CN113116895B - Quinoline derivatives for the treatment of neuroblastoma - Google Patents
Quinoline derivatives for the treatment of neuroblastoma Download PDFInfo
- Publication number
- CN113116895B CN113116895B CN202011629017.5A CN202011629017A CN113116895B CN 113116895 B CN113116895 B CN 113116895B CN 202011629017 A CN202011629017 A CN 202011629017A CN 113116895 B CN113116895 B CN 113116895B
- Authority
- CN
- China
- Prior art keywords
- neuroblastoma
- formula
- pharmaceutically acceptable
- compound
- acceptable salt
- 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.)
- Active
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—Heterocyclic 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
- A61K31/47—Quinolines; Isoquinolines
- A61K31/4709—Non-condensed quinolines and containing further heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/04—Antineoplastic agents specific for metastasis
Landscapes
- Health & Medical Sciences (AREA)
- Veterinary Medicine (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Epidemiology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Oncology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The invention relates to an application of quinoline derivatives in treating neuroblastoma, and belongs to the field of medicines. In particular, the invention relates to quinoline derivatives for use in the treatment of neuroblastoma. The advantages are as follows: an Luoti Ni can significantly inhibit the growth, migration and invasion of NB cell lines, induce cell cycle arrest thereof and promote apoptosis thereof.
Description
Technical Field
The invention belongs to the field of medicines, and particularly relates to application of quinoline derivatives in treatment of neuroblastoma.
Background
Neuroblastoma (neuroblastoma) is an embryonal tumor that originates from primitive neural crest cells, the most common extracranial solid tumor in children. Accounting for 8% -10% of all children malignant tumors, and the annual incidence rate is 0.3-5.5/10 ten thousand. Neuroblastomas are derived from undifferentiated sympathetic ganglion cells, so tumors can develop in any location where there are embryonal sympathetic ganglion cells. The suprarenal glands are the most common primary sites, followed by the abdominal, thoracic, cervical and pelvic sympathetic ganglia, about 1% of which fail to find the primary tumor. Neuroblastomas can metastasize to lymph nodes, bone marrow, bone, dura mater, orbit, liver and skin, and in a few cases also to the lungs and intracranial.
Prognosis of neuroblastoma is closely related to diagnosis age, clinical stage, tumor pathology type, copy number of N-myc gene and DNA index. The childhood tumor tissue (COG) divides neuroblastomas into low, medium and high risk groups according to the five indicators described above. The treatment difficulty of the childhood neuroblastoma is high, the single treatment prognosis is poor, and the current treatment of the neuroblastoma is multi-disciplinary combined treatment mainly comprising surgery, radiotherapy and chemotherapy and autologous stem cell transplantation. With the gradual improvement of diagnosis and treatment technology, the survival rate of children neuroblastoma in China is improved year by year, but the OS is still lower than that in European and American countries, 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 in the manufacture of a medicament for the treatment of neuroblastoma. The present application also provides a method of treating a 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 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 aspect, the present application provides a combination for use in the treatment of neuroblastoma 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 application also provides the use of a combination medicament in the manufacture of a medicament for the treatment of neuroblastoma. The application also provides the use of the combination medicament for treating neuroblastoma.
In a fourth aspect, the application also provides a method of treating neuroblastoma comprising administering to a patient in need thereof a combination of the application. The combination comprises (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 in the manufacture of a medicament for the treatment of neuroblastoma.
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, undifferentiated, differentiated, poorly differentiated neuroblastoma. In some embodiments, the neuroblastoma includes, but is not limited to, extracranial neuroblastoma.
In some embodiments, the neuroblastoma includes, but is not limited to, the following histopathological types: NB type (schwannian few matrix type), GNB hybrid (gamglioneurobola), GN mature (matrix), GNB nodular (noduliar).
In some embodiments, the neuroblastoma is a locally advanced, and/or metastatic neuroblastoma. In some embodiments, the neuroblastoma is metastatic neuroblastoma. In some embodiments, the neuroblastoma is a relapsed and/or refractory neuroblastoma. In some embodiments, the neuroblastoma is 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, a HRAS mutation, and/or a MYCN gene amplified neuroblastoma. In some embodiments, the neuroblastoma is 11q normal and differentiated neuroblastoma, or is 11q abnormal or undifferentiated neuroblastoma.
In the application, the neuroblastoma patient is a patient under 21 years old; in some embodiments, the patient with neuroblastoma is a patient under 18 years of age. In some embodiments, the neuroblastoma patient has previously received hematopoietic stem cell transplantation. In some embodiments, the neuroblastoma patient has previously received an autologous hematopoietic stem cell transplant.
In one embodiment of the application, the neuroblastoma patient has previously been treated with a first line therapy. In a specific embodiment of the present application, the neuroblastoma is a recurrent and/or refractory neuroblastoma that fails to undergo first-line therapy. In some embodiments, the neuroblastoma patient has previously received surgical resection, chemotherapy, radiation therapy, targeted therapy, immunotherapy, and/or hematopoietic stem cell transplantation. In some embodiments, the neuroblastoma is a chemotherapeutic, targeted, and/or immunotherapy failed neuroblastoma. In one embodiment of the application, the neuroblastoma patient has been previously subjected to chemotherapy. In one embodiment of the application, the neuroblastoma is a chemotherapy-failed neuroblastoma.
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 failed treatment with a chemotherapeutic regimen containing an anthracycline. In some specific embodiments, the neuroblastoma is a neuroblastoma that fails to be treated with one or more of a platinum-based drug, etoposide, vincristine, doxorubicin, cyclophosphamide, ifosfamide, topotecan, temozolomide, irinotecan, doxorubicin, mitoxantrone, methotrexate, cytarabine, procarbazine, gemcitabine, lenalidomide, rituximab (dinutuximab), dinutuximab beta, 13-cis-retinoic acid, granulocyte-macrophage colony stimulating factor GM-CSF, interleukin 2, ALK inhibitor, 131I-mig.
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 agent includes, but is not limited to, one or more of chemotherapeutic agents, targeted agents, immunotherapeutics, 13-cis-retinoic acid, granulocyte-macrophage colony stimulating factor GM-CSF, interleukin 2, 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 the group consisting of 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 agent, an immunotherapeutic agent.
In some embodiments, the second therapeutic agent is selected from one or more of platinum-based agents, etoposide, anthracyclines, vinca-alkaloids, granulocyte-macrophage colony stimulating factor GM-CSF, interleukin 2, 131I-migg, 13-cis-retinoic acid, mesna, cyclophosphamide, ifosfamide, rituximab, dinuteximab beta, busulfan, melphalan, 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, rituximab beta, vincristine, busulfan, melphalan, carboplatin, cisplatin, etoposide, 13-cis-retinoic acid, interleukin 2, GM-CSF, temozolomide;
Platinum-based drugs (e.g., carboplatin, cisplatin) and etoposide;
platinum-based 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 + rituximab;
an ALK inhibitor;
131I-MIBG;
an anti-GD-2 (anti-ganglioside 2) antibody; and
anti-GD-2 antibody + isotretinoin;
anti-GD-2 antibody + interleukin 2;
anti-tumor antibody drug + isotretinoin;
anti-tumor antibody drug + interleukin 2.
In a preferred embodiment, the second therapeutic agent is selected from irinotecan and temozolomide.
In a specific embodiment, the second therapeutic agent is selected from irinotecan and temozolomide, one 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/m 2 Is administered in a daily dose of (a); in some embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is present at 5-8mg/m 2 Is administered in a daily dose of (a); in some embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is present at 5-7mg/m 2 Is administered in a daily dose of (a);
in some specific embodiments, the second therapeutic agent is irinotecan or temozolomide, wherein the compound of formula I or a pharmaceutically acceptable salt thereof is at 7mg/m 2 Is administered on days 1-14; irinotecan at 50mg/m 2 Is administered on days 1-5; temozolomide at 100mg/m 2 Is administered on days 1-5.
In a second aspect, the present application provides a combination for use in the treatment of neuroblastoma 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 medicament for treating neuroblastoma comprising: (I) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) one or more of a chemotherapeutic drug, a targeted drug, immunotherapy, 13-cis-retinoic acid, granulocyte-macrophage colony-stimulating factor GM-CSF, interleukin 2, 131I-migg, and optionally in combination with radiotherapy.
In some embodiments, there is provided a combination medicament for treating neuroblastoma comprising: (I) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) one or more chemotherapeutic agents, 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-migg; in some embodiments, the chemotherapeutic agent is selected from the group consisting of platinum, temozolomide, etoposide, anthracyclines, vinblastine, camptothecins and analogs thereof, cyclophosphamide, ifosfamide, doxorubicin, topotecan, busulfanOne or more of melphalan. In some embodiments, the chemotherapeutic agent is selected from irinotecan and temozolomide; in some embodiments, one treatment cycle is every 3 weeks, wherein the compound of formula I, or a pharmaceutically acceptable salt thereof, is at 4-10mg/m 2 Is administered in a daily dose of (a); in some embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is present at 5-8mg/m 2 Is administered in a daily dose of (a); in some embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is present at 5-7mg/m 2 Is administered in a daily dose of (a); in some specific embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is at 7mg/m 2 Is administered on days 1-14; irinotecan at 50mg/m 2 Is administered on days 1-5; temozolomide at 100mg/m 2 Is administered on days 1-5.
In some embodiments, there is provided a combination medicament for treating neuroblastoma comprising: (I) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) at least one small molecule targeted anti-tumor drug, 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-migg; in some embodiments, the small molecule targeted anti-tumor drug is an ALK inhibitor.
In some embodiments, there is provided a combination medicament 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, 131I-migg. In some embodiments, the anti-tumor antibody drug is selected from anti-GD-2 antibodies; anti-tumor in some embodiments, the anti-tumor antibody drug is selected from the group consisting of rituximab, dinutuximab beta.
In some embodiments, there is provided a combination medicament for treating neuroblastoma comprising: (I) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) at least one immunotherapeutic 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-migg.
In some embodiments, there is provided a combination medicament 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, 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-migg.
In some embodiments, there is provided a combination medicament for treating neuroblastoma comprising: (I) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) camptothecin and analogs thereof and/or temozolomide, 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-migg.
In some embodiments, there is provided a combination medicament 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, 131I-migg.
In some embodiments, there is provided a combination medicament for treating neuroblastoma comprising: (I) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) platinum and/or 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, 131I-migg.
In some embodiments, there is provided a combination medicament for treating neuroblastoma comprising: (I) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) one or more of cyclophosphamide, vincristine, doxorubicin, 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, 131I-migg.
In some embodiments, there is provided a combination medicament for treating neuroblastoma comprising: (I) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) lenalidomide, and optionally in combination with radiation therapy. In some embodiments, there is provided a combination medicament for treating neuroblastoma comprising: (I) a compound of formula I or a pharmaceutically acceptable salt thereof; and (ii) one or both of gemcitabine, vinorelbine, and 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, rituximab beta, vincristine, busulfan, melphalan, carboplatin, cisplatin, etoposide, 13-cis-retinoic acid, interleukin 2, GM-CSF, temozolomide;
platinum-based drugs (e.g., carboplatin, cisplatin) and etoposide;
platinum-based 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 + rituximab;
an ALK inhibitor;
131I-MIBG;
an anti-GD-2 (anti-ganglioside 2) antibody; and
anti-GD-2 antibody + isotretinoin;
anti-GD-2 antibody + interleukin 2;
anti-tumor antibody drug + isotretinoin;
anti-tumor antibody drug + interleukin 2.
In a third aspect, the application also provides the use of a combination medicament in the manufacture of a medicament for the treatment of neuroblastoma.
In a fourth aspect, the application also provides a method of treating neuroblastoma comprising administering to a patient in need thereof a combination of the 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 suffering from neuroblastoma. In some aspects of the 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 again in disease progression after complete remission following chemotherapy and/or targeted and/or immunotherapeutic and/or radiation therapy. In some embodiments, the patient fails to completely or partially alleviate following chemotherapy and/or targeted and/or immunotherapeutic 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 previously untreated with a drug, the method comprising administering to a patient in need of treatment 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 previously treated with at least one chemotherapeutic agent, the method comprising administering to a patient in need of treatment 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 a neuroblastoma that fails a first line therapy, the method comprising administering to a patient in need of treatment 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 recurrent neuroblastoma comprising administering to a patient in need of treatment a compound of formula I, or a pharmaceutically acceptable salt thereof, and at least one second therapeutic agent.
In some aspects of the application, the patient has not previously received systemic treatment.
The method of administration can be determined comprehensively based on the activity, toxicity, tolerance of the patient, etc. In some embodiments of the 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 per day (bid), twice per week (bid), three times per day (tid), four times per day (qid), etc., daily (qd), every 3 days (q 3 d), every 4 days (q 4 d), every 5 days (q 5 d), weekly (q 1 w), every 2 weeks (q 2 w), every 3 weeks (q 3 w), or every 4 weeks (q 4 w). In some embodiments of the application, the second therapeutic agent may also be administered in a spaced-apart manner in the use or method of treatment. The intermittent administration includes an administration period and a withdrawal period, for example, the administration of the second therapeutic agent daily during the administration period, followed by withdrawal for a period of time during the withdrawal period, followed by the administration period, and then withdrawal, so that the cycle may be repeated a number of times.
In some embodiments of the application, the compounds of formula I or pharmaceutically acceptable salts thereof, including but not limited to, may be administered in a dosage of 8-12mg, administered continuously for 2 weeks, at a dosing regimen of 1 week; and/or, in a dosing regimen of 2 weeks, 2 weeks of continuous dosing.
In some embodiments, the compound of formula I, or a pharmaceutically acceptable salt thereof, is administered on days 1-14 of each cycle, one administration cycle every 21 days.
In some embodiments, the second therapeutic agent and the compound of formula I or a pharmaceutically acceptable salt thereof have the same or different treatment cycles, respectively. 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 both administered in a single treatment cycle every 3 weeks.
Optionally, the second therapeutic agent may be used in combination with a chemotherapeutic adjunct including, but not limited to, calcium leucovorin (CF), aldehyde hydrofolate, mesna, bisphosphonates, amifostine, hematopoietic cell 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 separately packaged, and instructions for treating neuroblastoma.
Neuroblastoma
In some embodiments of the application, the clinical stage of the neuroblastoma includes, but is not limited to, 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, undifferentiated, differentiated, poorly differentiated neuroblastoma. In some embodiments, the neuroblastoma includes, but is not limited to, extracranial neuroblastoma.
In some embodiments, the neuroblastoma includes, but is not limited to, the following histopathological types: NB type (schwannian few matrix type), GNB hybrid (gamglioneurobola), GN mature (matrix), GNB nodular (noduliar).
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, a HRAS mutation, and/or a MYCN gene amplified neuroblastoma. In some embodiments, the neuroblastoma is 11q normal and differentiated neuroblastoma, or is 11q abnormal or undifferentiated neuroblastoma.
In the application, the neuroblastoma patient is a patient under 21 years old; in some embodiments, the patient with neuroblastoma is a patient under 18 years of age.
In some embodiments, the neuroblastoma is a previously surgically resected neuroblastoma. In some embodiments, the neuroblastoma patient has previously received hematopoietic stem cell transplantation. In some embodiments, the neuroblastoma is a neuroblastoma that has previously received an autologous hematopoietic stem cell transplant.
In some embodiments, the neuroblastoma is a neuroblastoma that failed radiation therapy. In some embodiments, the neuroblastoma is a neuroblastoma that failed first line therapy. In some embodiments, the neuroblastoma is a failed neuroblastoma to be treated with a chemotherapeutic agent and/or a targeted agent. In some embodiments, the neuroblastoma is a neuroblastoma previously subjected to at least two chemotherapy regimens. In some embodiments, the neuroblastoma is a neuroblastoma that failed surgical and/or radiation and/or chemotherapy drug treatment. In some specific embodiments, the neuroblastoma is a neuroblastoma that fails to be treated with one or more of a platinum-based drug (e.g., carboplatin, cisplatin), an anthracycline antitumor drug, etoposide, vincristine, doxorubicin, cyclophosphamide, ifosfamide, topotecan, temozolomide, irinotecan, doxorubicin, mitoxantrone, methotrexate, cytarabine, procarbazine, gemcitabine, lenalidomide, rituximab, 13-cis-retinoic acid, granulocyte-macrophage colony-stimulating factor GM-CSF, interleukin 2, ALK inhibitor, 131I-migg, ibutenib.
Second therapeutic agent
In the present application, the second therapeutic agent includes, but is not limited to, one or more of a chemotherapeutic agent, a targeting agent, an immunotherapy, 13-cis-retinoic acid, granulocyte-macrophage colony stimulating factor GM-CSF, interleukin 2, 131I-MIBG. The targeting drugs comprise small molecule targeting antitumor drugs and antitumor antibody drugs.
The second therapeutic agent of the application comprises one or more of chemotherapeutic agents, small molecule targeted antitumor agents, immunotherapeutic agents and antitumor antibody agents.
In some embodiments, the second therapeutic agent is selected from chemotherapy and immunotherapy. In some embodiments, the second therapeutic agent is selected from the group consisting of 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-based anti-neoplastic agent, an alkylating agent, an anti-neoplastic antibody agent, an immunotherapeutic agent.
In some embodiments, the second therapeutic agent is selected from one or more of platinum-based agents, etoposide, anthracyclines, vinca-alkaloids, camptothecins and analogs thereof, granulocyte-macrophage colony stimulating factor GM-CSF, interleukin 2, 131I-migg, 13-cis-retinoic acid, mesna, cyclophosphamide, ifosfamide, rituximab, dinuteximab 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, doxorubicin, valrubicin, zorubicin, pitaxron, doxorubicin.
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, miltiplatin, triplatin tetranitrate, phenanthreneplatin, picoplatin, satraplatin, and leptoplatin.
In some embodiments, the second therapeutic agent is a fluoropyrimidine derivative including, but not limited to, one or more of gemcitabine, capecitabine, ancitabine, fluorouracil, bififluridine, doxifluridine, tegafur, carmofur, trifluoracetam.
In some embodiments, the second therapeutic agent is a camptothecin and analogs thereof; in some embodiments, the second therapeutic agent is camptothecins and analogs thereof, and anti-tumor antibody agents; the camptothecins and analogues thereof include, but are not limited to, one or more of camptothecins, hydroxycamptothecins, 9-aminocamptothecins, 7-ethylcamptothecins, irinotecan, 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, rituximab beta, vincristine, busulfan, melphalan, carboplatin, cisplatin, etoposide, 13-cis-retinoic acid, interleukin 2, GM-CSF, temozolomide;
platinum-based drugs (e.g., carboplatin, cisplatin) and etoposide;
platinum-based 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 + denox mab;
an ALK inhibitor;
131I-MIBG;
an anti-GD-2 (anti-ganglioside 2) antibody; and
anti-GD-2 antibody + isotretinoin;
anti-GD-2 antibody + interleukin 2;
anti-tumor antibody drug + isotretinoin;
anti-tumor antibody drug + interleukin 2.
Chemotherapeutic agents
In the present application, the chemotherapeutic agents include, but are not limited to, one or more of alkylating agents (e.g., platinum-based agents, busulfan, melphalan, cyclophosphamide, ifosfamide, procarbazine, dacarbazine, temozolomide, etc.), plant alkaloids (e.g., vinblastine-based agents, taxanes, podophyllotoxins (e.g., etoposide, teniposide), camptothecins, and analogs thereof), antibiotic antineoplastic agents (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.).
The chemotherapeutic drug of the application has the advantages of good therapeutic effect, examples that may be cited include, but are not limited to, platins (e.g., oxaliplatin, miboplatin, cisplatin, carboplatin, nedaplatin, dicycloplatin (dicycloplatin), leplatin (Lobaplatin), triplatin tetranitrate, phenanthriplatin, picoplatin, saplatin), fluoropyrimidine derivatives (e.g., gemcitabine, capecitabine, ancitabine, fluorouracil, bifurouracil, doxifluridine, tegafur, carmofur, trifluouridine), taxanes (e.g., paclitaxel, albumin-bound paclitaxel, and docetaxel), camptothecins and analogs thereof (e.g., camptothecins, hydroxycamptothecins, 9-aminocamptothecins, 7-ethylcamptothecins, irinotecan, topotecan), vinca (vinorelbine, vinblastine, vincristine, vindesine) vinflunine (vinflunine), norvinca alkaloid), anthracyclines (epirubicin, doxorubicin (doxorubicin), daunorubicin, pirarubicin, amrubicin, idarubicin, mitoxantrone, doxorubicin, valrubicin, zorubicin, pitaxadiol, doxorubicin), cytarabine, thioguanine, pemetrexed, carmustine, melphalan, etoposide, teniposide, mitomycin, ifosfamide, cyclophosphamide, azacytidine, methotrexate, bendamustine, pentoxifylline, carmustine, liposomal doxorubicin, actinomycin D (dactinomycin), bleomycin, pingamycin, temozolomide, amamide, pelomycin, pernicine, plinabulin (plinabulin), plilatin), sapacitabine, treosulfan, 153Sm-EDTMP, tiger, left-handed asparaginase, peganase, cephalotaxine, procarbazine, ARC-100, and encquidar.
In certain embodiments, the chemotherapeutic agent is one or more of the platinum group including, but not limited to, cisplatin, carboplatin, nedaplatin, oxaliplatin, miltiplatin, triplatin tetranitrate, phenanthreneplatin, picoplatin, satraplatin, leptoplatin, dicycloplatin (dicycloplatin), and the like.
Immunotherapeutic medicine
In the present application, the immunotherapeutic agent includes, but is not limited to, one or more of interferon (interferon alpha, interferon alpha-1 b, interferon alpha-2 b), interleukin, sirolimus (temsirolimus), everolimus (everolimus), everolimus (ridaforolimus), temsirolimus, lenalidomide.
Small molecule targeting antitumor drug
In the present application, the small molecule targeted antitumor drugs include, but are not limited to, protein kinase modulators. The action targets of the small molecule targeted antitumor drug include but are not limited to Fascin-1 protein, HDAC (histone deacetylase), proteasome, CD38, SLAMF7 (CS 1/CD 319/CRACC), proteasome, RANKL, EGFR (epidermal growth factor receptor), anaplastic Lymphoma (ALK), MET gene, ROS1 gene, HER2 gene, RET gene, BRAF gene, PI3K signal pathway, DDR2 (disc death receptor 2) gene, FGFR1 (fibroblast growth factor receptor 1), NTRK1 (neurotrophic tyrosine kinase type 1 receptor) gene and KRAS gene; targets for the small molecule targeted antitumor drug also include COX-2 (cyclooxygenase-2), APE1 (apE 1 (apurinic apyrimidinic endonuclease), VEGFR (vascular endothelial growth factor receptor), CXCR-4 (chemokine receptor-4), MMP (matrix metalloproteinase), IGF-1R (islet-like growth factor receptor), ezrin, PEDF (pigment epithelium derived factor), AS, ES, OPG (bone protecting factor), src, IFN, ALCAM (leukocyte activated adhesion factor), HSP, JIP1, GSK-3 (glycogen synthesis kinase 3 sugar), cyclin D1 (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 (CD 4), aurora a kinase, fusion proteins, PNP, cyclic depsipeptides, 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 inhibitors, serine and/or threonine kinase inhibitors, poly ADP ribose polymerase (PARP, poly ADP-ribose polymerase) inhibitors.
In some embodiments of the present invention, in some embodiments, the small molecule targeted antitumor drugs include, but are not limited to, imatinib, sunitinib, nilotinib (Nilotinib), bosutinib (bosutinib), celecoxib (Saracatinib), pazopanib, qu Bei-tide (Trabectedin), regafinib, ceridanib (celiranib), bortezomib, panobinostat (Panobinostat), carfilzomib), imatinib (ixazonib), apatinib, erlotinib, afatinib, crizotinib, ceritinib (Ceritinib), vemurafenib, dabrafenib (dabreninib), cabafatinib (cabozaninib), gefitinib, dacominib (Dacomitinib), oscitatinib (oscitatinib) Ai Leti (Alectinib), bragg, lagrantinib (Lorlatinib), trametinib (Trametinib), larotinib (Larotinib), ecotinib (icotinib), lapatinib (Lapatinib), vandatinib (Vandetanib), semetinib (Selumetinib), sorafenib (Sorafenib), omrotinib (Olmetinib), wo Liti (Savolitinib), fuscopinib (Fruquintinib), entriptinib (Entretinib), dasatinib (Dasatinib), ensartinib (Ensartinib), levalatinib (Lenvatinib), itacrinib, pyitinib), bimetinib (Binimet), erdaitinib (Erdaitinib), axitinib (Axitinib), cobicitinib (cobimatinib), acartinib (Acalabrutinib), famitinib (Famitinib), mosatinib (Masitinib), ibutetinib (ibutinib), rociletinib, nipanib (nintedanib), lenalidomide, LOXO-292, vorolanib, bemcentinib, capmatinib, entrectinib, TAK-931, ALT-803, palbociclib (palbociclib), famitinib L-map, LTT-462, BLU-667, ningetinib, tipifarnib, poziotinib, DS-1205c, capivasertib, SH-1028, metformin, seliciclib, OSE-2101, APL-101, berzosertib, idelalisib, lerociclib, ceralasertib, PLB-1003, tomivosertib, AST-2818, SKLB-1028, D-0316, LY-3023414, allitinib, MRTX-849, and combinations thereof AP-32788, AZD-4205, lifirafenib, vactosertib, mivebresib, napabucasin, sitravatinib, TAS-114, molibresib, CC-223, rivoceranib, CK-101, LXH-254, simotinib, GSK-3368715, TAS-0728, masitinib, tepotinib, HS-10296, AZD-4547, merestinib, olaptesedpegol, galunisertib, ASN-003, gedatolisib, defactinib, lazertinib, CKI-27, S-49076, BPI-9016M, RF-A-089, RMC-4630, AZD-3759, antroquinonol, SAF-189S, AT-101, TTI-101, antroquinonol, SAF-3794, HH-SCC-244, ASK-antroquinonol, SAF, CT-707, antroquinonol, SAF-1188-11, BPI-15000, antroquinonol, SAF, DV-281, antroquinonol, SAF-0250, GLG-801, ABTL-0812, bortezomib, antroquinonol, SAF-161, KML-001, ABTL-0812, bortezomib, belinostat (belinostat), pralatrexate (Pralatrexate), romidepsin (Romidepsin), cidamide (Chidamide), alisertib, duvelisib, denileukin-diftitox, forodesine, pitidepsin.
In some embodiments, the small molecule targeted anti-tumor drug is an ALK kinase inhibitor, and examples that may be cited include, but are not limited to, one or more of crizotinib, ceritinib, buzotinib, lazotinib, aletinib (alecitib), buginib, and ensartinib.
In some embodiments, the small molecule targeted anti-tumor drug is one or more of Bomacinib (abemaciclib), venetoclax, fenretinide, ABTL-0812, OTS-167, SF-1126, nifurtimox, eflornithine, ABTL-0812, CBL-0137, ONC-201, fadraciclib, LY-3295668 erbimine, rose bengal sodium salt (rose bengal sodium), venetoclax, copanlisib, tamibarotene, latifinib, ensartinib, entrectinib.
Antitumor antibody medicament
In the present application, the target point 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 (cytotoc T-lymphocyte antigen, ctla-4), platelet-derived growth factor receptor alpha (PDGFR- α), vascular Endothelial Growth Factor (VEGF), human epidermal growth factor receptor-2 (HER 2), 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 agents of the application are inhibitors of the CD20 (Cluster ofDifferentiation 20) antigen; in some embodiments, the anti-tumor antibody drug of the application is an MDM2 inhibitor; in some embodiments, the anti-tumor antibody drug of the application 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 (cytoxic 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 may be selected from any one or more of nano Wu Liyou mab (Nivolumab), palbociclizumab (Pembrolizumab), dewaruzumab (Durvalumab), terlipp Li Shan antibody (toripalimab, JS-001), singdi Li Shan antibody (IBI 308), karelizumab (Camrelizumab), tirelimumab (BGB-a 317), jenocarzumab (GB 226), lizumab (LZM 009), HLX-10, BAT-1306, AK103 (HX 008), AK104 (Kang Fang 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 (ZKAB 0011), 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 (Ipilimumab), tiximumab (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-pdgfrα antibody is an anti-pdgfrα monoclonal antibody. In some specific embodiments, the anti-PDGFR alpha antibody is olamumab (olokaumab).
In some specific embodiments, the anti-tumor antibody agent includes, but is not limited to, any one or more of naxitamab, omburtamab, OGD-201, APN-301, racotumomab, rituximab, dinutuximab beta, enoblituzumab, CD antagonist nivatronatamab.
A compound of formula I or a pharmaceutically acceptable salt thereof
The chemical name of the compound of formula I is 1- [ [ [4- (4-fluoro-2-methyl-1H-indol-5-yl) oxy-6-methoxyquinolin-7-yl ] oxy ] methyl ] cyclopropylamine having the structural formula:
in the present application, all the compounds of formula I refer to An Luoti Ni.
The compounds of formula I may be administered in its free base form, or in the form of salts, hydrates and prodrugs thereof, which are converted in vivo to the free base form of the compounds of formula I. For example, pharmaceutically acceptable salts of the compounds of formula I are within the scope of the application, which salts may be produced from different 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. In some embodiments, the compound of formula I is administered as the dihydrochloride salt. In some embodiments, the compound of formula I is administered in crystalline form as the hydrochloride salt. In certain embodiments, the compound of formula I is administered in crystalline form as the dihydrochloride salt of the compound.
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 the group consisting of: oral, parenteral, intraperitoneal, intravenous, intraarterial, transdermal, sublingual, intramuscular, rectocele, buccal, intranasal, inhalation, vaginal, intraocular, topical, subcutaneous, intrafat, intra-articular, intraperitoneal and intrathecal. In a particular embodiment, the administration is by oral administration.
The amount of the compound of formula I, or a pharmaceutically acceptable salt thereof, the second therapeutic agent administered may be determined based on the severity of the disease, the response of the disease, any treatment-related toxicity, the age and health of the patient.
In some embodiments, the daily dose of the compound of formula I, or a pharmaceutically acceptable salt thereof, is from 3 mg to 30 mg. In some embodiments, the daily dose of the compound of formula I, or a pharmaceutically acceptable salt thereof, is 5 mg to 20 mg. In some embodiments, the daily dose of the compound of formula I, or a pharmaceutically acceptable salt thereof, is 8 mg to 16 mg. In some embodiments, the daily dose of the compound of formula I, or a pharmaceutically acceptable salt thereof, is from 10 mg to 14 mg. In a particular embodiment, the daily dose of the compound of formula I or a pharmaceutically acceptable salt thereof is 8 mg. In a particular embodiment, the daily dose of the compound of formula I or a pharmaceutically acceptable salt thereof is 10 mg. In a particular embodiment, the daily dose of the compound of formula I or a pharmaceutically acceptable salt thereof is 12 mg. In the present application, for example, for tablets or capsules, "containing 12mg of the compound of formula I on a unit dose basis" means that each tablet or capsule finally produced contains 12mg of the compound of formula I.
In some embodiments, the administration of a compound of formula I or a pharmaceutically acceptable salt thereof may also be calculated from the body surface area of the patientAmount of the components. In some embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is present at 4-10mg/m 2 Is administered in a daily dose of (a); as will be appreciated by those skilled in the art, if the body surface area of the patient is 1m 2 If present, the compound of formula I or a pharmaceutically acceptable salt thereof is administered in an amount of 4-10mg. In some embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is present at 5-8mg/m 2 Is administered in a daily dose of (a); in some embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is present at 5-7mg/m 2 Is administered in a daily dose of (a); in some embodiments, the compound of formula I or a pharmaceutically acceptable salt thereof is present at 4, 5, 6, 7, 8mg/m 2 Is administered in a daily dose.
The compound of formula I, or a pharmaceutically acceptable salt thereof, and the second therapeutic agent may be administered one or more times daily. In some embodiments, the compound of formula I, or a pharmaceutically acceptable salt thereof, is administered once daily. In one embodiment, the solid dosage form is administered once daily as an oral solid dosage form.
In the above-mentioned therapeutic methods, the method of administration may be comprehensively determined according to the activity, toxicity, tolerance of the patient, and the like of the drug. Preferably, the compound of formula I or a pharmaceutically acceptable salt thereof is administered in a divided dosing regimen. The interval administration includes a dosing period and a withdrawal period, during which the compound of formula I or a pharmaceutically acceptable salt thereof may be administered one or more times per day. For example, the compound of formula I or a pharmaceutically acceptable salt thereof may be administered daily during a dosing period followed by a period of discontinuance of the dosing during a dosing period, followed by a dosing period, and then a dosing period, which may be repeated a number of 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 more preferably 2:0.5-1.
In some embodiments, administration is stopped for 2 weeks following 2 weeks of administration. In some embodiments, administration is 1 time per day for 14 days, followed by 14 days of discontinuation; the administration is then continued for 14 days and then for 14 days, 1 time per day, and the administration is repeated at intervals of 2 weeks for 2 weeks.
In some embodiments, administration is stopped 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 then continued for 14 days and then for 7 days 1 time per day, and the administration is repeated at intervals of 1 week for 2 weeks.
In some embodiments, administration is stopped for 2 days 5 days continuously. In some embodiments, administration is 1 time per day for 5 days, followed by 2 days of discontinuation; the administration is then continued 1 time per day for 5 days and then stopped for 2 days, so that the administration mode of stopping for 2 days at intervals of 5 days in succession can be repeated for a plurality of times.
In certain specific embodiments, the administration is orally at a dose of 12mg once daily for 2 weeks, followed by 1 week of administration.
Combined medicine
The components of the combination according to the application may optionally be combined with one or more pharmaceutically acceptable carriers, wherein the components may each independently, or some or all of them together comprise a pharmaceutically acceptable carrier and/or excipient. The combination according to the application may be formulated separately from each other or some or all of them may be formulated together. Preferably, the components of the combination are formulated separately or each as a suitable pharmaceutical composition. In some embodiments, the combination of the present application may be formulated into 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 application may each be administered alone or some or all of them may be co-administered. The components of the combination of the present application may be administered substantially simultaneously, or some or all of them may be administered substantially simultaneously.
The components of the combination of the application may each be administered independently, or some or all of them together, by suitable routes including, but not limited to, oral or parenteral (via intravenous, intramuscular, topical or subcutaneous routes). In some embodiments, the components of the combination of the application may each be administered orally or parenterally, e.g., intravenously or intraperitoneally, independently, or in combination with some or all of them.
The components of the combination of the present application may each independently, or some or all of them, be formulated into 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 sustained release formulations for oral or non-oral administration.
In some aspects of the 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 application 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 use in the treatment of neuroblastoma.
The application 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 the treatment of neuroblastoma in a divided dosing regimen. In some particular 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 the treatment of neuroblastoma in a divided dosing regimen. In some more typical embodiments, the neuroblastoma is a neuroblastoma that failed treatment with a chemotherapeutic and/or targeted drug. By "unit dose" is meant a pharmaceutical composition packaged in a single package for convenience of administration. Such as each tablet or capsule.
In some aspects of the 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 application, a 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 have been administered to the patient prior to the 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 the one or more second therapeutic agents are administered sequentially to the patient.
In some embodiments, there is also provided a kit of parts for use in the treatment of neuroblastoma comprising: (a) A pharmaceutical composition comprising as an active ingredient a compound of formula I or a pharmaceutically acceptable salt thereof; and (b) a pharmaceutical composition containing a chemotherapeutic agent as an active ingredient. In some embodiments, there is also provided a kit of parts for use in the treatment of neuroblastoma comprising: (a) A pharmaceutical composition comprising as an active ingredient a compound of formula I or a pharmaceutically acceptable salt thereof; the method comprises the steps of carrying out a first treatment on the surface of the 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 for use in a pharmaceutical composition for treating neuroblastoma, comprising: (a) A pharmaceutical composition comprising as an active ingredient a compound of formula I or a pharmaceutically acceptable salt thereof; and (b) a pharmaceutical composition containing an immunotherapeutic agent as an active ingredient. In some embodiments, there is also provided a kit for use in the treatment of a pharmaceutical combination of neuroblastoma comprising (a) a pharmaceutical composition comprising as an active ingredient a compound of formula I or a pharmaceutically acceptable salt thereof; and (b) a pharmaceutical composition containing the antibody drug as an active ingredient.
In some embodiments, there is also provided a kit of parts for use in the treatment of 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 containing an alkylating agent and/or a camptothecin and analogues thereof as active ingredients; in some embodiments, there is also provided a kit of parts for use in a pharmaceutical composition for the treatment of 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 containing temozolomide and/or camptothecin and analogues thereof as active ingredients; in some embodiments, there is also provided a kit of parts for use in a pharmaceutical composition for the treatment of 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 containing temozolomide and/or irinotecan as active ingredients.
The dosages and ranges provided herein are based on the molecular weight of the free base form of the compound of formula I, unless otherwise indicated.
For the purposes of the present application, the following terms, as used in the specification and claims, shall have the following meanings, unless otherwise indicated.
In the present application, all the compounds of formula I refer to An Luoti Ni.
"patient" means a mammal, preferably a human.
By "pharmaceutically acceptable" is meant that it is used to prepare a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes that it is acceptable for human pharmaceutical use.
"pharmaceutically acceptable salts" include, but are not limited to, acid addition salts with inorganic acids such as hydrochloric, hydrobromic, sulfuric, nitric, phosphoric and the like; or with organic acids such as acetic acid, trifluoroacetic acid, propionic acid, caproic acid, heptanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1, 2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, p-chlorobenzenesulfonic acid, p-toluenesulfonic acid, 3-phenylpropionic acid, trimethylacetic acid, t-butylacetic acid, dodecylsulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, and the like.
"treatment" means any administration of a therapeutically effective amount of a compound and includes:
(1) Inhibiting the disease in a human experiencing or exhibiting the pathology or symptomology of the disease (i.e., arresting further development of the pathology and/or symptomology), or
(2) Improving the disease in a human experiencing or exhibiting the pathology or symptomology of the disease (i.e., reversing the pathology and/or symptomology).
"failure of treatment" refers to the intolerance of toxic and side effects after dosage adjustment and the short-term development of disease progression during treatment. Intolerance includes, but is not limited to, the level of hematologic toxicity remaining after dose adjustment reaching level IV (thrombocytopenia level III and above), and the level of non-hematologic toxicity remaining reaching level III or above.
"CR" refers to complete remission. "PR" refers to partial relief. "PD" refers to disease progression. "SD" refers to disease stabilization.
By "about" in the present application is meant that the range of values given varies within + -5%, preferably within + -2%, more preferably within + -1%.
In this document, the terms "comprises," "comprising," and "includes," or equivalents thereof, unless otherwise specified, are open ended and mean that other unspecified elements, components, and steps are contemplated in addition to those listed.
"First-line treatment" or "First-line treatment regimen" as used herein is used interchangeably and refers to the initial, or First treatment regimen recommended to treat a disease; for cancer, first line therapy generally has a number of alternative treatment regimens.
In the application, an Luoti 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 retardation and promote the apoptosis. The growth of the neuroblastoma mouse model tumor is obviously inhibited, the curative effect is dose-dependent, and the high-dose safety is good. 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 capacity of the CD4 and CD 8T cells to secrete INF-gamma and TNF-alpha, improve the expression of MHC-I of the neuroblastoma tumor cells, namely the killing capacity of the CD4 and CD 8T cells, and enhance the antigen presenting function of the tumor cells. An Luoti Ni significantly up-regulates T cell activation, antigen presentation, etc. immune activation related genes and significantly down-regulates angiogenesis related genes.
All patents, patent applications, and other identified publications are expressly incorporated herein 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 or representation as to the contents of these documents are based on the information available to the applicant and do not constitute any admission as to the correctness of the dates or contents of these documents. Moreover, any reference to such publications in this document does not constitute an admission that the publications are part of the common general knowledge in the art, in any country.
Drawings
FIG. 1 inhibition of proliferation of neuroblastoma cell line by An Luoti Ni
FIG. 2 inhibition of the clonogenic effects of neuroblastoma cell lines by An Luoti Ni
FIG. 3 inhibition of neuroblastoma cell cycle by An Luoti Ni
FIG. 4 An Luoti Ni effect on promotion of apoptosis in neuroblastoma cell lines
FIG. 5 inhibition of neuroblastoma cell line migration by An Luoti Ni
FIG. 6 inhibition of neuroblastoma cell line invasion by An Luoti Ni
FIG. 7A An Luoti Ni inhibitory effect on mouse neuroblastoma
FIG. 7B An Luoti Ni inhibition of mouse neuroblastoma
FIG. 8A An Luoti Ni shows the activation of the mouse neuroblastoma immune microenvironment
FIG. 8B An Luoti Ni activation of the mouse neuroblastoma immune microenvironment
Detailed Description
The application will be further illustrated with reference to specific examples. It is understood that these examples are provided only for illustrating the present application and are not intended to limit the scope of the present application.
Example 1
1.1 Material cell lines 9464D and 975A2 were donated by Dr. Rimas Orentas (Seattle Children's Research Institute), both of which were derived from N-myc expanded C57BL/6 mice, which mimic the pathogenesis of neuroblastoma in the adrenal gland, and normal mouse immune function, largely mimicking the actual pathogenic background of the patient. Human neuroblastoma cell lines IMR-32 and SK-N-BE (2) were purchased from American type culture Collection (American Type Culture Collection, ATCC). C57BL/6 mice were purchased from Beijing Bei Fu Biotechnology Co.
1.2 reagents MEM (Medium), DMEM (Medium) and R1640 Medium were purchased from Gibco corporation of America. 0.25%, 0.05% trypsin was purchased from Gibco corporation of America. Fetal bovine serum was purchased from israel BI company. CCK8 kit, cell cycle kit and apoptosis kit were purchased from Beijing Soy Bao Biotechnology Co. Transwell cells (24-well plate cells, pore size 8 μm) and Mtrigel matrigel were purchased from Corning Inc. of America. Fluorescent labeled antibodies were purchased from BioLegend, usa, fluorescent assay primary antibodies were purchased from Abcam, uk, and secondary antibodies were purchased from CST, usa.
1.3 instrument constant temperature incubator (Thermo company, usa), flow cytometer (BD company, usa), inverted fluorescence microscope (Olympus company, japan), upright fluorescence microscope (Nikon company, thermo company, usa), enzyme-labeled instrument (Thermo company, germany), high-speed refrigerated centrifuge (Eppendorf company, germany), frozen microtome (Leica company, switzerland), refrigerator (Haier company, china), 80 ℃ low temperature refrigerator (Haier company, liquid nitrogen tank (Thermo company, usa), ultra clean bench (ESCO company, new slope).
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 were incubated at 37℃in a 5% CO2 incubator. When the cell fusion degree is about 80-90%, 9464D is digested with 0.25% pancreatin for 2 min, 975A2 is digested with 0.05% pancreatin for 2 min, 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 ℃ for 5min at 1000r/min, and subcultured.
1.4.2 cell proliferation assay (CCK 8 assay) 9464D, 975A2, IMR-32 and SK-N-BE (2) cells in the logarithmic growth phase were collected, counted, added to 96-well plates at 3500/100. Mu.L/well, cultured for 24 hours to adhere the cells, then treated with An Luoti Ni (0.25, 0.5, 1, 2, 4, 8, 16, 32, 64. Mu. Mol/L) at different concentrations for 24 hours, then 10. Mu.L of CCK8 solution was added to each well, the plates were incubated in an incubator for 1-4 hours, absorbance values at 450nm were measured with a microplate reader, and the cell inhibition was calculated.
1.4.3 plate cloning experiments the collected 9464D, 975A2, IMR-32 and SK-N-BE (2) cell pellets were resuspended and counted using the corresponding medium and each cell was seeded in 6-well plates at a cell density gradient of 50, 100, 200 cells per well. Cells were treated with An Luoti Ni (0, 1, 2. Mu. Mol/L) at various concentrations and cultured 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 was discarded, washed twice with PBS, and stained with 0.1% crystal violet stain for 15 minutes per well. Discarding the staining solution, washing twice with PBS, naturally airing, and photographing. The number of clone formations was counted visually, and the clone formation rate was calculated.
1.4.4 cell cycle experiments 9464D, 975A2, IMR-32 and SK-N-BE (2) cells were treated with different concentrations of An Luoti Ni (0, 1, 2, 4. Mu. Mol/L) respectively for 24 hours, and after harvesting the cells, the cells were 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 that, 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 that, PBS was washed once, 250. Mu.LPI (propidium iodide containing RNase) was added thereto, and the mixture was subjected to a light-shielding water bath at 37℃for 30 minutes. And can be stored at 4 ℃ in a dark place. Detection was performed using a flow cytometer for 24 hours. The analysis was performed using Flowjo software and the phase ratio was calculated for the cell cycle. The experiment was repeated three times.
1.4.5 apoptosis experiments 9464D, 975A2, IMR-32 and SK-N-BE (2) cells were treated with different concentrations of An Luoti Ni (0, 1, 2, 4. Mu. Mol/L) for 24 hours, respectively, and after harvesting the cells, the cells were centrifuged at 1000r/min for 5min and washed twice with PBS. The supernatant was discarded, 250. Mu.L of 1XAnnexinV Binding Buffer working solution was added to resuspend the cells, 2.5. Mu.L of Annexin V-APC and 2.5. Mu.L of PI staining solution were added to the cell suspension, and incubation was performed at room temperature for 15-20 minutes in the dark. Immediately after completion of the reaction, the reaction was examined by flow cytometry. The apoptotic cell fraction was calculated by analysis using Flowjo software. The experiment was repeated three times.
1.4.6 cell scratch experiments the collected 9464D, 975A2, IMR-32 and SK-N-BE (2) cell pellets were resuspended and counted using the corresponding medium, 6X 10 plates were plated per well 5 The individual cells were cultured in a constant temperature incubator. When the cell fusion rate reached 100%, the medium was discarded, scored with 200 μl sterile gun head perpendicular to the 6-well plate, and then washed twice with PBS. PBS was removed, serum-free medium was added, and the cells were treated with 1. Mu. Mol/L An Luoti Ni and placed in an incubator for further culture. Photographs were taken at fixed positions at 0, 24, 48 hours after scoring, 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 experiments Using manufacturer recommended DiluentThe Mtrigel matrix collagen was diluted (ice working), 100. Mu.L of the diluted solution was added to the upper chamber of the Transwell chamber, and the mixture was left in the incubator for 1 to 4 hours, after which the upper liquid medium was discarded. The collected 9464D, 975A2, IMR-32 and SK-N-BE (2) cell pellets were resuspended and counted 1X 10 in the corresponding serum-free medium 5 mu.L/well was added to the upper chamber and 600. Mu.L of the medium corresponding to 20% fetal bovine serum was added to the lower chamber. The Transwell chamber and 24-well plate contained An Luoti Ni (4. Mu. Mol/L) at the same concentration. Culturing 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 was added to the lower chamber to fix the cells for 15 minutes. The paraformaldehyde was discarded and washed twice with PBS. 600. Mu.L of 0.1% crystal violet staining solution is added to the lower chamber for 1 minute, PBS is used for washing twice, cells in the upper chamber are wiped off by a cotton stick, and the chamber is naturally air-dried. 5 visual fields are randomly taken from each cell under a microscope for photographing, and the cell invasion number is calculated by adopting ImageJ software for cell counting. The experiment was repeated three times.
1.4.8 mice subcutaneous Oncomelania animal experiments C57BL/6 mice of 5-6 weeks of age were selected for feeding, and were subjected to subcutaneous Oncomelania using two murine N-myc expanded neuroblastoma cell lines, 9464D and 975A 2. First, a tumor cell suspension was prepared at a rate of 1X 10 6 Cells/mouse were inoculated subcutaneously on the right back of mice diluted to 100 μl. After about 3-4 weeks after tumor inoculation, the tumor volume and the weight of the mice are measured every 3 days after the subcutaneous tumor volume can be measured, and a tumor growth curve is drawn according to 4 groups of volume fractions, namely a control group, a An Luoti-Ni treatment group, a Bomactinib treatment group and an Anrotinib combined Bomactinib treatment group, wherein each group comprises 6 mice, and then the stomach is irrigated for 21 days. After stopping the treatment, the mice were sacrificed by cervical dislocation, the surgical instruments were removed from the subcutaneous tumors and spleen, the tumors were weighed, photographed, recorded, and total spleen cell counts were performed. Statistics and mapping were performed using GraphpadPrism8 software.
1.4.9 flow cytometry analysis of tumor immune microenvironment after 21 days of treatment, mice were sacrificed in cervical dislocation, subcutaneous tumor tissue and spleen were removed with sterile surgical instruments and treated as single cell suspensions, and finally stained with fluorescent-labeled antibodies (CD 3, CD4, CD8, CD11b, CD11c, CD19, CD25, CD40, CD45, gr1, F4/80, NK1.1, NKp46, foxP3, PD1, ki67, tnfα, ifny, gzmB, H2, β2m, mult1, pdl 1). After completion of the reaction, detection was performed using a flow cytometer. And (3) analyzing by using Flowjo software, and calculating the ratio of various immune cells.
2. Experimental results
2.1 An Luoti Ny significantly inhibits neuroblastoma cell proliferation
The experimental results of CCK8 show that the higher the An Luoti Ni concentration, the higher the cell inhibition rate. And the inhibition rate of cells by An Luoti Ni at the same concentration was also enhanced with the time. The above results demonstrate that inhibition of the 9464D, 975A2, IMR-32 and SK-N-BE (2) cell lines by An Luoti is concentration and time dependent. The results of the plate cloning experiments show that An Luoti Ni can significantly inhibit the cloning of neuroblastoma cell lines (see FIG. 1 and FIG. 2).
IC 50 Cell line | 975A2 | 9464D | IMR-32 | SK-N-BE(2) |
24h (micromolar) | 23.49 | 15.72 | 24.71 | 23.27 |
48h (micromolar) | 6.683 | 4.091 | 6.452 | 6.396 |
72h (micromolar) | 2.770 | 2.051 | 2.514 | 2.320 |
2.2 An Luoti Ny significantly blocked the neuroblastoma cell cycle
Experimental results of An Luoti ni on the effects of 9464D, 975A2, IMR-32 and SK-N-BE (2) cell line cell cycle showed that An Luoti ni can significantly induce the neuroblastoma cell line to develop G2/M cycle arrest, and that the cell cycle arrest was stronger with increasing An Luoti ni concentration (fig. 3).
2.3 An Luoti Ny significantly promotes apoptosis of neuroblastoma cells
The effects of An Luoti on apoptosis of 9464D, 975A2, IMR-32 and SK-N-BE (2) cell lines were examined by flow cytometry. The results showed that An Luoti ni significantly promoted apoptosis in neuroblastoma cell lines, and that the higher the apoptosis rate with increasing concentration of amrotinib (fig. 4).
2.4 An Luoti Ny significantly inhibited neuroblastoma cell migration
The results of the scratch experiments showed that the migration capacity of 9464D, 975A2, IMR-32 and SKN-BE (2) cell lines were all significantly reduced after An Luoti Ni treatment, and the more significant the difference in cell mobility from the control group over time (FIG. 5).
2.5 An Luoti Ny significantly inhibits neuroblastoma cell invasion
Transwell cell invasion assay results showed that the invasion capacities of 9464D, 975A2, IMR-32 and SK-N-BE (2) cell lines were significantly reduced after An Luoti Ni treatment (FIG. 6).
2.6 An Luoti Nib significantly inhibited growth of mouse subcutaneous neuroblastoma
The experimental result of the mouse subcutaneous tumor-forming animal shows that An Luoti Ni obviously inhibits the growth of mouse subcutaneous neuroblastoma and reduces tumor load. The targeted drug, bomacinib, can enhance the therapeutic effect of An Luoti on neuroblastoma (fig. 7A and 7B), wherein NS refers to the control group, abe refers to Bomacinib, and Anlo refers to An Luoti.
2.7 An Luoti Nile significantly activates the neuroblastoma immune microenvironment
Flow cytometry analysis showed that An Luoti ni significantly increased the percentage of CD4 and CD 8T cell infiltration in mouse neuroblastoma and spleen and reduced the number of MDSCs. And simultaneously, the capacity of secreting INF-gamma and TNF-alpha of the CD4 and CD 8T cells is enhanced, namely, the killing capacity of the CD4 and CD 8T cells is enhanced, and the targeted drug, namely, the Bomacinib, can enhance the activation of An Luoti on the immune microenvironment of the neuroblastoma (fig. 8A and 8B).
EXAMPLE 2 clinical study treatment-refractory or recurrent neuroblastoma in children who had previously received first-line therapy
Group entry criteria:
distant metastasis or locally advanced and researchers judge neuroblastoma subjects unsuitable 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 were treated with at least one chemotherapeutic regimen (anthracycline-containing) and evaluated as disease progression or intolerance according to the efficacy evaluation criteria for solid tumors (RECIST 1.1); age of patients in group: under 21 years old, it can be used by both men and women; PS score: 0-1 (amputation subjects can relax to 2 minutes), with expected survival exceeding 12 weeks; patients who have previously received irinotecan or combination temozolomide without progression.
Specific dosing regimen: an Luoti Ni 7mg/m hydrochloric acid 2 The medicine is orally taken for 1 time a day, and is continuously taken for two weeks for stopping taking medicine for one week, namely d1-14 is taken, and every 3 weeks (21 days) is a treatment period;irinotecan 50mg/m 2 D 1-5, i.v.; temozolomide 100mg/m 2 D 1-5, and is orally taken. Every 3 weeks is a treatment cycle.
The main research index is as follows: an Luoti Ni hydrochloride and irinotecan in combination with temozolomide treat Progression Free Survival (PFS) of childhood refractory or recurrent neuroblastoma.
Secondary index: objective remission rate (ORR=CR+PR), disease control rate (DCR=CR+PR+SD), total survival (OS), quality of life score, safety index (adverse event AE) of An Luoti Nib hydrochloride, irinotecan in combination with temozolomide treatment were observed and evaluated.
Tumor imaging evaluation was performed according to RECIST v1.1, subject had to be accompanied by measurable tumor lesions at baseline. Efficacy assessment results were based on RECIST 1.1 criteria, including: complete Remission (CR), partial Remission (PR), stable (SD) and Progressive (PD), PFS, OS.
After the patient is aged 5 years for 7 months, the right retroperitoneal neuroblastoma is diagnosed for more than 1 year, 5 cycles of chemotherapy are performed before, the general anesthesia is performed, the right retroperitoneal tumor resection, the right nephrectomy and the bilateral retroperitoneal lymph node cleaning are performed, and the postoperative is performed for multiple cycles to assist the chemotherapy and the abdominal tumor area radiotherapy. After radiotherapy is finished, the infant has recurrent disease, blood NSE (neuron specific enolase)
393.2ug/L, significantly higher than normal, 55% of bone marrow anti-GD 2 staining positive cells, and cranium touching multiple bread blocks. Review PET-CT shows: the thickening of the soft tissues in the original operation area is increased more forward, the lymph nodes in the bilateral lock areas are metastasized, bone metastasis is multiple, subcutaneous nodules are multiple on the head, and the metastasis is suspected. The disease infant has 4 recurrent periods after the operation of the retroperitoneal neuroblastoma, and the diagnosis of the high-risk group is clear. After the chemotherapy is continued for 5 periods, the abdominal tumors of the children patients are not continuously reduced, NSE has a rising trend, the median PFS of the children patients with refractory or recurrent neuroblastoma is judged to be 3.3 months according to the previous study, the high risk of tumor recurrence is considered, the treatment is combined with An Luo tenny anti-vascular treatment for 5 periods, the NSE is regularly rechecked and is stable after being reduced, the optimal curative effect PR (reduced by 37%) is maintained, and the PFS is up to more than 8 months after the date of the special application is stopped.
Claims (9)
1. 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,
formula I.
2. The use of claim 1, wherein said pharmaceutical composition further comprises at least one second therapeutic agent.
3. Use of a combination pharmaceutical composition for the manufacture of a medicament for the treatment of neuroblastoma, wherein the combination pharmaceutical composition comprises: (I) a compound of formula I or a pharmaceutically acceptable salt thereof;
the compound of the formula I,
and (ii) at least one second therapeutic agent.
4. The use of any one of claims 1-3, wherein the neuroblastoma is advanced and/or metastatic neuroblastoma.
5. The use of any one of claims 1-3, wherein the neuroblastoma is a recurrent neuroblastoma.
6. The use of any one of claims 1-3, wherein the neuroblastoma is a high-risk neuroblastoma.
7. The use of any one of claims 1-3, wherein the neuroblastoma is a neuroblastoma that failed first line therapy.
8. The use of any one of claims 2-3, wherein the second therapeutic agent is selected from irinotecan and temozolomide.
9. The use of any one of claims 2-3, wherein the second therapeutic agent is selected from the group consisting of bomacenib.
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 CN113116895A (en) | 2021-07-16 |
CN113116895B true 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) |
Families Citing this family (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CA3232304A1 (en) * | 2021-09-26 | 2023-03-30 | Chen SHEN | Use of anti-cd40 antibody |
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 |
Citations (4)
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 |
-
2020
- 2020-12-31 CN CN202011629017.5A patent/CN113116895B/en active Active
Patent Citations (5)
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)
Title |
---|
Targeting angiogenesis for controlling neuroblastoma",subhasree roy choudhury et al.;subhasree roy choudhury et al.;《Journal of Oncology》;20110802;第2012卷;第7页左栏第4-5段 * |
Also Published As
Publication number | Publication date |
---|---|
CN113116895A (en) | 2021-07-16 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US20240293408A1 (en) | Methods of treating and preventing graft versus host disease | |
JP7506981B2 (en) | Combination drugs containing TLR7 agonists | |
RU2765997C2 (en) | Combination of anti-pd-l1 antibody and dna-pk inhibitor for treating malignant neoplasm | |
JP2019536805A (en) | Protecting the immune response during chemotherapy planning | |
JP2013543879A (en) | Volaceltiv alone or in combination with cytarabine to treat acute myeloid leukemia | |
US20180353602A1 (en) | Combination of hdac inhibitor and anti-pd-l1 antibody for treatment of cancer | |
TWI697329B (en) | Medicament for use in the treatment of a hematological cancer and use thereof | |
CN113116895B (en) | Quinoline derivatives for the treatment of neuroblastoma | |
CN114224889A (en) | Application of Xiaorony combined immune checkpoint inhibitor in antitumor therapy | |
CN113710658A (en) | Quinolines or pharmaceutically acceptable salts thereof for the treatment of ewing's sarcoma | |
CN118267469A (en) | Combined pharmaceutical composition for treating driving gene positive lung cancer | |
WO2020233602A1 (en) | Quinoline derivative used for combination treatment of small cell lung cancer | |
KR20210019422A (en) | Cancer treatment method | |
AU2023202746A1 (en) | Combination of a BCL-2 inhibitor and a MCL-1 inhibitor, uses and pharmaceutical compositions thereof | |
JP2021178825A (en) | Combination therapy with anti-her2 antibody-drug conjugate and bcl-2 inhibitor | |
WO2023034336A2 (en) | Improved treatments for advanced/metastatic cancers with checkpoint inhibitor resistance or resistance susceptibility | |
US11419862B2 (en) | Quinoline derivative for treatment of nasopharyngeal carcinoma | |
KR20200078483A (en) | Composition and method for treating liver cancer | |
CN101534860B (en) | Compositions and methods using anti-cs1 antibodies to treat multiple myeloma | |
CN114469949A (en) | Quinoline derivatives for the treatment of diffuse large B-cell lymphoma | |
WO2016172053A1 (en) | Methods of treating ibrutinib-resistant disease | |
CN113766917A (en) | Quinoline derivatives for the treatment of head and neck cancer | |
CN111821302A (en) | Quinolines for the combined treatment of chondrosarcoma | |
US20240150487A1 (en) | Combinations with an anti human cd39 antibody, an anti human pd-1 antibody, and chemotherapy in gastric cancer | |
RU2802962C2 (en) | Compositions and methods of treatment of liver cancer |
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 |