WO2022073515A1 - 抗pd-1抗体在联合用药中的应用 - Google Patents

抗pd-1抗体在联合用药中的应用 Download PDF

Info

Publication number
WO2022073515A1
WO2022073515A1 PCT/CN2021/122878 CN2021122878W WO2022073515A1 WO 2022073515 A1 WO2022073515 A1 WO 2022073515A1 CN 2021122878 W CN2021122878 W CN 2021122878W WO 2022073515 A1 WO2022073515 A1 WO 2022073515A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
seq
sequence
weeks
amino acid
Prior art date
Application number
PCT/CN2021/122878
Other languages
English (en)
French (fr)
Inventor
岳海涛
裴树军
苏紫琪
汤伟佳
俞金泉
李胜峰
Original Assignee
百奥泰生物制药股份有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 百奥泰生物制药股份有限公司 filed Critical 百奥泰生物制药股份有限公司
Priority to CN202180069405.9A priority Critical patent/CN116437926A/zh
Priority to EP21877032.9A priority patent/EP4226942A1/en
Priority to US18/030,886 priority patent/US20240002511A1/en
Publication of WO2022073515A1 publication Critical patent/WO2022073515A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/68033Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a maytansine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6855Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from breast cancer cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation

Definitions

  • the invention belongs to the field of biomedicine, and in particular relates to the application of anti-PD-1 antibody in combined medicine.
  • the incidence of cancer is increasing year by year, and the use of antitumor drugs is also increasing.
  • significant progress has been made in tumor treatment research, especially some antibody drugs have shown good efficacy in the treatment of malignant tumors.
  • the combined drug can be used when the anti-tumor drug is used.
  • PD-1 is an immunosuppressive receptor expressed on activated T cells, B cells and myeloid cells and is a member of the CD28 immunoglobulin superfamily.
  • PD-1 is a 55 kDa type I transmembrane glycoprotein containing a ligand-binding Ig variable domain and a cytoplasmic tail responsible for binding signaling molecules.
  • the PD-1 cytoplasmic tail contains two tyrosine-based signal transduction motifs, ITIM (Immunoreceptor Tyrosine Inhibitory Motif) and ITSM (Immunoreceptor Tyrosine Switching Motif).
  • anti-PD-1 antibodies play an important role in the combination of anti-tumor drugs.
  • the present invention discloses a method or application of anti-PD-1 antibody for combined treatment of tumor or cancer.
  • an anti-PD-1 antibody and a therapeutic agent are used in combination to treat a tumor or cancer.
  • the therapeutic agent is an antibody or antibody drug conjugate.
  • the anti-PD-1 antibody comprises at least HCDR1 set forth in SEQ ID NO:1, HCDR2 set forth in SEQ ID NO:2, HCDR3 set forth in SEQ ID NO:3, SEQ ID NO:4 One or more of LCDR1 shown in SEQ ID NO: 5, LCDR2 shown in SEQ ID NO: 6, and LCDR3 shown in SEQ ID NO: 6.
  • the anti-PD-1 antibody comprises HCDR1 shown in SEQ ID NO:1, HCDR2 shown in SEQ ID NO:2, HCDR3 shown in SEQ ID NO:3, HCDR3 shown in SEQ ID NO:4 LCDR1 shown in SEQ ID NO:5, LCDR2 shown in SEQ ID NO:5, and LCDR3 shown in SEQ ID NO:6.
  • the heavy chain variable region of the anti-PD-1 antibody comprises the sequence set forth in SEQ ID NO:7, a sequence that is at least 80% identical to the sequence set forth in SEQ ID NO:7, or An amino acid sequence with one or more conservative amino acid substitutions compared to the sequence shown in SEQ ID NO: 7; and/or
  • the light chain variable region of the anti-PD-1 antibody comprises the sequence shown in SEQ ID NO:8, a sequence with at least 80% identity to the sequence shown in SEQ ID NO:8, or a sequence shown in SEQ ID NO:8.
  • the indicated sequences are compared to amino acid sequences with one or more conservative amino acid substitutions.
  • the heavy chain variable region of the anti-PD-1 antibody comprises the sequence shown in SEQ ID NO:7
  • the light chain variable region of the anti-PD-1 antibody comprises the sequence shown in SEQ ID NO:8 the sequence shown.
  • the heavy chain of the anti-PD-1 antibody comprises the sequence set forth in SEQ ID NO:9, a sequence that is at least 80% identical to the sequence set forth in SEQ ID NO:9, or the sequence set forth in SEQ ID NO:9 The amino acid sequence with one or more conservative amino acid substitutions compared to the sequence shown in 9; and/or
  • the light chain of the anti-PD-1 antibody comprises the sequence shown in SEQ ID NO: 10, a sequence with at least 80% identity to the sequence shown in SEQ ID NO: 10, or a sequence similar to the sequence shown in SEQ ID NO: 10 An amino acid sequence with one or more conservative amino acid substitutions.
  • the anti-PD-1 antibody is Antibody A
  • the heavy chain of Antibody A comprises the sequence shown in SEQ ID NO:9
  • the light chain of Antibody A comprises the sequence shown in SEQ ID NO:10
  • Antibody A contains two heavy chains with the same sequence and two light chains with the same sequence.
  • the therapeutic agent is selected from the group consisting of antibodies or antibody drug conjugates (ADCs) against the following targets: EGFR (epidermal growth factor receptor), VEGF (vascular endothelial growth factor), VEGFR2 (vascular endothelial growth factor) factor receptor 2), CTLA4 (cytotoxic T lymphocyte-associated protein 4), PD-L1, HER2 (human epidermal growth factor receptor 2), CD20 (cluster of differentiation 20), Trop2 (human trophoblast cell surface antigen 2) , Lag3 (lymphocyte activation gene-3 molecule), TIGIT (T cell Ig and ITIM domain), CD27 (cluster of differentiation 27), OX40 (tumor necrosis factor receptor superfamily member 4), ICOS (inducible costimulator), BTLA ( B and T lymphocyte attenuating factor), TIM3 (T cell immunoglobulin mucin 3), BCMA (B cell maturation antigen), c-MET (mesenchymal epidermal transforming factor) and TAA
  • the therapeutic agent is selected from the group consisting of: anti-EGFR antibody, anti-VEGF antibody, anti-VEGFR2 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, anti-HER2 antibody, anti-CD20 antibody, anti-Trop2 antibody , anti-TIGIT antibody, anti-OX40 antibody or anti-ICOS antibody.
  • the therapeutic agent is an anti-CTLA4 antibody.
  • the anti-CTLA4 antibody is Antibody C
  • the heavy chain of Antibody C comprises the sequence shown in SEQ ID NO: 15
  • the light chain of Antibody C comprises the sequence shown in SEQ ID NO: 16
  • Antibody C comprises both sequences The same heavy chain and two light chains with the same sequence.
  • Antibody C is expressed by a-(1,6)-fucosyltransferase knockout CHO cells, such as the CHO-BAT-KF cell line disclosed in PCT/CN2018/100008.
  • the total amount of high mannose glycoforms in the Fc region of Antibody C is ⁇ 5% and/or the total amount of sialylated glycoforms is ⁇ 3%. In some embodiments, the total amount of high mannose glycoforms in the Fc region of Antibody C is about 0.1%, about 0.3%, about 0.9%, about 1.18%, about 1.7%, about 2.6%, about 3.3%, about 4.1% %, about 4.9%, about 4.99%, or a range (including endpoints) between any two of these values, or any value therein.
  • the total amount of sialylated glycoforms in the Fc region of the antibody C is about 0.1%, 0.2%, about 0.36%, about 0.8%, about 1.5%, about 2.2%, about 2.7%, about 2.9% %, 2.99%, or a range (including endpoints) between any two of these values or any value therein.
  • the total amount of high mannose glycoforms in the Fc region of the antibody C is ⁇ 2% and/or the total amount of sialylated glycoforms is ⁇ 1%.
  • the level of fucosylation in the Fc region of the antibody C is 0%-10%. In some embodiments, the level of fucosylation in the Fc region of the antibody C is 0%-5%. In some embodiments, the fucosylation content of the Fc region in the antibody C is about 0, about 0.1%, about 0.3%, about 0.4%, about 0.6%, about 1.3%, about 1.9%, about 2.2% %, about 2.8%, about 3.3%, about 3.7%, about 4.1%, about 4.5%, about 5%, or a range (including endpoints) between any two of these values or any value therein.
  • the therapeutic agent is an anti-HER2 antibody drug conjugate or an anti-Trop2 antibody drug conjugate. In some embodiments, the therapeutic agent is an anti-HER2-antibody drug conjugate (HER2-ADC). In some embodiments, the therapeutic agent is an anti-Trop2-antibody drug conjugate (Trop2-ADC).
  • the Trop2-ADC is a conjugate of BAT0807 or BAT0808 of patent application CN109078181A and a drug.
  • the therapeutic agent is an antibody drug conjugate (ADC) of Formula I or a pharmaceutically acceptable salt thereof:
  • Abu is an anti-HER2 antibody or an anti-Trop2 antibody, and p is selected from 1-10;
  • X is -H or halogen
  • R 2 is -H or C1-C6 alkyl
  • R 4 is -OH or -SH
  • R 5 is C1-C6 alkyl or benzyl
  • R 6 is C1-C6 alkyl, phenyl or benzyl
  • R 7 is -H, C1-C6 alkyl or amino acid side chain
  • R 8 is -H or C1-C6 alkyl
  • Z is independently -H or a C1-C4 hydrocarbyl group, or two Z and the carbon atom to which they are attached form a carbonyl group;
  • L is -( CH2 ) m- , wherein m is an integer and m is 1-20. In some embodiments, m is 1-10. In some embodiments, m is 5-10. In some embodiments, m is 1, 2, 3, 4, 5, 7, 9, 11, 12, 13, 15, 16, 18, 19, or 20, or a range between any two of these values (including endpoints) or any of these values.
  • R 11 is selected from -H, alkyl, aryl, cycloalkyl, cycloalkenyl, heteroaryl, and heterocycle, or two R 11 and nitrogen form a heterocycle, wherein in the heterocycle The carbons can be optionally replaced with one or two oxygens.
  • the therapeutic agent is an antibody drug conjugate (ADC) of Formula II or a pharmaceutically acceptable salt thereof:
  • Abu is an anti-HER2 antibody or an anti-Trop2 antibody
  • p is selected from 1-10.
  • p is the number of small molecule drugs bound by an antibody in the ADC, that is, the number of drug binding of the antibody, or the drug-antibody coupling ratio (DAR). In some embodiments, p is about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, or about 10. In some embodiments, p is 2-3. In some embodiments, p is about 2.
  • p is the average number of small molecule drugs bound, ie, the average number of drug binding of the antibody, or the average drug-to-antibody coupling ratio (average DAR). In some embodiments, p is 1-10, and p can be a non-integer. In some embodiments, p is about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, or a range between any two of these values (including endpoints) or any of these values. In some embodiments, p is selected from 2-8. In some embodiments, p is selected from 3-5. In some embodiments, p is selected from 3.3-3.7. In some embodiments, p is about 3.5. In some embodiments, p is about 2.1.
  • the heavy chain of the anti-HER2 antibody comprises the sequence set forth in SEQ ID NO:11, a sequence that is at least 80% identical to the sequence set forth in SEQ ID NO:11, or the sequence set forth in SEQ ID NO:11
  • the indicated sequences are compared to amino acid sequences with one or more conservative amino acid substitutions; and/or
  • the light chain of the anti-HER2 antibody comprises the sequence shown in SEQ ID NO: 12, a sequence that is at least 80% identical to the sequence shown in SEQ ID NO: 12, or has a sequence that is at least 80% identical to the sequence shown in SEQ ID NO: 12 An amino acid sequence with one or more conservative amino acid substitutions.
  • the anti-HER2 antibody is Antibody B
  • the heavy chain of Antibody B comprises the sequence set forth in SEQ ID NO:11
  • the light chain of Antibody B comprises the sequence set forth in SEQ ID NO:12.
  • the anti-Trop2 antibody is BAT0807 or BAT0808 in CN109078181A patent application. In some embodiments, p is about 2. In some embodiments, p is about 2.1.
  • the HER2-ADC is HER2-ADC B
  • HER2-ADC B is a compound represented by formula II
  • the antibody in HER2-ADC B is antibody B
  • antibody B contains two heavy chains with the same sequence and Two light chains with identical sequences; p-value 3.3-3.7, or about 3.5.
  • Antibody protein can be expressed in CHO cells by genetic engineering and obtained by purification; purification can be carried out by conventional methods, such as centrifuging the cell suspension and collecting the supernatant, and centrifuging again to further remove impurities. Methods such as Protein A affinity columns and ion exchange columns can be used to purify antibody proteins.
  • the method or use comprises administering to a patient in need thereof an effective amount of an anti-PD-1 antibody and a therapeutic agent.
  • the anti-PD-1 antibody is Antibody A.
  • the therapeutic agent is HER2- ADCB .
  • the effective dose of anti-PD-1 antibody administered is about 50 mg to 600 mg per dose.
  • the effective dose of HER2-ADC administered is about 70 mg to 400 mg per dose.
  • the therapeutic agent is an anti-CTLA4 antibody.
  • the therapeutic agent is Antibody C.
  • the effective dose of anti-CTLA4 administered is about 6 mg to 600 mg per dose.
  • the anti-PD-1 antibody and the therapeutic agent are separate dosing units for combined administration.
  • the anti-PD-1 antibody can be administered prior to administration of the therapeutic agent, after administration of the anti-therapeutic agent, or concurrently with the administration of the therapeutic agent.
  • the anti-PD-1 antibody and the therapeutic agent form a combined administration unit at the same time, and are administered in combination.
  • tumors or cancers include, but are not limited to: head and neck tumors, Hodgkin's lymphoma, non-Hodgkin's lymphoma, pre-lymphoblastic lymphoma, small non-cleaved cell lymphoma, Burkitt's lymphoma tumor, non-Burkitt lymphoma, diffuse large B-cell lymphoma, anaplastic large cell lymphoma, renal tumor, Wilms tumor, Wilms tumor, renal clear cell carcinoma, renal rhabdoid tumor, renal clear cell sarcoma, Renal primitive neuroectodermal tumor, neuroblastoma, ganglioneuroblastoma, ganglioneuroma, extracranial germ cell tumor, mature teratoma, immature teratoma, endodermal sinus tumor, yolk sac tumor , seminoma, dysgerminoma, choriocarcinoma, embryonal carcinoma
  • the present invention discloses a method for treating a tumor or cancer in a patient in need thereof, comprising administering an effective amount of an anti-PD-1 antibody and a therapeutic agent, wherein the effective amount of anti-PD-1 administered is About 50mg to 600mg per treatment cycle.
  • a treatment cycle is 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 5 weeks, 6 weeks, 7 weeks, or a range between any two of these values (including endpoint) or any of its values.
  • the anti-PD-1 antibody is Antibody A.
  • the therapeutic agent is a monoclonal antibody (anti-HER2 antibody) that specifically binds to the extracellular dimerization domain (subdomain II) of epidermal growth factor receptor 2 (HER2), such as Pertuzumab, which has a molecular weight of about 148 kDa.
  • Pertuzumab can be expressed in cells (such as CHO) by genetic engineering and obtained by purification; the purification can be carried out by conventional methods, such as centrifuging the cell suspension and collecting the supernatant, and centrifuging again to further remove impurities. Methods such as Protein A affinity columns and ion exchange columns can be used to purify antibodies.
  • the therapeutic agent is Pertuzumab, which includes Pertuzumab Or its biosimilar or ADCC effect-enhancing monoclonal antibody or defucosylated monoclonal antibody.
  • the effective amount of Pertuzumab administered is about 40 mg to 900 mg per treatment cycle.
  • a treatment cycle is 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 5 weeks, 6 weeks, 7 weeks, or a range between any two of these values (including endpoint) or any of its values.
  • the effective amount of Pertuzumab administered is about 840 mg initially, followed by 420 mg every 3 weeks.
  • the therapeutic agent is a recombinant humanized immunoglobulin G1 (IgG1) monoclonal antibody that binds VEGF-A and inhibits its binding to VEGF receptor-2 (VEGFR-2) (anti-VEGF receptor-2).
  • VEGF antibody such as bevacizumab.
  • Bevacizumab can be expressed in cells (such as CHO) by genetic engineering and obtained by purification; purification can be carried out by conventional methods, such as centrifuging the cell suspension and collecting the supernatant, and centrifuging again to further remove impurities. Methods such as Protein A affinity columns and ion exchange columns can be used to purify antibodies.
  • the therapeutic agent is bevacizumab, which includes or its biosimilars, such as Or BAT1706, or ADCC effect-enhancing mAb or defucosylated mAb.
  • the effective amount of bevacizumab administered is about 50 mg to 400 mg per treatment cycle.
  • a treatment cycle is 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 5 weeks, 6 weeks, 7 weeks, or a range between any two of these values (including endpoint) or any of its values.
  • the effective amount of bevacizumab administered is about 5 mg/kg to 15 mg/kg every 2 weeks or every 3 weeks.
  • the effective amount of bevacizumab administered is about 5 mg/kg, 7.5 mg/kg, 10 mg/kg, or 15 mg/kg every 2 weeks or every 3 weeks.
  • the effective amount of bevacizumab administered is about 5 mg/kg every 2 weeks, 10 mg/kg every 2 weeks, 7.5 mg/kg every 3 weeks, 15 mg/kg every 3 weeks 1 time a week.
  • the therapeutic agent is an antibody targeting CD20 (anti-CD20 antibody), such as ofatumumab, which is a fully human IgG1 kappa monoclonal antibody, or obinutuzumab.
  • anti-CD20 antibody such as ofatumumab, which is a fully human IgG1 kappa monoclonal antibody, or obinutuzumab.
  • Ofatumumab and obinutuzumab can be expressed in cells (such as CHO) through genetic engineering and obtained by purification; purification can be carried out by conventional methods, such as first centrifuging the cell suspension and collecting the supernatant, and centrifuging again to further remove impurities. Methods such as Protein A affinity columns and ion exchange columns can be used to purify antibodies.
  • the therapeutic agent is ofatumumab, which comprises ofatumumab or Its biosimilar or ADCC effect-enhancing monoclonal antibody or defucosylated monoclonal antibody, such as BAT4406F disclosed in CN109096399A.
  • ofatumumab is administered in an effective amount of about 10 mg to 2000 mg per treatment cycle.
  • a treatment cycle is 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 5 weeks, 6 weeks, 7 weeks, or a range between any two of these values (including endpoint) or any of its values.
  • the effective amount of ofatumumab administered is about 20 mg weekly or monthly.
  • the effective amount of ofatumumab administered is about 300 mg initially, 1000 mg after 1 week, and then 1000 mg every 4 weeks or every 8 weeks. In some embodiments, the effective amount of ofatumumab administered is about 300 mg initially, 2000 mg after 1 week, and then 2000 mg every 1 week or every 4 weeks.
  • the therapeutic agent is obinutuzumab, including Its biosimilar, or ADCC effect-enhancing monoclonal antibody or defucosylated monoclonal antibody, such as BAT4306F described in CN109096399A.
  • the effective amount of obinutuzumab administered is about 10 mg to 2000 mg per treatment cycle.
  • a treatment cycle is 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 5 weeks, 6 weeks, 7 weeks, or a range between any two of these values (including endpoint) or any of its values.
  • the effective amount of obinutuzumab administered is 100 mg on day 1, 900 mg on day 2, 1,000 mg on days 8, 15, and 1,000 mg per course thereafter.
  • the effective amount of obinutuzumab administered is 1,000 mg each on day 1, day 8, day 15, and then 1,000 mg per course of treatment thereafter. Each course of treatment can be 1 month or 2 months.
  • the therapeutic agent is an antibody targeting T lymphocyte-associated antigen 4 (CTLA4) (anti-CTLA4 antibody), such as ipilimumab, which is an IgG1 kappa immunoglobulin with a molecular weight of about 148 kDa.
  • CTL4 T lymphocyte-associated antigen 4
  • Ipilimumab can be expressed in cells (such as CHO) through genetic engineering and obtained by purification; the purification can be carried out by conventional methods, such as centrifuging the cell suspension and collecting the supernatant, and centrifuging again to further remove impurities. Methods such as Protein A affinity columns and ion exchange columns can be used to purify antibodies.
  • the therapeutic agent is ipilimumab, ipilimumab including Yervoy TM or its biosimilar or ADCC effect enhancing mAb or defucosylated ipilimumab, as described in WO2014089113 .
  • the effective amount of ipilimumab administered is about 30 mg to 300 mg per treatment cycle.
  • a treatment cycle is 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 5 weeks, 6 weeks, 7 weeks, or a range between any two of these values (including endpoint) or any of its values.
  • the effective amount of ipilimumab administered is 1 mg/kg, 3 mg/kg, or 10 mg/kg every 3, 6, or 12 weeks. In some embodiments, the effective amount of ipilimumab administered is 1 mg/kg every 3 or 6 weeks. In some embodiments, the effective amount of ipilimumab administered is 3 mg/kg every 3 or 6 weeks. In some embodiments, the effective amount of ipilimumab administered is 10 mg/kg every 3 weeks or every 12 weeks.
  • the therapeutic agent is a Trop2-ADC described herein.
  • the Trop2-ADC is administered in an effective amount of about 60 mg to 400 mg per treatment cycle.
  • a treatment cycle is 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 5 weeks, 6 weeks, 7 weeks, or a range between any two of these values (including endpoint) or any of its values.
  • the Trop2-ADC is administered in an effective amount of 0.2, 0.5, 1, 2, 4, 6, 8, or 10 mg/kg weekly to once every 3 weeks.
  • the effective amount of Trop2-ADC administered is 0.2, 0.5, 1, 2, 4, 6, 8 or 10 mg/kg weekly or once every 3 weeks.
  • the therapeutic agent is a HER2-ADC.
  • the effective amount of HER2-ADC administered is about 70 mg to 400 mg per treatment cycle.
  • a treatment cycle is 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 5 weeks, 6 weeks, 7 weeks, or a range between any two of these values (including endpoint) or any of its values.
  • the HER2-ADC is HER2- ADCB .
  • an anti-PD-1 antibody and a therapeutic agent can be separately formulated as a pharmaceutical composition and administered to a variety of forms suitable for the chosen route of administration Patient administration, eg, by parenteral, intravenous (iv), intramuscular, topical or subcutaneous routes.
  • the anti-PD-1 antibody and therapeutic agent can be administered separately by intravenous infusion.
  • anti-PD-1 antibody and therapeutic agent will depend on the nature of the drug, the extent to which internalization, transport and release of the drug is triggered on the cell surface, the disease being treated, and the condition of the patient (eg, age, gender, weight, etc.).
  • the anti-PD-1 antibody is administered at about 1 mg/kg to 10 mg/kg per administration or a formulation containing such dose of the anti-PD-1 antibody.
  • the anti-PD-1 antibody is about 1 mg/kg, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg per administration , about 8 mg/kg, about 9 mg/kg, about 10 mg/kg, or a range between any two of these values (including endpoints) or any value therein, or a formulation containing this dose of anti-PD-1 antibody.
  • therapeutically effective amounts of Pertuzumab and anti-PD-1 antibody are administered to the subject patient separately or simultaneously.
  • the dosing cycle of Pertuzumab and anti-PD-1 antibody may be the same or different.
  • the Pertuzumab per administration is about 1 mg/kg to 12 mg/kg or a formulation containing this dose of Pertuzumab. In some embodiments, the Pertuzumab per administration is about 1 mg/kg, about 1.2 mg/kg, about 2 mg/kg, about 2.4 mg/kg, about 3 mg/kg, about 3.6 mg/kg, about 4mg/kg, about 4.8mg/kg, about 5mg/kg, about 5.5mg/kg, about 6mg/kg, about 6.9mg/kg, about 7mg/kg, about 8.4mg/kg, about 9mg/kg, about 11mg /kg, about 12 mg/kg, or a range between any two of these values (including the endpoints) or any value therein, or a formulation containing Pertuzumab at this dose.
  • a therapeutically effective amount of bevacizumab and an anti-PD-1 antibody is administered to the subject patient separately or simultaneously.
  • the dosing cycle of bevacizumab and anti-PD-1 antibody may be the same or different.
  • the bevacizumab per administration is about 1 mg/kg to 9 mg/kg or a formulation containing bevacizumab at this dose. In some embodiments, the bevacizumab per administration is about 1 mg/kg, about 1.2 mg/kg, about 2 mg/kg, about 2.4 mg/kg, about 3 mg/kg, about 3.6 mg/kg, about 4 mg/kg, about 4.8 mg/kg, about 5 mg/kg, about 5.5 mg/kg, about 6 mg/kg, about 6.9 mg/kg, about 7 mg/kg, about 8.4 mg/kg, about 9 mg/kg, or these A range (including endpoints) between any two values of a numerical value, or any value therein, or a formulation containing bevacizumab at that dose.
  • therapeutically effective amounts of ofatumumab and anti-PD-1 antibody are administered to the subject patient separately or simultaneously.
  • the dosing cycle of ofatumumab and anti-PD-1 antibody may be the same or different.
  • ofatumumab per administration is about 0.5 mg/kg to 18 mg/kg or a formulation containing such dose of ofatumumab. In some embodiments, ofatumumab per administration is about 0.5 mg/kg, about 1 mg/kg, about 1.2 mg/kg, about 2 mg/kg, about 2.4 mg/kg, about 3 mg/kg, about 3.6mg/kg, about 4mg/kg, about 4.8mg/kg, about 5mg/kg, about 5.5mg/kg, about 6mg/kg, about 6.9mg/kg, about 7mg/kg, about 8.4mg/kg, about 9 mg/kg, about 11 mg/kg, about 13 mg/kg, about 14 mg/kg, about 15 mg/kg, about 17 mg/kg, about 18 mg/kg, or a range between any two of these values (including endpoints) or any of these values, or a formulation containing this dose of ofatumumab.
  • a therapeutically effective amount of obinutuzumab and an anti-PD-1 antibody is administered to the subject patient separately or simultaneously.
  • the dosing cycle of obinutuzumab and the anti-PD-1 antibody can be the same or different.
  • obinutuzumab per administration is about 0.5 mg/kg to 15 mg/kg or a formulation containing this dose of ofatumumab.
  • ofatumumab per administration is about 0.5 mg/kg, about 1 mg/kg, about 1.2 mg/kg, about 2 mg/kg, about 2.4 mg/kg, about 3 mg/kg, about 3.6mg/kg, about 4mg/kg, about 4.8mg/kg, about 5mg/kg, about 5.5mg/kg, about 6mg/kg, about 6.9mg/kg, about 7mg/kg, about 8.4mg/kg, about 9 mg/kg, about 11 mg/kg, about 13 mg/kg, about 14 mg/kg, about 15 mg/kg, or a range between any two of these values (including endpoints) or any value therein, or a dose containing Formulations of falimumab.
  • a therapeutically effective amount of an anti-CTLA4 antibody eg, ipilimumab or defucosylated ipilimumab
  • an anti-PD-1 antibody is administered separately or simultaneously to the subject patient.
  • the anti-CTLA4 antibody eg, ipilimumab or defucosylated ipilimumab
  • the anti-PD-1 antibody may be administered on the same or different cycles.
  • the anti-CTLA4 antibody (eg, ipilimumab or defucosylated ipilimumab) is about 0.1 mg/kg to 10 mg/kg, or about 0.5 mg/kg to 10 mg per administration /kg or a formulation containing this dose of an anti-CTLA4 antibody such as ipilimumab or defucosylated ipilimumab.
  • the ipilimumab is about 0.5 mg/kg, about 1 mg/kg, about 1.2 mg/kg, about 2 mg/kg, about 2.4 mg/kg, about 3 mg/kg, about 3.6 mg/kg per administration mg/kg, about 4mg/kg, about 4.8mg/kg, about 5mg/kg, about 5.5mg/kg, about 6mg/kg, about 6.9mg/kg, about 7mg/kg, about 8.4mg/kg, about 9mg /kg, about 10 mg/kg, or a range between any two of these values (including the endpoints) or any value therein, or a formulation containing ipilimumab at this dose.
  • therapeutically effective amounts of Trop2-ADC and anti-PD-1 antibody are administered to the subject patient separately or simultaneously.
  • the dosing cycle of Trop2-ADC and anti-PD-1 antibody can be the same or different.
  • the Trop2-ADC per administration is about 0.5 mg/kg to 7 mg/kg or a formulation containing this dose of Trop2-ADC. In some embodiments, the Trop2-ADC per administration is about 1 mg/kg, about 1.2 mg/kg, about 2 mg/kg, about 2.4 mg/kg, about 3 mg/kg, about 3.6 mg/kg, about 4 mg/kg kg, about 4.8 mg/kg, about 5 mg/kg, about 5.5 mg/kg, about 7 mg/kg, or a range between any two of these values (including endpoints) or any value therein, or containing such a dose Trop2 - Formulation of ADC.
  • a therapeutically effective amount of the HER2-ADC and anti-PD-1 antibody is administered to the subject patient separately or simultaneously.
  • the dosing cycle of the HER2-ADC and the anti-PD-1 antibody can be the same or different.
  • the HER2-ADC per administration is about 1 mg/kg to 6 mg/kg or a formulation containing this dose of HER2-ADC. In some embodiments, the HER2-ADC per administration is about 1 mg/kg, about 1.2 mg/kg, about 2 mg/kg, about 2.4 mg/kg, about 3 mg/kg, about 3.6 mg/kg, about 4 mg/kg kg, about 4.8 mg/kg, about 5 mg/kg, about 5.5 mg/kg, about 6 mg/kg, or a range between any two of these values (including endpoints) or any value therein, or a dose containing HER2 - Formulation of ADC.
  • the present invention discloses a method of treating a tumor or cancer, comprising administering to a patient in need thereof an effective amount of an anti-PD-1 antibody (or formulation) and a therapeutic agent (or formulation); wherein the anti-PD-1 antibody (or formulation) is administered in an effective amount.
  • An effective amount of PD-1 antibody is about 50 mg to 600 mg (or a preparation containing such dose of anti-PD-1 antibody) in a single administration.
  • the therapeutic agent is a HER2-ADC.
  • the effective amount of HER2-ADC is about 70 mg to 400 mg (or formulation containing such dose of HER2-ADC) in a single administration.
  • the therapeutic agent is an anti-CTLA4 antibody.
  • the anti-CTLA4 antibody is Antibody C.
  • the effective amount of anti-CTLA4 antibody is about 6 mg to 600 mg (or a formulation containing such dose of anti-CTLA4 antibody) in a single administration. Dosage schedule and mode of administration depend on benefit-risk assessment and general clinical practice guidelines for anti-PD-1 antibody (or formulation), HER2-ADC (or formulation), or anti-CTLA4 antibody (or formulation) in certain patient populations .
  • the effective amount of the anti-PD-1 antibody administered to the patient per treatment cycle is about 50 mg to 600 mg of the anti-PD-1 antibody (or a formulation containing this dose of the anti-PD-1 antibody) per treatment cycle for the patient
  • the effective amount of HER2-ADC administered in a cycle is about 70 mg to 400 mg (or a formulation containing this dose of HER2-ADC).
  • the effective amount of the anti-PD-1 antibody administered to the patient per treatment cycle is about 50 mg to 600 mg of the anti-PD-1 antibody (or a formulation containing this dose of the anti-PD-1 antibody) per treatment cycle for the patient
  • An effective amount of anti-CTLA4 administered in a cycle is about 6 mg to 600 mg (or a formulation containing this dose of anti-CTLA4 antibody).
  • the effective amount of the anti-PD-1 antibody administered to the patient per treatment cycle is about 50 mg, about 60 mg, about 80 mg, about 120 mg, about 200 mg, about 250 mg, about 290 mg, about 300 mg, about 330 mg, about 380 mg, about 400 mg, about 434 mg, about 480 mg, about 500 mg, about 567 mg, about 580 mg, about 600 mg, or a range between any two of these values (including endpoints) or any value therein, or containing such a dose of anti-PD -1 Antibody preparation.
  • one treatment cycle is administered once every 1 week to 7 weeks.
  • the effective amount of anti-PD-1 antibody administered in each treatment cycle is 100 mg to 200 mg, or a formulation containing such dose of anti-PD-1 antibody; wherein one treatment cycle is about 1 week, about 2 weeks , about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, or a range (including endpoints) between any two of these values or any value therein. In some embodiments, one treatment cycle is about 1 week, about 2 weeks, about 3 weeks, or about 4 weeks.
  • the patient is administered an effective amount of anti-PD-1 antibody in each treatment cycle of about 200 mg to about 300 mg, or a formulation containing such dose of anti-PD-1 antibody; wherein one treatment cycle is about 1 week, About 2 weeks, about 3 weeks, or about 4 weeks.
  • the effective amount of the anti-PD-1 antibody administered to the patient per treatment cycle is about 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, about 200 mg, or a range between any two of these values (including endpoints) or any value therein, or a formulation containing such doses of anti-PD-1 antibody; wherein one treatment cycle is about 1 week, about 2 weeks, about 3 weeks, or about 4 weeks.
  • the effective amount of anti-PD-1 antibody administered to the patient per treatment cycle is about 100 mg, or a formulation containing such dose of anti-PD-1 antibody; wherein one treatment cycle is about 1 week, about 2 weeks , about 3 weeks or about 4 weeks.
  • the patient is administered an effective amount of anti-PD-1 antibody per treatment cycle of about 90 mg to 110 mg, or a formulation containing such dose of anti-PD-1 antibody; such as about 100 mg administered once.
  • the effective amount of anti-PD-1 antibody administered to the patient per treatment cycle is about 120 mg, or a formulation containing this dose of anti-PD-1 antibody; wherein one treatment cycle is about 1 week, about 2 weeks , about 3 weeks or about 4 weeks.
  • the patient is administered an effective amount of anti-PD-1 antibody per treatment cycle of about 100 mg to 140 mg, or a formulation containing such dose of anti-PD-1 antibody; such as about 120 mg administered once.
  • the effective amount of anti-PD-1 antibody administered to the patient per treatment cycle is about 160 mg, or a formulation containing this dose of anti-PD-1 antibody; wherein one treatment cycle is about 1 week, about 2 weeks , about 3 weeks or about 4 weeks.
  • the patient is administered an effective amount of anti-PD-1 antibody per treatment cycle of about 150 mg to 190 mg, or a formulation containing such dose of anti-PD-1 antibody; such as about 160 mg administered once.
  • the effective amount of anti-PD-1 antibody administered to the patient per treatment cycle is about 200 mg, or a formulation containing such dose of anti-PD-1 antibody; wherein one treatment cycle is about 1 week, about 2 weeks , about 3 weeks or about 4 weeks.
  • the patient is administered an effective amount of anti-PD-1 antibody per treatment cycle of about 190 mg to 230 mg, or a formulation containing such dose of anti-PD-1 antibody; such as about 200 mg administered once.
  • the effective amount of anti-PD-1 antibody is about 100 mg to 600 mg every 3 weeks. In some embodiments, the effective amount of anti-PD-1 antibody is about 100 mg, about 200 mg, about 300 mg, about 400 mg, about 500 mg, or about 600 mg every 3 weeks. In some embodiments, the effective amount of anti-PD-1 antibody is about 100 mg, about 300 mg, or about 600 mg every 3 weeks.
  • the effective amount of HER2-ADC administered to the patient per treatment cycle is about 70 mg, about 90 mg, about 120 mg, about 160 mg, about 180 mg, about 200 mg, about 230 mg, about 250 mg, about 280 mg, about 300 mg, about 310 mg, about 334 mg, about 350 mg, about 370 mg, about 380 mg, about 390 mg, about 400 mg, or a range between any two of these values (including endpoints) or any value therein, or a HER2-ADC containing such dose preparation.
  • one treatment cycle is administered once every 1 week to 4 weeks.
  • the effective amount of HER2-ADC administered in each treatment cycle is 100 mg to 200 mg, or a formulation containing such dose of HER2-ADC; wherein one treatment cycle is about 1 week, about 2 weeks, about 3 weeks , about 4 weeks, or a range (including endpoints) between any two of these values or any value therein.
  • the patient is administered an effective amount of HER2-ADC per treatment cycle of about 200 mg to about 300 mg, or a formulation containing such dose of HER2-ADC; wherein one treatment cycle is about 1 week, about 2 weeks, About 3 weeks, or about 4 weeks.
  • the effective amount of HER2-ADC administered to the patient per treatment cycle is about 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, about 200 mg, or a range between any two of these values (including endpoints) or any value therein, or a formulation containing this dose of HER2-ADC; wherein one treatment cycle is about 1 week, about 2 weeks, about 3 weeks or about 4 weeks.
  • the patient is administered an effective amount of HER2-ADC per treatment cycle of about 100 mg, or a formulation containing such dose of HER2-ADC; wherein one treatment cycle is about 1 week, about 2 weeks, about 3 weeks or about 4 weeks.
  • the patient is administered an effective amount of HER2-ADC per treatment cycle of about 90 mg to 110 mg, or a formulation containing such dose of HER2-ADC; such as about 100 mg administered once.
  • the effective amount of HER2-ADC administered to the patient per treatment cycle is about 130 mg, or a formulation containing this dose of HER2-ADC; wherein one treatment cycle is about 1 week, about 2 weeks, about 3 weeks or about 4 weeks.
  • the patient is administered an effective amount of HER2-ADC per treatment cycle of about 100 mg to 140 mg, or a formulation containing such dose of HER2-ADC; such as about 130 mg administered once.
  • the patient is administered an effective amount of HER2-ADC per treatment cycle of about 170 mg, or a formulation containing this dose of HER2-ADC; wherein one treatment cycle is about 1 week, about 2 weeks, about 3 weeks or about 4 weeks.
  • the patient is administered an effective amount of HER2-ADC per treatment cycle of about 150 mg to 190 mg, or a formulation containing such dose of HER2-ADC; such as about 170 mg administered once.
  • the patient is administered an effective amount of HER2-ADC per treatment cycle of about 200 mg, or a formulation containing such dose of HER2-ADC; wherein one treatment cycle is about 1 week, about 2 weeks, about 3 weeks or about 4 weeks.
  • the patient is administered an effective amount of HER2-ADC per treatment cycle of about 190 mg to 230 mg, or a formulation containing such dose of HER2-ADC; such as about 200 mg administered once.
  • the effective amount of the anti-CTLA4 antibody administered to the patient per treatment cycle is about 6 mg, about 8 mg, about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 50 mg, about 60 mg, about 80 mg, about 120 mg, about 200 mg, about 250 mg, about 290 mg, about 300 mg, about 330 mg, about 380 mg, about 400 mg, about 434 mg, about 480 mg, about 500 mg, about 567 mg, about 580 mg, about 600 mg, or these A range (including endpoints) between any two values of a numerical value, or any value therein, or a formulation containing such dose of anti-CTLA4 antibody.
  • one treatment cycle is administered once every 1 week to 7 weeks.
  • the effective amount of anti-CTLA4 antibody administered per treatment cycle is 6 mg to 80 mg, or a formulation containing such dose of anti-CTLA4 antibody; wherein one treatment cycle is about 1 week, about 2 weeks, about 3 weeks , about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, or a range (including endpoints) between any two of these values, or any value therein.
  • the effective amount of anti-CTLA4 antibody administered in each treatment cycle is 100 mg to 200 mg, or a formulation containing such dose of anti-CTLA4 antibody; wherein one treatment cycle is about 1 week, about 2 weeks, about 3 weeks , about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, or a range (including endpoints) between any two of these values, or any value therein. In some embodiments, one treatment cycle is about 1 week, about 2 weeks, about 3 weeks, or about 4 weeks.
  • the patient is administered an effective amount of anti-CTLA4 antibody in each treatment cycle of about 200 mg to about 300 mg, or a formulation containing such dose of anti-CTLA4 antibody; wherein one treatment cycle is about 1 week, about 2 weeks, About 3 weeks, or about 4 weeks.
  • the effective amount of the anti-CTLA4 antibody administered to the patient per treatment cycle is about 6 mg, about 8 mg, about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 50 mg , about 60 mg, 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, or a range (including endpoints) between any two of these values or any value therein, or A preparation containing this dose of anti-CTLA4 antibody; wherein, one treatment cycle is about 1 week, about 2 weeks, about 3 weeks or about 4 weeks.
  • the effective amount of anti-CTLA4 antibody administered to the patient per treatment cycle is about 6 mg, or a formulation containing such dose of anti-CTLA4 antibody; wherein one treatment cycle is about 1 week, about 2 weeks, about 3 weeks or about 4 weeks. In some embodiments, the patient is administered about 6 mg once per treatment cycle.
  • the effective amount of anti-CTLA4 antibody administered to the patient per treatment cycle is about 15 mg, or a formulation containing such dose of anti-CTLA4 antibody; wherein one treatment cycle is about 1 week, about 2 weeks, about 3 weeks or about 4 weeks. In some embodiments, the patient is administered about 15 mg once per treatment cycle.
  • the patient is administered an effective amount of anti-CTLA4 antibody in each treatment cycle of about 23 mg, or a formulation containing such dose of anti-CTLA4 antibody; wherein one treatment cycle is about 1 week, about 2 weeks, about 3 weeks or about 4 weeks. In some embodiments, the patient is administered about 23 mg once per treatment cycle.
  • the effective amount of anti-CTLA4 antibody administered to the patient per treatment cycle is about 60 mg, or a formulation containing such dose of anti-CTLA4 antibody; wherein one treatment cycle is about 1 week, about 2 weeks, about 3 weeks or about 4 weeks. In some embodiments, the patient is administered about 60 mg once per treatment cycle.
  • the effective amount of the anti-CTLA4 antibody is about 6 mg to 600 mg once every 3 weeks. In some embodiments, the effective amount of the anti-CTLA4 antibody is about 6 mg, about 20 mg, about 30 mg, about 100 mg, about 200 mg, about 230 mg, about 300 mg, about 400 mg, about 500 mg, or about 600 mg every 3 weeks. In some embodiments, the effective amount of the anti-CTLA4 antibody is about 6 mg, about 30 mg, or about 60 mg every 3 weeks.
  • the patient is administered an anti-PD-1 antibody and a therapeutic agent (or a combination of an anti-PD-1 antibody and a therapeutic agent) once per treatment cycle.
  • the anti-PD-1 antibody and therapeutic agent or combination of anti-PD-1 antibody and therapeutic agent
  • the patient is only dosed 1 or 4 times per treatment cycle. In some embodiments, the patient is treated with one treatment cycle. In some embodiments, the patient is treated with multiple (eg, 2, 3, or 4) treatment cycles. In some embodiments, the patient receives treatment until the condition resolves and no longer requires treatment. In some embodiments, the patient receives multiple cycles of anti-PD-1 antibody therapy and 4 cycles of anti-CTLA4 antibody therapy.
  • a method for treating a tumor or cancer comprising: administering to a patient in need thereof about 50 mg to 200 mg, about 200 mg to 300 mg, about 300 mg to 400 mg, or about 400 mg to 600 mg, such as about 100 mg, about 120 mg, about 200 mg, or about 400 mg of an anti-PD-1 antibody, a formulation containing the anti-PD-1 antibody at this dose; also about 70 mg to 100 mg, about 100 mg to 200 mg, about 200 mg to 300 mg, or About 300 mg to 400 mg, such as about 100 mg, about 120 mg, about 150 mg, or about 200 mg of HER2-ADC, formulations containing this dose of HER2-ADC.
  • the patient is treated with a single dose of anti-PD-1 antibody, and a single dose of HER2-ADC. In some embodiments, the patient is treated with a single dose of an anti-PD-1 antibody and a HER2-ADC composition.
  • the anti-PD-1 antibody 200 mg and the HER2-ADC 2.4 mg/kg are administered once every 3 weeks. In some embodiments, the anti-PD-1 antibody is administered at 200 mg every 3 weeks and the HER2-ADC is administered at 3.6 mg/kg every 3 weeks.
  • a method for treating a tumor or cancer comprising: administering to a patient in need thereof about 50 mg to 200 mg, about 200 mg to 300 mg, about 300 mg to 400 mg, or about 400 mg to 600 mg, such as about 100 mg, about 120 mg, about 200 mg, or about 400 mg of an anti-PD-1 antibody, a formulation containing this dose of an anti-PD-1 antibody; also administering about 6 mg to 80 mg, about 80 mg to 200 mg, about 200 mg to 300 mg, or About 300 mg to 600 mg, such as about 6 mg, about 12 mg, about 25 mg, about 35 mg, about 50 mg, about 75 mg, about 125 mg, or about 200 mg of anti-CTLA4 antibody, formulations containing this dose of anti-CTLA4 antibody.
  • the patient is treated with a single dose of an anti-PD-1 antibody, and a single dose of an anti-CTLA4 antibody. In some embodiments, the patient is treated with a single dose of the anti-PD-1 antibody and anti-CTLA4 antibody combination.
  • the anti-PD-1 antibody 200 mg and the anti-CTLA4 antibody 0.1 mg/kg are administered once every 3 weeks. In some embodiments, the anti-PD-1 antibody is administered at 200 mg every 3 weeks and the anti-CTLA4 antibody is administered at 1 mg/kg every 3 weeks. In some embodiments, the patient's symptoms are relieved after a single dose is administered. In some embodiments, after a single dose is administered, the patient's symptoms are not relieved as expected, and the patient is administered about 50 mg to 600 mg of the anti-PD-1 antibody and about 70 mg to 400 mg of the HER2-ADC, respectively.
  • the patient is further administered a composition of about 50 mg to 600 mg of the anti-PD-1 antibody and about 70 mg to 400 mg of the HER2-ADC. In some embodiments, after a single dose is administered and the patient's symptoms are not relieved as expected, the patient is administered about 50 mg to 600 mg of anti-PD-1 antibody and about 6 mg to 600 mg of anti-CTLA4 antibody, respectively. In some embodiments, after a single dose is administered and the patient's symptoms are not relieved as expected, the patient is administered a combination of about 50 mg to 600 mg of anti-PD-1 antibody and about 6 mg to 600 mg of anti-CTLA4 antibody.
  • the anti-PD-1 antibody (or formulation), HER2-ADC (or formulation), anti-CTLA4 antibody (or formulation) is administered by subcutaneous (s.c.) injection, intraperitoneal (i.p.) injection, parenteral injection, Administration by intra-arterial injection or intravenous (i.v.) injection.
  • the anti-PD-1 antibody (or formulation), HER2-ADC (or formulation) is administered by infusion.
  • the anti-PD-1 antibody (or formulation), HER2-ADC (or formulation) is administered as a bolus injection.
  • the anti-PD-1 antibody (or formulation), the anti-CTLA4 antibody (or formulation) are administered by infusion.
  • the anti-PD-1 antibody (or formulation), anti-CTLA4 antibody (or formulation) is administered as a bolus injection.
  • the anti-PD-1 antibody (or formulation), HER2-ADC (or formulation), anti-CTLA4 antibody (or formulation) is administered by intravenous (i.v.) infusion.
  • the duration of the intravenous infusion is about 50 minutes, about 55 minutes, about 60 minutes, about 65 minutes, about 70 minutes, about 75 minutes, about 81 minutes, about 87 minutes, about 90 minutes, about 95 minutes minutes, or the range (including the endpoints) between any two of these values, or any value therein.
  • anti-PD-1 antibodies or formulations
  • therapeutic agents or formulations
  • other therapeutic methods for the treatment of tumors or cancers such as chemotherapy, radiotherapy, surgery, and the like.
  • the present invention discloses the use of anti-PD-1 antibodies and therapeutic agents in the preparation of medicaments for treating tumors or cancers.
  • the therapeutic agent is selected from the group consisting of: anti-EGRR antibody, anti-VEGF antibody, anti-VEGFR2 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, anti-HER2 antibody, anti-CD20 antibody, anti-Trop2 antibody , anti-TIGIT antibody, anti-OX40 antibody and anti-ICOS antibody, anti-HER2 antibody-drug conjugate and anti-Trop2 antibody-drug conjugate.
  • a drug for treating a tumor or cancer includes an anti-PD-1 antibody and a therapeutic agent.
  • the anti-PD-1 antibody is Antibody A.
  • the therapeutic agent is a HER2-ADC.
  • the HER2-ADC is HER2- ADCB .
  • the therapeutic agent is an anti-CTLA4 antibody.
  • the anti-CTLA4 antibody is Antibody C.
  • the present invention also discloses a kit comprising an anti-PD-1 antibody (or preparation), a therapeutic agent (or preparation) and an anti-PD-1 antibody (or preparation) for instructing a patient in need to administer the anti-PD-1 antibody (or preparation) ) and instructions for the therapeutic agent (or preparation).
  • the present invention also discloses a kit comprising a composition (or preparation) of an anti-PD-1 antibody and a therapeutic agent and used for guiding the administration of the anti-PD-1 antibody and treatment to a patient in need thereof Instructions for the composition (or formulation) of the agent.
  • the present invention also discloses a pharmaceutical composition suitable for injection comprising an anti-PD-1 antibody and a therapeutic agent, such as a bolus injection type pharmaceutical composition or an infusion (drip) type pharmaceutical composition.
  • a pharmaceutical composition suitable for injection comprising an anti-PD-1 antibody and a therapeutic agent, such as a bolus injection type pharmaceutical composition or an infusion (drip) type pharmaceutical composition.
  • the pharmaceutical composition comprises at least 0.1% anti-PD-1 antibody and 0.1% therapeutic agent.
  • the percentages of antibody and therapeutic agent can vary and are between about 2% and about 90% by weight of a given dosage form.
  • the amount of anti-PD-1 antibody and therapeutic agent in such a therapeutically useful pharmaceutical composition can be an effective amount for administration.
  • the present invention also discloses a preparation method of the above-mentioned pharmaceutical composition: respectively combining the anti-PD-1 antibody and the therapeutic agent (or the composition of the anti-PD-1 antibody and the therapeutic agent) described herein with a pharmaceutically acceptable suitable injection carrier (such as water for injection, physiological saline, etc.).
  • a pharmaceutically acceptable suitable injection carrier such as water for injection, physiological saline, etc.
  • the present invention uses anti-PD-1 antibody (or preparation) and therapeutic agent (or preparation) in tumor or cancer treatment, which can alleviate symptoms.
  • Figure 1 shows that the combined administration of anti-PD-1 antibody and HER2-ADC inhibits the proliferation of tumor cells.
  • Figure 2 shows the change in tumor volume after the combined administration of Antibody A and Antibody C; the mean body weight is expressed as mean ⁇ SEM.
  • Figure 3 shows tumor weights at day 27 after co-administration of Antibody A and Antibody C; data are presented as mean ⁇ SEM.
  • vs G1 except for the G5 group, the test method for other groups was the independent sample T test. Due to the non-normal distribution of the data in the G5 group, the test method was two independent samples test, Mann-Whitney test. * means P ⁇ 0.05, ** means P ⁇ 0.01, *** means P ⁇ 0.001.
  • Figure 4 shows the changes in the body weight of mice after co-administration of Antibody A and Antibody C.
  • an entity refers to one or more of such entities, eg "an antibody” should be understood to mean one or more antibodies, thus the term “an” (or “an” ), “one or more” and “at least one” are used interchangeably herein.
  • compositions, methods and the like include the recited elements, such as components or steps, but do not exclude others.
  • Consisting essentially of means that the compositions and methods exclude other elements that have an essential effect on the characteristics of the combination, but do not exclude elements that have no essential effect on the compositions or methods.
  • Consisting of means excluding elements not specifically recited.
  • polypeptide is intended to encompass the singular “polypeptide” as well as the plural “polypeptide”, and refers to a molecule formed from amino acid monomers linked linearly by amide bonds (also known as peptide bonds).
  • polypeptide refers to any single chain or chains of two or more amino acids, and does not refer to a particular length of the product.
  • the definition of “polypeptide” includes a peptide, dipeptide, tripeptide, oligopeptide, "protein”, “amino acid chain” or any other term used to refer to two or more amino acid chains, and the term “polypeptide” may Used in place of, or used interchangeably with, any of the above terms.
  • polypeptide is also intended to refer to the product of post-expression modifications of the polypeptide, including but not limited to glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage or non-native Amino acid modifications that occur.
  • a polypeptide may be derived from a natural biological source or produced by recombinant techniques, but it need not be translated from a given nucleic acid sequence, and it may be produced by any means including chemical synthesis.
  • Amino acid refers to an organic compound containing both an amino group and a carboxyl group, such as an alpha-amino acid, which can be encoded by a nucleic acid directly or in a precursor form.
  • a single amino acid is encoded by a nucleic acid consisting of three nucleotides, so-called codons or base triplets. Each amino acid is encoded by at least one codon. The same amino acid is encoded by different codons called “degeneracy of the genetic code”.
  • Amino acids include natural amino acids and unnatural amino acids.
  • Natural amino acids include alanine (three-letter code: ala, one-letter code: A), arginine (arg, R), asparagine (asn, N), aspartic acid (asp, D), cysteine Amino acid (cys, C), glutamine (gln, Q), glutamic acid (glu, E), glycine (gly, G), histidine (his, H), isoleucine (ile, I) ), leucine (leu, L), lysine (lys, K), methionine (met, M), phenylalanine (phe, F), proline (pro, P), serine (ser, S), threonine (thr, T), tryptophan (trp, W), tyrosine (tyr, Y) and valine (val, V).
  • alanine three-letter code: ala, one-letter code: A
  • arginine arg, R
  • asparagine asparag
  • Constant amino acid substitution refers to the replacement of one amino acid residue by another amino acid residue containing a side chain (R group) of similar chemical properties (eg, charge or hydrophobicity). In general, conservative amino acid substitutions will not substantially alter the functional properties of the protein.
  • amino acid classes containing chemically similar side chains include: 1) aliphatic side chains: glycine, alanine, valine, leucine, and isoleucine; 2) aliphatic hydroxyl side chains: serine and threonine 3) Amide-containing side chains: asparagine and glutamine; 4) Aromatic side chains: phenylalanine, tyrosine and tryptophan; 5) Basic side chains: lysine, Arginine and histidine; 6) Acidic side chains: aspartic acid and glutamic acid.
  • the number of amino acids for "conservative amino acid substitutions of VL and VH" can be about 1, about 2, about 3, about 4, about 5, about 6, about 8, about 9, about 10, About 11, about 13, about 14, about 15 conservative amino acid substitutions, or a range (including endpoints) between any two of these values, or any value therein.
  • the number of amino acids for a "conservative amino acid substitution of a heavy or light chain” can be about 1, about 2, about 3, about 4, about 5, about 6, about 8, about 9, about 10 about 11, about 13, about 14, about 15, about 18, about 19, about 22, about 24, about 25, about 29, about 31, about 35, About 38, about 41, about 45 conservative amino acid substitutions, or a range (including endpoints) between any two of these values, or any value therein.
  • encoding when applied to a polynucleotide refers to a polynucleotide referred to as “encoding” a polypeptide, transcribed and/or in its native state or when manipulated by methods well known to those skilled in the art Or translation can yield the polypeptide and/or fragments thereof.
  • recombinant refers to a polypeptide or polynucleotide and means a form of the polypeptide or polynucleotide that does not occur in nature, non-limiting examples may be combined to produce polynucleotides that do not normally exist or peptide.
  • Homology refers to the sequence similarity between two peptides or between two nucleic acid molecules. Homology can be determined by comparing the positions within each sequence that can be aligned. A molecule is homologous when a position in the sequences being compared is occupied by the same base or amino acid. The degree of homology between sequences is a function of the number of matches or homologous positions shared by the sequences.
  • At least 80% identity is about 80% identity, about 81% identity, about 82% identity, about 83% identity, about 85% identity, about 86% identity, about 87% identity, about 88% identity, about 90% identity, about 91% identity, about 92% identity, about 94% identity, about 95% identity, about 98% identity, about 99% identity, or these A range (including endpoints) between any two values in a numerical value or any value therein.
  • a polynucleotide or polynucleotide sequence has a certain percentage (eg, 90%, 95%, 98% or 99%) "identity" or “sequence identity” to another sequence "Sex” means that when the sequences are aligned, the percentage of bases (or amino acids) in the two sequences being compared are identical.
  • the percent alignment or sequence identity can be determined using visual inspection or software programs known in the art, such as those described by Ausubel et al. eds. (2007) in Current Protocols in Molecular Biology. Alignments are preferably performed using default parameters.
  • Biologically equivalent polynucleotides are polynucleotides that have the above-specified percentages of identity and encode polypeptides having the same or similar biological activity.
  • Antibody refers to a polypeptide or polypeptide complex that specifically recognizes and binds an antigen.
  • Antibodies can be whole antibodies and any antigen-binding fragments thereof or single chains thereof.
  • the term “antibody” thus includes any protein or peptide in the molecule that contains at least a portion of an immunoglobulin molecule that has the biological activity of binding to an antigen.
  • Antibodies and antigen-binding fragments include, but are not limited to, the complementarity determining regions (CDRs), heavy chain variable regions (VH), light chain variable regions (VL), heavy chain constant regions of heavy or light chains or ligand binding portions thereof (CH), a light chain constant region (CL), a framework region (FR), or any portion thereof, or at least a portion of a binding protein.
  • the CDR regions include the CDR regions of the light chain (LCDR1-3) and the CDR regions of the heavy chain (HCDR1-3).
  • antibody includes a wide variety of biochemically distinguishable polypeptides. Those of skill in the art will appreciate that classes of heavy chains include gamma, mu, alpha, delta, or epsilon (gamma, mu, alpha, delta, epsilon), with some subclasses (eg, gamma1-gamma4). The nature of this chain determines the "class” of the antibody as IgG, IgM, IgA, IgG or IgE, respectively. Immunoglobulin subclasses (isotypes), eg, IgGl, IgG2, IgG3, IgG4, IgG5, etc., are well characterized and the functional specificities conferred are known.
  • the immunoglobulin molecule is of the IgG class.
  • the four chains are connected by disulfide bonds in a "Y" configuration, where the light chain begins at the "Y" mouth and continues through the variable region surrounding the heavy chain.
  • Antibodies, antigen-binding fragments or derivatives disclosed herein include, but are not limited to, polyclonal, monoclonal, multispecific, fully human, humanized, primatized, chimeric antibodies, single chain antibodies, epitope binding Fragments (eg, Fab-like, Fab'-like, and F(ab') 2 ), single-chain-like Fvs (scFv).
  • Light chains can be classified as kappa ( ⁇ ) or lambda ( ⁇ ). Each heavy chain can bind to a kappa or lambda light chain.
  • kappa
  • lambda
  • Each heavy chain can bind to a kappa or lambda light chain.
  • immunoglobulins are produced by hybridomas, B cells or genetically engineered host cells, their light and heavy chains are joined by covalent bonds, and the "tail" portion of the two heavy chains is joined by a covalent disulfide bond or non-covalent bond.
  • the amino acid sequence extends from the N-terminus of the forked terminus in the Y configuration to the C-terminus at the bottom of each chain.
  • the variable region of immunoglobulin kappa light chain is V ⁇ ; the variable region of immunoglobulin lambda light chain is V ⁇ .
  • variable regions of the light (VL) and heavy (VH) chain portions determine antigen recognition and specificity.
  • the constant region (CL) of the light chain and the constant region (CH) of the heavy chain confer important biological properties such as secretion, transplacental movement, Fc receptor binding, complement fixation, and the like. By convention, the numbering of constant regions increases as they become further from the antigen binding site or amino terminus of the antibody.
  • the N-terminal portion is the variable region and the C-terminal portion is the constant region; the CH3 and CL domains actually comprise the carboxy-terminus of the heavy and light chains, respectively.
  • CDR complementarity determining region
  • CDRs as defined by Kabat and Chothia include overlaps or subsets of amino acid residues when compared to each other. Nonetheless, it is within the scope of the invention to apply either definition to refer to the CDRs of an antibody or variant thereof.
  • the exact residue numbers encompassing a particular CDR will vary depending on the sequence and size of the CDR. Those skilled in the art can usually determine which specific residues the CDRs contain based on the amino acid sequence of the variable region of the antibody.
  • Kabat et al. also define a numbering system applicable to variable region sequences of any antibody.
  • One of ordinary skill in the art can apply this "Kabat numbering" system to any variable region sequence independent of experimental data other than the sequence itself.
  • Kabat Numbering means the numbering system proposed by Kabat et al., U.S. Dept. of Health and Human Services in "Sequence of Proteins of Immunological Interest” (1983).
  • Antibodies may also use the EU or Chothia numbering system.
  • antibody drug conjugate refers to an antibody or antigen-binding fragment thereof chemically linked to one or more chemical agents, which may optionally be therapeutic or cytotoxic agents.
  • the ADC includes an antibody, cytotoxic or therapeutic drug, and a linker that enables the drug to be attached or conjugated to the antibody.
  • ADCs typically have 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 number of drugs conjugated to the antibody.
  • Drugs that can be included in ADCs are, but are not limited to: mitotic inhibitors, antitumor antibiotics, immunomodulators, vectors for gene therapy, alkylating agents, antiangiogenic agents, antimetabolites, boron-containing agents, chemoprotective agents, Hormones, antihormones, corticosteroids, photoactive therapeutics, oligonucleotides, radionuclide agents, topoisomerase inhibitors, tyrosine kinase inhibitors and radiosensitizers.
  • the drug included in the ADC can be a maytansinoid drug.
  • the drug included in the ADC can be a compound of Formula I as described herein, or a pharmaceutically acceptable salt thereof.
  • the antibody in the ADC, is conjugated to the drug through self-cysteine or sulfylated amino acids, such as sulfylated lysine, to form a disulfide bond.
  • alkyl and alkylene as used herein are meant to include branched and straight chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms.
  • C1-C6 in “C1-C6 alkyl” includes groups having 1, 2, 3, 4, 5 or 6 carbon atoms arranged in a straight or branched chain.
  • C1-C6 specifically includes methyl, ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, isobutyl, pentyl (including 8 isomers), hexyl ( including 23 isomers).
  • cycloalkyl refers to a monocyclic saturated aliphatic hydrocarbon group having the specified number of carbon atoms.
  • cycloalkyl includes cyclopropyl, methyl-cyclopropyl, 2,2-dimethyl-cyclobutyl, 2-ethyl-cyclopentyl, cyclohexyl, and the like.
  • halogen as used herein includes fluorine, chlorine, bromine and iodine.
  • amino acid side chain refers to a substituent that replaces a certain group (such as a hydrogen atom) in an amino acid; for example, a glycine side chain is a substituent formed by replacing a hydrogen atom on a methylene group of glycine.
  • amino acid side chains include, but are not limited to, natural amino acid side chains.
  • Antibody drug conjugates can form a wide variety of pharmaceutically acceptable salts including, but not limited to, acid addition salts with organic acids, including but not limited to aliphatic mono- and dicarboxylic acids , phenyl-substituted alkanoic acids, hydroxyalkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, amino acids, etc., such as acetic acid, trifluoroacetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, horse Lactic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, etc.; and inorganic Acid addition salts formed by the reaction of acids, such as inorganic acids including hydro
  • Treatment means therapeutic treatment and prophylactic or prophylactic measures, the purpose of which is to prevent, slow, ameliorate and stop adverse physiological changes or disorders, such as the progression of disease, including but not limited to the following whether detectable or undetectable As a result, alleviation of symptoms, reduction of disease severity, stabilization of disease state (ie, no worsening), delay or slowdown of disease progression, improvement or alleviation of disease state, alleviation or disappearance (whether in part or in whole), prolongation and Expected duration of survival when not receiving treatment, etc.
  • a patient in need of treatment includes a patient already suffering from a condition or disorder, a patient susceptible to a condition or disorder, or a patient in need of prevention of the condition or disorder, may or may be expected from administration of the antibodies or compositions disclosed herein for detection, Patients who benefit from the diagnostic process and/or treatment.
  • Patient refers to any mammal in need of diagnosis, prognosis, or treatment, including humans, dogs, cats, guinea pigs, rabbits, rats, mice, horses, cattle, and the like.
  • Effective amount refers to the amount of active compound or agent that elicits a biological or medical response in a tissue, system, animal, individual, or human; an effective amount is sought by the researcher, veterinarian, physician, or other clinician.
  • the phrase "in need” means that a patient has been identified as in need of a particular method or treatment. In some embodiments, identification can be made by any diagnostic means. In any of the methods and treatments described herein, the patient may need.
  • Antibody-encoding DNA can be designed and synthesized according to the antibody amino acid sequences described herein by conventional methods, inserted into an expression vector, then transfected into host cells, and the transfected host cells are cultured in culture to produce monoclonal antibodies.
  • an antibody expression vector includes at least one promoter element, an antibody coding sequence, a transcription termination signal, and a polyA tail. Other elements include enhancers, Kozak sequences, and donor and acceptor sites for RNA splicing flanking the inserted sequence.
  • Efficient transcription can be achieved by the early and late promoters of SV40, long terminal repeats from retroviruses such as RSV, HTLV1, HIVI, and the early promoter of cytomegalovirus, and other cellular promoters such as muscle can be used.
  • Suitable expression vectors may include pIRES1neo, pRetro-Off, pRetro-On, PLXSN, or Plncx, pcDNA3.1(+/-), pcDNA/Zeo(+/-), pcDNA3.1/Hygro(+/-), PSVL, PMSG, pRSVcat, pSV2dhfr, pBC12MI and pCS2 etc.
  • Commonly used mammalian cells include 293 cells, Cos1 cells, Cos7 cells, CV1 cells, mouse L cells and CHO cells.
  • EC50 the concentration for 50% of maximal effect (EC50), refers to the concentration that elicits 50% of the maximal effect.
  • the DNA sequences of the light and heavy chains are constructed according to the amino acid sequences of the heavy and light chains of the antibody.
  • the 5' and 3' ends of the DNA sequence were modified with PCR primers, the primers were designed to add appropriate leader sequences to each chain, and then cloned into the existing recombinant antibody expression vector, and the correct construction of the vector was verified by sequencing analysis.
  • the expression vector was introduced into CHO cells for expression and obtained by purification; the preparation method of antibody A can refer to the invention patent application with the application number of PCT/CN2020/083954; the amino acid sequences of antibody A, antibody B and antibody C are shown in Table 1, The nucleic acid sequences of Antibody A and Antibody C are shown in Table 2.
  • Antibody C was prepared by using CHO-BAT-KF cells, and the glycoform content of antibody C was measured after purification, as shown in Table 3.
  • Example 3 Combined administration of antibody A and HER2-ADC B in vitro inhibits the proliferation of NCI-N87 cells (human gastric cancer cells)
  • the concentration of antibody A was fixed (25 ⁇ g/mL), the concentration of HER2-ADC B was changed (10 nM starting, 3-fold serial dilution), PBMC cells (Reid Biotechnology Co., Ltd.) were added, and SEB (staphylococcal enterotoxin) was added at the same time.
  • B Academy of Military Medical Sciences SL008 activated PBMC cells, and after culturing for 72 hours, the inhibitory effect of the combined drug on the proliferation of HER2 and PD-L1 double positive cells in vitro was determined by the CCK method (CCK8 kit from Dojindo):
  • Example 4 Combined administration of antibody A and antibody C inhibits the proliferation of cancer cells
  • This example evaluates the efficacy of Antibody A and Antibody C (antibody C is expressed by CHO-BAT-KF cells) in an immune checkpoint humanized mouse C57BL/6-hPD1/hCTLA4 subcutaneously inoculated MC38 colon cancer tumor model.
  • Murine colon cancer cell MC38 cells were recovered (Jiangsu Jicui Yaokang Biotechnology Co., Ltd.), and MC38 cells in logarithmic growth phase were collected (Jiangsu Jicui Yaokang Biotechnology Co., Ltd.), and the culture medium was removed and washed twice with PBS before inoculation (loaded The cell survival rates before tumor and after tumor bearing were: 99.6% and 97.8% respectively), the inoculation amount was 1 ⁇ 10 6 /100 ⁇ L/cell, and the cells were injected intraperitoneally under the armpit.
  • mice On the 6th day after inoculation, when the average tumor volume reached 89.19 mm3 , 60 female mice were randomly divided into 6 groups of 10 mice according to the tumor volume.
  • the day of grouping was defined as day D0, and the administration started on day D0; the administration schedule for grouping was shown in Table 4, and the administration dates were: D0, D4, D7, D11, D14, and D18.
  • mice Following cell inoculation, tumor effects on the animals' normal behavior were routinely monitored weekly. Specific indicators include mouse activity, food and water intake, weight gain or loss, eyes, coat and other abnormalities.
  • mice The experimental results such as tumor volume, mouse body weight, and tumor weight of mice in each group are expressed as mean ⁇ standard error (mean ⁇ SEM).
  • the independent sample T test was used to compare whether there were significant differences between the different administration groups and the control group. Data were analyzed using SPSS. P ⁇ 0.05 means there is a significant difference.
  • TGItv inhibition rate relative to tumor volume
  • RTVn Vnt / Vn0 ; Vnt : tumor volume of mouse number n on day t, Vn0 : tumor volume of mouse number n on day 0, RTVn: mouse number n Relative tumor volume in mice on day t
  • TGItv (1-(mean RTV administration group)/(mean RTV control group )) ⁇ 100%; mean RTV administration group: average RTV of administration group, mean RTV control group : average RTV of control group;
  • TGItw (1-(mean TW administration group)/(mean TW control group )) ⁇ 100%;
  • Mean TW control group control The mean value of tumor weight at the end point treatment of mice in the group.
  • N is the number of animals; BIW x 3W, administered twice a week for 3 weeks; intraperitoneally administered 6 times; G5 and G6 groups are combined administration groups, antibody A is administered first, and administered for about 2 hours Antibody C, the administration concentration was not doubled and was consistent with the concentration of the single drug group.
  • single-drug antibody A has a significant anti-tumor effect
  • the high-dose group of antibody C shows a significant anti-tumor effect
  • antibody C shows a certain dose-dependent effect
  • the high-dose combination administration of antibody A and antibody C had significant anti-tumor efficacy
  • the anti-tumor effect was better than the anti-tumor effect of single-drug antibody A and single-drug antibody C (G1, G2 groups did not find that the mice had complete tumor Regressed, the tumors of 2 mice in G3 and G6 groups each completely regressed, the tumors of 1 mouse in G4 group completely regressed, and the tumors of 6 mice in G5 group completely regressed); as shown in Figure 4, each group No significant difference was found in the body weight of the mice between them, and no mouse body weight was lower than 10% of the body weight of the mice on the day of grouping during the experiment, indicating that the mice tolerated the drugs in the current system well

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Oncology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Cell Biology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

本发明公开了抗PD-1抗体在联合用药中的应用,治疗方法包括向有需要的患者给药有效量的抗PD-1抗体和治疗剂。

Description

抗PD-1抗体在联合用药中的应用 技术领域
本发明属于生物医药领域,尤其涉及抗PD-1抗体在联合用药中的应用。
背景技术
肿瘤的发病率逐年上升,抗肿瘤药物的使用也逐渐增多。近年来,肿瘤治疗研究取得了重大进展,尤其是一些抗体药物在恶性肿瘤治疗中显示出了良好疗效。然而,只有少部分患者可达到长期生存。为了进一步提高药物的抗肿瘤治疗的临床疗效,在使用抗肿瘤药物时,可采用联合用药。
程序性死亡受体-1(PD-1)是在活化的T细胞、B细胞和髓样细胞上表达的免疫抑制性受体,是CD28免疫球蛋白超家族成员。PD-1为55kDa的I型跨膜糖蛋白,其含有与配体结合的Ig可变型结构域和负责结合信号转导分子的胞质尾区。PD-1胞质尾区含有两个基于酪氨酸的信号转导模体ITIM(免疫受体酪氨酸抑制基序)和ITSM(免疫受体酪氨酸转换基序)。
迄今有大量的研究显示PD-1和PD-L1(程序性死亡配体1)之间的相互作用导致渗入肿瘤的淋巴细胞减少、T细胞受体介导的增殖减少和癌细胞的免疫逃避。阻断PD-1与PD-L1之间的相互作用可增加T细胞增殖和细胞因子产生,提高肿瘤特异性CD8 +T细胞的免疫性。
因此,抗PD-1抗体在抗肿瘤的联合用药中发挥着重要作用。
发明内容
本发明公开了抗PD-1抗体用于联合治疗肿瘤或癌症的方法或用途。在一些实施方案中,抗PD-1抗体和治疗剂联合用于治疗肿瘤或癌症。在一些实施方案中,所述治疗剂为抗体或抗体药物偶联物。
在一些实施方案中,所述抗PD-1抗体至少包含SEQ ID NO:1所示的HCDR1、SEQ ID NO:2所示的HCDR2、SEQ ID NO:3所示的HCDR3、SEQ ID NO:4所示的LCDR1、SEQ ID NO:5所示的LCDR2、SEQ ID NO:6所示的LCDR3中一个或多个。
在一些实施方案中,所述抗PD-1抗体包含SEQ ID NO:1所示的HCDR1、SEQ ID NO:2所示的HCDR2、SEQ ID NO:3所示的HCDR3、SEQ ID NO:4所示的LCDR1、 SEQ ID NO:5所示的LCDR2和SEQ ID NO:6所示的LCDR3。
在一些实施方案中,所述抗PD-1抗体的重链可变区包含SEQ ID NO:7所示的序列,与SEQ ID NO:7所示序列具有至少80%同一性的序列,或与SEQ ID NO:7所示序列相比具有一或多个保守氨基酸取代的氨基酸序列;和/或
所述抗PD-1抗体的轻链可变区包含SEQ ID NO:8所示的序列,与SEQ ID NO:8所示序列具有至少80%同一性的序列,或与SEQ ID NO:8所示序列相比具有一或多个保守氨基酸取代的氨基酸序列。
在一些实施方案中,所述抗PD-1抗体的重链可变区包含SEQ ID NO:7所示的序列,所述抗PD-1抗体的轻链可变区包含SEQ ID NO:8所示的序列。
在一些实施方案中,所述抗PD-1抗体的重链包含SEQ ID NO:9所示的序列,与SEQ ID NO:9所示序列具有至少80%同一性的序列,或与SEQ ID NO:9所示序列相比具有一或多个保守氨基酸取代的氨基酸序列;和/或
所述抗PD-1抗体的轻链包含SEQ ID NO:10所示的序列,与SEQ ID NO:10所示序列具有至少80%同一性的序列,或与SEQ ID NO:10所示序列相比具有一或多个保守氨基酸取代的氨基酸序列。
在一些实施方案中,所述抗PD-1抗体为抗体A,抗体A的重链包含如SEQ ID NO:9所示序列,所述抗体A的轻链包含如SEQ ID NO:10所示序列;抗体A含有两条序列相同的重链和两条序列相同的轻链。
在一些实施方案中,所述治疗剂选自针对以下靶点的抗体或抗体药物偶联物(ADC):EGFR(表皮生长因子受体)、VEGF(血管内皮生长因子)、VEGFR2(血管内皮生长因子受体2)、CTLA4(细胞毒性T淋巴细胞相关蛋白4)、PD-L1、HER2(人表皮生长因子受体2)、CD20(分化簇20)、Trop2(人滋养层细胞表面抗原2)、Lag3(淋巴细胞活化基因-3分子)、TIGIT(T cell Ig and ITIM domain)、CD27(分化簇27)、OX40(肿瘤坏死因子受体超家族成员4)、ICOS(inducible costimulator)、BTLA(B和T淋巴细胞弱化因子)、TIM3(T细胞免疫球蛋白黏液素3)、BCMA(B细胞成熟抗原)、c-MET(间质表皮转化因子)和TAA-1/2/3(肿瘤相关抗原)。在一些实施方案中,所述抗体为抑制型抗体或激动型抗体。
在一些实施方案中,所述治疗剂选自以下组成的组:抗EGFR抗体、抗VEGF抗体、抗VEGFR2抗体、抗CTLA4抗体、抗PD-L1抗体、抗HER2抗体、抗CD20抗体、抗Trop2抗体、抗TIGIT抗体、抗OX40抗体或抗ICOS抗体。
在一些实施方案中,所述治疗剂为抗CTLA4抗体。在一些实施方案中,抗CTLA4抗体为抗体C,抗体C的重链包含如SEQ ID NO:15所示序列,抗体C轻链包含如SEQ  ID NO:16所示序列;抗体C含有两条序列相同的重链和两条序列相同的轻链。在一些实施方案中,抗体C由α-(1,6)-岩藻糖基转移酶基因敲除的CHO细胞表达,如PCT/CN2018/100008公开的CHO-BAT-KF细胞系。
在一些实施方案中,抗体C中Fc区的高甘露糖糖型总量<5%和/或唾液酸化糖型总量<3%。在一些实施方案中,抗体C中Fc区的高甘露糖糖型总量为约0.1%、约0.3%、约0.9%、约1.18%、约1.7%、约2.6%、约3.3%、约4.1%、约4.9%、约4.99%,或这些数值中任何两个值之间的范围(包括端点)或其中任何值。在一些实施方案中,所述抗体C中Fc区的唾液酸化糖型总量为约0.1%、0.2%、约0.36%、约0.8%、约1.5%、约2.2%、约2.7%、约2.9%、2.99%,或这些数值中任何两个值之间的范围(包括端点)或其中任何值。
在一些实施方案中,所述抗体C中Fc区的高甘露糖糖型总量<2%和/或唾液酸化糖型总量<1%。
在一些实施方案中,所述抗体C中Fc区的岩藻糖基化水平为0%-10%。在一些实施方案中,所述抗体C中Fc区的岩藻糖基化水平为0%-5%。在一些实施方案中,所述抗体C中Fc区的岩藻糖基化含量为约0、约0.1%、约0.3%、约0.4%、约0.6%、约1.3%、约1.9%、约2.2%、约2.8%、约3.3%、约3.7%、约4.1%、约4.5%、约5%,或这些数值中任何两个值之间的范围(包括端点)或其中任何值。
在一些实施方案中,所述治疗剂为抗HER2抗体药物偶联物或抗Trop2抗体药物偶联物。在一些实施方案中,所述治疗剂为抗HER2-抗体药物偶联物(HER2-ADC)。在一些实施方案中,所述治疗剂为抗Trop2-抗体药物偶联物(Trop2-ADC)。
在一些实施方案中,Trop2-ADC为CN109078181A专利申请中的BAT0807或BAT0808与药物的偶联物。
在一些实施方案中,所述治疗剂为如式I所示的抗体药物偶联物(ADC)或其药学上可接受的盐:
Figure PCTCN2021122878-appb-000001
其中,Abu为抗HER2抗体或抗Trop2抗体,p选自1-10;
X为-H或卤素基;
Y选自-H、C1-C6烷基、C3-C6环烷基或-C(=O)R 5
R 1选自-H、-OH、-OC(=O)R 5或-OR 5基团;
R 2为-H或C1-C6烷基;
R 3为甲基﹑-CH 2OH或-CH 2OC(=O)R 6
R 4为-OH或-SH;
R 5为C1-C6烷基或苄基;
R 6为C1-C6烷基﹑苯基或苄基;
R 7为-H、C1-C6烷基或氨基酸侧链;
R 8为-H或者C1-C6烷基;
Z独立地为-H或C1-C4烃基,或两个Z与其所连接的碳原子形成一个羰基;
L选自任选被取代的C1-C20亚烃基或C3-C8环亚烃基,其中一个或多个-CH 2-基团独立地任选地被选自以下的基团所替代:C3-C8环亚烃基、-O-、-S-﹑-NR 8-﹑-C(=O)-﹑-C(=O)NR 8-﹑-NR 8C(=O)-﹑-SO 2NR 8-或-NR 8SO 2-。
在一些实施方案中,L为-(CH 2) m-,其中m为整数,m为1-20。在一些实施方案中,m为1-10。在一些实施方案中,m为5-10。在一些实施方案中,m为1、2、3、4、5、7、9、11、12、13、15、16、18、19或20,或这些数值中任何两个值之间的范围(包括端点)或其中任何值。
在一些实施方式中,L中所述C1-C20亚烃基被1到4个-SO 3H﹑-P(=O)(OH) 2或R 23取代,其中R 23各自独立地为任选地被独立地选自-SH﹑-S-C 1-4烷基﹑-CONR 11R 11以及-NR 11R 11的一个或两个基团取代的C1-C6烷基。在一些实施方案中,R 11选自-H、烷基、芳基、环烷基、环烯基、杂芳基和杂环,或者两个R 11和氮形成一个杂环,其中杂环中的碳可以选择用一个或两个氧取代。
在一些实施方案中,所述治疗剂为如式II所示的抗体药物偶联物(ADC)或其药 学上可接受的盐:
Figure PCTCN2021122878-appb-000002
其中,Abu为抗HER2抗体或抗Trop2抗体,p选自1-10。
p为ADC中一个抗体结合的小分子药物的个数,即抗体的药物结合数,或称为药物抗体偶联比(DAR)。在一些实施方式中,p为约1、约2、约3、约4、约5、约6、约7、约8、约9或约10。在一些实施方案中,p为2-3。在一些实施方案中,p为约2。
在一些实施方案中,p为小分子药物的平均结合个数,即抗体的药物平均结合数,或称为平均药物抗体偶联比(平均DAR)。在一些实施方案中,p为1-10,p可以为非整数。在一些实施方案中,p为约1、约2、约3、约4、约5、约6、约7、约8、约9、约10,或这些数值中任何两个值之间的范围(包括端点)或其中任何值。在一些实施方案中,p选自2-8。在一些实施方案中,p选自3-5。在一些实施方案中,p选自3.3-3.7。在一些实施方案中,p为约3.5。在一些实施方案中,p为约2.1。
在一些实施方案中,所述抗HER2抗体的重链包含SEQ ID NO:11所示的序列,与SEQ ID NO:11所示序列具有至少80%同一性的序列,或与SEQ ID NO:11所示序列相比具有一或多个保守氨基酸取代的氨基酸序列;和/或
所述抗HER2抗体的轻链包含SEQ ID NO:12所示的序列,与SEQ ID NO:12所示序列具有至少80%同一性的序列,或与SEQ ID NO:12所示序列相比具有一或多个保守氨基酸取代的氨基酸序列。
在一些实施方案中,所述抗HER2抗体为抗体B,抗体B的重链包含如SEQ ID NO:11所示序列,抗体B的轻链包含如SEQ ID NO:12所示序列。
在一些实施方案中,抗Trop2抗体为CN109078181A专利申请中的BAT0807或BAT0808。在一些实施方案中,p为约2。在一些实施方案中,p为约2.1。
在一些实施方案中,所述HER2-ADC为HER2-ADC B,HER2-ADC B为式II所示的化合物,HER2-ADC B中抗体为抗体B,抗体B含有两条序列相同的重链和两条序列相同的轻链;p值为3.3-3.7,或约3.5。
抗体蛋白可以通过基因工程在CHO细胞中表达,并通过纯化获得;纯化可以采用常规方法进行,例如先离心细胞悬液并收集上清液,再次离心进一步去除杂质。Protein A亲和柱和离子交换柱等方法可以用于纯化抗体蛋白。
在一些实施方案中,所述方法或用途包括:向有需要的患者施用有效量的抗PD-1抗体和治疗剂。在一些实施方案中,所述抗PD-1抗体为抗体A。在一些实施方案中,所述治疗剂为HER2-ADC B。在一些实施方案中,抗PD-1抗体施用的有效剂量为每剂约50mg至600mg。在一些实施方案中,HER2-ADC施用的有效剂量为每剂约70mg至400mg。在一些实施方案中,所述治疗剂为抗CTLA4抗体。在一些实施方案中,所述治疗剂为抗体C。在一些实施方案中,抗CTLA4施用的有效剂量为每剂约6mg至600mg。
在一些实施方案中,所述抗PD-1抗体和所述治疗剂分别为独立的给药单元,联合用药。在一些实施方案中,所述抗PD-1抗体可以在施加所述治疗剂之前给药,也可以在施加抗所述治疗剂之后给药,也可以在施加所述治疗剂的同时给药。
在一些实施方案中,所述抗PD-1抗体和所述治疗剂同时形成组合给药单元,联合用药。
在一些实施方案中,患者患有肿瘤或癌症。在一些实施方案中,肿瘤或癌症包括但不限于:头颈部肿瘤、霍奇金淋巴瘤、非霍奇金淋巴瘤、前淋巴母细胞淋巴瘤、小无裂细胞淋巴瘤、伯基特淋巴瘤、非伯基特淋巴瘤、弥漫性大B细胞淋巴瘤、间变性大细胞淋巴瘤、肾脏肿瘤、肾母细胞瘤、Wilms瘤、肾透明细胞癌、肾横纹肌样瘤、肾透明细胞肉瘤、肾原始神经外胚叶瘤、神经母细胞瘤、节细胞神经母细胞瘤、节细胞神经瘤、颅外生殖细胞瘤、成熟畸胎瘤、未成熟畸胎瘤、内胚窦瘤、卵黄囊瘤、精原细胞瘤、无性细胞瘤、绒毛膜上皮癌、胚胎癌、骨肉瘤、软骨肉瘤、横纹肌肉瘤、软组织肉瘤、纤维肉瘤、恶性纤维组织细胞瘤、脂肪肉瘤、平滑肌肉瘤、血管肉瘤、淋巴管肉瘤、恶性神经鞘瘤、腺泡状软组织肉瘤、上皮样肉瘤、透明细胞肉瘤、恶性黑色素瘤、滑膜肉瘤、促纤维增生性小圆细胞瘤、尤文氏肉瘤、原始神经外胚叶瘤、肝脏肿瘤、肝母细胞瘤、视网膜母细胞瘤、后颅窝髓母细胞瘤、胸腺瘤、肺母细胞瘤、胰母细胞瘤、胰岛细胞瘤、回盲部类癌、间皮瘤、黑色素瘤、间质细胞瘤、骨髓瘤、脑星形细胞瘤、鼻咽癌、甲状腺乳头状癌、肠癌、乳癌、胃癌、肝癌、前列腺癌、乳腺癌、肺癌、宫颈癌、卵巢癌、肾癌、淋巴瘤、白血病、皮肤癌和食管鳞癌等。
在一些实施方案中,本发明公开了一种用于治疗有需要患者的肿瘤或癌症的方法,其包括施用有效量的抗PD-1抗体和治疗剂,其中抗PD-1施用的有效量为约50mg至600mg每个治疗周期。在一些实施方案中,一个治疗周期为1周、2周、3周、4周、1个月、5周、6周、7周,或这些数值中的任何两个值之间的范围(包括端点)或其 中任何值。在一些实施方案中,所述抗PD-1抗体为抗体A。
在一些实施方案中,所述治疗剂为与表皮生长因子受体2(HER2)的细胞外二聚化结构域(亚结构域II)发生特异性结合的单克隆抗体(抗HER2抗体),如帕妥珠单抗,其分子量为约148kDa。帕妥珠单抗可以通过基因工程在细胞(如CHO)中表达,并通过纯化获得;纯化可以采用常规方法进行,例如先离心细胞悬液并收集上清液,再次离心进一步去除杂质。Protein A亲和柱和离子交换柱等方法可以用于纯化抗体。
在一些实施方案中,所述治疗剂为帕妥珠单抗,帕妥珠单抗包括
Figure PCTCN2021122878-appb-000003
或其生物类似物或ADCC效应增强单抗或去岩藻糖基化单抗。在一些实施方案中,帕妥珠单抗施用的有效量为约40mg至900mg每个治疗周期。在一些实施方案中,一个治疗周期为1周、2周、3周、4周、1个月、5周、6周、7周,或这些数值中的任何两个值之间的范围(包括端点)或其中任何值。在一些实施方案中,帕妥珠单抗施用的有效量为约初始840mg,之后420mg每3周1次。
在一些实施方案中,所述治疗剂是一种重组人源化免疫球蛋白G1(IgG1)单克隆抗体,可以结合VEGF-A,抑制其与VEGF受体-2(VEGFR-2)结合(抗VEGF抗体),如贝伐珠单抗。贝伐珠单抗可以通过基因工程在细胞(如CHO)中表达,并通过纯化获得;纯化可以采用常规方法进行,例如先离心细胞悬液并收集上清液,再次离心进一步去除杂质。Protein A亲和柱和离子交换柱等方法可以用于纯化抗体。
在一些实施方案中,所述治疗剂为贝伐珠单抗,贝伐珠单抗包括
Figure PCTCN2021122878-appb-000004
或其生物类似物,如
Figure PCTCN2021122878-appb-000005
或BAT1706,或ADCC效应增强单抗或去岩藻糖基化单抗。
在一些实施方案中,贝伐珠单抗施用的有效量为约50mg至400mg每个治疗周期。在一些实施方案中,一个治疗周期为1周、2周、3周、4周、1个月、5周、6周、7周,或这些数值中的任何两个值之间的范围(包括端点)或其中任何值。在一些实施方案中,贝伐珠单抗施用的有效量为约5mg/kg到15mg/kg每2周或每3周1次。在一些实施方案中,贝伐珠单抗施用的有效量为约5mg/kg,7.5mg/kg,10mg/kg,或15mg/kg每2周或每3周1次。在一些实施方案中,贝伐珠单抗施用的有效量为约5mg/kg每2周1次,10mg/kg每2周1次,7.5mg/kg每3周1次,15mg/kg每3周1次。
在一些实施方案中,所述治疗剂为是靶向CD20抗体(抗CD20抗体),如奥法木单抗,其为全人源IgG1kappa单克隆抗体,或obinutuzumab。奥法木单抗及obinutuzumab可以通过基因工程在细胞(如CHO)中表达,并通过纯化获得;纯化可以采用常规方 法进行,例如先离心细胞悬液并收集上清液,再次离心进一步去除杂质。Protein A亲和柱和离子交换柱等方法可以用于纯化抗体。
在一些实施方案中,所述治疗剂为奥法木单抗,奥法木单抗包括
Figure PCTCN2021122878-appb-000006
Figure PCTCN2021122878-appb-000007
其生物类似物或ADCC效应增强单抗或去岩藻糖基化单抗,如CN109096399A公开的BAT4406F。在一些实施方案中,奥法木单抗施用的有效量为约10mg至2000mg每个治疗周期。在一些实施方案中,一个治疗周期为1周、2周、3周、4周、1个月、5周、6周、7周,或这些数值中的任何两个值之间的范围(包括端点)或其中任何值。在一些实施方案中,奥法木单抗施用的有效量为约20mg每周或每月1次。在一些实施方案中,奥法木单抗施用的有效量为约初始300mg,1周后1000mg,之后1000mg每4周或每8周1次。在一些实施方案中,奥法木单抗施用的有效量为约初始300mg,1周后2000mg,之后2000mg每1周或每4周1次。
在一些实施方案中,所述治疗剂为obinutuzumab,包括
Figure PCTCN2021122878-appb-000008
其生物类似物,或ADCC效应增强单抗或去岩藻糖基化单抗,如CN109096399A所述BAT4306F。在一些实施方案中,obinutuzumab施用的有效量为约10mg至2000mg每个治疗周期。在一些实施方案中,一个治疗周期为1周、2周、3周、4周、1个月、5周、6周、7周,或这些数值中的任何两个值之间的范围(包括端点)或其中任何值。在一些实施方案中,obinutuzumab施用的有效量为第1天100mg,第2天900mg,第8天、第15天1,000mg,之后1,000mg每疗程。在一些实施方案中,obinutuzumab施用的有效量为第1天、第8天、第15天各1,000mg,之后1,000mg每疗程。每疗程可以是1个月或2个月。
在一些实施方案中,所述治疗剂为靶向T淋巴细胞相关抗原4(CTLA4)抗体(抗CTLA4抗体),如伊匹单抗,其为IgG1kappa免疫球蛋白,其分子量约为148kDa。伊匹单抗可以通过基因工程在细胞(如CHO)中表达,并通过纯化获得;纯化可以采用常规方法进行,例如先离心细胞悬液并收集上清液,再次离心进一步去除杂质。Protein A亲和柱和离子交换柱等方法可以用于纯化抗体。
在一些实施方案中,所述治疗剂为伊匹单抗,伊匹单抗包括Yervoy TM或其生物类似物或ADCC效应增强单抗或去岩藻糖基化伊匹单抗,如WO2014089113所述。在一些实施方案中,伊匹单抗施用的有效量为约30mg至300mg每个治疗周期。在一些实施方案中,一个治疗周期为1周、2周、3周、4周、1个月、5周、6周、7周,或这些数值中的任何两个值之间的范围(包括端点)或其中任何值。在一些实施方案中,伊匹单抗施用的有效量为1mg/kg,3mg/kg或10mg/kg每3周、6周、或12周1次。在一些实施方案中,伊匹单抗施用的有效量为1mg/kg每3周或6周1次。在一些实施方案中,伊匹单抗施用的有效量为3mg/kg每3周或6周1次。在一些实施方案中, 伊匹单抗施用的有效量为10mg/kg每3周或每12周1次。
在一些实施方案中,所述治疗剂为本文所述Trop2-ADC。在一些实施方案中,Trop2-ADC施用的有效量为约60mg至400mg每个治疗周期。在一些实施方案中,一个治疗周期为1周、2周、3周、4周、1个月、5周、6周、7周,或这些数值中的任何两个值之间的范围(包括端点)或其中任何值。在一些实施方案中,Trop2-ADC施用的有效量为0.2,0.5,1,2,4,6,8或10mg/kg每周到每3周1次。在一些实施方案中,Trop2-ADC施用的有效量为0.2,0.5,1,2,4,6,8或10mg/kg每周或每3周1次。
在一些实施方案中,所述治疗剂为HER2-ADC。在一些实施方案中,HER2-ADC施用的有效量为约70mg至400mg每个治疗周期。在一些实施方案中,一个治疗周期为1周、2周、3周、4周、1个月、5周、6周、7周,或这些数值中的任何两个值之间的范围(包括端点)或其中任何值。在一些实施方案中,所述HER2-ADC为HER2-ADC B
在一些实施方案中,可以分别将抗PD-1抗体和治疗剂(或PD-1抗体和治疗剂的组合物)配制成药物组合物,并以适合于所选给药途径的多种形式向患者给药,例如通过肠胃外、静脉内(iv)、肌肉内、局部或皮下途径。在一些实施方案中,可以分别将抗PD-1抗体和治疗剂(或PD-1抗体和治疗剂的组合物)静脉输注。抗PD-1抗体和治疗剂的量将取决于药物的性质,细胞表面触发药物的内在化,运输和释放的程度,所治疗的疾病,患者的状况(如年龄,性别,体重等)。
在一些实施方案中,每次施用的抗PD-1抗体约1mg/kg至10mg/kg或含此剂量抗PD-1抗体的制剂。在一些实施方案中,每次施用的抗PD-1抗体为约1mg/kg,约2mg/kg,约3mg/kg,约4mg/kg,约5mg/kg,约6mg/kg,约7mg/kg,约8mg/kg,约9mg/kg,约10mg/kg,或这些数值中任何两个值之间的范围(包括端点)或其中任何值,或含此剂量抗PD-1抗体的制剂。
在一些实施方案中,采用治疗有效量的帕妥珠单抗和抗PD-1抗体分别或者同时施加在受试患者上。帕妥珠单抗和抗PD-1抗体的给药周期可以相同或者不同。
在一些实施方案中,每次施用的帕妥珠单抗为约1mg/kg至12mg/kg或含此剂量帕妥珠单抗的制剂。在一些实施方案中,每次施用的帕妥珠单抗为约1mg/kg,约1.2mg/kg,约2mg/kg,约2.4mg/kg,约3mg/kg,约3.6mg/kg,约4mg/kg,约4.8mg/kg,约5mg/kg,约5.5mg/kg,约6mg/kg,约6.9mg/kg,约7mg/kg,约8.4mg/kg,约9mg/kg,约11mg/kg,约12mg/kg,或这些数值中任何两个值之间的范围(包括端点)或其中任何值,或含此剂量帕妥珠单抗的制剂。
在一些实施方案中,采用治疗有效量的贝伐珠单抗和抗PD-1抗体分别或者同时施加在受试患者上。贝伐珠单抗和抗PD-1抗体的给药周期可以相同或者不同。
在一些实施方案中,每次施用的贝伐珠单抗为约1mg/kg至9mg/kg或含此剂量贝伐珠单抗的制剂。在一些实施方案中,每次施用的贝伐珠单抗为约1mg/kg,约1.2mg/kg,约2mg/kg,约2.4mg/kg,约3mg/kg,约3.6mg/kg,约4mg/kg,约4.8mg/kg,约5mg/kg,约5.5mg/kg,约6mg/kg,约6.9mg/kg,约7mg/kg,约8.4mg/kg,约9mg/kg,或这些数值中任何两个值之间的范围(包括端点)或其中任何值,或含此剂量贝伐珠单抗的制剂。
在一些实施方案中,采用治疗有效量的奥法木单抗和抗PD-1抗体分别或者同时施加在受试患者上。奥法木单抗和抗PD-1抗体的给药周期可以相同或者不同。
在一些实施方案中,每次施用的奥法木单抗为约0.5mg/kg至18mg/kg或含此剂量奥法木单抗的制剂。在一些实施方案中,每次施用的奥法木单抗为约0.5mg/kg,约1mg/kg,约1.2mg/kg,约2mg/kg,约2.4mg/kg,约3mg/kg,约3.6mg/kg,约4mg/kg,约4.8mg/kg,约5mg/kg,约5.5mg/kg,约6mg/kg,约6.9mg/kg,约7mg/kg,约8.4mg/kg,约9mg/kg,约11mg/kg,约13mg/kg,约14mg/kg,约15mg/kg,约17mg/kg,约18mg/kg,或这些数值中任何两个值之间的范围(包括端点)或其中任何值,或含此剂量奥法木单抗的制剂。
在一些实施方案中,采用治疗有效量的obinutuzumab和抗PD-1抗体分别或者同时施加在受试患者上。obinutuzumab和抗PD-1抗体的给药周期可以相同或者不同。
在一些实施方案中,每次施用的obinutuzumab为约0.5mg/kg至15mg/kg或含此剂量奥法木单抗的制剂。在一些实施方案中,每次施用的奥法木单抗为约0.5mg/kg,约1mg/kg,约1.2mg/kg,约2mg/kg,约2.4mg/kg,约3mg/kg,约3.6mg/kg,约4mg/kg,约4.8mg/kg,约5mg/kg,约5.5mg/kg,约6mg/kg,约6.9mg/kg,约7mg/kg,约8.4mg/kg,约9mg/kg,约11mg/kg,约13mg/kg,约14mg/kg,约15mg/kg,或这些数值中任何两个值之间的范围(包括端点)或其中任何值,或含此剂量奥法木单抗的制剂。
在一些实施方案中,采用治疗有效量的抗CTLA4抗体(如伊匹单抗或去岩藻糖基化伊匹单抗)和抗PD-1抗体分别或者同时施加在受试患者上。抗CTLA4抗体(如伊匹单抗或去岩藻糖基化伊匹单抗)和抗PD-1抗体的给药周期可以相同或者不同。
在一些实施方案中,每次施用的抗CTLA4抗体(如伊匹单抗或去岩藻糖基化伊匹单抗)为约0.1mg/kg至10mg/kg,或约0.5mg/kg至10mg/kg或含此剂量抗CTLA4抗体(如伊匹单抗或去岩藻糖基化伊匹单抗)的制剂。在一些实施方案中,每次施用的伊匹单抗为约0.5mg/kg,约1mg/kg,约1.2mg/kg,约2mg/kg,约2.4mg/kg,约 3mg/kg,约3.6mg/kg,约4mg/kg,约4.8mg/kg,约5mg/kg,约5.5mg/kg,约6mg/kg,约6.9mg/kg,约7mg/kg,约8.4mg/kg,约9mg/kg,约10mg/kg,或这些数值中任何两个值之间的范围(包括端点)或其中任何值,或含此剂量伊匹单抗的制剂。
在一些实施方案中,采用治疗有效量的Trop2-ADC和抗PD-1抗体分别或者同时施加在受试患者上。Trop2-ADC和抗PD-1抗体的给药周期可以相同或者不同。
在一些实施方案中,每次施用的Trop2-ADC为约0.5mg/kg至7mg/kg或含此剂量Trop2-ADC的制剂。在一些实施方案中,每次施用的Trop2-ADC为约1mg/kg,约1.2mg/kg,约2mg/kg,约2.4mg/kg,约3mg/kg,约3.6mg/kg,约4mg/kg,约4.8mg/kg,约5mg/kg,约5.5mg/kg,约7mg/kg,或这些数值中任何两个值之间的范围(包括端点)或其中任何值,或含此剂量Trop2-ADC的制剂。
在一些实施方案中,采用治疗有效量的HER2-ADC和抗PD-1抗体分别或者同时施加在受试患者上。HER2-ADC和抗PD-1抗体的给药周期可以相同或者不同。
在一些实施方案中,每次施用的HER2-ADC为约1mg/kg至6mg/kg或含此剂量HER2-ADC的制剂。在一些实施方案中,每次施用的HER2-ADC为约1mg/kg,约1.2mg/kg,约2mg/kg,约2.4mg/kg,约3mg/kg,约3.6mg/kg,约4mg/kg,约4.8mg/kg,约5mg/kg,约5.5mg/kg,约6mg/kg,或这些数值中任何两个值之间的范围(包括端点)或其中任何值,或含此剂量HER2-ADC的制剂。
在一些实施方案中,本发明公开了一种治疗肿瘤或癌症的方法,其包括向有需要的患者施用有效量的抗PD-1抗体(或制剂)和治疗剂(或制剂);其中,抗PD-1抗体的有效量为单次给药约50mg至600mg(或含此剂量抗PD-1抗体的制剂)。在一些实施方案中,治疗剂为HER2-ADC。在一些实施方案中,HER2-ADC的有效量为单次给药约70mg至400mg(或含此剂量HER2-ADC的制剂)。在一些实施方案中,治疗剂为抗CTLA4抗体。在一些实施方案中,抗CTLA4抗体为抗体C。在一些实施方案中,抗CTLA4抗体的有效量为单次给药约6mg至600mg(或含此剂量抗CTLA4抗体的制剂)。剂量时间表和给药方式取决于某些患者群中的抗PD-1抗体(或制剂)、HER2-ADC(或制剂)或抗CTLA4抗体(或制剂)的获益风险评估和一般临床实践指南。
在一些实施方案中,患者每个治疗周期内抗PD-1抗体施用的有效量为约50mg至600mg的抗PD-1抗体(或含此剂量抗PD-1抗体的制剂),患者每个治疗周期内HER2-ADC施用的有效量为约70mg至400mg(或含此剂量HER2-ADC的制剂)。
在一些实施方案中,患者每个治疗周期内抗PD-1抗体施用的有效量为约50mg至600mg的抗PD-1抗体(或含此剂量抗PD-1抗体的制剂),患者每个治疗周期内抗 CTLA4施用的有效量为约6mg至600mg(或含此剂量抗CTLA4抗体的制剂)。
在一些实施方案中,患者每个治疗周期内施用抗PD-1抗体的有效量是约50mg、约60mg、约80mg、约120mg、约200mg、约250mg、约290mg、约300mg、约330mg、约380mg、约400mg、约434mg、约480mg、约500mg、约567mg、约580mg、约600mg,或这些数值中任何两个值之间的范围(包括端点)或其中任何值,或含此剂量抗PD-1抗体的制剂。在一些实施方案中,一个治疗周期为1周至7周给药1次。在一些实施方案中,每个治疗周期内施用抗PD-1抗体的有效量是100mg至200mg,或含此剂量抗PD-1抗体的制剂;其中,一个治疗周期为约1周、约2周、约3周、约4周、约5周、约6周、约7周,或这些数值中的任何两个值之间的范围(包括端点)或其中任何值。在一些实施方案中,一个治疗周期为约1周、约2周、约3周或约4周。在一些实施方案中,患者每个治疗周期内施用抗PD-1抗体的有效量为约200mg至约300mg,或含此剂量抗PD-1抗体的制剂;其中,一个治疗周期为约1周、约2周、约3周、或约4周。在一些实施方案中,患者每个治疗周期内施用抗PD-1抗体的有效量为约100mg、约110mg、约120mg、约130mg、约140mg、约150mg、约160mg、约170mg、约180mg、约190mg、约200mg,或这些数值中的任何两个值之间的范围(包括端点)或其中任何值,或含此剂量抗PD-1抗体的制剂;其中,一个治疗周期为约1周、约2周、约3周或约4周。
在一些实施方案中,患者每个治疗周期内施用抗PD-1抗体的有效量为约100mg,或含此剂量抗PD-1抗体的制剂;其中,一个治疗周期为约1周、约2周、约3周或约4周。在一些实施方案中,患者每个治疗周期内施用抗PD-1抗体的有效量为约90mg至110mg,或含此剂量抗PD-1抗体的制剂;比如约100mg给药1次。
在一些实施方案中,患者每个治疗周期内施用抗PD-1抗体的有效量为约120mg,或含此剂量抗PD-1抗体的制剂;其中,一个治疗周期为约1周、约2周、约3周或约4周。在一些实施方案中,患者每个治疗周期内施用抗PD-1抗体的有效量为约100mg至140mg,或含此剂量抗PD-1抗体的制剂;比如约120mg给药1次。
在一些实施方案中,患者每个治疗周期内施用抗PD-1抗体的有效量为约160mg,或含此剂量抗PD-1抗体的制剂;其中,一个治疗周期为约1周、约2周、约3周或约4周。在一些实施方案中,患者每个治疗周期内施用抗PD-1抗体的有效量为约150mg至190mg,或含此剂量抗PD-1抗体的制剂;比如约160mg给药1次。
在一些实施方案中,患者每个治疗周期内施用抗PD-1抗体的有效量为约200mg,或含此剂量抗PD-1抗体的制剂;其中,一个治疗周期为约1周、约2周、约3周或约4周。在一些实施方案中,患者每个治疗周期内施用抗PD-1抗体的有效量为约190mg至230mg,或含此剂量抗PD-1抗体的制剂;比如约200mg给药1次。
在一些实施方案中,抗PD-1抗体的有效量为约100mg到600mg每3周一次。在一些实施方案中,抗PD-1抗体的有效量为约100mg、约200mg、约300mg、约400mg、约500mg或约600mg每3周一次。在一些实施方案中,抗PD-1抗体的有效量为约100mg、约300mg或约600mg每3周一次。
在一些实施方案中,患者每个治疗周期内施用HER2-ADC的有效量是约70mg、约90mg、约120mg、约160mg、约180mg、约200mg、约230mg、约250mg、约280mg、约300mg、约310mg、约334mg、约350mg、约370mg、约380mg、约390mg、约400mg,或这些数值中任何两个值之间的范围(包括端点)或其中任何值,或含此剂量HER2-ADC的制剂。在一些实施方案中,一个治疗周期为1周至4周给药1次。在一些实施方案中,每个治疗周期内施用HER2-ADC的有效量是100mg至200mg,或含此剂量HER2-ADC的制剂;其中,一个治疗周期为约1周、约2周、约3周、约4周,或这些数值中的任何两个值之间的范围(包括端点)或其中任何值。在一些实施方案中,患者每个治疗周期内施用HER2-ADC的有效量为约200mg至约300mg,或含此剂量HER2-ADC的制剂;其中,一个治疗周期为约1周、约2周、约3周、或约4周。在一些实施方案中,患者每个治疗周期内施用HER2-ADC的有效量为约100mg、约110mg、约120mg、约130mg、约140mg、约150mg、约160mg、约170mg、约180mg、约190mg、约200mg,或这些数值中的任何两个值之间的范围(包括端点)或其中任何值,或含此剂量HER2-ADC的制剂;其中,一个治疗周期为约1周、约2周、约3周或约4周。
在一些实施方案中,患者每个治疗周期内施用HER2-ADC的有效量为约100mg,或含此剂量HER2-ADC的制剂;其中,一个治疗周期为约1周、约2周、约3周或约4周。在一些实施方案中,患者每个治疗周期内施用HER2-ADC的有效量为约90mg至110mg,或含此剂量HER2-ADC的制剂;比如约100mg给药1次。
在一些实施方案中,患者每个治疗周期内施用HER2-ADC的有效量为约130mg,或含此剂量HER2-ADC的制剂;其中,一个治疗周期为约1周、约2周、约3周或约4周。在一些实施方案中,患者每个治疗周期内施用HER2-ADC的有效量为约100mg至140mg,或含此剂量HER2-ADC的制剂;比如约130mg给药1次。
在一些实施方案中,患者每个治疗周期内施用HER2-ADC的有效量为约170mg,或含此剂量HER2-ADC的制剂;其中,一个治疗周期为约1周、约2周、约3周或约4周。在一些实施方案中,患者每个治疗周期内施用HER2-ADC的有效量为约150mg至190mg,或含此剂量HER2-ADC的制剂;比如约170mg给药1次。
在一些实施方案中,患者每个治疗周期内施用HER2-ADC的有效量为约200mg,或含此剂量HER2-ADC的制剂;其中,一个治疗周期为约1周、约2周、约3周或约 4周。在一些实施方案中,患者每个治疗周期内施用HER2-ADC的有效量为约190mg至230mg,或含此剂量HER2-ADC的制剂;比如约200mg给药1次。
在一些实施方案中,患者每个治疗周期内施用抗CTLA4抗体的有效量是约6mg、约8mg、约10mg、约15mg、约20mg、约25mg、约30mg、约35mg、约40mg、约50mg、约60mg、约80mg、约120mg、约200mg、约250mg、约290mg、约300mg、约330mg、约380mg、约400mg、约434mg、约480mg、约500mg、约567mg、约580mg、约600mg,或这些数值中任何两个值之间的范围(包括端点)或其中任何值,或含此剂量抗CTLA4抗体的制剂。在一些实施方案中,一个治疗周期为1周至7周给药1次。在一些实施方案中,每个治疗周期内施用抗CTLA4抗体的有效量是6mg至80mg,或含此剂量抗CTLA4抗体的制剂;其中,一个治疗周期为约1周、约2周、约3周、约4周、约5周、约6周、约7周,或这些数值中的任何两个值之间的范围(包括端点)或其中任何值。在一些实施方案中,每个治疗周期内施用抗CTLA4抗体的有效量是100mg至200mg,或含此剂量抗CTLA4抗体的制剂;其中,一个治疗周期为约1周、约2周、约3周、约4周、约5周、约6周、约7周,或这些数值中的任何两个值之间的范围(包括端点)或其中任何值。在一些实施方案中,一个治疗周期为约1周、约2周、约3周或约4周。在一些实施方案中,患者每个治疗周期内施用抗CTLA4抗体的有效量为约200mg至约300mg,或含此剂量抗CTLA4抗体的制剂;其中,一个治疗周期为约1周、约2周、约3周、或约4周。在一些实施方案中,患者每个治疗周期内施用抗CTLA4抗体的有效量为约约6mg、约8mg、约10mg、约15mg、约20mg、约25mg、约30mg、约35mg、约40mg、约50mg、约60mg、100mg、约110mg、约120mg、约130mg、约140mg、约150mg、约160mg、约170mg,或这些数值中的任何两个值之间的范围(包括端点)或其中任何值,或含此剂量抗CTLA4抗体的制剂;其中,一个治疗周期为约1周、约2周、约3周或约4周。
在一些实施方案中,患者每个治疗周期内施用抗CTLA4抗体的有效量为约6mg,或含此剂量抗CTLA4抗体的制剂;其中,一个治疗周期为约1周、约2周、约3周或约4周。在一些实施方案中,患者每个治疗周期内约6mg给药1次。
在一些实施方案中,患者每个治疗周期内施用抗CTLA4抗体的有效量为约15mg,或含此剂量抗CTLA4抗体的制剂;其中,一个治疗周期为约1周、约2周、约3周或约4周。在一些实施方案中,患者每个治疗周期内约15mg给药1次。
在一些实施方案中,患者每个治疗周期内施用抗CTLA4抗体的有效量为约23mg,或含此剂量抗CTLA4抗体的制剂;其中,一个治疗周期为约1周、约2周、约3周或约4周。在一些实施方案中,患者每个治疗周期内约23mg给药1次。
在一些实施方案中,患者每个治疗周期内施用抗CTLA4抗体的有效量为约60mg,或含此剂量抗CTLA4抗体的制剂;其中,一个治疗周期为约1周、约2周、约3周或约4周。在一些实施方案中,患者每个治疗周期内约60mg给药1次。
在一些实施方案中,抗CTLA4抗体的有效量为约6mg到600mg每3周一次。在一些实施方案中,抗CTLA4抗体的有效量为约6mg、约20mg、约30mg、约100mg、约200mg、约230mg、约300mg、约400mg、约500mg、或约600mg每3周一次。在一些实施方案中,抗CTLA4抗体的有效量为约6mg、约30mg或约60mg每3周一次。
在一些实施方案中,患者每个治疗周期内分别给药一次抗PD-1抗体和治疗剂(或给药一次抗PD-1抗体和治疗剂的组合物)。在一些实施方案中,每个治疗周期内多次分别给药抗PD-1抗体和治疗剂(或抗PD-1抗体和治疗剂的组合物),例如2次、3次、4次或5次。
在一些实施方案中,患者每个治疗周期只能给药1次或4次。在一些实施方案中,患者接受一个治疗周期治疗。在一些实施方案中,患者接受多个(例如2个、3个或4个)治疗周期治疗。在一些实施方案中,患者接受治疗直至病症得到缓解而不再需要治疗。在一些实施方案中,患者接受多个抗PD-1抗体治疗周期,4个抗CTLA4抗体治疗周期。
在一些实施方案中,一种用于治疗肿瘤或癌症的方法,所述方法包括:向有需要的患者给予约50mg至200mg、约200mg至300mg、约300mg至400mg或约400mg至600mg,比如约100mg、约120mg、约200mg或约400mg的抗PD-1抗体,含此剂量抗PD-1抗体的制剂;还向有需要的患者给予约70mg至100mg、约100mg至200mg、约200mg至300mg或约300mg至400mg,比如约100mg、约120mg、约150mg或约200mg的HER2-ADC,含此剂量HER2-ADC的制剂。在一些实施方案中,患者接受单剂量抗PD-1抗体的治疗,以及单剂量HER2-ADC的治疗。在一些实施方案中,患者接受单剂量抗PD-1抗体和HER2-ADC组合物的治疗。
在一些实施方案中,每3周一次给药抗PD-1抗体200mg和HER2-ADC 2.4mg/kg。在一些实施方案中,每3周一次给药抗PD-1抗体200mg和每3周一次给药HER2-ADC3.6mg/kg。
在一些实施方案中,一种用于治疗肿瘤或癌症的方法,所述方法包括:向有需要的患者给予约50mg至200mg、约200mg至300mg、约300mg至400mg或约400mg至600mg,比如约100mg、约120mg、约200mg或约400mg的抗PD-1抗体,含此剂量抗PD-1抗体的制剂;还向有需要的患者给予约6mg至80mg、约80mg至200mg、 约200mg至300mg或约300mg至600mg,比如约6mg、约12mg、约25mg、约35mg、约50mg、约75mg、约125mg或约200mg的抗CTLA4抗体,含此剂量抗CTLA4抗体的制剂。在一些实施方案中,患者接受单剂量抗PD-1抗体的治疗,以及单剂量抗CTLA4抗体的治疗。在一些实施方案中,患者接受单剂量抗PD-1抗体和抗CTLA4抗体组合物的治疗。
在一些实施方案中,每3周一次给药抗PD-1抗体200mg和抗CTLA4抗体0.1mg/kg。在一些实施方案中,每3周一次给药抗PD-1抗体200mg和每3周一次给药抗CTLA4抗体1mg/kg。在一些实施方案中,单剂量给药后,患者的症状得到缓解。在一些实施方案中,单剂量给药后,患者后的症状未得到预期缓解,再对患者分别给药约50mg至600mg抗PD-1抗体和约70mg至400mg HER2-ADC。在一些实施方案中,单剂量给药后,患者后的症状未得到预期缓解,再对患者给药约50mg至600mg抗PD-1抗体和约70mg至400mg HER2-ADC的组合物。在一些实施方案中,单剂量给药后,患者后的症状未得到预期缓解,再对患者分别给药约50mg至600mg抗PD-1抗体和约6mg至600mg抗CTLA4抗体。在一些实施方案中,单剂量给药后,患者后的症状未得到预期缓解,再对患者给药约50mg至600mg抗PD-1抗体和约6mg至600mg抗CTLA4抗体的组合物。
在一些实施方案中,抗PD-1抗体(或制剂)、HER2-ADC(或制剂)、抗CTLA4抗体(或制剂)是通过皮下(s.c.)注射、腹膜内(i.p.)注射、肠胃外注射、动脉内注射或静脉内(i.v.)注射等方式进行给药。在一些实施方案中,抗PD-1抗体(或制剂)、HER2-ADC(或制剂)是输液方式进行给药。在一些实施方案中,抗PD-1抗体(或制剂)、HER2-ADC(或制剂)是推注方式进行给药。在一些实施方案中,抗PD-1抗体(或制剂)、抗CTLA4抗体(或制剂)是输液方式进行给药。在一些实施方案中,抗PD-1抗体(或制剂)、抗CTLA4抗体(或制剂)是推注方式进行给药。
在一些实施方案中,抗PD-1抗体(或制剂)、HER2-ADC(或制剂)、抗CTLA4抗体(或制剂)是通过静脉内(i.v.)输液方式进行给药。在一些实施方案中,静脉内输液持续时间为约50分钟、约55分钟、约60分钟、约65分钟、约70分钟、约75分钟、约81分钟、约87分钟、约90分钟、约95分钟,或这些数值中任何两个值之间的范围(包括端点)或其中任何值。
在一些实施方案中,抗PD-1抗体(或制剂)、治疗剂(或制剂)与其他治疗方法联合使用用于治疗肿瘤或癌症,例如化疗、放疗和手术治疗等。
另一方面,本发明公开了抗PD-1抗体和治疗剂在制备用于治疗肿瘤或癌症的药物中的应用。在一些实施方案中,所述治疗剂选自以下组成的组:抗EGRR抗体、抗VEGF 抗体、抗VEGFR2抗体、抗CTLA4抗体、抗PD-L1抗体、抗HER2抗体、抗CD20抗体、抗Trop2抗体、抗TIGIT抗体、抗OX40抗体和抗ICOS抗体、抗HER2抗体药物偶联物和抗Trop2抗体药物偶联物。在一些实施方案中,用于治疗肿瘤或癌症的药物包括抗PD-1抗体和治疗剂。在一些实施方案中,抗PD-1抗体为抗体A。在一些实施方案中,治疗剂为HER2-ADC。在一些实施方案中,HER2-ADC为HER2-ADC B。在一些实施方案中,治疗剂为抗CTLA4抗体。在一些实施方案中,抗CTLA4抗体为抗体C。
另一方面,本发明还公开了一种试剂盒,试剂盒包含抗PD-1抗体(或制剂)、治疗剂(或制剂)和用于指导有需要患者给药抗PD-1抗体(或制剂)和治疗剂(或制剂)的说明书。在一些实施方案中,本发明还公开了一种试剂盒,试剂盒包含抗PD-1抗体和治疗剂的组合物(或制剂)以及用于指导有需要患者给药抗PD-1抗体和治疗剂的组合物(或制剂)的说明书。
另一方面,本发明还公开了包含抗PD-1抗体和治疗剂的适合注射用的药物组合物,如推注型药物组合物或输液(滴注)型药物组合物。在一些实施方案中,药物组合物至少包含0.1%的抗PD-1抗体和0.1%的治疗剂。抗体和治疗剂的百分比可以变化,并且为给定剂型重量的约2%约90%之间。这种治疗上有用的药物组合物中抗PD-1抗体和治疗剂的量可以为给药的有效量。
另一方面,本发明还公开了上述药物组合物的制备方法:分别将本文所述的抗PD-1抗体和治疗剂(或抗PD-1抗体和治疗剂的组合物)与药学上可接受的适合注射用的载体(例如注射用水,生理盐水等)混合。上述抗PD-1抗体和治疗剂与药学上可接受的载体的混合方法是本领域通常已知的。
本发明将抗PD-1抗体(或制剂)和治疗剂(或制剂)用于肿瘤或癌症治疗中,可以减缓症状。
附图说明
图1显示抗PD-1抗体和HER2-ADC联合给药抑制肿瘤细胞的增殖。
图2显示抗体A和抗体C联合给药后,肿瘤体积的变化;平均体重以mean±SEM表示。
图3显示抗体A和抗体C联合给药后,第27天时肿瘤的重量;数据以mean±SEM表示。vs G1,除G5组外,其他组别的检验方法为独立样本T检验,G5组由于数据非正态分布,其检测方法为两个独立样本检验,Mann-Whitney检测。*表示P<0.05,**表示P<0.01,***表示P<0.001。
图4显示抗体A和抗体C联合给药后,小鼠体重的变化。
术语
除非另作说明,否则下列的每一个术语应当具有下文所述的含义。
定义
应当注意的是,术语“一种”实体是指一种或多种该实体,例如“一种抗体”应当被理解为一种或多种抗体,因此,术语“一种”(或“一个”)、“一种或多种”和“至少一种”可以在本文中互换使用。
本文所用的术语“包含”或“包括”意味着组合物和方法等包括所列举的元素,例如组份或步骤,但不排除其它。“基本上由……组成”意味着组合物和方法排除对组合的特征有根本影响的其它元素,但不排除对组合物或方法无本质上影响的元素。“由……组成”意味着排除未特别列举的元素。
术语“多肽”旨在涵盖单数的“多肽”以及复数的“多肽”,并且是指由通过酰胺键(也称为肽键)线性连接的氨基酸单体形成的分子。术语“多肽”是指两个或更多个氨基酸的任何单条链或多条链,并且不涉及产物的特定长度。因此,“多肽”的定义中包括肽、二肽、三肽、寡肽、“蛋白质”、“氨基酸链”或用于指两个或多个氨基酸链的任何其他术语,并且术语“多肽”可以用来代替上述任何一个术语,或者与上述任何一个术语交替使用。术语“多肽”也意在指多肽表达后修饰的产物,包括但不限于糖基化、乙酰化、磷酸化、酰胺化、通过已知的保护/封闭基团衍生化、蛋白水解切割或非天然发生的氨基酸修饰。多肽可以源自天然生物来源或通过重组技术产生,但其不必从指定的核酸序列翻译所得,它可能以包括化学合成的任何方式产生。
“氨基酸”是指既含氨基又含羧基的有机化合物,比如α-氨基酸,其可直接或以前体的形式由核酸编码。单个氨基酸由三个核苷酸(所谓的密码子或碱基三联体)组成的核酸编码。每一个氨基酸由至少一个密码子编码。相同氨基酸由不同密码子编码称为“遗传密码的简并性”。氨基酸包括天然氨基酸和非天然氨基酸。天然氨基酸包括丙氨酸(三字母代码:ala,一字母代码:A)、精氨酸(arg,R)、天冬酰胺(asn,N)、天冬氨酸(asp,D)、半胱氨酸(cys,C)、谷氨酰胺(gln,Q)、谷氨酸(glu,E)、甘氨酸(gly,G)、组氨酸(his,H)、异亮氨酸(ile,I)、亮氨酸(leu,L)、赖氨酸(lys,K)、甲硫氨酸(met,M)、苯丙氨酸(phe,F)、脯氨酸(pro,P)、丝氨酸(ser,S)、苏氨酸(thr,T)、色氨酸(trp,W)、酪氨酸(tyr,Y)和缬氨酸(val,V)。
“保守氨基酸取代”是指一个氨基酸残基被另一个含有化学性质(例如电荷或疏水性)相似的侧链(R基团)的氨基酸残基所取代。一般而言,保守氨基酸取代不大会在实质上改变蛋白质的功能性质。含有化学性质相似侧链的氨基酸类别的实例包括:1)脂族侧链:甘氨酸、丙氨酸、缬氨酸、亮氨酸和异亮氨酸;2)脂族羟基侧链:丝氨酸和苏氨酸;3)含酰胺的侧链:天冬酰胺和谷氨酰胺;4)芳族侧链:苯丙氨酸、酪氨 酸和色氨酸;5)碱性侧链:赖氨酸、精氨酸和组氨酸;6)酸性侧链:天冬氨酸和谷氨酸。
“VL、VH的保守氨基酸取代”的氨基酸数目可为约1个、约2个、约3个、约4个、约5个、约6个、约8个、约9个、约10个、约11个、约13个、约14个、约15个保守氨基酸取代,或这些数值中的任何两个值之间的范围(包括端点)或其中任何值。“重链或轻链的保守氨基酸取代”的氨基酸数目可为约1个、约2个、约3个、约4个、约5个、约6个、约8个、约9个、约10个、约11个、约13个、约14个、约15个、约18个、约19个、约22个、约24个、约25个、约29个、约31个、约35个、约38个、约41个、约45个保守氨基酸取代,或这些数值中的任何两个值之间的范围(包括端点)或其中任何值。
术语“编码”应用于多聚核苷酸时,是指被称为“编码”多肽的多聚核苷酸,在其天然状态或当通过本领域技术人员公知的方法操作时,经转录和/或翻译可以产生该多肽和/或其片段。
术语“重组”涉及多肽或多聚核苷酸,意指天然不存在的多肽或多聚核苷酸的形式,不受限制的实施例可以通过组合产生通常并不存在的多聚核苷酸或多肽。
“同源性”或“同一性”或“相似性”是指两个肽之间或两个核酸分子之间的序列相似性。可以通过比较每个序列中可以比对的位置来确定同源性。当被比较的序列中的位置被相同的碱基或氨基酸占据时,则分子在该位置是同源的。序列之间的同源程度是由序列共有的匹配或同源位置的数目组成的一个函数。
“至少80%同一性”为约80%同一性、约81%同一性、约82%同一性、约83%同一性、约85%同一性、约86%同一性、约87%同一性、约88%同一性、约90%同一性、约91%同一性、约92%同一性、约94%同一性、约95%同一性、约98%同一性、约99%同一性,或这些数值中的任何两个值之间的范围(包括端点)或其中任何值。
多聚核苷酸或多聚核苷酸序列(或多肽或抗体序列)与另一序列有具有一定百分比(例如90%、95%、98%或者99%)的“同一性”或“序列同一性”是指当序列比对时,所比较的两个序列中该百分比的碱基(或氨基酸)相同。可以使用目测或本领域已知的软件程序来确定该比对同一性百分比或序列同一性,比如Ausubel et al.eds.(2007)在Current Protocols in Molecular Biology中所述的软件程序。优选使用默认参数进行比对。其中一种比对程序是使用默认参数的BLAST,例如BLASTN和BLASTP,两者使用下列默认参数:Geneticcode=standard;filter=none;strand=both;cutoff=60;expect=10;Matrix=BLOSUM62;Descriptions=50sequences;sortby=HIGHSCORE;Databases=non-redundant;GenBank+EMBL+DDBJ+PDB+GenBankCDStranslations+Swi ssProtein+SPupdate+PIR。生物学上等同的多聚核苷酸是具有上述指定百分比的同一性并编码具有相同或相似生物学活性的多肽的多聚核苷酸。
“抗体”、“抗原结合片段”是指特异性识别和结合抗原的多肽或多肽复合物。抗体可以是完整的抗体及其任何抗原结合片段或其单链。因此术语“抗体”包括分子中含有具有与抗原结合的生物学活性的免疫球蛋白分子的至少一部分的任何蛋白质或肽。抗体和抗原结合片段包括但不局限重链或轻链或其配体结合部分的互补决定区(CDR)、重链可变区(VH)、轻链可变区(VL)、重链恒定区(CH)、轻链恒定区(CL)、框架区(FR)或其任何部分,或结合蛋白的至少一部分。CDR区包括轻链的CDR区(LCDR1-3)和重链的CDR区(HCDR1-3)。
术语“抗体”包括可以在生物化学上区分的各种广泛种类的多肽。本领域技术人员将会理解,重链的类别包括gamma、mu、alpha、delta或epsilon(γ、μ、α、δ、ε),其中还有一些亚类(例如γ1-γ4)。该链的性质决定了抗体的“种类”分别为IgG、IgM、IgA、IgG或IgE。免疫球蛋白亚类(同种型),例如IgG1、IgG2、IgG3、IgG4、IgG5等已被充分表征并且赋予的功能特异性也已知。所有的免疫球蛋白种类都在本发明公开的保护范围内。在一些实施方案中,免疫球蛋白分子为IgG种类。这四条链通过二硫键以“Y”构型连接,其中轻链从“Y”口开始并延续通过可变区包围重链。
本发明公开的抗体、抗原结合片段或衍生物包括但不限于多克隆、单克隆、多特异性、全人源、人源化、灵长类化、嵌合抗体、单链抗体、表位结合片段(例如类Fab、类Fab'和类F(ab') 2)、类单链Fvs(scFv)。
轻链可以分为kappa(κ)或lambda(λ)。每个重链可以与κ或λ轻链结合。一般来说,当由杂交瘤,B细胞或基因工程宿主细胞生产免疫球蛋白时,其轻链和重链通过共价键结合,两条重链的“尾巴”部分通过共价二硫键或非共价键结合。在重链中,氨基酸序列从Y构型的叉状末端的N末端延伸至每条链底部的C末端。免疫球蛋白κ轻链可变区为Vκ;免疫球蛋白λ轻链可变区为V λ
轻链(VL)和重链(VH)链部分的可变区决定了抗原识别和特异性。轻链的恒定区(CL)和重链的恒定区(CH)赋予重要的生物学性质,如分泌、经胎盘移动、Fc受体结合、补体结合等。按照惯例,恒定区的编号随着它们变得更远离抗体的抗原结合位点或氨基末端而增加。N端部分是可变区,C端部分是恒定区;CH3和CL结构域实际上分别包含重链和轻链的羧基端。
在本领域中使用和/或接受的术语有两个或多个定义的情况下,除非明确地对立指出,否则本文使用的术语的定义包括所有这些含义。一个具体的例子是使用“互补决定区”(“CDR”)一词来描述在重链和轻链多肽的可变区内发现的非连续的抗原结合位点。 这一特定区域在Kabat et al.,U.S.Dept.of Health and Human Services,Sequences of Proteins of Immunological Interest(1983)和Chothia等在J.Mol.Biol.196:901-917(1987)有相关描述,其通过引用全部并入本文。
根据Kabat和Chothia定义的CDR包括相互比较时的氨基酸残基的重叠或子集。尽管如此,应用任一定义来指代抗体或其变体的CDR都在本发明范围内。包含特定CDR的确切残基编号将根据CDR的序列和大小而变化。本领域技术人员通常可以根据抗体的可变区氨基酸序列确定出CDR包含哪些特定的残基。
Kabat等人还定义了适用于任何抗体的可变区序列的编号系统。本领域普通技术人员可以不依赖于序列本身以外的其他实验数据将该“Kabat编号”系统应用到任何可变区序列。“Kabat编号”是指由Kabat et al.,U.S.Dept.of Health and Human Services在“Sequence of Proteinsof Immunological Interest”(1983)提出的编号系统。抗体还可以用EU或Chothia编号系统。
本发明中术语“抗体药物偶联物”或“ADC”是指与一个或多个化学药物(其可以任选地是治疗剂或细胞毒性剂)化学连接的抗体或其抗原结合片段。在一些实施方案中,ADC包括抗体、细胞毒性或治疗药物和使得药物能够与抗体连接或偶联的接头。ADC通常具有与抗体偶联的1、2、3、4、5、6、7、8、9或10个数的药物。可以包括在ADC中的药物有但不限于:有丝分裂抑制剂、抗肿瘤抗生素、免疫调节剂、基因治疗的载体、烷化剂、抗血管生成剂、抗代谢物、含硼试剂、化疗保护剂、激素、抗激素剂、皮质类固醇、光活性治疗剂、寡核苷酸、放射性核素试剂、拓扑异构酶抑制剂、酪氨酸激酶抑制剂和放射致敏剂。在一些实施方案中,包括在ADC中的药物可以是类美登素药物。在一些实施方案中,包括在ADC中的药物可以是如本申请所述的如式Ⅰ所示的化合物或其药学上可接受的盐。在一些实施方案中,在ADC中,抗体通过自身半胱氨酸或疏基化的氨基酸如疏基化赖氨酸,形成二硫键,与药物偶联。
本文所用术语“烷基”和“亚烷基”意指包括具有特定碳原子数目的支链的和直链的饱和脂肪烃基。例如,“C1-C6烷基”中“C1-C6”的定义包括以直链或支链排列的具有1、2、3、4、5或6个碳原子的基团。例如,“C1-C6”具体包括甲基、乙基、正丙基、异丙基、正丁基、叔丁基、异丁基、戊基(包括8种同分异构体)、己基(包括23种同分异构体)。术语“环烷基”指具有特定碳原子数目的单环饱和脂肪烃基。例如“环烷基”包括环丙基、甲基-环丙基、2,2-二甲基-环丁基、2-乙基-环戊基、环己基等。
本文中所用术语“卤素”包括氟、氯、溴和碘。
本文中所用术语“氨基酸侧链”指取代氨基酸中某一个基团(如氢原子)的取代 基;如甘氨酸侧链,为取代甘氨酸亚甲基上一个氢原子形成的取代基。氨基酸侧链实例包括,但不限于天然氨基酸侧链。
抗体药物偶联物可以形成广泛多种医药学上可接受的盐,包括但不限于:与有机酸形成的酸加成盐,这种有机酸包括但不限于脂肪族单羧酸和二羧酸、苯基取代的烷酸、羟基烷酸、烷二酸、芳香酸、脂肪族和芳香族磺酸、氨基酸等等,例如乙酸、三氟乙酸、丙酸、乙醇酸、丙酮酸、草酸、马来酸、丙二酸、琥珀酸、富马酸、酒石酸、柠檬酸、苯甲酸、肉桂酸、扁桃酸、甲磺酸、乙磺酸、对甲苯磺酸、水杨酸,等等;与无机酸反应形成的酸加成盐,这种无机酸包括盐酸、氢溴酸、硫酸、硝酸、磷酸、氢碘酸、氢氟酸、亚磷酸,等等;及与金属离子(例如,碱金属离子(例如钠或钾)、碱土金属离子(例如钙或镁)或铝离子)或者与有机碱如二乙醇胺、三乙醇胺、N-甲基葡糖胺等等形成的盐。本文中所述抗体药物偶联物包括其医药学上可接受的盐。
“治疗”是指治疗性治疗和预防性或防治性措施,其目的是预防、减缓、改善和停止不良的生理改变或紊乱,例如疾病的进程,包括但不限于以下无论是可检测还是不可检测的结果,症状的缓解、疾病程度的减小、疾病状态的稳定(即不恶化)、疾病进展的延迟或减缓、疾病状态的改善或缓和,减轻或消失(无论是部分还是全部)、延长与不接受治疗时预期的生存期限等。需要治疗的患者包括已经患有病症或紊乱的患者,容易患有病症或紊乱的患者,或者需要预防该病症或紊乱的患者,可以或预期从施用本发明公开的抗体或组合物用于检测、诊断过程和/或治疗中受益的患者。
“患者”指需要诊断、预后或治疗的任何哺乳动物,包括人类、狗、猫、豚鼠、兔子、大鼠、小鼠、马、牛等。
“约”指相关技术领域技术人员容易知道的相应数值的常规误差范围。在一些实施方式中,本文中提到“约”指所描述的数值以及其±10%、±5%或±1%的范围。
“有效量”是指活性化合物或药剂的量,其能引起组织、系统、动物、个体或人类的生物学或医学反应;有效量由研究人员、兽医、医生或其他临床医生寻求的。
如本文所用,短语“有需要”是指已将患者鉴定为需要特定方法或治疗。在一些实施例中,可以通过任何诊断方式进行识别。在本文描述的任何方法和治疗中,患者可能需要。
可以按常规方法根据本文所述抗体氨基酸序列设计合成编码抗体的DNA,将其置入表达载体中,然后转染宿主细胞,在培养基中培养被转染的宿主细胞产生单克隆抗体。在一些实施方案中,表达抗体载体包括至少一个启动子元件,抗体编码序列,转录终止信号和polyA尾。其他元件包括增强子,Kozak序列及插入序列两侧RNA剪接的供体和受体位点。可以通过SV40的前期和后期启动子,来自逆转录病毒的长末端 重复序列如RSV、HTLV1、HIVI及巨细胞病毒的早期启动子来获得高效的转录,也可应用其它一些细胞的启动子如肌动蛋白启动子。合适的表达载体可包括pIRES1neo,pRetro-Off,pRetro-On,PLXSN,或者Plncx,pcDNA3.1(+/-),pcDNA/Zeo(+/-),pcDNA3.1/Hygro(+/-),PSVL,PMSG,pRSVcat,pSV2dhfr,pBC12MI和pCS2等。常使用的哺乳动物细胞包括293细胞,Cos1细胞,Cos7细胞,CV1细胞,鼠L细胞和CHO细胞等。
“EC 50”即半最大效应浓度(concentration for 50%of maximal effect,EC50)是指能引起50%最大效应的浓度。
具体实施方式
以下通过具体的实施例进一步说明本发明的技术方案,具体实施例不代表对本发明保护范围的限制。其他人根据本发明理念所做出的一些非本质的修改和调整仍属于本发明的保护范围。
下述实施例中所用的材料、试剂等,如无特殊说明,均可从商业途径得到。
实施例1 抗体的制备方法
1)根据抗体的重链和轻链氨基酸序列构建轻链和重链的DNA序列。用PCR引物修饰DNA序列的5’和3’端,所述引物设计成为各链增加合适的前导序列,再克隆到现有重组抗体表达载体上,通过测序分析验证载体构建正确。将表达载体导入CHO细胞中进行表达,并通过纯化获得;抗体A的制备方法可参照申请号为PCT/CN2020/083954的发明专利申请;抗体A、抗体B和抗体C的氨基酸序列见表1,抗体A和抗体C的核酸序列见表2。
表1 氨基酸序列
Figure PCTCN2021122878-appb-000009
Figure PCTCN2021122878-appb-000010
Figure PCTCN2021122878-appb-000011
表2 抗体A和抗体C的核酸序列
Figure PCTCN2021122878-appb-000012
Figure PCTCN2021122878-appb-000013
2)采用CHO-BAT-KF细胞制备抗体C,抗体C纯化后并测定其糖型含量,如表3所示。
表3 部分糖型百分含量
Figure PCTCN2021122878-appb-000014
Figure PCTCN2021122878-appb-000015
实施例2 HER2-ADC B的制备方法
HER2-ADC B的制备方法可参照公开号为WO2014094527A的发明专利的实施例1-3和实施例9等。
实施例3 抗体A与HER2-ADC B体外联合给药抑制NCI-N87细胞(人胃癌细胞)的增殖
固定抗体A的浓度(25μg/mL),改变HER2-ADC B的浓度(10nM起始,3倍梯度稀释),加入PBMC细胞(雷德生物科技有限公司),并同时加入SEB(葡萄球菌肠毒素B,军事医学科学院SL008)激活PBMC细胞,培养72h后通过CCK法(CCK8试剂盒来源于Dojindo)测定体外联合用药对HER2和PD-L1双阳性细胞的增殖抑制作用:
1)96孔板细胞铺板,NCI-N87细胞调整密度为2.0×10 5个/ml,铺板10000个/孔,每孔50μL;
2)细胞培养3-5h后加入PMBC细胞,用含100ng/mL的SEB的培养基调整PBMC密度为8.0×10 5个/ml(PBMC细胞与NCI-N87细胞的比例为4:1),体积50μL;
3)梯度稀释HER2-ADC B,10nM起始,3倍梯度稀释,共9个梯度(10nM,3.3nM,1.1nM,0.37nM,0.12nM,0.04nM,0.01nM,0.004nM,0.001nM),第10孔浓度为0,每孔50μl;上面三排加入25μg/mL抗体A,每孔50μl,下面三排加培养基作为对照,每孔50μl,布板如下表;抗体A和HER2-ADC B采用RPMI1640(Gibco)+10%FBS(Excell Bio)稀释。
4)37℃培养箱,5%CO 2培养约72h,去除上清,加入含10%的CCK8的培养基。
5)37℃孵育4h;酶标仪在吸光度450nm读板。
结果如图1所示,在PBMC存在的条件下,抗体A(抗PD-1抗体)能一定程度的增强HER2-ADC B对NCI-N87肿瘤细胞的增殖抑制作用;通过计算,抗体A和HER2-ADC B联合给药的EC 50为0.032nM,HER2-ADC B单独给药的EC 50为0.045nM。
实施例4 抗体A与抗体C联合给药抑制癌细胞的增殖
本实施例评价了抗体A和抗体C(抗体C由CHO-BAT-KF细胞表达)在免疫检查点人源化小鼠C57BL/6-hPD1/hCTLA4皮下接种MC38结肠癌肿瘤模型中的药效。
1)肿瘤细胞接种
复苏小鼠结肠癌细胞MC38细胞(江苏集萃药康生物科技有限公司),收集对数生长期的MC38细胞(江苏集萃药康生物科技有限公司),去除培养液并用PBS洗两次后接种(荷瘤前、荷瘤后细胞存活率分别为:99.6%及97.8%),接种量为1×10 6/100μL/只,腋下腹腔注射。
2)分组给药
接种后第6天,平均肿瘤体积达到89.19mm 3时,60只雌性小鼠根据肿瘤体积随机分成6组,每组10只。分组当天定义为D0天,并于D0天开始给药;分组给药方案见表4,给药日期为:D0、D4、D7、D11、D14、D18。
3)实验观察和数据采集
细胞接种后,每周常规监测肿瘤对动物正常行为的影响。具体指标包括小鼠的活动性,摄食和饮水情况,体重增加或降低情况,眼睛、被毛及其它异常情况。
开始给药后,于D0、D3、D6、D10、D13、D17、D20、D24、D27观测肿瘤大小并称量小鼠体重。肿瘤体积计算方式为:肿瘤体积(mm 3)=0.5×(肿瘤长径×肿瘤短径 2)。
4)统计
各组小鼠的肿瘤体积、小鼠体重、肿瘤重量等实验结果以平均值±标准误差(mean±SEM)表示。采用独立样本T检验比较不同给药组与对照组相比有无显著性差异。数据采用SPSS进行分析。P<0.05为具有显著性差异。
TGItv(相对肿瘤体积的抑制率)计算公式:
RTV n=V nt/V n0;V nt:编号为n的小鼠在第t天的肿瘤体积,V n0:编号为n的小鼠在第0天的肿瘤体积,RTV n:编号为n的小鼠在第t天的肿瘤相对体积
TGItv=(1-(mean RTV给药组)/(mean RTV 对照组))×100%;mean RTV给药组:给药组RTV平均值,mean RTV 对照组:对照组RTV平均值;
TGItw(肿瘤重量的抑制率)计算公式:
TGItw=(1-(mean TW给药组)/(mean TW 对照组))×100%;Mean TW给药组:给药组小鼠终点处理时肿瘤重量的平均值,Mean TW 对照组:对照组小鼠终点处理时肿瘤重量的平均值。
表4 给药方案
Figure PCTCN2021122878-appb-000016
备注:N为动物只数;BIW x 3W,每周两次给药,给药3周;腹腔给药6次;G5和G6组为联合给药组,先给抗体A,2小时左右给药抗体C,给药浓度不加倍且和单药组的浓度保持一致。
如图2、图3和表5、表6所示,单药抗体A有显著的抗肿瘤效果,抗体C高剂量组显示出显著的抗肿瘤药效,且抗体C呈现出一定的剂量依赖效果,抗体A和抗体C高剂量联合给药组有显著的抗肿瘤药效,且抗肿瘤效果优于单药抗体A和单药抗体C的抗肿瘤效果(G1、G2组未发现小鼠肿瘤完全消退,G3、G6组各有2只小鼠的肿瘤完全消退,G4组有1只小鼠的肿瘤完全消退,G5组有6只小鼠的肿瘤完全消退);如图4所示,各组之间的小鼠体重未发现明显差异,且实验过程中没有小鼠体重低于分组当天小鼠体重的10%,表明小鼠对当前体系中的药物耐受性良好。
表5 不同组别肿瘤体积的抑制率(TGItv)
Figure PCTCN2021122878-appb-000017
表6 第27天时不同组别肿瘤重量的抑制率(TGItw)及P值统计分析(vs G1)
组别 肿瘤重量(g) TGItw P值
G1 1.4662±0.2701 - -
G2 0.7601±0.1302 48.16% 0.030*
G3 0.3144±0.1050 78.56% 0.002**
G4 1.0236±0.2542 30.19% 0.248
G5 0.0796±0.0500 94.57% <0.001***
G6 0.9008±0.2827 38.57% 0.165

Claims (25)

  1. 一种用于治疗肿瘤或癌症的方法,其特征在于,所述方法包括:向有需要的患者给药有效量的抗PD-1抗体和治疗剂;
    所述抗PD-1抗体包含SEQ ID NO:1所示的HCDR1、SEQ ID NO:2所示的HCDR2、SEQ ID NO:3所示的HCDR3、SEQ ID NO:4所示的LCDR1、SEQ ID NO:5所示的LCDR2和SEQ ID NO:6所示的LCDR3。
  2. 如权利要求1所述的方法,其特征在于,所述抗PD-1抗体的重链可变区包含SEQ ID NO:7所示的序列,与SEQ ID NO:7所示序列具有至少80%同一性的序列,或与SEQ ID NO:7所示序列相比具有一或多个保守氨基酸取代的氨基酸序列;和/或
    所述抗PD-1抗体的轻链可变区包含SEQ ID NO:8所示的序列,与SEQ ID NO:8所示序列具有至少80%同一性的序列,或与SEQ ID NO:8所示序列相比具有一或多个保守氨基酸取代的氨基酸序列。
  3. 如权利要求1所述的方法,其特征在于,所述抗PD-1抗体的重链可变区包含SEQ ID NO:7所示的序列,所述抗PD-1抗体的轻链可变区包含SEQ ID NO:8所示的序列。
  4. 如权利要求1所述的方法,其特征在于,所述抗PD-1抗体的重链包含SEQ ID NO:9所示的序列,与SEQ ID NO:9所示序列具有至少80%同一性的序列,或与SEQ ID NO:9所示序列相比具有一或多个保守氨基酸取代的氨基酸序列;和/或
    所述抗PD-1抗体的轻链包含SEQ ID NO:10所示的序列,与SEQ ID NO:10所示序列具有至少80%同一性的序列,或与SEQ ID NO:10所示序列相比具有一或多个保守氨基酸取代的氨基酸序列。
  5. 如权利要求4所述的方法,其特征在于,所述抗PD-1抗体的重链包含如SEQ ID NO:9所示序列,所述抗PD-1抗体的轻链包含如SEQ ID NO:10所示序列。
  6. 如权利要求1-5任一项所述的方法,其特征在于,所述治疗剂选自针对以下靶点的抗体或抗体药物偶联物:EGFR、VEGF、VEGFR2、CTLA4、PD-L1、HER2、CD20、Trop2、Lag3、TIGIT、CD27、OX40、ICOS、BTLA、TIM3、BCMA、c-MET和TAA-1/2/3。
  7. 如权利要求1-5任一项所述的方法,其特征在于,所述治疗剂为如式II所示的抗抗体药物偶联物或其药学上可接受的盐:
    Figure PCTCN2021122878-appb-100001
    其中,Abu为抗HER2抗体,p选自1-10;优选的,p为3.3-3.7;
    或Abu为抗Trop2抗体,p选自1-10;优选的,p为2-3。
  8. 如权利要求7所述的方法,其特征在于,所述抗HER2抗体的重链包含SEQ ID NO:11所示的序列,与SEQ ID NO:11所示序列具有至少80%同一性的序列,或与SEQ ID NO:11所示序列相比具有一或多个保守氨基酸取代的氨基酸序列;和/或
    所述抗HER2抗体的轻链包含SEQ ID NO:12所示的序列,与SEQ ID NO:12所示序列具有至少80%同一性的序列,或与SEQ ID NO:12所示序列相比具有一或多个保守氨基酸取代的氨基酸序列。
  9. 如权利要求8所述的方法,其特征在于,所述抗HER2抗体的重链包含SEQ ID NO:11所示的序列,所述抗HER2抗体的轻链包含SEQ ID NO:12所示的序列。
  10. 如权利要求1-6任一项所述的方法,其特征在于,每个治疗周期内抗PD-1抗体给药的有效量为50mg至600mg。
  11. 如权利要求7所述的方法,其特征在于,每个治疗周期内抗HER2抗体药物偶联物给药的有效量为70mg至400mg。
  12. 如权利要求1-6任一项所述的方法,其特征在于,抗PD-1抗体的每次给药量为1-10mg/kg。
  13. 如权利要求7所述的方法,其特征在于,抗HER2抗体药物偶联物的每次给药量为1-6mg/kg。
  14. 如权利要求1-6任一项所述的方法,其特征在于,所述治疗剂为抗TIGIT抗体。
  15. 如权利要求14所述的方法,其特征在于,所述抗CTLA4抗体重链包含如SEQ ID NO:15所示序列,与SEQ ID NO:15所示序列具有至少80%同一性的序列,或与SEQ ID NO:15所示序列相比具有一或多个保守氨基酸取代的氨基酸序列;和/或
    所述抗CTLA4抗体的轻链包含SEQ ID NO:16所示的序列,与SEQ ID NO:16所示序列具有至少80%同一性的序列,或与SEQ ID NO:16所示序列相比具有一或多个保守氨基酸取代的氨基酸序列。
  16. 如权利要求15所述的方法,其特征在于,所述抗CTLA4抗体重链包含如SEQ ID NO:15所示序列,所述抗CTLA4抗体的轻链包含SEQ ID NO:16所示的序列。
  17. 如权利要求14-16任一项所述的方法,其特征在于,所述抗CTLA4抗体由α-(1,6)-岩藻糖基转移酶基因敲除的CHO细胞系表达。
  18. 如权利要求6所述的方法,其特征在于,每个治疗周期内抗CTLA4抗体给药的有效量为6mg至600mg。
  19. 如权利要求6所述的方法,其特征在于,抗CTLA4抗体的每次给药量为0.1-10mg/kg。
  20. 如权利要求1-19任一项所述的方法,其特征在于,患者给药的一个治疗周期为1周、2周、3周、4周、1个月、5周、6周或7周。
  21. 如权利要求1-20任一项所述的方法,其特征在于,患者接受多个治疗周期治疗,直至病症得到缓解而不再需要治疗。
  22. 如权利要求1-20任一项所述的方法,患者通过静脉输液方式进行给药。
  23. 一种试剂盒,其特征在于,包含抗PD-1抗体、治疗剂和用于指导有需要患者给药PD-1抗体和治疗剂的说明书;
    所述抗PD-1抗体包含SEQ ID NO:1所示的HCDR1、SEQ ID NO:2所示的HCDR2、SEQ ID NO:3所示的HCDR3、SEQ ID NO:4所示的LCDR1、SEQ ID NO:5所示的LCDR2和SEQ ID NO:6所示的LCDR3。
  24. 包含抗PD-1抗体和治疗剂的药物组合物,其特征在于,所述抗PD-1抗体包含SEQ ID NO:1所示的HCDR1、SEQ ID NO:2所示的HCDR2、SEQ ID NO:3所示的HCDR3、SEQ ID NO:4所示的LCDR1、SEQ ID NO:5所示的LCDR2和SEQ ID NO:6 所示的LCDR3。
  25. 如权利要求23所述的试剂盒或24所述的药物组合物,其特征在于,所述治疗剂选自针对以下靶点的抗体或抗体药物偶联物:EGFR、VEGF、VEGFR2、CTLA4、PD-L1、HER2、CD20、Trop2、Lag3、TIGIT、CD27、OX40、ICOS、BTLA、TIM3、BCMA、c-MET和TAA-1/2/3。
PCT/CN2021/122878 2020-10-11 2021-10-09 抗pd-1抗体在联合用药中的应用 WO2022073515A1 (zh)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CN202180069405.9A CN116437926A (zh) 2020-10-11 2021-10-09 抗pd-1抗体在联合用药中的应用
EP21877032.9A EP4226942A1 (en) 2020-10-11 2021-10-09 Use of anti-pd-1 antibody in combination medication
US18/030,886 US20240002511A1 (en) 2020-10-11 2021-10-09 Use of anti-pd-1 antibody in combination therapy

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CNPCT/CN2020/120262 2020-10-11
CN2020120262 2020-10-11

Publications (1)

Publication Number Publication Date
WO2022073515A1 true WO2022073515A1 (zh) 2022-04-14

Family

ID=81125643

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/122878 WO2022073515A1 (zh) 2020-10-11 2021-10-09 抗pd-1抗体在联合用药中的应用

Country Status (4)

Country Link
US (1) US20240002511A1 (zh)
EP (1) EP4226942A1 (zh)
CN (1) CN116437926A (zh)
WO (1) WO2022073515A1 (zh)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014089113A1 (en) 2012-12-03 2014-06-12 Bristol-Myers Squibb Company Enhancing anti-cancer activity of immunomodulatory fc fusion proteins
WO2014094527A1 (en) 2012-12-21 2014-06-26 Bio-Thera Solutions, Ltd. Compounds and methods for the treatment of erb b2/neu positive diseases
WO2018160538A1 (en) * 2017-02-28 2018-09-07 Mersana Therapeutics, Inc. Combination therapies of her2-targeted antibody-drug conjugates
CN109078181A (zh) 2017-08-11 2018-12-25 百奥泰生物科技(广州)有限公司 Trop2阳性疾病治疗的化合物及方法
CN109096399A (zh) 2017-08-11 2018-12-28 百奥泰生物科技(广州)有限公司 一种由基因组被编辑的cho宿主细胞产生的具有独特糖谱的重组抗体及其制备方法
CN111808190A (zh) * 2019-04-10 2020-10-23 百奥泰生物制药股份有限公司 结合pd-1的抗体
CN111973753A (zh) * 2019-05-24 2020-11-24 百奥泰生物制药股份有限公司 抗肿瘤的联合用药物及其应用

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014089113A1 (en) 2012-12-03 2014-06-12 Bristol-Myers Squibb Company Enhancing anti-cancer activity of immunomodulatory fc fusion proteins
WO2014094527A1 (en) 2012-12-21 2014-06-26 Bio-Thera Solutions, Ltd. Compounds and methods for the treatment of erb b2/neu positive diseases
CN104530235A (zh) * 2012-12-21 2015-04-22 百奥泰生物科技(广州)有限公司 一种抑制肿瘤生长的抗体药物衍生物及其制备方法和用途
WO2018160538A1 (en) * 2017-02-28 2018-09-07 Mersana Therapeutics, Inc. Combination therapies of her2-targeted antibody-drug conjugates
CN109078181A (zh) 2017-08-11 2018-12-25 百奥泰生物科技(广州)有限公司 Trop2阳性疾病治疗的化合物及方法
CN109096399A (zh) 2017-08-11 2018-12-28 百奥泰生物科技(广州)有限公司 一种由基因组被编辑的cho宿主细胞产生的具有独特糖谱的重组抗体及其制备方法
CN111808190A (zh) * 2019-04-10 2020-10-23 百奥泰生物制药股份有限公司 结合pd-1的抗体
CN111973753A (zh) * 2019-05-24 2020-11-24 百奥泰生物制药股份有限公司 抗肿瘤的联合用药物及其应用

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
"Current Protocols in Molecular Biology", 2007
CHOTHIA ET AL., J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
JU YEON LEE, HYUN TAE LEE, WOORI SHIN, JONGSEOK CHAE, JAEMO CHOI, SUNG HYUN KIM, HEEJIN LIM, TAE WON HEO, KYEONG YOUNG PARK, YEON : "Structural basis of checkpoint blockade by monoclonal antibodies in cancer immunotherapy", NATURE COMMUNICATIONS, vol. 7, pages 13354, XP055438772, DOI: 10.1038/ncomms13354 *
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1983, U.S. DEPT. OF HEALTH AND HUMAN SERVICES

Also Published As

Publication number Publication date
EP4226942A1 (en) 2023-08-16
US20240002511A1 (en) 2024-01-04
CN116437926A (zh) 2023-07-14

Similar Documents

Publication Publication Date Title
JP2022177090A (ja) 腫瘍壊死因子 (tnf) スーパーファミリー受容体結合分子およびその使用
AU2015229591A1 (en) Anti-EGFRvlll antibodies and uses thereof
WO2000044788A1 (en) Production of tetravalent antibodies
KR102608723B1 (ko) 항―pd-1 항체 및 그의 용도
AU2021273240A1 (en) Anti-tigit antibody and preparation method and application thereof
JP2023527583A (ja) Lag3に結合する抗体およびその使用
JP2022179623A (ja) 抗ox40抗体及びその用途
JP2023037000A (ja) 抗cd40抗体及びその使用
US11667721B2 (en) CD30 bispecific antibodies and method of immunotherapy of CD30+ malignancies
CN113227142B (zh) 抗pd-1抗体及其用途
WO2022184067A1 (zh) 抗tigit抗体在联合用药中的应用
IL293118A (en) Methods for treating cancer by anti-oxo40 antibody in combination with anti-tigit antibody
JP7036471B2 (ja) デスレセプター4及びデスレセプター5に結合する非常に強力な抗体
WO2022073515A1 (zh) 抗pd-1抗体在联合用药中的应用
WO2023001118A1 (zh) 抗ox40抗体在联合用药中的应用
JP2023504630A (ja) 腫瘍治療のための二重特異性融合タンパク質
WO2022242757A1 (zh) 抗pd-1抗体的应用
WO2022105839A1 (zh) 抗ox40抗体在治疗肿瘤或癌症中的应用
WO2024046389A1 (zh) 抗tigit抗体和抗ctla-4抗体联合用于治疗肿瘤中的应用
CN114641500B (zh) 使用抗ox40抗体与抗tim3抗体的组合治疗癌症的方法
WO2022184068A9 (zh) 抗tigit抗体在治疗肿瘤或癌症中的应用
WO2023221936A9 (zh) 双功能蛋白质及其制剂和用途
WO2021098749A1 (en) Methods of cancer treatment with anti-ox40 antibody in combination with radiation
CN116333123A (zh) 抗tnfr2抗体及其用途

Legal Events

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

Ref document number: 21877032

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2021877032

Country of ref document: EP

Effective date: 20230511