CN113117072A - Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody - Google Patents

Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody Download PDF

Info

Publication number
CN113117072A
CN113117072A CN202110026242.8A CN202110026242A CN113117072A CN 113117072 A CN113117072 A CN 113117072A CN 202110026242 A CN202110026242 A CN 202110026242A CN 113117072 A CN113117072 A CN 113117072A
Authority
CN
China
Prior art keywords
treatment
antibody
seq
dose
human
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202110026242.8A
Other languages
Chinese (zh)
Inventor
马益华
黄小换
廉凯洋
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
Original Assignee
Chia Tai Tianqing Pharmaceutical Group Co Ltd
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 filed Critical Chia Tai Tianqing Pharmaceutical Group Co Ltd
Publication of CN113117072A publication Critical patent/CN113117072A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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

Landscapes

  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Mycology (AREA)
  • Endocrinology (AREA)
  • Oncology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

There is provided a pharmaceutical combination of a quinoline derivative and PD-1 mab which comprises a tyrosine kinase inhibitor and an immune checkpoint inhibitor, wherein the tyrosine kinase inhibitor is a compound of formula I or a pharmaceutically acceptable salt thereof. The medicine composition has good anti-esophageal cancer activity.

Description

Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody
Technical Field
The application belongs to the technical field of medicines and relates to a combined treatment for resisting tumors. In particular, the application relates to a combination based on quinoline derivatives and PD-1 monoclonal antibody and application thereof in anti-esophageal cancer.
Background
Tyrosine kinases are a group of enzymes catalyzing phosphorylation of tyrosine residues of proteins, play important roles in signal transduction in cells, participate in regulation, signal transmission and development of normal cells, and are closely related to proliferation, differentiation, migration and apoptosis of tumor cells. Many receptor tyrosine kinases are involved in tumor formation and are classified into Epidermal Growth Factor Receptor (EGFR), platelet-derived growth factor receptor (PDGFR), Vascular Endothelial Growth Factor Receptor (VEGFR), Fibroblast Growth Factor Receptor (FGFR), and the like according to their extracellular domain structures.
Anlotinib (Anlotinib) is a quinoline derivative tyrosine kinase inhibitor, and plays a role in influencing tumor angiogenesis and proliferation signal transduction as a multi-target Tyrosine Kinase Inhibitor (TKI), and main targets comprise: receptor tyrosine kinases Vascular Endothelial Growth Factor Receptors (VEGFR)1 to 3, Epidermal Growth Factor Receptors (EGFR), Fibroblast Growth Factor Receptors (FGFR)1 to 4, Platelet Derived Growth Factor Receptors (PDGFR) α and β, and Stem Cell Factor Receptors (SCFR)7, 8, and 9. A phase 2 trial showed that erlotinib improved progression-free survival with potential benefits for overall survival (Han B, et al Br J cancer.2018; 118 (5): 654-. A multicenter, double-blind, phase 3 randomized clinical trial showed that, among Chinese patients, Arotinib resulted in prolonged overall and progression-free survival, which indicated that Arotinib was well tolerated and is a potential three-line or further treatment for advanced NSCLC patients (Han B, et al, JAMA Oncol.2018 Nov; 4 (11): 1569-.
Document WO2008112407 discloses in example 24 a quinoline derivative tyrosine kinase inhibitor 1- [ [ [4- (4-fluoro-2-methyl-1H-indol-5-yl) oxy-6-methoxyquinolin-7-yl ] oxy ] methyl ] cyclopropylamine and a process for its preparation, which has the formula shown in formula I:
Figure BDA0002890379420000011
PD-1(programmed death-1) is a key immune checkpoint receptor expressed by activated T and B lymphocytes and mediates immunosuppression, and its ligands include at least PD-L1 and PD-L2. PD-L1(Programmed death-ligand 1), also known as CD274 or B7-H1, is a 40kDa type 1 transmembrane protein encoded by the CD274 gene and is a ligand for PD-1. Both PD-L1 and PD-1 belong to the immunoglobulin superfamily and both consist of two extracellular Ig domains, an N-terminal V domain and a C-terminal constant domain. The binding interface of PD-L1 to programmed death receptor-1 (PD-1) and B7-1(CD80) is on an IgV-like domain (Lin et al (2008) PNAS 105: 3011-3016). PD-L1 contains a conserved short intracellular tail region (about 30 amino acids), and PD-1 contains two cytoplasmic tyrosine-based signaling motifs, an immunoreceptor tyrosine-based inhibitory motif (ITIM) and an immunoreceptor tyrosine-based switching motif (ITSM). Following T cell stimulation, PD-1 recruits the tyrosine phosphatase SHP-2 to the ITSM motif in its cytoplasmic tail, resulting in dephosphorylation of effector molecules involved in the CD3+ T cell signaling cascade, such as CD3 ζ, PKC θ and ZAP70 (Freeman et al (2000) J Exp Med 192: 1027-34; Latchman et al (2001) Nat Immunol 2: 261-8; Carter et al (2002) Eur J Immunol 32: 634-43). PD-L1 is widely distributed not only on leukocytes and nonhematopoietic cells in lymphoid and non-lymphoid tissues, but also in various cancer cells, is highly expressed on the surface of various tumor cells, and the degree of malignancy and poor prognosis of tumors are closely related to the expression level of PD-L1. There are clinical data indicating that high tumor expression of PD-L1 is associated with increased tumor invasiveness and poor prognosis. The formation of the PD-1/PD-L1 complex transmits inhibitory signals and negatively regulates T cell immune responses; it inhibits TCR-mediated T cell activation, cytokine production and T cell proliferation (Fife et al (2011) Nature Immunology 10: 1185-1193); induction of depletion or anergy in cognate antigen-specific T cells (Hofmeyer et al (2011) Journal of Biomedicine and Biotechnology 2011: 1-9); promote the differentiation of Th1 cells into Foxp3+ regulatory T cells (Armanath et al (2011) Science TransMed 3: 1-13; Francisco et al (2009) J. exp. Med.206: 3015-; and inducing apoptosis of effector T cells. Disruption of the PD-L1 gene resulted in an upregulated T cell response and the generation of autoreactive T cells (Latchman et al (2004) PNAS 101: 10691-10696). Antibody blockade of PD-1 or PD-L1 resulted in increased anti-tumor immunity (Iwai et al (2002) PNAS 99: 12293-12297).
Chinese patent document CN106977602A discloses a PD-1 monoclonal antibody 14C12H1L1, which can effectively block the combination of PD1 and PDL1 and shows good antitumor activity.
The biggest challenge in the course of tumor immunotherapy in foreigners is poor treatment due to tumor immune tolerance and escape. Therefore, the small-molecule anti-tumor compound and the anti-PD-1/PD-L1 antibody are used in combination to break the immune tolerance established by the body to the tumor cells, and the method has important theoretical significance and application value.
Disclosure of Invention
It is an object of the present invention at least to provide a pharmaceutical combination comprising a tyrosine kinase inhibitor and a human PD-1 antibody, said human PD-1 antibody comprising a light chain and a heavy chain, wherein said light chain comprises light chain complementarity determining regions LCDR1, LCDR2 and LCDR3, said light chain complementarity determining regions consisting of the amino acid sequences shown in SEQ ID No. 1, SEQ ID No. 2 and SEQ ID No. 3, respectively, and wherein said heavy chain comprises heavy chain complementarity determining regions HCDR1, HCDR2 and HCDR3, said heavy chain complementarity determining regions consisting of the amino acid sequences shown in SEQ ID No. 4, SEQ ID No. 5 and SEQ ID No. 6, respectively.
In some embodiments, the tyrosine kinase inhibitor is a compound of formula I or a pharmaceutically acceptable salt thereof, and in some particular embodiments, the tyrosine kinase inhibitor is a hydrochloride salt of a compound of formula I, i.e., angutinib hydrochloride.
In some embodiments, the human PD-1 antibody comprises a light chain variable region as set forth in amino acid sequence SEQ ID NO. 7 and a heavy chain variable region as set forth in SEQ ID NO. 8.
In some embodiments, the human PD-1 antibody is 14C12H1L 1.
In some embodiments, the compound of formula I may be present as a pharmaceutically acceptable salt or a pharmaceutically acceptable formulation thereof, preferably as its hydrochloride salt.
In some embodiments, the compound is the hydrochloride salt of 1- [ [ [4- (4-fluoro-2-methyl-1H-indol-5-yl) oxy-6-methoxyquinolin-7-yl ] oxy ] methyl ] cyclopropylamine, namely, nilotinib hydrochloride.
In some embodiments, the pharmaceutical combination comprises: a compound of formula I or a hydrochloride salt thereof (e.g., dihydrochloride); and 14C12H1L1 monoclonal antibody or antigen-binding fragment thereof.
It is also an object of the present invention at least to provide the use of a pharmaceutical combination comprising a tyrosine kinase inhibitor and a human PD-1 antibody, said human PD-1 antibody comprising a light chain and a heavy chain, wherein said light chain comprises light chain complementarity determining regions LCDR1, LCDR2 and LCDR3, said light chain complementarity determining regions consisting of the amino acid sequences shown in SEQ ID No. 1, SEQ ID No. 2 and SEQ ID No. 3, respectively, and wherein said heavy chain comprises heavy chain complementarity determining regions HCDR1, HCDR2 and HCDR3, said heavy chain complementarity determining regions consisting of the amino acid sequences shown in SEQ ID No. 4, SEQ ID No. 5 and SEQ ID No. 6, respectively, for the treatment of a tumor.
In some embodiments, the tyrosine kinase inhibitor is a compound of formula I or a hydrochloride salt thereof.
In some embodiments, the human PD-1 antibody comprises a light chain variable region as set forth in amino acid sequence SEQ ID NO. 7 and a heavy chain variable region as set forth in SEQ ID NO. 8. In some embodiments, the human PD-1 antibody is 14C12H1L 1.
The invention also provides a method for treating a subject having esophageal cancer comprising administering to the subject a therapeutically effective amount of a tyrosine kinase inhibitor and a therapeutically effective amount of a human PD-1 antibody, said human PD-1 antibody comprising a light chain and a heavy chain, wherein the light chain comprises light chain complementarity determining regions LCDR1, LCDR2 and LCDR3, said light chain complementarity determining regions consisting of the amino acid sequences set forth in SEQ ID No. 1, SEQ ID No. 2 and SEQ ID No. 3, respectively, and wherein the heavy chain comprises heavy chain complementarity determining regions HCDR1, HCDR2 and HCDR3, said heavy chain complementarity determining regions consisting of the amino acid sequences set forth in SEQ ID No. 4, SEQ ID No. 5 and SEQ ID No. 6, respectively. In some embodiments, the tyrosine kinase inhibitor is a compound of formula I or a hydrochloride salt thereof. In some embodiments, the human PD-1 antibody comprises a light chain variable region as set forth in amino acid sequence SEQ ID NO. 7 and a heavy chain variable region as set forth in SEQ ID NO. 8. In some embodiments, the human PD-1 antibody is 14C12H1L 1.
The invention also provides a combination therapy for treating a subject having esophageal cancer, the method comprising administering to the subject a therapeutically effective amount of a tyrosine kinase inhibitor alone and a therapeutically effective amount of a human PD-1 antibody alone, the human PD-1 antibody comprising a light chain and a heavy chain, wherein the light chain comprises light chain complementarity determining regions LCDR1, LCDR2 and LCDR3, the light chain complementarity determining regions consisting of the amino acid sequences set forth in SEQ ID No. 1, SEQ ID No. 2 and SEQ ID No. 3, respectively, and wherein the heavy chain comprises heavy chain complementarity determining regions HCDR1, HCDR2 and HCDR3, the heavy chain complementarity determining regions consisting of the amino acid sequences set forth in SEQ ID No. 4, SEQ ID No. 5 and SEQ ID No. 6, respectively.
The invention also provides a method of treating a subject having a cancer or tumor that is esophageal cancer, comprising: (i) measuring the level of PD-1 and/or PD-L1 in a sample of the subject, wherein the subject is PD-1 and/or PD-L1 positive, and (ii) administering to the subject a therapeutically effective amount of an anti-PD-1 and/or PD-L1 antibody or antigen-binding portion thereof.
The invention also provides a method for treating a subject having a cancer or tumor. In certain embodiments, the subject is a patient diagnosed with esophageal cancer, e.g., esophageal cancer diagnosed as late-stage recurrent or metastatic esophageal cancer, or unresectable esophageal cancer, or the subject is not suitable for receiving curative treatment, e.g., curative chemoradiotherapy and/or surgical treatment. In certain embodiments, the subject is diagnosed with, or unresectable, advanced relapsed or metastatic esophageal cancer, or the subject is not suitable for receiving curative treatment, such as advanced relapsed or metastatic esophageal cancer that is treated with curative chemoradiotherapy and/or surgery. In certain embodiments, the subject is a patient diagnosed with esophageal squamous cell carcinoma, e.g., advanced relapsed or metastatic esophageal squamous cell carcinoma, or unresectable esophageal squamous cell carcinoma, or is not suitable for receiving curative treatment, such as curative chemoradiotherapy and/or surgical treatment. In certain embodiments, the subject has not previously received systemic treatment or has received systemic treatment. In certain embodiments, the methods are for first line treatment of esophageal cancer.
In some versions of the invention, the subject has previously undergone surgery, chemotherapy, and/or radiation therapy. In some embodiments, the subject has re-developed disease progression after achieving complete remission following surgery, chemotherapy, and/or radiation therapy. In some embodiments, the subject has failed to complete remission or failed to partial remission following surgery, chemotherapy, and/or radiation therapy.
In some embodiments of the invention, the subject has not previously received systemic chemotherapy. In some embodiments, the subject has previously received surgical treatment, radiation therapy, induction chemotherapy and/or adjuvant chemotherapy, or the subject has received concurrent chemotherapy. In some embodiments, the subject has not previously received systemic chemotherapy, but has received surgical treatment, radiation therapy, induction chemotherapy and/or adjuvant chemotherapy, or will receive concurrent chemotherapy. In some embodiments, the subject has complete remission following surgical treatment, radiation therapy, induction chemotherapy, concurrent chemotherapy, and/or adjuvant chemotherapy before disease progression occurs again. In some embodiments, the subject has failed to complete remission or failed to partial remission following surgical treatment, radiation therapy, induction chemotherapy, concurrent chemotherapy, and/or adjuvant chemotherapy. In some embodiments, the subject undergoes metastasis following surgical treatment, radiation treatment, induction chemotherapy, concurrent chemotherapy, and/or adjuvant chemotherapy.
In some embodiments of the present application, the pharmaceutical 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.
In some embodiments of the present application, the pharmaceutical combination is a non-fixed combination. In some embodiments, the human PD-1 antibody and the compound of formula I in the non-fixed combination are each in the form of a pharmaceutical composition.
It is also an object of the present application to at least provide a pharmaceutical pack comprising separately packaged pharmaceutical compositions in separate containers, wherein in one container a pharmaceutical composition comprising a compound of formula I or a pharmaceutically acceptable salt thereof is contained and in a second container a pharmaceutical composition comprising a human PD-1 antibody is contained.
In some embodiments of the present application, the pharmaceutical composition comprises a compound of formula I in an amount of 6-168 mg. In some embodiments, the pharmaceutical composition comprises a compound of formula I in an amount selected from 6mg, 8mg, 10mg, 12mg, 15mg, 20mg, 30mg, 50mg, 56mg, 70mg, 84mg, 112mg, 140mg, 168mg, or any range formed by any of the foregoing. In some embodiments, the pharmaceutical composition comprises a compound of formula I in an amount from 8mg to 12 mg. In some embodiments, the pharmaceutical composition comprises a compound of formula I in an amount of 8mg or 10 mg. In some embodiments, the pharmaceutical composition comprises a compound of formula I in an amount of 12 mg.
In some embodiments, the human PD-1 antibody is administered in one or more uniform doses effective to treat the cancer. In some embodiments, the uniform dose is in the range of about 10mg to about 1000mg of human PD-1 antibody. In some embodiments, the unitary dose is selected from about 100mg, about 150mg, about 200mg, about 250mg, about 300mg, about 350mg, about 400mg, about 450mg, about 500mg, about 600mg, about 700mg, about 800mg, about 900mg, or about 1000mg of the human PD-1 antibody. In some embodiments, the unitized dose is selected from about 200mg of human PD-1 antibody.
In some embodiments, the treatment for administration of the human PD-1 antibody is administered intravenously to the human PD-1 antibody on the first day (D1) of each cycle for 2 weeks (14 days) or 3 weeks (21 days). That is, the anti-PD-1 antibody is administered at a frequency of once every two weeks (q2w) or once every three weeks (q3 w).
It is also an object of the present application to provide a unit formulation, wherein the unit formulation comprises: a compound component, 6-12 mg of a compound of formula I or a hydrochloride thereof; and an antibody component, 50-350mg of a human PD-1 antibody or antigen-binding fragment thereof; wherein the compound component and the antibody component are packaged separately.
In some embodiments, the unit formulation comprises: compound component, 8mg, 10mg or 12mg of a compound of formula I or its hydrochloride salt; and an antibody component, 100mg or 200mg of a human PD-1 antibody or antigen-binding fragment thereof; wherein the compound component and the antibody component are packaged separately.
It is also an object of the present application to provide a method for preventing or treating cancer or tumor, wherein one or more of the above-described unit preparations are administered to a subject in need thereof. Preferably, the compound component and the antibody component of the unit formulation are each administered separately. Preferably, the cancer or tumor is esophageal cancer.
In some embodiments of the present application, the administration of erlotinib in combination with 14C12H1L1, every 21 days for one treatment cycle, is administered as follows: anrotinib 10 mg/time or 8 mg/time on days 1-14, 14C12H1L 1200 mg on day 1.
In some embodiments of the present application, the administration of erlotinib in combination with 14C12H1L1, wherein the administration of the erlotinib hydrochloride capsule is 1 time daily, 8mg or 10mg each time. The oral administration is continued for 2 weeks and stopped for 1 week, i.e. 3 weeks (21 days) are a treatment period, and the 14C12H1L1 is administered by 1 time every 3 weeks at 200 mg/time and by intravenous infusion for 60 + -10 min.
The medicine composition has good curative effect on esophageal cancer, and can inhibit growth and proliferation of esophageal cancer cells.
Arotinib
As used herein, the chemical name of said nilotinib (i.e., the compound of formula I) is 1- [ [ [4- (4-fluoro-2-methyl-1H-indol-5-yl) oxy-6-methoxyquinolin-7-yl ] oxy ] methyl ] cyclopropylamine, which has the following structural formula:
Figure BDA0002890379420000051
as used herein, the nilotinib includes its non-salt forms (e.g., free acid or free base), as well as its pharmaceutically acceptable salts, which are all included within the scope of the present application. For example, the pharmaceutically acceptable salt of the nilotinib can be the hydrochloride salt or the dihydrochloride salt. The dosage of the nilotinib or salt thereof referred to herein is calculated based on the free base of the nilotinib, unless otherwise indicated.
14C12H1L1
As used herein, 14C12H1L1 or14C12H1L1 monoclonal antibody,is an anti-PD-1 monoclonal antibody, and the sequence and the structure of the monoclonal antibody can be found in the literature (CN 106977602A). In the 14C12H1L1 monoclonal antibody, LCDR1 comprises the sequence QDINTY (SEQ ID NO:1), LCDR2 comprises the sequence RAN (SEQ ID NO:2), LCDR3 comprises the sequence LQYDEFPLT (SEQ ID NO:3), HCDR1 comprises the sequence GFAFSSYD (SEQ ID NO:4), HCDR2 comprises the sequence ISGGGRYT (SEQ ID NO:5), and HCDR3 comprises the sequence ANRYGEAWFAY (SEQ ID NO: 6).
The amino acid sequence of the light chain variable region is as follows:
DIQMTQSPSSMSASVGDRVTFTCRASQDINTYLSWFQQKPGKSPKTLIYRANRLVSGVPSRFSGSGSGQDYTLTISSLQPEDMATYYCLQYDEFPLTFGAGTKLELK(SEQ ID NO:7)。
the amino acid sequence of the heavy chain variable region is as follows:
EVQLVESGGGLVQPGGSLRLSCAASGFAFSSYDMSWVRQAPGKGLDWVATISGGGRYTYYPDSVKGRFTISRDNSKNNLYLQMNSLRAEDTALYYCANRYGEAWFAYWGQGTLVTVSS(SEQ ID NO:8)。
definitions and explanations
The following terms used in the present application have the following meanings, unless otherwise specified. A particular term should not be considered as ambiguous or unclear without special definition, but rather construed according to ordinary meaning in the art. When a trade name appears in this application, it is intended to refer to its corresponding commodity, composition, or active ingredient thereof.
As used herein, the term "antibody" refers to an antigen binding protein having at least one antigen binding domain. The antibodies and fragments thereof of the present application can be whole antibodies or any fragment thereof. Thus, the antibodies and fragments thereof of the present application include monoclonal antibodies or fragments thereof and antibody variants or fragments thereof, as well as immunoconjugates. Examples of antibody fragments include Fab fragments, Fab 'fragments, f (ab)' fragments, Fv fragments, isolated CDR regions, single chain Fv molecules (scFv), and other antibody fragments known in the art. Antibodies and fragments thereof can also include recombinant polypeptides, fusion proteins, and bispecific antibodies. The anti-PD-L1 antibodies and fragments thereof disclosed herein may be of the IgG1, IgG2, IgG3, or IgG4 isotype.
The term "isotype" refers to the class of antibodies encoded by the heavy chain constant region gene. In one embodiment, the anti-PD-1/PD-L1 antibodies and fragments thereof disclosed herein are of the IgG1 or IgG4 isotype. The anti-PD-1/PD-L1 antibodies and fragments thereof of the present application may be derived from any species, including but not limited to mouse, rat, rabbit, primate, llama, and human. The PD-1/PD-L1 antibody and fragments thereof may be chimeric, humanized or fully human.
The term "humanized antibody" refers to antibodies in which the antigen binding site is derived from a non-human species and the variable region framework is derived from human immunoglobulin sequences. Humanized antibodies may comprise substitutions in the framework regions such that the framework may not be an exact copy of the expressed human immunoglobulin or germline gene sequence.
By "isolated antibody" is meant an antibody that: it is substantially free of other antibodies having different antigen specificities (e.g., an isolated antibody that specifically binds PD-1/PD-L1 is substantially free of antibodies that specifically bind antigens other than PD-1/PD-L1). However, an isolated antibody that specifically binds PD-1/PD-L1 may have cross-reactivity with other antigens (such as PD-1/PD-L1 molecules from different species). Furthermore, the isolated antibody may be substantially free of other cellular material and/or chemicals.
An "antigen-binding portion" (also referred to as an "antigen-binding fragment") of an antibody refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen bound by an intact antibody.
As used herein, the term "derived" when used in reference to a molecule or polypeptide relative to a reference antibody or other binding protein means a molecule or polypeptide that is capable of specifically binding the same epitope as the reference antibody or other binding protein.
As used herein, the term "EC 50" refers to the effective concentration, 50% of the maximal response of an antibody. As used herein, the term "IC 50" refers to the inhibitory concentration, 50% of the maximal response of an antibody. Both EC50 and IC50 may be measured by ELISA or FACS analysis or any other method known in the art.
The term "treatment" generally refers to an act of obtaining a desired pharmacological and/or physiological effect. The effect may be prophylactic, in terms of preventing the disease or its symptoms, in whole or in part; and/or may be therapeutic in terms of partially or completely stabilizing or curing the disease and/or side effects due to the disease. As used herein, "treatment" encompasses any treatment of a disease in a patient, including: (a) preventing a disease or condition in a patient susceptible to the disease or condition but not yet diagnosed as having the disease; (b) inhibiting the symptoms of the disease, i.e., arresting its development; or (c) alleviating the symptoms of the disease, i.e., causing regression of the disease or symptoms.
As used herein, the term "systemic treatment" refers to treatment in which a drug substance is transported through the bloodstream to reach and affect cells throughout the body.
As used herein, the term "systemic chemotherapy" refers to systemic chemotherapy that does not include chemotherapy for locally advanced disease as one of the links of multimodal treatment, wherein chemotherapy for locally advanced disease includes induction chemotherapy, concurrent chemotherapy with radiotherapy, and adjuvant chemotherapy.
As used herein, the term "systemic treatment" refers to systemic chemotherapy, systemic or local radiotherapy.
As used herein, the term "first line treatment" refers to treatment with a drug that may be selected first or by criteria, depending on the patient's condition.
As used herein, the term "subject" means a mammal, such as a rodent, feline, canine, and primate. Preferably, the subject according to the present application is a human.
By "administering" is meant physically introducing a composition comprising a therapeutic agent to a subject using any of a variety of methods and delivery systems known to those skilled in the art. Routes of administration of immune checkpoint inhibitors (e.g., anti-PD-1 antibodies or anti-PD-L1 antibodies) include intravenous, intramuscular, subcutaneous, intraperitoneal, spinal, or other parenteral routes of administration, e.g., by injection or infusion. The phrase "parenteral administration" as used herein refers to modes of administration other than enteral and topical administration, typically by injection, and includes, but is not limited to, intravenous, intramuscular, intraarterial, intrathecal, intralymphatic, intralesional, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion, and in vivo electroporation. In certain embodiments, the immune checkpoint inhibitor (e.g., an anti-PD-1 antibody or an anti-PD-L1 antibody) is administered by a non-parenteral route, and in certain embodiments, orally. Other non-parenteral routes include topical, epidermal or mucosal routes of administration, e.g., intranasally, vaginally, rectally, sublingually or topically. Administration may also be performed, for example, once, multiple times, and/or over one or more extended periods of time.
As used herein, an "adverse event" (AE) is any adverse and often unintended or undesirable sign (including abnormal laboratory findings), symptom or disease associated with the use of medical therapy. For example, an adverse event can be associated with activation of the immune system or expansion of cells of the immune system (e.g., T cells) in response to a treatment. The medical treatment may have one or more related AEs, and each AE may have the same or different severity level. Reference to a method capable of "altering an adverse event" refers to a treatment regimen that reduces the incidence and/or severity of one or more AEs associated with the use of a different treatment regimen.
As used herein, "dosing interval" refers to the amount of time that elapses between multiple doses of a formulation disclosed herein administered to a subject. The dosing interval may thus be indicated as a range.
The term "dosing frequency" as used herein means the frequency of doses administered of a formulation disclosed herein over a given time. The frequency of administration may be indicated as the number of administrations per given time, e.g. 1 or 1 in 2 weeks per week.
The use of the term "flat dose" refers to a dose that is administered to a patient without regard to the weight or Body Surface Area (BSA) of the patient. Thus, a uniform dose is defined as the absolute amount of the agent (e.g., anti-PD-1 antibody) rather than the mg/kg dose. For example, a 60kg human and a 100kg human will receive the same dose of antibody (e.g., 240mg anti-PD-1 antibody).
The use of the term "fixed dose" in reference to a composition of the present application means that two or more different antibodies in a single composition are present in the composition in a specific (fixed) ratio to each other. In certain embodiments, the fixed dose is based on the weight of the antibody (e.g., mg). In certain embodiments, the fixed dose is based on the concentration of the antibody (e.g., mg/ml). In certain embodiments, the ratio of mg of the first antibody to mg of the second antibody is at least about 1:1, about 1:2, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:15, about 1:20, about 1:30, about 1:40, about 1:50, about 1:60, about 1:70, about 1:80, about 1:90, about 1:100, about 1:120, about 1:140, about 1:160, about 1:180, about 1:200, about 200:1, about 180:1, about 160:1, about 140:1, about 120:1, about 100:1, about 90:1, about 80:1, about 70:1, about 60:1, about 50:1, about 40:1, about 30:1, about 20:1, about 15:1, about 10:1, about 1: 8:1, about 1:1, about 5: 7, about 1:1, about 1: 6:1, about 1:1, about, About 4:1, about 3:1, or about 2: 1. For example, a 3:1 ratio of primary and secondary antibodies may mean that the vial may contain about 240mg of primary and 80mg of secondary antibody, or about 3mg/ml of primary and 1mg/ml of secondary antibody.
The term "weight-based dose" as referred to herein refers to a dose administered to a patient that is calculated based on the weight of the patient. For example, when a patient with a weight of 60kg requires 3mg/kg of anti-PD-1 antibody and 1mg/kg of anti-CTLA-4 antibody, one can extract appropriate amounts of anti-PD-1 antibody (i.e., 180mg) and anti-CTLA-4 antibody (i.e., 60mg) at a time from a 3:1 ratio fixed dose formulation of anti-PD-1 antibody and anti-CTLA-4 antibody.
The term "immunotherapy" means the treatment of a subject having a disease or at risk of infection or of suffering from a relapse of a disease by a method that includes inducing, enhancing, suppressing or otherwise altering an immune response. By "treatment" or "therapy" of a subject is meant any type of intervention or process performed on the subject, or the administration of an active agent to a subject, with the purpose of reversing, alleviating, ameliorating, inhibiting, slowing, or preventing the onset, progression, severity, or recurrence of a symptom, complication, or condition, or biochemical indicator associated with the disease.
As used herein, "PD 1/PD-L1 positive" may be used interchangeably with "at least about 1% PD-1/PD-L1 expression". In one embodiment, PD-1/PD-L1 expression may be used by any method known in the art. In another embodiment, PD-1/PD-L1 expression is measured by automated IHC. In certain embodiments, "PD-1/PD-L1 positive" means that there are at least 100 cells expressing PD-1/PD-L1 on the cell surface.
"programmed death receptor-1 (PD-1)" means an immunosuppressive receptor belonging to the CD28 family. PD-1 is expressed predominantly on previously activated T cells in vivo and binds to both ligands PD-L1 and PD-L2. The term "PD-1" as used herein includes variants, homologs, and species homologs of human PD-1(hPD-1), hPD-1, and analogs having at least one common epitope with hPD-1.
"programmed death ligand-1 (PD-L1)" is one of two cell surface glycoprotein ligands for PD-1 (the other being PD-L2) that down-regulates T cell activation and cytokine secretion upon binding to PD-1.
"subject" includes any human or non-human animal. The term "non-human animal" includes, but is not limited to, vertebrates such as non-human primates, sheep, dogs, and rodents such as mice, rats, and guinea pigs. In certain embodiments, the subject is a human. The terms "subject," "subject," and "patient" are used interchangeably herein in certain contexts.
A "therapeutically effective amount" or "therapeutically effective dose" of a drug or therapeutic agent is any amount of drug that, when used alone or in combination with another therapeutic agent, protects a subject from the onset of a disease or promotes disease regression as evidenced by a reduction in the severity of disease symptoms, an increase in the frequency and duration of disease symptom-free stages, or prevention of injury or disability caused by the affliction of the disease. The ability of a therapeutic agent to promote disease regression can be evaluated using a variety of methods known to skilled practitioners, such as in human subjects during clinical trials, in animal model systems predicting efficacy for humans, or by determining the activity of the agent in an in vitro assay.
As used herein, a "sub-therapeutic dose" refers to a dose of a therapeutic compound (e.g., an antibody) that is lower than the usual or typical dose of the therapeutic compound when administered alone for the treatment of a hyperproliferative disease (e.g., cancer).
As an example, an "anti-cancer drug" promotes cancer regression in a subject or prevents further tumor growth. In certain embodiments, the therapeutically effective amount of the drug promotes regression of the cancer to the point of eliminating the cancer. By "promoting cancer regression" is meant the administration of an effective amount of a drug, alone or in combination with an anti-neoplastic agent, resulting in the reduction of tumor growth or size, necrosis of the tumor, a reduction in the severity of at least one disease symptom, an increase in the frequency and duration of disease symptom-free stages, or the prevention of injury or disability resulting from the affliction of the disease. Furthermore, the terms "effective" and "effectiveness" with respect to treatment include pharmacological effectiveness and physiological safety. Pharmacological efficacy refers to the ability of a drug to promote cancer regression in a patient. Physiological safety means the level of toxicity or other adverse physiological effects (adverse effects) at the cellular, organ and/or organism level resulting from drug administration.
As an example for treating a tumor, a therapeutically effective amount of an anti-cancer agent can inhibit cell growth or tumor growth by at least about 10%, at least about 20%, at least about 40%, at least about 60%, or at least about 80% relative to an untreated subject, or, in certain embodiments, relative to a patient treated with standard of care therapy. In other embodiments of the present application, tumor regression may be observed for a period of at least about 20 days, at least about 40 days, or at least about 60 days. Despite these final measures of therapeutic effectiveness, the evaluation of immunotherapeutic drugs must also take into account "immune-related" response patterns.
By "immune-related" response pattern is meant the clinical response pattern often observed in cancer patients treated with immunotherapeutic agents that produce an anti-tumor effect by inducing a cancer-specific immune response or by altering the innate immune process. This response pattern is characterized by beneficial therapeutic effects following an initial increase in tumor burden or the appearance of new lesions, which would be classified as disease progression and would be synonymous with drug failure in the evaluation of traditional chemotherapeutic agents. Thus, proper evaluation of immunotherapeutic agents may require long-term monitoring of the effect of these agents on the target disease.
A therapeutically effective amount of a drug includes a "prophylactically effective amount," which is any amount of drug that inhibits the occurrence or recurrence of cancer when administered, alone or in combination with an anti-neoplastic agent, to a subject at risk of developing cancer (e.g., a subject with a premalignant condition) or a subject at risk of cancer recurrence. In certain embodiments, the prophylactically effective amount completely prevents the occurrence or recurrence of cancer. By "inhibiting" the occurrence or recurrence of cancer is meant reducing the likelihood of occurrence or recurrence of cancer, or completely preventing the occurrence or recurrence of cancer.
A "recurrent" cancer is one that regenerates at the primary site or a distant site in response to an initial treatment (e.g., surgery). A "locally recurrent" cancer is one that occurs at the same location after treatment as a previously treated cancer.
A "non-resectable" cancer is one that cannot be removed by surgery.
"metastatic" cancer refers to cancer that spreads from one part of the body (e.g., the lungs) to another part of the body.
The use of alternatives (e.g., "or") should be understood to refer to either, both, or any combination of alternatives. The indefinite articles "a" or "an" as used herein shall be understood to mean "one or more" of any listed or enumerated component.
By "failure of treatment with a platinum-containing chemotherapeutic regimen" is meant disease progression or toxic side effects that are not tolerated during or after treatment with first-line chemotherapy or radiotherapy with a platinum-containing regimen.
The definition of "failure of a systemic standard chemotherapy" is: disease progression during or after the last treatment, or intolerance during treatment due to toxic side effects.
The definition of "failure of chemotherapy at or above one line" is: disease progression during or after the last treatment; or is intolerable in the treatment process due to toxic and side effects.
Low grade (G1) defined as nuclear fission image number <2/10 high power field [ HPF ] and/or Ki-67 differentiation index < 3%; the medium grade (G2) is defined as the number of nuclear fission images 2-20/10 high power field [ HPF ] and/or Ki-67 differentiation index 3-20%. If the nuclear division image and the Ki-67 index of the same tumor tissue correspond to different grades, the compliance is higher.
The terms "about," about, "or" consisting essentially of mean a value or composition within an acceptable error range for the particular value or composition as determined by one of ordinary skill in the art, which will depend in part on how the value or composition is measured or determined, i.e., the limitations of the measurement system. For example, "about," about, "or" consisting essentially of can mean within 1 or more than 1 standard deviation, as practiced in the art. Alternatively, "about" or "consisting essentially of may refer to a range that differs by up to 10% or 20% (i.e., ± 10% or ± 20%) from the parameter or value modified thereby. For example, about 3mg may include any number between 2.7mg to 3.3mg (for 10%) or between 2.4mg to 3.6mg (for 20%). Furthermore, particularly with respect to biological systems or processes, the term may refer to up to an order of magnitude or up to at most 5 times the numerical value. Where a particular value or composition is provided in the application and claims, unless otherwise stated, the meaning of "about" or "consisting essentially of" should be assumed to be within an acceptable error range for that particular value or composition.
As used herein, the terms "about once per week", "about once per two weeks" or any other similar dosing interval term refer to approximations. "about once per week" may include every 7 days ± 1 day, i.e., every 6 days to every 8 days. "about once every two weeks" may include every 14 days ± 3 days, i.e., every 11 days to every 17 days. Similar approximations apply, for example, about once every 3 weeks, about once every 4 weeks, about once every 5 weeks, about once every 6 weeks, and about once every 12 weeks. In certain embodiments, a dosing interval of about once every 6 weeks or about once every 12 weeks refers to that a first dose may be administered on any day of the first week, and then a second dose may be administered on any day of the sixth or twelfth week, respectively. In other embodiments, a dosing interval of about once every 6 weeks or about once every 12 weeks refers to administration of a first dose on a particular day of the first week (e.g., monday) followed by administration of a second dose on the same day of the sixth or twelfth week (i.e., monday), respectively. Similar principles apply to phrases including, but not limited to, "about 1 every 2 weeks," "about 1 every month," etc. … ….
As used herein, any concentration range, percentage range, ratio range, or integer range should be understood to include the value of any integer within the recited range, and when appropriate, to include fractions thereof (such as tenths and hundredths of integers), unless otherwise indicated.
Unless specifically stated otherwise, "about" or "approximately" in this application means within + -5% of the specified numerical range given, preferably within + -2% of the specified numerical range given, and more preferably within + -1% of the specified numerical range given. For example, a pH of about 5.5 means a pH of 5.5. + -. 5%, preferably a pH of 5.5. + -. 2%, more preferably a pH of 5.5. + -. 1%.
The term "pharmaceutically acceptable" is intended to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The term "pharmaceutically acceptable salt" includes salts of the base ion with the free acid or salts of the acid ion with the free base, including, for example, hydrochloride, hydrobromide, nitrate, sulfate, phosphate, formate, acetate, trifluoroacetate, fumarate, oxalate, maleate, citrate, succinate, methanesulfonate, benzenesulfonate or p-methylbenzenesulfonate, preferably hydrochloride, hydrobromide, sulfate, formate, acetate, trifluoroacetate, fumarate, maleate, methanesulfonate, p-methylbenzenesulfonate, sodium salt, potassium salt, ammonium salt, amino acid salt and the like. In the present application, when forming a pharmaceutically acceptable salt, the molar amount of free acid to base ion is about 1:0.5 to 1:5, preferably 1:0.5, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, or 1: 8. In the present application, when forming a pharmaceutically acceptable salt, the molar ratio of the free base to the acid ion is about 1:0.5 to 1:5, preferably 1:0.5, 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7 or 1: 8.
The term "fixed combination" means that the active ingredients (e.g. an anti-PD-1 antibody or a compound of formula I) are administered to a subject simultaneously in a fixed total dose or dose ratio, or in the form of a single entity, pharmaceutical composition or formulation.
The term "non-fixed combination" means that two or more active ingredients are administered to a subject as separate entities (e.g. pharmaceutical compositions, formulations) simultaneously, concurrently or sequentially and without specific time constraints, wherein the active ingredients are administered to the subject to a therapeutically effective amount level. An example of an unfixed combination is cocktail therapy, e.g. 3 or more active ingredients are administered. In a non-fixed combination, the individual active ingredients may be packaged, sold or administered as a completely separate pharmaceutical composition. The term "non-fixed combination" also includes the use of "fixed combinations" in between, or "fixed combinations" in combination with, any one or more of the individual entities of the active ingredients.
As used herein, "in combination" or "in combination" means that two or more active substances may be administered to a subject together in a mixture, simultaneously as a single formulation, or sequentially in any order as a single formulation.
The term "pharmaceutical composition" refers to a mixture of one or more of the active ingredients of the present application (e.g., an anti-PD-1 antibody or a compound of formula I) or a pharmaceutical combination thereof with pharmaceutically acceptable excipients. The purpose of the pharmaceutical composition is to facilitate administration of the compounds of the present application, or a pharmaceutical combination thereof, to a subject.
The term "synergistic effect" refers to a simple addition of two or more components (e.g., an anti-PD-1 antibody or a compound of formula I) that produces an effect (e.g., inhibiting the growth of colon cancer, or ameliorating symptoms of colon cancer) that is greater than the effect of the components when administered alone.
In one aspect of the present application, the following is also provided:
1. use of a pharmaceutical combination for the manufacture of a medicament for the treatment or prevention of esophageal cancer, the pharmaceutical combination comprising:
a) a human PD-1 antibody that comprises a light chain and a heavy chain, wherein the light chain comprises light chain complementarity determining regions LCDR1, LCDR2 and LCDR3 consisting of the amino acid sequences set forth in SEQ ID NO. 1, SEQ ID NO. 2 and SEQ ID NO. 3, respectively, and wherein the heavy chain comprises heavy chain complementarity determining regions HCDR1, HCDR2 and HCDR3 consisting of the amino acid sequences set forth in SEQ ID NO. 4, SEQ ID NO. 5 and SEQ ID NO. 6, respectively, and
b) a tyrosine kinase inhibitor, wherein the tyrosine kinase inhibitor is a compound of formula I or a pharmaceutically acceptable salt thereof,
Figure BDA0002890379420000111
2. the use according to item 1, wherein the pharmaceutically acceptable salt of the compound of formula I is the hydrochloride salt of 1- [ [ [4- (4-fluoro-2-methyl-1H-indol-5-yl) oxy-6-methoxyquinolin-7-yl ] oxy ] methyl ] cyclopropylamine, preferably the dihydrochloride.
3. The use according to any one of items 1-2, wherein the human PD-1 antibody comprises a light chain variable region having an amino acid sequence as set forth in SEQ ID NO. 7 and a heavy chain variable region as set forth in SEQ ID NO. 8.
4. The use according to any one of items 1-3, wherein the human PD-1 antibody is 14C12H1L 1.
5. The use according to any one of claims 1 to 4, wherein the pharmaceutical combination is a non-fixed combination.
6. The use according to item 5, the human PD-1 antibody and the compound of formula I or a pharmaceutically acceptable salt thereof in the non-fixed combination each being in the form of a pharmaceutical composition.
7. The use according to item 6, for the treatment of esophageal cancer that did not receive a curative treatment, or that had advanced relapse or metastasis, or that had not previously received a systemic treatment.
8. The use according to any one of claims 6 to 7, for the treatment of esophageal squamous cell carcinoma.
9. The use according to any one of items 6 to 8, in combination for first line treatment of esophageal squamous cell carcinoma.
10. The use according to any one of items 6-9, for the treatment of advanced recurrent or metastatic esophageal squamous cell carcinoma, or unresectable esophageal squamous cell carcinoma, or esophageal squamous cell carcinoma for which the subject is not amenable to curative treatment radically, or esophageal squamous cell carcinoma for which the subject has not previously received systemic treatment or has received systemic treatment.
11. The use according to any one of claims 6 to 10, wherein the compound of formula I hydrochloride is administered in a uniform dose of about 8 mg/dose or 10 mg/dose and 14C12H1L1 is administered in a uniform dose of about 200 mg/dose.
12. The use according to any one of claims 6 to 10, wherein the hydrochloride salt of the compound of formula I is administered 1 time a day, 8mg or 10mg each time, 2 weeks for 1 week on continuous oral administration, and 14C12H1L1 is administered 1 time every 3 weeks, 200 mg/time.
13. An article of manufacture comprising a container containing a fixed dose of a hydrochloride salt of a compound of formula I selected from about 8mg, about 10mg, and about 12mg, and a 14C12H1L1 antibody, a fixed dose of 14C12H1L1 antibody selected from about 100mg and about 200 mg.
Mode of administration
The following is not intended to limit the mode of administration of the pharmaceutical combinations of the present application.
The components of the pharmaceutical combination of the present application may be formulated separately from each other or some or all of them may be co-formulated. In one embodiment, the pharmaceutical combination of the present application may be formulated as a pharmaceutical composition suitable for single or multiple administration.
The components of the pharmaceutical combination of the present application may each be administered separately, or some or all of them may be co-administered. The components of the pharmaceutical 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 pharmaceutical combination of the present application may be administered independently of each other, or some or all of them together in a variety of routes as appropriate, including, but not limited to, oral or parenteral (by intravenous, intramuscular, topical or subcutaneous routes). In some embodiments, the components of the pharmaceutical combination of the present application may be administered orally or parenterally, such as intravenously or intraperitoneally, each independently, or together with some or all of them.
The components of the pharmaceutical combination of the present application may each independently, or some or all of them together be in a suitable dosage form, 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.
The components of the pharmaceutical combination of the present application may each independently, or some or all of them together, contain a pharmaceutically acceptable carrier and/or excipient.
The pharmaceutical combination of the present application may also comprise additional therapeutic agents. In one embodiment, the additional therapeutic agent may be a cancer therapeutic agent known in the art.
Detailed Description
The present application is further described below with reference to specific examples, which, however, are only for illustration and do not limit the scope of the present application. Likewise, the present application is not limited to any particular preferred embodiment described herein. It should be understood by those skilled in the art that equivalent substitutions for the technical features of the present application or corresponding improvements are still within the scope of the present application. The reagents used in the following examples are commercially available products, and the solutions can be prepared by techniques conventional in the art, except where otherwise specified.
TABLE 1 abbreviation table
Figure BDA0002890379420000121
Figure BDA0002890379420000131
"14C 12H1L1 injectable solution" refers to a medical formulation for injection containing the 14C12H1L1 monoclonal antibody, which is usually administered to a patient by intravenous infusion route. In a particular embodiment, the expression "injection of 14C12H1L1, 200 mg/time" may be understood in the usual manner in the art as a liquid injectable medical formulation containing 200mg of 14C12H1L1 monoclonal antibody per administration to a patient.
Example a clinical study protocol-study criteria and endpoints
1.1 inclusion exclusion criteria
And (3) inclusion standard: the following candidates can be put into the test
All conditions of any one of the following queues are met:
1) age: 18-75 years old, male and female;
2) the advanced esophageal squamous carcinoma which can not be radically cured is confirmed by histology or cytology;
3) not previously receiving systemic treatment;
4) KPS score is more than or equal to 70 points;
5) measurable lesions with a double check irrecist criteria;
6) the estimated life time is more than or equal to 3 months;
7) the main organs have normal functions and no serious abnormal blood, heart, lung, liver and kidney functions and immunodeficiency diseases;
8) patients voluntarily join the study and sign informed consent, and compliance is good.
Exclusion criteria: subjects presenting any of the following subjects would not be able to enter the study
1) Those who have previously been treated with chemotherapeutic drugs/targeted drugs/immunotherapy, or who have received chemotherapy within 6 months of first-time administration;
2) known people allergic to the hydrochloric acid anrotinib, 14C12H1L1 injection or any component;
3) imaging (CT or MRI) shows that tumor invasion into the adjacent organs of the esophageal lesion (aorta or trachea) results in patients with higher risk of retention and fistulas;
4) immunosuppressive drugs were used within 14 days prior to the first use of the 14C12H1L1 injection, excluding nasal spray and inhalation corticosteroids or physiologic doses of systemic steroid hormones (i.e., no more than 10 mg/day prednisolone or other corticosteroids at equivalent physiologic doses);
5) live attenuated vaccines were administered within 4 weeks prior to the first dose or scheduled for the study period;
6) patients with hypertension or coronary heart disease, arrhythmia and cardiac insufficiency;
7) those with various factors affecting oral medication (e.g., inability to swallow, post-gastrointestinal resection, chronic diarrhea, ileus, etc.);
8) invading or poorly demarcating the great vessels;
9) significant surgical treatment, incisional biopsy or overt traumatic injury was received within 28 days prior to the first dose;
10) patients with arterial/venous thrombotic events, such as cerebrovascular accidents (including transient ischemic attacks), deep vein thrombosis and pulmonary embolism, occurring within 6 months prior to the first administration;
11) those who have a history of abuse of psychotropic drugs and are unable to abstain or have a psychotic disorder;
12) at the discretion of the investigator, those with concomitant disease who severely compromise patient safety or affect the patient's completion of the study
13) Other cases considered unsuitable by the investigator for inclusion.
1.2 Exit Standard
1) Disease progression occurred and the investigator judged that the subject would not benefit from continued treatment;
2) adverse events occurred, were not tolerated, not alleviated;
3) subjects who had a severe adverse event and were not eligible to continue the study;
4) those who deviate or violate a protocol severely and impact drug safety or efficacy assessments;
5) subject withdrawal of informed consent;
6) follow-up cannot be continued on time for various reasons.
1.3 study endpoint
Primary endpoint
Objective Remission Rate (ORR)
Secondary endpoint
Disease Control Rate (DCR), duration of remission (DOR), Progression Free Survival (PFS), Total survival (OS), and the like
The incidence and severity of Adverse Events (AEs) and Severe Adverse Events (SAEs), and abnormal laboratory test indices.
Example two clinical trial design
Open, single-center clinical studies were employed.
2.1 sample size
30 cases were planned into the group (adjusted according to the specific test results).
2.2 image evaluation design
The primary efficacy endpoint of this study was ORR, using results assessed by researchers at various study centers. An independent imaging group is additionally arranged in the research to carry out imaging curative effect evaluation and recheck.
2.3 dosing regimen design
Anrotinib hydrochloride capsules:
1 daily (recommended infusion starting at 14C12H1L1 injection + -60min hollow abdominal administration), 8mg each time (final dose determined by safe lead-in period, temporary 8 mg). 2 weeks and 1 week, i.e. 3 weeks (21 days) for a treatment period. If the administration period is missed, the time for next administration is shorter than 12 hours, and the medicine is not taken again.
14C12H1L1 injecta:
administered 1 time every 3 weeks at 200 mg/time, and administered by intravenous infusion. The infusion time was 60. + -.10 min.
The medication period is as follows:
every 21 days is a treatment period, and other anti-tumor treatments can not be carried out during the administration period; patients in disease control (CR + PR + SD) and with tolerable adverse effects continue to be dosed until disease progression or intolerance.
Efficacy was assessed every 2 cycles and every 4 cycles after 16 cycles. Patients with disease control (CR + PR + SD) and tolerable adverse reactions continue to take their medications until clinical benefit is lost, toxicity is not tolerable, efficacy is assessed as PD, and the study is concluded when the investigator deems it inappropriate to continue taking medications.
Adjusting the dosage of the hydrochloric acid Arotinib capsule:
in the research process, after the medicine is taken for a period of time, if a researcher judges that the disease is possible to progress and the safety of a patient is stable, the dosage can be adjusted up once. Each patient can only make one dose up-regulation at most and cannot adjust across dose groups.
14C12H1L1 injection fixed dose, without a dose down-regulation scheme.
2.4 disease progression and efficacy assessment
In the period of clinical application of the same kind of 14C12H1L1 injection, the subjects have false progress, and the therapeutic effect evaluation standard of the study is based on RECIST 1.1 (therapeutic effect evaluation standard of solid tumors). The efficacy was also confirmed using the irrecist criteria (tumor immunotherapy-related evaluation criteria). That is, subjects judged to be disease Progression (PD) according to RECIST 1.1 criteria were further confirmed according to irrecist criteria to determine whether to further observe the drug.
Efficacy was assessed every 2 cycles, 16 cycles, and 4 cycles, starting on the first day of the first cycle, until subject development of tumor-image confirmed disease progression, which did not change the frequency of assessment as subjects delayed or discontinued treatment. If the subject terminates study treatment for reasons other than disease progression, it is still necessary to continue receiving the frequent tumor imaging assessments until the subject begins new anti-tumor therapy, or imaging-evidence of disease progression, or the subject actively withdraws, or the subject dies, whichever comes first.
The imaging evaluation mode of the tumor can adopt CT or MRI, but the evaluation method, machine and technical parameters are consistent during the whole research period; if no contraindications are indicated, contrast agents should be used. If tumor assessments were made within 14 days prior to the first dose and the same procedure was used in the same hospital, this could be taken as a baseline tumor assessment. Baseline tumor assessments should include CT or MRI of the chest, abdomen and pelvic cavities. During the screening period, craniocerebral flat scan and enhancement/enhancement MRI examination are required. All suspicious lesion sites should be examined imagewise. For patients with bone metastases, bone scans should be used to follow up on the lesions. For patients with bone metastasis, if no clinical symptoms are aggravated, the tumor evaluation is not required to be performed for recheck every time, and if the clinical symptoms are aggravated, the patient should be rechecked in time. For cases suspected of disease progression before the start of the next evaluation in the plan, an unplanned tumor assessment should be performed. During the test period, only the imaging examination of the focus part is performed, and during the test period, the image of the corresponding part can be examined if the suspicious part exists.
EXAMPLE III Collection of biological samples
3.1 serum anti-14C 12H1L1 antibody (ADA) assay
The immunogenicity monitoring time point is based on the administration time of the 14C12H1L1 injection; when 14C12H1L1 was given late, there was a corresponding delay in immunogenic blood sampling. If the subject is detected to be positive for ADA, the neutralizing antibody is measured.
Collection was taken before dosing (-60min) at cycles 1, 2, 4, 8 and every 6 cycles thereafter. Simultaneous collection was done 30min (+ -5 min) after the end of infusion in cycle 1 and cycle 8 and 30 days (+ -7 days), 90 days (+ -7 days) after the last dose. 5mL of venous blood is required to be collected each time, the venous blood is placed in a blood collection tube containing procoagulant separation gel for 30min at room temperature, after natural coagulation, 3000g of the venous blood is centrifuged for 10min, the venous blood is averagely distributed into 4 freezing storage tubes (3 parts of detection tubes and 1 part of backup tube, wherein each detection tube is not less than 0.5mL), and serum is taken as a marker and then stored in a refrigerator at the temperature of-40 to-80 ℃ for detecting immunogenicity and 14C12H1L1 blood concentration.
Unexpected adverse events related to immunity occurred during the test, and additional 1 blood sampling was required to test immunogenicity and 14C12H1L1 blood concentration after the adverse event was confirmed, but the time from the last blood sampling was less than 24H and the sampling could not be performed.
Note that: based on the ADA results, neutralizing antibodies are subsequently tested if necessary.
3.2 biomarker assays
Participation in this study required the provision of tumor tissue specimens for biomarker studies, including PD-L1 expression, mismatch repair/microsatellite instability (MMR/MSI) detection, and the like.
The samples for biomarker detection were selected from fresh biopsy samples within 1 month prior to group entry. When a fresh tissue sample is taken, 1 needle or more is punctured through the skin. If a fresh biopsy tissue sample cannot be obtained, the biopsy tissue sample can be collected, used and filed, 10 unstained pathological tissue slices (anti-alopecia) are taken, the thickness is 4-6 mu m, white slices prepared by fresh cutting are taken, and wax sealing treatment is needed if the white slices cannot be sent out in time (within one month).
Example four evaluation of effectiveness and safety
4.1. Analysis of the main efficacy index
4.1.1. Objective Remission Rate (ORR):
the ratio of the number of objective remission cases (PR + CR) in each group to the total number of cases in each group and 95% CI were calculated. The 95% CI for ORR was calculated based on the exact binomial method of F distribution.
4.1.2. Analysis of secondary efficacy index
4.1.2.1. Progression Free Survival (PFS)
And estimating the median PFS by adopting a Kaplan-Meier method, and drawing a survival curve graph.
4.1.2.2. Overall lifetime (OS)
And estimating the median OS by adopting a Kaplan-Meier method, and drawing a survival curve graph.
4.1.2.3. Duration of remission (DOR)
The median PFS and 95% CI thereof are estimated by adopting a Kaplan-Meier method, and a survival curve graph is drawn.
4.1.2.4. Disease Control Rate (DCR):
the ratio of the number of disease control cases (CR + PR + SD) to the total number of cases and 95% CI were calculated. The 95% CI for the DCR was calculated based on the exact binomial method of F distribution.
4.2. Evaluation of safety
4.2.1. Drug exposure and compliance
The drug exposure is described by mean, standard deviation, maximum, minimum, median.
Summary of subject exposure to study drug treatment, patient cycle count, dose adjustments during treatment, cumulative number of doses adjusted during treatment, etc.
Statistical descriptions of study drug treatment time, study drug total dose and daily average dose and study drug dose compliance over the treatment period are presented.
Study drug dose compliance will be calculated based on the actual daily study drug total dose and the regimen prescribed study drug total dose recorded by the eCRF.
The comparison of the treatment time of each study drug, the total dose and the daily average dose of the study drug and the study drug dose compliance adopts one-factor analysis of variance, and the comparison of the compliance classification adopts chi-square test or Fisher accurate probability method.
4.2.2. Adverse events
Summary adverse events, adverse events prior to first administration, adverse events during treatment, unexpected adverse events during treatment, significant adverse events during treatment, adverse events of particular interest during treatment, adverse events of grade 3 and above during treatment, severe adverse events during treatment, adverse events related to study drug during treatment, SAEs related to study drug during treatment, instances, number of instances, and incidence of adverse events that lead to dose adjustments during treatment, permanent cessation of treatment, termination of trial, patient death were categorized and summarized according to SOC, PT.
Adverse events with incidence rate of more than or equal to 5% during treatment and drug-related adverse events are summarized according to PT classification.
Drug-related adverse events with CTC AEs graded as grade 3 or 4 during treatment were summarized by PT classification.
Adverse events with incidence rate of more than or equal to 10% during treatment and drug-related adverse events are summarized according to PT classification.
The median first occurrence time of adverse events of particular concern.
4.2.3. Vital signs
Mean ± standard deviation, maximum, minimum, median are used to describe the measurements and changes before and after treatment.
4.2.4. Laboratory test index
Table 2 examination item table
Figure BDA0002890379420000171
Figure BDA0002890379420000181
The measured values and the changed values before and after treatment are described by mean values +/-standard deviation, maximum values, minimum values and median values for blood routine, blood biochemistry, thyroid function, blood coagulation function, amylase and lipase, and the paired t test is adopted for group comparison. And describing normal and abnormal change conditions before and after treatment by adopting a cross classification table.
The routine of urine: and describing normal and abnormal change conditions before and after treatment by adopting a cross classification table.
And (3) conventionally: and describing normal and abnormal change conditions before and after treatment by adopting a cross classification table.
Describes the proportion of "abnormal, clinically significant" in subjects with abnormal changes, where the presence or absence of an abnormality is judged by the investigator.
4.2.5. Electrocardiogram
Electrocardiogram: according to the normal and abnormal conditions judged by the researchers, the change conditions of the normal and abnormal conditions before and after treatment are described.
The heart rate, PR interval, QRS interval, QT interval and QTc describe the measured values and the variation values before and after administration by using the mean value plus or minus standard deviation, the maximum value, the minimum value and the median. The electrocardiogram total evaluation result adopts a cross classification table to describe normal and abnormal change conditions before and after administration. The proportion of "clinically significant" abnormalities in subjects who describe abnormal changes, where the presence or absence of abnormalities is judged by the investigator. The list presents a post-administration exception list.
4.2.6. Physical examination
The changes of normal and abnormal before and after treatment are described.
While the compositions and methods of this application have been described in terms of preferred embodiments in light of the present disclosure, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the application.
The disclosures of all documents cited herein are incorporated by reference herein, to the extent that they provide exemplary, procedural and other details supplementary to those set forth herein.
The medicine composition has good curative effect on esophageal cancer, and can inhibit growth and proliferation of esophageal cancer cells.
SEQUENCE LISTING
<110> Ningda Ningqing pharmaceutical industry group, Inc
<120> pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody
<130> 2021.01.05
<160> 8
<170> PatentIn version 3.5
<210> 1
<211> 6
<212> PRT
<213> Synthetic sequence
<400> 1
Gln Asp Ile Asn Thr Tyr
1 5
<210> 2
<211> 3
<212> PRT
<213> Synthetic sequence
<400> 2
Arg Ala Asn
1
<210> 3
<211> 9
<212> PRT
<213> Synthetic sequence
<400> 3
Leu Gln Tyr Asp Glu Phe Pro Leu Thr
1 5
<210> 4
<211> 8
<212> PRT
<213> Synthetic sequence
<400> 4
Gly Phe Ala Phe Ser Ser Tyr Asp
1 5
<210> 5
<211> 8
<212> PRT
<213> Synthetic sequence
<400> 5
Ile Ser Gly Gly Gly Arg Tyr Thr
1 5
<210> 6
<211> 11
<212> PRT
<213> Synthetic sequence
<400> 6
Ala Asn Arg Tyr Gly Glu Ala Trp Phe Ala Tyr
1 5 10
<210> 7
<211> 107
<212> PRT
<213> Synthetic sequence
<400> 7
Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Met Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Phe Thr Cys Arg Ala Ser Gln Asp Ile Asn Thr Tyr
20 25 30
Leu Ser Trp Phe Gln Gln Lys Pro Gly Lys Ser Pro Lys Thr Leu Ile
35 40 45
Tyr Arg Ala Asn Arg Leu Val Ser Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Gln Asp Tyr Thr Leu Thr Ile Ser Ser Leu Gln Pro
65 70 75 80
Glu Asp Met Ala Thr Tyr Tyr Cys Leu Gln Tyr Asp Glu Phe Pro Leu
85 90 95
Thr Phe Gly Ala Gly Thr Lys Leu Glu Leu Lys
100 105
<210> 8
<211> 118
<212> PRT
<213> Synthetic sequence
<400> 8
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Ala Phe Ser Ser Tyr
20 25 30
Asp Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Asp Trp Val
35 40 45
Ala Thr Ile Ser Gly Gly Gly Arg Tyr Thr Tyr Tyr Pro Asp Ser Val
50 55 60
Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn Asn Leu Tyr
65 70 75 80
Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Leu Tyr Tyr Cys
85 90 95
Ala Asn Arg Tyr Gly Glu Ala Trp Phe Ala Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115

Claims (10)

1. Use of a pharmaceutical combination for the manufacture of a medicament for the treatment or prevention of esophageal cancer, the pharmaceutical combination comprising:
a) a human PD-1 antibody that comprises a light chain and a heavy chain, wherein the light chain comprises light chain complementarity determining regions LCDR1, LCDR2 and LCDR3 consisting of the amino acid sequences set forth in SEQ ID NO. 1, SEQ ID NO. 2 and SEQ ID NO. 3, respectively, and wherein the heavy chain comprises heavy chain complementarity determining regions HCDR1, HCDR2 and HCDR3 consisting of the amino acid sequences set forth in SEQ ID NO. 4, SEQ ID NO. 5 and SEQ ID NO. 6, respectively, and
b) a tyrosine kinase inhibitor, wherein the tyrosine kinase inhibitor is a compound of formula I or a pharmaceutically acceptable salt thereof,
Figure FDA0002890379410000011
2. use according to claim 1, wherein the pharmaceutically acceptable salt of the compound of formula I is the hydrochloride salt of 1- [ [ [4- (4-fluoro-2-methyl-1H-indol-5-yl) oxy-6-methoxyquinolin-7-yl ] oxy ] methyl ] cyclopropylamine, preferably the dihydrochloride.
3. The use of any one of claims 1-2, wherein the human PD-1 antibody comprises a light chain variable region having an amino acid sequence as set forth in SEQ ID No. 7 and a heavy chain variable region as set forth in SEQ ID No. 8.
4. The use of any one of claims 1-3, wherein the human PD-1 antibody is 14C12H1L 1.
5. The use according to any one of claims 1 to 4, wherein the pharmaceutical combination is a non-fixed combination.
6. The use according to claim 5, wherein the human PD-1 antibody 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.
7. Use according to claim 6, for the treatment of esophageal cancer that is not receiving a curative treatment, or that is late-relapsed or metastatic, or that is not previously receiving a systemic treatment.
8. Use according to any one of claims 6 to 7, for the treatment of esophageal squamous cell carcinoma.
9. Use according to any one of claims 6 to 8, for the treatment of advanced recurrent or metastatic esophageal squamous cell carcinoma, or unresectable esophageal squamous cell carcinoma, or esophageal squamous cell carcinoma for which the subject is not amenable to curative treatment, or esophageal squamous cell carcinoma for which the subject has not previously received systemic treatment, or has received systemic treatment.
10. The use according to any one of claims 6 to 9, wherein the hydrochloride salt of the compound of formula I is administered in a single dose of about 8 mg/dose or 10 mg/dose and the 14C12H1L1 is administered in a single dose of about 200 mg/dose.
CN202110026242.8A 2020-01-14 2021-01-08 Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody Pending CN113117072A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2020100378275 2020-01-14
CN202010037827 2020-01-14

Publications (1)

Publication Number Publication Date
CN113117072A true CN113117072A (en) 2021-07-16

Family

ID=76772242

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110026242.8A Pending CN113117072A (en) 2020-01-14 2021-01-08 Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody

Country Status (1)

Country Link
CN (1) CN113117072A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023030532A1 (en) * 2021-09-06 2023-03-09 正大天晴药业集团股份有限公司 Drug combination for treating esophageal cancer

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023030532A1 (en) * 2021-09-06 2023-03-09 正大天晴药业集团股份有限公司 Drug combination for treating esophageal cancer

Similar Documents

Publication Publication Date Title
KR102662228B1 (en) Combination of PD-1 antagonists and VEGFR/FGFR/RET tyrosine kinase inhibitors to treat cancer
TWI780994B (en) Anti-b7-h1 and anti-ctla-4 antibodies for treating non-small cell lung cancer
KR20170074962A (en) Combination therapy for cancer
EP3842070A1 (en) Drug combination of quinoline derivative and antibody
US11572405B2 (en) Combination therapy with anti-IL-8 antibodies and anti-PD-1 antibodies for treating cancer
TW201827076A (en) Methods of treating cancer using anti-pd-l1 antibodies and antiandrogens
CN111617243A (en) Pharmaceutical combination of quinoline derivatives and antibodies
CN113613674A (en) Combined pharmaceutical composition for treating small cell lung cancer
CN113939315B (en) Combined pharmaceutical composition for treating melanoma
US20240327519A1 (en) Methods and compositions for treating cancer
CN112915203A (en) Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody
TW202137984A (en) Combination of a pd-1 antagonist, a vegfr/fgfr/ret tyrosine kinase inhibitor and a cbp/beta-catenin inhibitor for treating cancer
CN113117072A (en) Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody
US20240317854A1 (en) Methods for treating cancer with anti-ilt3 antibodies
CN112915202A (en) Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody
CN114432438A (en) Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody
CN114470190A (en) Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody
CN114667159B (en) Pharmaceutical combination of quinoline derivative and PD-1 monoclonal antibody
CN114470191A (en) Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody
CN113164599B (en) Use of anti-PD-L1 monoclonal antibodies for the treatment of cancer
CN118176017A (en) Combined medicine for treating tumor
CN115779095A (en) Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody for treating colorectal 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