouse neuroblastoma and spleen, and decreased the number of MDSCs. The ability of CD4 and CD 8T cells to secrete INF-gamma and TNF-alpha is enhanced simultaneously, namely the killing ability of CD4 and CD 8T cells is enhanced, and the targeted drug Bomaciclib can enhance the activation effect of the immune microenvironment of the neuroblastoma by the antrocin (fig. 8A and 8B).
Example 2 clinical study of refractory or recurrent neuroblastoma in children who had received first-line treatment
Grouping standard:
a neuroblastoma subject that is distant metastasis or locally advanced and that the investigator decides is not suitable for surgical treatment; tumor imaging evaluation is carried out according to RECIST v1.1, and at least 1 double-diameter measurable focus is judged according to CT or MRI; patients who have been treated with at least one chemotherapeutic regimen (containing anthracyclines) and who have been evaluated as progressing or intolerant of the disease according to the criteria for efficacy assessment of solid tumors (RECIST 1.1); age of patients in group: under the age of 21 years, both men and women can use the Chinese medicinal composition; PS scoring: 0-1 (amputated subjects can be relaxed to 2 points), and the life cycle is expected to exceed 12 weeks; patients who had previously received no progression of irinotecan or in combination with temozolomide.
The specific administration scheme is as follows: 7mg/m of erlotinib hydrochloride 21 time daily, orally, and stopping administration for two weeks, namely d1-14 administration, wherein each 3 weeks (21 days) is a treatment period; irinotecan 50mg/m2D 1-5, i.v.; temozolomide 100mg/m2D 1-5, oral administration. Every 3 weeks is one treatment cycle.
The main research indexes are as follows: arotinib hydrochloride, irinotecan in combination with temozolomide treat progression-free survival (PFS) in refractory or relapsed neuroblastoma in children.
Secondary indexes are as follows: objective remission rate (ORR ═ CR + PR), disease control rate (DCR ═ CR + PR + SD), Overall Survival (OS), quality of life score, safety index (adverse event AE) were observed and evaluated for ambrotinib hydrochloride, irinotecan in combination with temozolomide.
For tumor imaging evaluation according to RECIST v1.1, the subject must be accompanied by measurable tumor lesions at baseline. Efficacy assessment results were according to RECIST 1.1 criteria, including: complete Remission (CR), Partial Remission (PR), Stable (SD) and Progression (PD), PFS, OS.
For the sick children, the aged 5 is 7 months old, the neuroblastoma behind the right peritoneum is diagnosed for more than 1 year, the resection of the tumor behind the right peritoneum under the general anesthesia, the right nephrectomy, the bilateral retroperitoneal lymph node cleaning and the multi-cycle auxiliary chemotherapy and the abdominal tumor area radiotherapy are carried out after the previous 5-cycle chemotherapy. Recurrence of disease, blood NSE (neuron specific enolase) in the infant after 1 month after the radiotherapy is finished
393.2ug/L, significantly higher than normal, 55% of bone marrow stained positive cells against GD2, touching the skull with multiple masses. Review PET-CT shows: the thickening of soft tissues in the original operation area is increased and enlarged more than before, the lymph nodes in the bilateral clavicle area are metastasized, the bone metastasis is frequently generated, the nodules are frequently generated under the head skin, and the metastasis is suspicious. 4-stage postoperative recurrence of right retroperitoneal neuroblastoma of the infant patient is clear and definite in diagnosis of high-risk groups. After the chemotherapy is continuously performed for 5 weeks, abdominal tumors of children are not continuously reduced, NSE has a rising trend, median PFS of children who are difficult to treat or relapse neuroblastoma is judged to be 3.3 months according to previous research, the tumor relapse risk is considered to be higher, the children are subjected to the chemotherapy and the anitinib anti-vascular therapy for 5 periods, NSE is periodically rechecked and kept stable after being reduced, the best curative effect PR (reduced by 37 percent), and the PFS reaches more than 8 months by the date of patent application.

Claims (17)

1. Use of a pharmaceutical composition comprising a compound of formula I or a pharmaceutically acceptable salt thereof and at least one pharmaceutically acceptable carrier for the manufacture of a medicament for the treatment of neuroblastoma,
Figure FDA0002878117630000011
2. the use of claim 1, wherein said pharmaceutical composition further comprises at least one second therapeutic agent.
3. Use of a combination for the preparation of a medicament for the treatment of neuroblastoma, wherein said combination comprises: (i) a compound of formula I or a pharmaceutically acceptable salt thereof;
Figure FDA0002878117630000012
and (ii) at least one second therapeutic agent.
4. The use according to any one of claims 1 to 3, wherein the neuroblastoma is a locally advanced, and/or advanced and/or metastatic neuroblastoma.
5. The use according to any one of claims 1 to 4, wherein the neuroblastoma is a relapsed and/or refractory neuroblastoma.
6. The use according to any one of claims 1 to 5, wherein the neuroblastoma is a high risk neuroblastoma.
7. The use of any one of claims 1-6, wherein the neuroblastoma is an ALK gene mutation, a PHOX2B gene mutation, a neuroblastoma deleted at positions 1p36 or 11q 14-23, a TP53-R337H mutation, a CDKN1C mutation, a HRAS mutation, and/or an MYCN gene amplification; alternatively, the neuroblastoma is 11q normal differentiated neuroblastoma, or 11q abnormal or undifferentiated neuroblastoma.
8. The use of any one of claims 1-7, wherein the neuroblastoma is a neuroblastoma that fails a first line therapy.
9. The use of any one of claims 2-8, wherein the second therapeutic agent is selected from one or more of a chemotherapeutic agent, a targeted agent, an immunotherapy, 13-cis retinoic acid, GM-CSF, interleukin 2, 131I-MIBG.
10. The use of claim 9, wherein the chemotherapeutic agent is selected from one or more of platinum group drugs, fluoropyrimidine derivatives, taxanes, camptothecins and their analogues, vinblastines, anthracyclines, cytarabine, thioguanine, pemetrexed, carmustine, melphalan, etoposide, tennixin, mitomycin, ifosfamide, cyclophosphamide, azacitidine, methotrexate, bendamustine, pentostatin, carmustine, liposomal doxorubicin, actinomycin D, bleomycin, pingomycin, temozolomide, amiloride, pellomycin, eribulin, plin, Sapacitabine, troxsuprane, 153Sm-EDTMP, tegafur, levoasparaginase, pemetrexed, cephalotaxine, procarbazine, ARC-100 and encedar.
11. The use of claim 10, wherein the chemotherapeutic agent is selected from the group consisting of irinotecan and temozolomide.
12. The use of claim 9, wherein the targeted drug is selected from the group consisting of small molecule targeted antineoplastic and antineoplastic antibody drugs.
13. The use of claim 12, wherein the small molecule targeted antineoplastic agent is selected from ALK inhibitors.
14. The use as claimed in claim 12, wherein the small molecule targeted antineoplastic agent is selected from one or more of bemaciclib, venetochlax, fenretinide, ABTL-0812, OTS-167, SF-1126, nifurtimox, eflornithine, ABTL-0812, CBL-0137, ONC-201, fadraciclib, LY-3295668erbumine, rose sodium salt, venetochlax, copanlisib, tamibarotene, loratinib, enaritinib, and entrecttinib.
15. The use of claim 14, wherein the second therapeutic agent is selected from Bomaciclib.
16. The use of claim 9, wherein the second therapeutic agent is selected from one or more of the group consisting of:
one or more of cyclophosphamide, doxorubicin, dexuximab, dinutuximab beta, vincristine, busulfan, melphalan, carboplatin, cisplatin, etoposide, 13-cis-retinoic acid, IL-2, GM-CSF, and temozolomide;
platinum drugs and etoposide;
platinum drugs, etoposide and melphalan;
vincristine + doxorubicin + cyclophosphamide + mesna;
vincristine + cyclophosphamide + mesna;
topotecan;
topotecan + etoposide;
topotecan + vincristine + doxorubicin;
topotecan + temozolomide;
topotecan + cyclophosphamide;
temozolomide;
temozolomide + irinotecan;
irinotecan;
carboplatin + irinotecan and/or temozolomide;
busulfan + melphalan;
temozolomide + irinotecan + dexuximab;
an ALK inhibitor;
131 I-MIBG;
anti-GD-2 (anti-ganglioside 2) antibody; and
anti-GD-2 antibody + isotretinoin;
anti-GD-2 antibody + IL-2;
anti-tumor antibody drugs + isotretinoin;
antitumor antibody drug + IL-2.
17. A method of treating neuroblastoma, comprising administering to a patient in need thereof a pharmaceutical composition comprising (I) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) at least one second therapeutic agent,
Figure FDA0002878117630000031
CN202011629017.5A 2020-12-31 2020-12-31 Quinoline derivatives for the treatment of neuroblastoma Active CN113116895B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202011629017.5A CN113116895B (en) 2020-12-31 2020-12-31 Quinoline derivatives for the treatment of neuroblastoma

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202011629017.5A CN113116895B (en) 2020-12-31 2020-12-31 Quinoline derivatives for the treatment of neuroblastoma

Publications (2)

Publication Number Publication Date
CN113116895A true CN113116895A (en) 2021-07-16
CN113116895B CN113116895B (en) 2023-08-18

Family

ID=76772357

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011629017.5A Active CN113116895B (en) 2020-12-31 2020-12-31 Quinoline derivatives for the treatment of neuroblastoma

Country Status (1)

Country Link
CN (1) CN113116895B (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115389766A (en) * 2022-08-29 2022-11-25 深圳市瑞格生物科技有限公司 Marker for diagnosing whether neuroblastoma has bone marrow infiltration or not and application thereof
WO2023046131A1 (en) * 2021-09-26 2023-03-30 正大天晴药业集团股份有限公司 Use of anti-cd40 antibody

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101730697A (en) * 2007-03-21 2010-06-09 惠氏公司 Imidazolopyrimidine analogs and their use as pi3 kinase and mtor inhibitors
CN102344438A (en) * 2010-08-01 2012-02-08 江苏正大天晴药业股份有限公司 Crystals of quinoline derivative and preparation methods thereof
CN111643503A (en) * 2019-03-04 2020-09-11 正大天晴药业集团股份有限公司 Quinoline derivatives for the treatment of non-small cell lung cancer
CN111973747A (en) * 2019-05-23 2020-11-24 正大天晴药业集团股份有限公司 Quinoline derivatives for the combined treatment of ovarian cancer

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101730697A (en) * 2007-03-21 2010-06-09 惠氏公司 Imidazolopyrimidine analogs and their use as pi3 kinase and mtor inhibitors
CN102344438A (en) * 2010-08-01 2012-02-08 江苏正大天晴药业股份有限公司 Crystals of quinoline derivative and preparation methods thereof
CN103664892A (en) * 2010-08-01 2014-03-26 正大天晴药业集团股份有限公司 Crystal of quinoline derivative
CN111643503A (en) * 2019-03-04 2020-09-11 正大天晴药业集团股份有限公司 Quinoline derivatives for the treatment of non-small cell lung cancer
CN111973747A (en) * 2019-05-23 2020-11-24 正大天晴药业集团股份有限公司 Quinoline derivatives for the combined treatment of ovarian cancer

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SUBHASREE ROY CHOUDHURY ET AL.: "Targeting angiogenesis for controlling neuroblastoma",subhasree roy choudhury et al.", 《JOURNAL OF ONCOLOGY》 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023046131A1 (en) * 2021-09-26 2023-03-30 正大天晴药业集团股份有限公司 Use of anti-cd40 antibody
CN115389766A (en) * 2022-08-29 2022-11-25 深圳市瑞格生物科技有限公司 Marker for diagnosing whether neuroblastoma has bone marrow infiltration or not and application thereof
CN115389766B (en) * 2022-08-29 2023-09-22 深圳市瑞格生物科技有限公司 Marker for diagnosing whether neuroblastoma is subjected to bone marrow infiltration and application thereof

Also Published As

Publication number Publication date
CN113116895B (en) 2023-08-18

Similar Documents

Publication Publication Date Title
JP7465305B2 (en) Methods for Treating and Preventing Graft-Versus-Host Disease - Patent application
CN110996952A (en) Methods for treating cancer
JP2020528880A (en) Combination cancer therapy
US20180353602A1 (en) Combination of hdac inhibitor and anti-pd-l1 antibody for treatment of cancer
CA2982435C (en) Treatment method by combined use of mdm2 inhibitor and btk inhibitor
WO2023035611A1 (en) Pharmaceutical composition and use thereof
CN109819649B (en) Solid forms of aminopurine compounds and methods of use thereof
CN113116895B (en) Quinoline derivatives for the treatment of neuroblastoma
KR20140040728A (en) Methods of treating mesothelioma with a pi3k inhibitor compound
CN113710658A (en) Quinolines or pharmaceutically acceptable salts thereof for the treatment of ewing's sarcoma
CN111643503A (en) Quinoline derivatives for the treatment of non-small cell lung cancer
KR20210019422A (en) Cancer treatment method
WO2023093663A1 (en) Pharmaceutical composition and use thereof
AU2023202746A1 (en) Combination of a BCL-2 inhibitor and a MCL-1 inhibitor, uses and pharmaceutical compositions thereof
WO2020233602A1 (en) Quinoline derivative used for combination treatment of small cell lung cancer
US11419862B2 (en) Quinoline derivative for treatment of nasopharyngeal carcinoma
CN114469949A (en) Quinoline derivatives for the treatment of diffuse large B-cell lymphoma
WO2016172053A1 (en) Methods of treating ibrutinib-resistant disease
CN111821302A (en) Quinolines for the combined treatment of chondrosarcoma
CN117797151A (en) Quinoline derivatives combined with chemotherapeutics for treating non-small cell lung cancer
CN114288303B (en) Antineoplastic pharmaceutical composition containing piperazines and application thereof
WO2024059962A1 (en) Pharmaceutical composition and use thereof
JP2015509967A (en) Novel combination for the treatment of acute or chronic myeloid leukemia
CN113750096A (en) Quinoline derivatives for the treatment of peripheral T cell lymphoma
EP4095137A1 (en) Use of pyrido[1,2-a]pyrimidinone compound in treating lymphoma

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant