CN113244388A - Application of anti-human VEGF antibody and chemical drug combination in preparation of drugs for treating ovarian cancer - Google Patents

Application of anti-human VEGF antibody and chemical drug combination in preparation of drugs for treating ovarian cancer Download PDF

Info

Publication number
CN113244388A
CN113244388A CN202110742169.4A CN202110742169A CN113244388A CN 113244388 A CN113244388 A CN 113244388A CN 202110742169 A CN202110742169 A CN 202110742169A CN 113244388 A CN113244388 A CN 113244388A
Authority
CN
China
Prior art keywords
seq
ovarian cancer
sequence
antibody
tumor
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202110742169.4A
Other languages
Chinese (zh)
Inventor
薛黎婷
杨文清
冯晓飞
张子婧
任晋生
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shandong Simcere Bio Pharmaceutical Co ltd
Original Assignee
Jiangsu Simcere Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Jiangsu Simcere Pharmaceutical Co Ltd filed Critical Jiangsu Simcere Pharmaceutical Co Ltd
Priority to CN202211290780.9A priority Critical patent/CN115814076A/en
Priority to CN202110742169.4A priority patent/CN113244388A/en
Publication of CN113244388A publication Critical patent/CN113244388A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies

Landscapes

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

Abstract

The invention relates to an application of a combination of an anti-human VEGF antibody and a chemical drug in preparation of a drug for treating ovarian cancer, in particular to a combination of the anti-human VEGF antibody and a chemotherapeutic drug, an ovarian cancer treatment method and an application of the combination in preparation of the drug for treating ovarian cancer. The invention is beneficial to increasing the selection of ovarian cancer patients, improving the treatment effect of ovarian cancer and benefiting more ovarian cancer patients.

Description

Application of anti-human VEGF antibody and chemical drug combination in preparation of drugs for treating ovarian cancer
Technical Field
The invention relates to the field of medicines, in particular to application of an anti-human VEGF antibody and a chemical drug in preparation of a drug for treating ovarian cancer.
Background
Ovarian cancer is a malignant tumor of epithelial origin, including epithelial ovarian cancer, fallopian tube cancer and primary peritoneal cancer, the second most common and most mortality gynecological malignancy. Ovarian cancer has no specific tumor markers, is difficult to diagnose early and is usually diagnosed at a later stage. Thus, the prognosis for ovarian cancer is poor, with a 5-year survival rate of only 37.6% in Europe (Luigi Rossi et al, Oncotarget, 2016.8: p.12389-12405). Ovarian cancer is probably around 52100 new cases in china, and the rate of new cases increases by about 2% each year. Annual death cases are probably around 22500 (Chen W. et al, Cancer statistics in China, 2015. CA Cancer J Clin, 2016.66 (2): p. 115-32).
The accepted standard treatments for ovarian cancer are cytoreductive surgery and postoperative platinum-and paclitaxel-based chemotherapy. Despite the high response rate of first-line chemotherapy, up to 70-75% of patients still experience relapse (Luigi Rossi et al, Oncotarget, 2016.8: p.12389-. Ovarian cancer patients relapse within 6 months after treatment with platinum-based drugs is defined as platinum-based resistance. After the first relapse, approximately 25% of patients will develop platinum-based resistance, and eventually almost all relapsing patients will develop platinum-based resistance. In this case, the choice of effective treatment is very few and the prognosis is also poor. There is therefore a need for new therapeutic strategies to improve their clinical prognosis (Pujade-Lauraine, E., et al., J Clin Oncol, 2014.32 (13): p.1302).
Vascular Endothelial Growth Factor (VEGF) is a multifunctional cytokine, is a currently known angiogenesis promoting factor with the strongest effect and the highest specificity, and participates in the generation and development of solid tumors by promoting tumor angiogenesis, increasing Vascular permeability, regulating host anti-tumor immune response and other mechanisms. Studies have shown that anti-human VEGF antibodies have the effect of inhibiting the growth of solid tumors.
The BD0801 antibody is a humanized rabbit anti-human VEGF monoclonal antibody, and the scheme of treating ovarian cancer by the BD0801 antibody is researched and discussed, so that the selection of patients is increased, the treatment effect of ovarian cancer is improved, and more ovarian cancer patients are benefited.
In view of the above, the present invention is particularly proposed.
Disclosure of Invention
The invention provides a combination of an anti-human VEGF antibody and a chemotherapeutic drug, a method for treating ovarian cancer by using the combination or application of the combination in preparing a drug for treating ovarian cancer.
In a first aspect, the present invention provides the use of an anti-human VEGF antibody in combination with a chemotherapeutic agent selected from a taxane compound or a camptothecin compound, in the manufacture of a medicament for the treatment of ovarian cancer, the antibody comprising a heavy chain variable region comprising HCDR1, HCDR2 and HCDR3 and a light chain variable region comprising LCDR1, LCDR2 and LCDR3, wherein,
the sequence of the HCDR1 is shown as SEQ ID NO. 5;
the sequence of the HCDR2 is shown as SEQ ID NO. 8;
the sequence of the HCDR3 is shown as SEQ ID NO. 11;
the sequence of the LCDR1 is shown as SEQ ID NO. 13;
the sequence of the LCDR2 is shown as SEQ ID NO. 15; and the combination of (a) and (b),
the sequence of LCDR3 is shown in SEQ ID NO. 17.
In some embodiments, the heavy chain variable region has the sequence shown in SEQ ID NO. 1 and the light chain variable region has the sequence shown in SEQ ID NO. 3.
In some embodiments, the antibody further comprises a heavy chain constant region having the sequence set forth in SEQ ID NO. 2, and a light chain constant region having the sequence set forth in SEQ ID NO. 4.
In some embodiments, the ovarian cancer is epithelial ovarian cancer, fallopian tube cancer, or primary peritoneal cancer.
In some embodiments, the ovarian cancer is platinum-resistant recurrent ovarian cancer.
In some embodiments, the pathological type of ovarian cancer is serous adenocarcinoma, clear cell adenocarcinoma, or mixed epithelial carcinoma.
In some embodiments, the taxane compound is paclitaxel and the camptothecin compound is topotecan hydrochloride.
In some embodiments, the effective amount of the anti-human VEGF antibody administered is 0.5-2 mg/kg.
In some embodiments, the effective amount of the anti-human VEGF antibody administered is 0.5mg/kg, 1mg/kg, 1.5mg/kg, or 2 mg/kg.
In some embodiments, the dosage form of the anti-human VEGF antibody is a single dose dosage form, each dose containing an amount of the antibody effective for administration to a patient.
In some embodiments, the single dose dosage form contains 25-150 mg of the anti-human VEGF antibody.
In some embodiments, the single dose dosage form contains 25mg, 50mg, 80mg, 100mg, or 150mg of the anti-human VEGF antibody.
In some embodiments, the effective amount of paclitaxel administered is 80mg/m2(ii) a The effective application amount of the topotecan hydrochloride is 4 mg/m2
In a second aspect, the present invention provides the use of an anti-human VEGF antibody in combination with a chemotherapeutic selected from a taxane compound or a camptothecin compound, for the manufacture of a medicament for the treatment of ovarian cancer, said antibody comprising a heavy chain variable region comprising HCDR1, HCDR2 and HCDR3 and a light chain variable region comprising LCDR1, LCDR2 and LCDR3, wherein:
the HCDR1 is selected from a sequence shown in any one of SEQ ID NO 5-7;
the HCDR2 is selected from a sequence shown in any one of SEQ ID NO 8-10;
the HCDR3 is selected from a sequence shown in any one of SEQ ID NO 11-12;
the LCDR1 is selected from a sequence shown in any one of SEQ ID NO. 13-14;
the LCDR2 is selected from a sequence shown in any one of SEQ ID NO. 15-16; and the combination of (a) and (b),
the LCDR3 is selected from a sequence shown in SEQ ID NO. 17.
In some embodiments, the heavy chain variable region has the sequence shown as SEQ ID No. 1, or a sequence that is at least 95%, 96%, 97%, 98%, or 99% identical to SEQ ID No. 1; the light chain variable region has a sequence as shown in SEQ ID NO 3, or a sequence that is at least 95%, 96%, 97%, 98% or 99% identical to SEQ ID NO 3.
In some embodiments, the antibody further comprises a heavy chain constant region having a sequence as set forth in SEQ ID No. 2, or a sequence at least 95%, 96%, 97%, 98%, or 99% identical to SEQ ID No. 2; the antibody also includes a light chain constant region having a sequence as set forth in SEQ ID NO. 4, or a sequence that is at least 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO. 4.
In some embodiments, the ovarian cancer is epithelial ovarian cancer, fallopian tube cancer, or primary peritoneal cancer.
In some embodiments, the ovarian cancer is platinum-resistant recurrent ovarian cancer.
In some embodiments, the pathological type of ovarian cancer is serous adenocarcinoma, clear cell adenocarcinoma, or mixed epithelial carcinoma.
In some embodiments, the taxane compound is paclitaxel and the camptothecin compound is topotecan hydrochloride.
In some embodiments, the effective amount of the anti-human VEGF antibody administered is 0.5-2 mg/kg, preferably 0.5mg/kg, 1mg/kg, 1.5mg/kg or 2 mg/kg.
In some embodiments, the anti-human VEGF antibody is provided in a single dosage form, each containing an amount of antibody effective for administration to a patient, preferably, the single dosage form contains 25 to 150mg of the anti-VEGF antibody, more preferably 25mg, 50mg, 80mg, 100mg, or 150 mg.
In some embodiments, the effective amount of paclitaxel administered is 80mg/m2
In some embodiments, the effective amount of topotecan hydrochloride administered is 4 mg/m2
In a third aspect, the invention also provides a method of treating ovarian cancer, the method comprising administering to an ovarian cancer patient an effective amount of an anti-human VEGF antibody in combination with a chemotherapeutic selected from a taxane or a camptothecin compound, the antibody comprising a heavy chain variable region comprising HCDR1, HCDR2 and HCDR3, and a light chain variable region comprising LCDR1, LCDR2 and LCDR3, wherein:
the HCDR1 is selected from a sequence shown in any one of SEQ ID NO 5-7;
the HCDR2 is selected from a sequence shown in any one of SEQ ID NO 8-10;
the HCDR3 is selected from a sequence shown in any one of SEQ ID NO 11-12;
the LCDR1 is selected from a sequence shown in any one of SEQ ID NO. 13-14;
the LCDR2 is selected from a sequence shown in any one of SEQ ID NO. 15-16; and the combination of (a) and (b),
the LCDR3 is selected from a sequence shown in SEQ ID NO. 17.
In some embodiments, the heavy chain variable region has the sequence shown as SEQ ID No. 1, or a sequence that is at least 95%, 96%, 97%, 98%, or 99% identical to SEQ ID No. 1; the light chain variable region has a sequence as shown in SEQ ID NO 3, or a sequence that is at least 95%, 96%, 97%, 98% or 99% identical to SEQ ID NO 3.
In some embodiments, the antibody further comprises a heavy chain constant region having a sequence as set forth in SEQ ID No. 2, or a sequence at least 95%, 96%, 97%, 98%, or 99% identical to SEQ ID No. 2; the antibody also includes a light chain constant region having a sequence as set forth in SEQ ID NO. 4, or a sequence that is at least 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO. 4.
In some embodiments, the ovarian cancer is epithelial ovarian cancer, fallopian tube cancer, or primary peritoneal cancer.
In some embodiments, the ovarian cancer is platinum-resistant recurrent ovarian cancer.
In some embodiments, the pathological type of ovarian cancer is serous adenocarcinoma, clear cell adenocarcinoma, or mixed epithelial carcinoma.
In some embodiments, the taxane compound is paclitaxel and the camptothecin compound is topotecan hydrochloride.
In some embodiments, the effective amount of the anti-human VEGF antibody administered is 0.5-2 mg/kg, preferably 0.5mg/kg, 1mg/kg, 1.5mg/kg or 2 mg/kg.
In some embodiments, the effective amount of paclitaxel administered is 80mg/m2
In some embodiments, the effective amount of topotecan hydrochloride administered is 4 mg/m2
In some embodiments, the anti-human VEGF antibody is administered 1 time every 2 weeks.
In some embodiments, the paclitaxel is administered 1 time per week.
In some embodiments, the topotecan hydrochloride is administered 3 times every 4 weeks, preferably 1 time per week for the first 3 weeks.
In some embodiments, the combination is administered for a period of 28 days, the anti-human VEGF antibody is administered 1 time each on days 1 and 15, the paclitaxel is administered 1 time each on days 1, 8, 15 and 22, and the topotecan hydrochloride is administered 1 time each on days 1, 8 and 15.
In a fourth aspect, the invention also provides a combination of an anti-human VEGF antibody and a chemotherapeutic selected from a taxane or camptothecin compound for use in the treatment of ovarian cancer, the antibody comprising a heavy chain variable region comprising HCDR1, HCDR2 and HCDR3 and a light chain variable region comprising LCDR1, LCDR2 and LCDR3, wherein:
the HCDR1 is selected from a sequence shown in any one of SEQ ID NO 5-7;
the HCDR2 is selected from a sequence shown in any one of SEQ ID NO 8-10;
the HCDR3 is selected from a sequence shown in any one of SEQ ID NO 11-12;
the LCDR1 is selected from a sequence shown in any one of SEQ ID NO. 13-14;
the LCDR2 is selected from a sequence shown in any one of SEQ ID NO. 15-16; and the combination of (a) and (b),
the LCDR3 is selected from a sequence shown in SEQ ID NO. 17.
In some specific embodiments, the heavy chain variable region has the sequence shown as SEQ ID No. 1, or a sequence that is at least 95%, 96%, 97%, 98% or 99% identical to SEQ ID No. 1; the light chain variable region has a sequence as shown in SEQ ID NO 3, or a sequence that is at least 95%, 96%, 97%, 98% or 99% identical to SEQ ID NO 3.
In some specific embodiments, the antibody further comprises a heavy chain constant region having the sequence shown as SEQ ID No. 2, or a sequence that is at least 95%, 96%, 97%, 98%, or 99% identical to SEQ ID No. 2; the antibody also includes a light chain constant region having a sequence as set forth in SEQ ID NO. 4, or a sequence that is at least 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO. 4.
In some embodiments, the ovarian cancer is epithelial ovarian cancer, fallopian tube cancer, or primary peritoneal cancer.
In some embodiments, the ovarian cancer is platinum-resistant recurrent ovarian cancer.
In some embodiments, the pathological type of ovarian cancer is serous adenocarcinoma, clear cell adenocarcinoma, or mixed epithelial carcinoma.
In some embodiments, the taxane compound is paclitaxel and the camptothecin compound is topotecan hydrochloride.
In some embodiments, the effective amount of the anti-human VEGF antibody administered is 0.5-2 mg/kg, preferably 0.5mg/kg, 1mg/kg, 1.5mg/kg or 2 mg/kg.
In some specific embodiments, the dosage form of the anti-human VEGF antibody is a single dosage form, each containing an amount of the antibody effective for administration to a patient, preferably, the single dosage form contains 25 to 150mg of the anti-VEGF antibody, more preferably 25mg, 50mg, 80mg, 100mg, or 150 mg.
In some specific embodiments, the effective amount of paclitaxel administered is 80mg/m2
In some specific embodiments, the effective amount of topotecan hydrochloride administered is 4 mgm2
Definition and description of terms
Unless defined otherwise herein, scientific and technical terms used in connection with the present invention shall have the meanings that are understood by those of ordinary skill in the art.
Furthermore, unless otherwise indicated herein, singular terms herein shall include the plural and plural terms shall include the singular. More specifically, as used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the content clearly dictates otherwise.
The terms "comprising," "including," and "having," used interchangeably herein, are intended to be inclusive and mean that there may be additional elements other than the listed elements. It should also be understood that the use of "including," "comprising," and "having" descriptions herein also provides a "consisting of … …" solution.
The term "and/or" as used herein includes the meanings of "and", "or" and "all or any other combination of elements linked by the term.
The term "antibody" is used herein in the broadest sense and refers to a polypeptide or combination of polypeptides that comprises sufficient sequence from an immunoglobulin heavy chain variable region and/or sufficient sequence from an immunoglobulin light chain variable region to be capable of specifically binding to an antigen. Herein, "antibody" encompasses various forms and various structures as long as they exhibit the desired antigen-binding activity, and exemplarily includes whole antibodies as well as antibody fragments having binding antigen-binding activity. The term "desired antigen binding activity" as used herein means, for example, that an antibody typically binds specifically to an antigen and to substantially the same antigen with high affinity, but does not bind to an unrelated antigen with high affinity. Affinity is usually reflected in an equilibrium dissociation constant (KD), where a lower KD indicates a higher affinity. Illustratively, high affinity generally means having about 10-7M or less, about 10-8M or less, about 1X 10-9M or less, about 1X 10-10M or less,1×10-11M or less or 1X 10-12M or lower KD. KD is calculated as follows: KD = KD/Ka, where KD represents the dissociation rate and Ka represents the association rate. The equilibrium dissociation constant KD can be measured using methods well known in the art, such as surface plasmon resonance (e.g., Biacore) or equilibrium dialysis assay.
The term "antibody" herein includes a typical "four-chain antibody" belonging to an immunoglobulin composed of two Heavy Chains (HC) and two Light Chains (LC); heavy chain refers to a polypeptide chain consisting of, in the N-terminal to C-terminal direction, a heavy chain variable region (VH), a heavy chain constant region CH1 domain, a Hinge Region (HR), a heavy chain constant region CH2 domain, a heavy chain constant region CH3 domain; and, when the full-length antibody is of IgE isotype, optionally further comprising a heavy chain constant region CH4 domain; a light chain is a polypeptide chain consisting of a light chain variable region (VL) and a light chain constant region (CL) in the N-terminal to C-terminal direction; the heavy chains are connected with each other and the heavy chains are connected with each other through disulfide bonds to form a Y-shaped structure. Because of the differences in the amino acid composition and arrangement of the constant regions of immunoglobulin heavy chains, their antigenicity also differs. Accordingly, the term "immunoglobulin" is used herein to refer to five classes, or isotypes called immunoglobulins, namely IgM, IgD, IgG, IgA and IgE, with the corresponding heavy chains being the μ, δ, γ, α and ε chains, respectively. The same class of igs can be divided into different subclasses according to differences in amino acid composition of the hinge region and the number and position of heavy chain disulfide bonds, for example, iggs can be classified into IgG1, IgG2, IgG3 and IgG4, and IgA can be classified into IgA1 and IgA 2. Light chains are classified as either kappa or lambda chains by differences in the constant regions. Each of the five classes of Ig may have either a kappa chain or a lambda chain.
The "antibody" herein may be derived from any animal including, but not limited to, humans and non-human animals which may be selected from primates, mammals, rodents and vertebrates, such as camelids, llamas, prosiches, alpacas, sheep, rabbits, mice, rats or chondroiidae (e.g. shark).
The term "humanized antibody" herein refers to a non-human antibody that has been genetically engineered to have an amino acid sequence modified to increase homology to the sequence of a human antibody. Generally, all or a portion of the CDR regions of a humanized antibody are derived from a non-human antibody (donor antibody), and all or a portion of the non-CDR regions (e.g., variable region FR and/or constant regions) are derived from a human immunoglobulin (acceptor antibody). Humanized antibodies typically retain or partially retain the desired properties of the donor antibody, including, but not limited to, antigen specificity, affinity, reactivity, the ability to increase immune cell activity, the ability to enhance an immune response, and the like.
The term "variable region" herein refers to the region of an antibody heavy chain or light chain involved in binding an antibody to an antigen, the "heavy chain variable region" being used interchangeably with "VH", "HCVR" and the "light chain variable region" being used interchangeably with "VL", "LCVR". The variable domains of the heavy and light chains of natural antibodies (VH and VL, respectively) generally have similar structures, each domain comprising four conserved Framework Regions (FR) and three hypervariable regions (HVRs). See, e.g., Kindt et al, Kuby Immunology,6th ed., w.h.freeman and co., p.91 (2007). A single VH or VL domain may be sufficient to confer antigen binding specificity.
The terms "complementarity determining region" and "CDR" are used interchangeably herein and generally refer to the hypervariable region (HVR) of a heavy chain variable region (VH), which may be abbreviated as HCDR, or a light chain variable region (VL), which is also referred to as a complementarity determining region because it is sterically complementary to an epitope precisely. The terms "framework region" or "FR region" are used interchangeably herein to refer to those amino acid residues in an antibody heavy chain variable region or light chain variable region other than the CDRs. Generally, a typical antibody variable region consists of 4 FR regions and 3 CDR regions in the following order: FR1-CDR1-FR2-CDR2-FR3-CDR3-FR 4.
For further description of the CDRs, reference is made to Kabat et al, J. biol. chem.,252: 6609-; kabat et Al, U.S. department of health and public service, "Sequences of proteins of immunological interest" (1991), Chothia et Al, J.mol. biol. 196:901-917 (1987); Al-Lazikani B. et Al, J.mol. biol. 273: 927-948 (1997); MacCallum et Al, J.mol. biol. 262:732-745 (1996); Abhinandan and Martin, mol. Immunol. 45: 3832-3839 (2008); Lefranc M.P. et Al, Dev. Comp. Immunol., 27: 55-77 (2003)), and Honegger and Pluckthun, J.mol. biol. 309:657 (670); J.mol. biol. 2001). The "CDRs" herein may be labeled and defined by means known in the art, including but not limited to Kabat numbering system, Chothia numbering system, or IMGT numbering system, using tool sites including but not limited to AbRSA sites (http:// cao. lab share. cn/AbRSA/CDRs. php), abYsis sites (www.abysis.org/analysis/sequence _ input/key _ annotation. cgi), and IMGT sites (http:// www.imgt.org/3D structure-DB/cgi/DomainGalign. cgi # results).
Illustratively, SEQ ID NO:1 or 3 are as shown in the following table: wherein Kabat and Chothia use the abYsis website and IMGT uses the IMGT website.
Watch (A)
Figure 352993DEST_PATH_IMAGE001
Anti-human VEGF antibody heavy chain and light chain CDR region amino acid sequence table
Figure 473396DEST_PATH_IMAGE003
The term "heavy chain constant region" herein refers to the carboxy-terminal portion of an antibody heavy chain that is not directly involved in binding of the antibody to an antigen, but exhibits effector functions, such as interaction with an Fc receptor, which has a more conserved amino acid sequence relative to the variable domain of the antibody. The "heavy chain constant region" comprises at least: a CH1 domain, a hinge region, a CH2 domain, a CH3 domain, or a variant or fragment thereof. The "heavy chain constant region" includes a "full-length heavy chain constant region" having a structure substantially similar to a natural antibody constant region, and a "heavy chain constant region fragment" including only a portion of the full-length heavy chain constant region. Illustratively, a typical "full-length antibody heavy chain constant region" consists of the CH1 domain-hinge region-CH 2 domain-CH 3 domain; when the antibody is IgE, it further comprises a CH4 domain; when the antibody is a heavy chain antibody, it does not include a CH1 domain. Exemplary "heavy chain constant region fragments" may be selected from the CH1, Fc, or CH3 domains.
The term "light chain constant region" herein refers to the carboxy-terminal portion of an antibody light chain that is not directly involved in binding of the antibody to an antigen, which light chain constant region may be selected from a constant kappa domain or a constant lambda domain.
The term "Fc" herein refers to the carboxy-terminal portion of an antibody that is papain-hydrolyzed from an intact antibody, typically comprising the CH3 and CH2 domains of the antibody. Fc regions include, for example, native sequence Fc regions, recombinant Fc regions, and variant Fc regions. Although the boundaries of the Fc region of an immunoglobulin heavy chain may vary slightly, the Fc region of a human IgG heavy chain is generally defined as extending from the amino acid residue at position Cys226 or from Pro230 to its carboxy terminus. The C-terminal lysine of the Fc region (residue 447 according to the Kabat numbering system) may be removed, for example, during production or purification of the antibody, or by recombinant engineering of the nucleic acid encoding the heavy chain of the antibody, and thus, the Fc region may or may not include Lys 447.
The term "identity" herein can be calculated by: to determine the percent "identity" of two amino acid sequences or two nucleic acid sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of the first and second amino acid sequences or nucleic acid sequences for optimal alignment or non-homologous sequences can be discarded for comparison purposes). The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position.
The percent identity between two sequences varies with the same position shared by the sequences, taking into account the number of gaps that need to be introduced and the length of each gap for optimal alignment of the two sequences.
The term "drug" herein refers to a substance used for the prevention, treatment and diagnosis of a disease, including "pharmaceutical compositions" or "drug combinations", illustratively, the drug also includes a cell therapeutic agent, such as a therapeutic CAR-T cell or CAR-NK cell. By "pharmaceutical composition" is meant a formulation that is present in a form effective to allow the biological activity of the active ingredient contained therein, and that does not contain additional ingredients that have unacceptable toxicity to the subject to which the pharmaceutical composition is administered. The term "combination" or "pharmaceutical combination" as used herein refers to an unfixed combination, wherein the active agent and the at least one further active agent may be administered separately, either simultaneously or within time intervals, in particular where these time intervals allow the combination partners to show a cooperative (e.g. synergistic) effect. The term "non-fixed combination" means that the active ingredients (e.g., one active agent and at least one additional active agent) are both administered to a patient as separate entities either simultaneously or sequentially without specific time constraints, wherein such administration provides therapeutically effective levels of both compounds in the patient.
The term "treatment" herein refers to surgery or drug treatment (therapeutic or therapeutic treatment) with the purpose of preventing, slowing (reducing) the progression of an undesired physiological change or pathology, such as a cancer or tumor, in a subject being treated. Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or complete), whether detectable or undetectable. Subjects in need of treatment include subjects already suffering from a condition or disease as well as subjects susceptible to a condition or disease or subjects for whom prevention of a condition or disease is intended. When terms such as slow, etc., are referred to, their meanings also include the elimination, disappearance, absence, etc.
The term "patient" or "subject" herein refers to an organism that is receiving treatment for a particular disease or disorder as described herein. Examples of "patients" or "subjects" include mammals, such as humans, primates (e.g., monkeys), or non-primate mammals, that are being treated for a disease or disorder.
The term "effective amount" herein refers to an amount of a therapeutic agent that is effective to prevent or ameliorate a disease condition or the progression of the disease when administered alone or in combination with another therapeutic agent to a cell, tissue, or subject. An "effective amount" also refers to an amount of a compound or cell having a therapeutic effect (e.g., CAR-T cells, CAR-NK cells) sufficient to alleviate symptoms, e.g., to treat, cure, prevent, or alleviate a related medical condition, or to treat, cure, prevent, or alleviate an increased rate of such a condition. When the active ingredient is administered alone to an individual, a therapeutically effective dose refers to the ingredient alone.
The term "cancer" herein refers to or describes a physiological condition in mammals that is typically characterized by unregulated cell growth. Included in this definition are benign and malignant cancers. The term "tumor" or "neoplasm" herein refers to all neoplastic (neoplastic) cell growth and proliferation, whether malignant or benign, and all pre-cancerous (pre-cancerous) and cancerous cells and tissues. The terms "cancer" and "tumor" are not mutually exclusive when referred to herein.
The term "patient" or "subject" herein refers to an organism that is receiving treatment for a particular disease or disorder as described herein. Examples of "patients" or "subjects" include mammals, such as humans, primates (e.g., monkeys), or non-primate mammals, that are being treated for a disease or disorder.
Drawings
FIG. 1 anti-tumor effects of BD0801, Avastin and Paclitaxel in human ovarian cancer OVCAR-8 nude mouse graft tumor model.
FIG. 2 anti-tumor effects of BD0801, Avastin and Paclitaxel in combination and single drug in human ovarian cancer OVCAR-8 nude mouse transplantation tumor model.
FIG. 3 anti-tumor effects of BD0801, Avastin and Paclitaxel as single drugs and in combination in a human ovarian cancer SK-OV-3 nude mouse transplantation tumor model.
FIG. 4. BD0801 in combination with PTX subjects Objective Remission Rate (ORR).
FIG. 5 is a graph of Objective Remission Rate (ORR) of BD0801 combined PTX/TOPO ovarian cancer patients versus BD0801 AUC.
Figure 6, median Progression Free Survival (PFS) profile in subjects overall.
FIG. 7 BD0801 in combination with PTX exposure as a function of percentage reduction of the sum of tumor longest diameter (SLD).
Detailed Description
The invention will be further described with reference to specific embodiments, and the advantages and features of the invention will become apparent as the description proceeds. The examples, in which specific conditions are not specified, were conducted under conventional conditions or conditions recommended by the manufacturer. The reagents or instruments used are not indicated by the manufacturer, and are all conventional products available commercially.
The examples are exemplary only and do not limit the scope of the invention in any way. It will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the spirit and scope of the invention, and that such changes and modifications may be made without departing from the spirit and scope of the invention.
The BD0801 antibody used in the following examples was produced and supplied by Shandong Mitsui Bio-pharmaceuticals, Inc. and stored at 2-8 ℃. The sequence information of the BD0801 antibody is shown below.
Heavy chain variable region (SEQ ID NO: 1):
EVQLVESGGGLVKPGGSLRLSCAASGFSFSNNDVMCWVRQAPGKGLEWIGCIMTTDVVTEYANWAKSRFTVSRDSAKNSVYLQMNSLRAEDTAVYFCARDSVGSPLMSFDLWGPGTLVTVSS;
heavy chain constant region (SEQ ID NO: 2):
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK;
light chain variable region (SEQ ID NO: 3):
DIQMTQSPSSLSASVGDRVTINCQASQSIYNNNELSWYQQKPGKPPKLLIYRASTLASGVPSRFSGSGSGTDFTLTISSLQPEDVATYYCGGYKSYSNDGNGFGGGTKVEIK;
light chain constant region (SEQ ID NO: 4):
RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC。
example 1 evaluation of therapeutic Effect of BD0801 on human ovarian cancer nude mouse transplanted tumor model
1.1 test materials and tumor evaluation methods (unless otherwise specified, this example uses the test materials and tumor evaluation methods described in 1.1)
The tested drugs are:
BD0801 is manufactured and provided by tsukusan shou bio-pharmaceuticals, inc;
bevacizumab (Avastin) was purchased from Roche (Roche);
paclitaxel (Paclitaxel) was purchased from Beijing synergia.
The preparation method of the medicine comprises the following steps: the drugs were prepared and diluted with sterile normal saline (purchased from Jiangsu Huai' an double crane, pharmaceutical Co., Ltd.).
OVCAR-8 cells: national Institute of Health (NIH).
SK-OV-3 cells: shanghai national institute of Life sciences (SIBS), Chinese academy of sciences.
Reagent: RPMI 1640: gibco, cat number: c22400500 BT;
fetal bovine serum: gibco, cat number: 10099-141C;
PBS: hyclone, cat # s: SH 30256.01;
matrix glue: corning, cargo number: 354234.
the tumor evaluation method comprises the following steps:
tumor volume V = 0.5 × a × b2Wherein a is the major diameter of the tumor and b is the minor diameter of the tumor;
relative tumor volume RTV = Vt / V0In which V is0Tumor volume of the animal at the time of grouping; vtIs the tumor volume of the animal after treatment.
Relative tumor proliferation rate T/C (%) = TRTV/CRTVX 100% where TRTVMean RTV for treatment groups; cRTVThe negative control group mean RTV.
Tumor growth inhibition ratio TGI (%) = (1-T/C) × 100%.
Single medicine for evaluating curative effect of human ovarian cancer OVCAR-8 nude mouse transplantation tumor model
Experimental animals: BALB/c Nude mice, female, 6-8 weeks, and 18.5-25.5 g in weight, purchased from Jiangsu Jiejiaokang Biotech, Inc., production license number: SCXK (su) 2018-: 201911201. a breeding environment: SPF grade.
OVCAR-8 cells were cultured in RPMI1640 medium containing 10% fetal bovine serum. Collecting cells in exponential growth phase, 1X 107OVCAR-8 cells were resuspended in 1: 1 (0.1 ml/mouse) in a matrix gel to inoculate the cells subcutaneously in the experimental mice. When the tumor grows to the average volume of 158.62 mm3Group administration, the day of group administration being defined as day 0 (D0), mice were randomized into 8 groups (n = 8), and each group of mice was administered saline (vehicle), 0.8 mg/kg, 2.5mg/kg or 7.5mg/kg of the test drug BD0801, 0.8 mg/kg, 2.5mg/kg or 7.5mg/kg of the control drug Avastin, or 15 mg/kg of the positive control drug Paclitaxel, respectively, via the tail vein. BD0801 and Avastin, administered 2 times per week for about 6 weeks, Paclitaxel administered 1 time every four days for 4 times. Tumor volume was measured 2 times per week, mice were weighed, data was recorded, and tumor size change and mouse weight change were counted over about 6 weeks. Therefore, in the human ovarian cancer OVCAR-8 nude mouse transplantation tumor model, the BD0801 can obviously inhibit the tumor growth at 2.5mg/kg and 7.5mg/kg, and the tumor inhibition effect is better than Avastin.
On day 38 after administration, the tumor volumes of the 0.8 mg/kg BD0801 group were not statistically different from those of the Vehicle control group, and the tumor volumes of the 2.5mg/kg and 7.5mg/kg BD0801 groups showed significant tumor suppression effects compared with those of the control group. The tumor volumes of the 0.8 mg/kg, 2.5mg/kg and 7.5mg/kg Avastin groups were not statistically different compared to the Vehicle control group. The tumor inhibition effect of BD0801 is better than that of Avastin under the same dosage, and the statistical difference exists between the two dosages of 0.8 mg/kg and 2.5 mg/kg. No weight loss or other significant toxic response was seen in the animals for the different doses of BD0801 and Avastin. 15 mg/kg of Paclitaxel also produced significant tumor suppression, but the drug had a significant toxic response, with 2 of 8 mice dying. See table 2 and figure 1 for details. The result shows that in the human ovarian cancer OVCAR-8 nude mouse transplantation tumor model, BD0801 can obviously inhibit the tumor growth at 2.5mg/kg and 7.5mg/kg, and the tumor inhibition effect is superior to Avastin.
TABLE 2 tumor inhibiting Effect of test substances on OVCAR-8 xenograft tumor model
Figure 971242DEST_PATH_IMAGE004
Note: A. data are shown as mean ± Standard Deviation (SD).
B. PValues obtained from analysis of relative tumor volumes using the two-way ANOVA, shownPThe values are compared between each group and the vehicle control group.
C. The 2 tumor-bearing mice of the Paclixel 15 mg/kg group were found dead on days 5 and 12, respectively, and none of these two mice were found in the tumor volume and corresponding calculation on day 38.
Evaluation of efficacy of human ovarian cancer OVCAR-8 nude mouse transplantation tumor in combination with chemotherapy
Experimental animals: BALB/c Nude mice, female, 6-8 weeks, body weight 16.2-21.5 g, purchased from Jiangsu Jiejiaokang Biotech, Inc., production license number: SCXK (su) 2018-: 202002168. a breeding environment: SPF grade.
OVCAR-8 cells were cultured in RPMI1640 medium containing 10% fetal bovine serum. Collecting cells in exponential growth phase, 1X 107OVCAR-8 cells were resuspended in 1: 1 (0.1 ml/mouse) in a matrix gel to inoculate the cells subcutaneously in the experimental mice. When the tumor grows to the average volume of 159.42 mm3Group administration, defined as day 0 on the day of group administration (D0), mice were randomized into 6 groups (n = 8), and each group of mice was given saline (Vehicle) via the tail vein, 2.5mg/kg of the test drug BD0801, 2.5mg/kg of the control drug Avastin, 10 mg/kg of the positive control drug Paclitaxel, 2.5mg/kg of BD0801 in combination with 10 mg/kg of Paclitaxel, or 2.5mg/kg of Avastin in combination with 10 mg/kg of Paclitaxel, respectively. BD0801 and Avastin administered 2 times per week for 4 weeks, Paclitaxel administered 1 time every four days for 4 times. Tumor volume was measured 2 times per week, mice were weighed, data was recorded, and tumor size change and mouse weight change were counted over about 4 weeks.
On the 24 th day after the administration, 2.5mg/kg BD0801 or 2.5mg/kg Avastin can obviously inhibit the growth of tumors, and the tumor inhibition effect is equivalent. The 2.5mg/kg Avastin and 10 mg/kg Paclitaxel combination group did not show significantly enhanced anti-tumor effects relative to the Avastin monotherapy group; the combination of 2.5mg/kg BD0801 and 10 mg/kg Paclixel showed significantly enhanced tumor suppression compared with the two single drug groups, and the tumor suppression effect of the combined group tended to be superior to that of the Avastin and Paclixel combined group. The tumor-bearing mice were resistant to treatment with BD0801 alone at 2.5mg/kg, Avastin at 2.5mg/kg, Paclitaxel 10 mg/kg, and BD 08012.5 mg/kg in combination with Paclitaxel 10 mg/kg, Avastin at 2.5mg/kg in combination with Paclitaxel 10 mg/kg. See table 3 and figure 2 for details. The result shows that in a human ovarian cancer OVCAR-8 nude mouse transplantation tumor model, 2.5mg/kg BD0801 single drug treatment has obvious tumor inhibition effect, and when the drug is combined with 10 mg/kg Paclixel, the drug shows obviously enhanced tumor inhibition effect relative to two single drug groups, and has the trend of obviously better antitumor effect than the combined group of the same dose of Avastin and Paclixel.
TABLE 3 tumor-inhibiting Effect of test substances on OVCAR-8 xenograft tumor model
Figure 632031DEST_PATH_IMAGE005
Note: A. data are shown as mean ± SD.
B. PValues obtained from analysis of relative tumor volumes using the two-way ANOVA, shownPThe values are compared between each group and the vehicle control group.
Evaluation of curative effect on human ovarian cancer SK-OV-3 nude mouse transplantation tumor-combined chemotherapy
Experimental animals: BALB/c Nude mice, female, 6-8 weeks, and 18.0-23.5 g in weight, purchased from Jiangsu Jiejiaokang Biotech, Inc., production license number: SCXK (su) 2018-: 202002168. a breeding environment: SPF grade.
SK-OV-3 cells were cultured in McCoy's 5a medium (McCoy's 5 a: Gibco, cat # 12330-031) containing 10% fetal bovine serum (Isuase, cat # FND 500). Collecting cells in exponential growth phase, 1X 107SK-OV-3 cells were resuspended in 1: 1 (0.1 ml/mouse) in a matrix gel to inoculate the cells subcutaneously in the experimental mice. When the tumor grows to the average volume of 135.48 mm3Group administration, the day of group administration being defined as day 0 (D0), the mice were randomized into 8 groups (n = 8), each group of mice being administered saline (vehicle) via the tail vein, 2.5mg/kg or 7.5mg/kg of the test drug BD0801, 2.5mg/kg or 7.5mg/kg of the control drug Avastin, 10 mg/kg of the positive control drug Paclitaxel, 10 mg/kg of Paclitaxel in combination with 2.5mg/kg BD0801, or 10 mg/kg of Paclitaxel in combination with 2.5mg/kg Avastin, respectively. BD0801 and Avastin, administered 2 times per week for 4 weeks, Paclitaxel administered 1 time every four days for 4 times. Tumor volume was measured 2 times per week, mice were weighed, data was recorded, and tumor size change and mouse weight change were counted over about 4 weeks.
On day 24 after administration, all the administration groups showed significant tumor growth inhibitory activity compared to the control group. The test drug BD0801 showed very good dose dependence, whereas the control drug Avastin showed a slightly weaker dose dependence. The tumor inhibition effect of BD0801 is equivalent to that of Avastin at the dose of 2.5mg/kg, and the tumor inhibition effect of BD0801 is obviously superior to that of Avastin at the dose of 7.5 mg/kg. The 2.5mg/kg Avastin and 10 mg/kg Paclitaxel combination group did not show significantly enhanced anti-tumor effects relative to the Avastin monotherapy group; while the combination of 2.5mg/kg BD0801 and 10 mg/kg Paclixel showed significantly enhanced tumor suppression relative to both single drug groups. The tumor-bearing mice were tolerated by treatment with BD0801 at 2.5mg/kg and 7.5mg/kg, respectively, with Avastin administered alone at 2.5mg/kg and 7.5mg/kg, respectively, and with Paclitaxel at a dose of 10 mg/kg, and with BD 08012.5 mg/kg administered in combination with Paclitaxel at a dose of 10 mg/kg, and with Avastin at 2.5mg/kg administered in combination with Paclitaxel at a dose of 10 mg/kg. See table 4 and figure 3 for details. The result shows that in a human ovarian cancer SK-OV-3 nude mouse transplantation tumor model, BD0801 and Avastin treatment have obvious tumor inhibition effect on an SK-OV-3 mouse ovarian cancer model, BD0801 shows a very good dose response relationship, and the Avastin dose response relationship is slightly weak. BD0801 showed significantly better tumor suppression at high doses of 7.5mg/kg than Avastin at the same dose, and significantly enhanced tumor suppression at 2.5mg/kg in combination with Paclitaxel relative to both single drug groups.
TABLE 4 tumor-inhibiting Effect of test substances on SK-OV-3 xenograft tumor model
Figure 915245DEST_PATH_IMAGE006
Note: A. data are shown as mean ± SD.
B. PValues obtained from analysis of relative tumor volumes using the two-way ANOVA, shownPThe values are compared between each group and the vehicle control group.
Example 2 clinical trials of BD0801 combination chemotherapy in ovarian cancer patients
Purpose of study
BD0801 monoclonal antibody was evaluated for safety and tolerability, pharmacokinetic profile and anti-tumor efficacy in combination with chemotherapy for platinum-resistant relapsed epithelial ovarian cancer, fallopian tube cancer and primary peritoneal carcinoma patients.
Design of research
Adopts a multicenter, open and BD0801 monoclonal antibody dosage increasing design and combines chemotherapy to treat patients with platinum drug-resistant recurrent epithelial ovarian cancer, salpingemphraxis and primary peritoneal carcinoma.
The test is divided into two parts: the first stage is a dose increasing stage, the BD0801 monoclonal antibody dose increment is calculated to be 0.5mg/kg, 1mg/kg, 1.5mg/kg and 2mg/kg, the administration is carried out once every 2 weeks, the dose is increased gradually so as to complete the dose-limiting toxicities (DLTs) occurring in 2 administration cycles (two BD0801 monoclonal antibody combination chemotherapies, generally for 28 days, and the maximum dose does not exceed 56 days) as a basis, and blood sample specimens are collected during the period for determining the drug concentration. The chemotherapeutic drug selected for combination was paclitaxel (once weekly) or topotecan hydrochloride (3 times every 4 weeks). After treatment is complete, efficacy is assessed and subjects with clinical benefit can continue to take the drug at the original dose and frequency until disease progression or intolerance of adverse reactions occurs or informed consent/loss is withdrawn or a new anti-tumor treatment is initiated.
The second stage is an extension into the group stage to further investigate the safety and effectiveness of the product. If no DLT was observed in the first 3 subjects of each dose group at the end of the first 2 treatment cycles (week 4) or only 1 subject observed DLT at the end of the first 2 treatment cycles (week 4), then the cohort was allowed to continue to 8-12. In addition, a particular dose group can be selected to extend up to 40 subjects, discussed collectively by investigators and sponsors. The administration method and blood sampling method are the same as those of the subjects in the dose group.
Dose escalation and termination criteria:
no dose escalation was performed in the subject individuals. Dose escalation between groups will be guided by the occurrence of dose-limiting toxicity (DLT) over 2 dosing cycles (two administrations of BD0801 mab, typically 28 days, up to 56 days). Dose escalation was performed by the traditional "3 + 3" method, and each dose group was first administered to 1 subject, and after 1 administration of BD0801 mab (d 14), 2 subjects were administered again if DLT did not occur. At least 3 subjects completed treatment and evaluation on cycles 1 and 2 per dose group and were available for DLT assessment, and were escalated to the next dose in the absence of DLT; 1 patient presented with DLT as in the previous 3, and 3 additional patients enrolled into the dose group, with dose escalation stopped if subjects > 2/6 presented with DLT; as in the previous 3 patients with 2 patients who developed DLT, dose escalation was stopped.
MTD (maximum tolerated dose) is defined as the highest dose at which less than one third of the subjects develop DLT within a specified time. The determination of MTD is based on the occurrence of DLT within 2 administration cycles (BD 0801 monoclonal antibody is administered twice, generally for 28 days, and the maximum duration is not more than 56 days).
Population of subjects
Patients with platinum-based drug-resistant recurrent epithelial ovarian cancer, fallopian tube cancer, and primary peritoneal cancer.
And (3) inclusion standard:
1. age: is more than or equal to 18 years old;
2. histologically or cytologically confirmed platinum-based drug-resistant epithelial ovarian cancer, fallopian tube cancer or primary peritoneal cancer of the types: non-specific adenocarcinoma (NOS), clear cell adenocarcinoma, endometrial adenocarcinoma, malignant brenner's tumor, mixed epithelial carcinoma, mucinous adenocarcinoma, serous adenocarcinoma, transitional cell carcinoma, and undifferentiated carcinoma;
3. at least one measurable tumor lesion (according to RECIST 1.1);
ECOG PS score 0-1;
5. subjects must have proper organ function and meet all the following laboratory test results before enrollment:
a) bone marrow reserves were essentially normal (no transfusions and blood products, no correction with G-CSF and other hematopoietic stimulators within 14 days prior to screening): ANC ≥ 1.5 × 109/L,HB≥90 g/L,PLT≥100×109/L;
b) Liver function is essentially normal: TBIL is less than or equal to 1.5 multiplied by ULN, AST is less than or equal to 2.5 multiplied by ULN, and alkaline phosphatase is less than or equal to 2.5 multiplied by ULN;
c) renal function is basically normal: the serum creatinine is less than or equal to ULN or the GFR (glomerular clearance) is more than or equal to 60mL/min, and the serum creatinine is calculated according to a Cockroft-Gault formula: male GFR = (140-age) BW/(Scr × (72), female GFR = male 0.85;
d) the blood coagulation function is basically normal: INR < 1.5 (if the patient receives a stable dose of warfarin to manage venous thrombosis, INR should range from 2-3), APTT < 1.2 × ULN;
6. disease progression occurs within 6 months after completion of at least 4 cycles of platinum-containing therapy;
7. the estimated survival time is more than or equal to 12 weeks;
8. at least 4 weeks from the last chemotherapy, and if the anti-tumor biological product is received, at least 4 half-lives of the elution phase are required;
9. the toxic and side effects caused by the prior treatment need to be recovered to the grade less than or equal to 1 (NCI CTC 4.03);
10. subjects had to give informed consent to the study prior to the trial and voluntarily signed a written informed consent form;
11. the subjects were able to communicate well with the investigator and completed the study in accordance with the study prescription.
Exclusion criteria:
1. has previously received >2 anti-tumor treatment regimens;
2. patients were refractory to platinum-based disease, which was defined as progression of the disease during previous platinum-containing therapy;
3. ovarian tumors with low malignant potential, such as borderline tumors;
4. patients with aggressive malignancies (except for non-melanoma skin cancers) or previous malignant tumor treatments are contraindicated by current treatment regimens;
5. the patient received any pelvic or abdominal radiotherapy;
6. patients are associated with severe, non-healing wounds, ulcers or fractures;
7. patients with a history of intestinal obstruction (including occlusive disease) had a history of peritoneal fistula, gastrointestinal perforation, and peritoneal abscess. In the screening period, pelvic cavity examination and rectum sigmoid colon invasion are carried out, or a patient with intestinal invasion is found by CT examination, or intestinal obstruction with clinical symptoms is carried out;
severe infections require intravenous infusion of antibiotics or hospitalization;
9. the disease history of thromboembolism or hemorrhagic diseases is within 6 months before the treatment;
10. CNS diseases unrelated to tumors, symptomatic brain metastases;
11. cardiovascular diseases with clinical significance:
a) uncontrollable hypertension (systolic pressure is more than or equal to 150 mmHg and/or diastolic pressure is more than or equal to 90 mmHg);
b) a history of myocardial infarction or unstable angina within 6 months prior to group entry;
c) congestive heart failure or new york heart disease society (NYHA) grade ii heart failure;
d) cardiac arrhythmias in severe need of drug therapy; asymptomatic atrial fibrillation without a controllable heart rate;
12. left ventricular ejection fraction below the lower limit of normal;
13. neuropathy more than or equal to CTCAE 2 level exists in the past (only limited to a taxol + BD0801 monoclonal antibody group);
14. known to be severely allergic to the therapeutic drugs or adjuvants used in the regimen;
15. pregnant or lactating women;
16. patients with proteinuria (urinary protein >1+ found in screening phase exam or proteinuria of 1+, not returning to normal within 24 hours);
17. those who have been treated with anti-VEGF protein drugs such as bevacizumab and BD0801 monoclonal antibody;
18. the patient undergoes major surgery or is expected to undergo surgical treatment:
a) large surgical operations or obvious trauma were performed within 28 days before group entry;
b) major surgical procedures are expected to be performed during the study, including but not limited to abdominal surgery (open or laparoscopic surgery) prior to disease progression;
c) open biopsy was performed within 7 days before group entry;
19. treatment with other study drugs is currently or within 30 days prior to group entry.
Test drug
The BD0801 antibody was produced and supplied by Shandong Mitsui Bio-pharmaceuticals, Inc., and stored at 2-8 ℃.
Paclitaxel injection: the manufacturer: beijing collaborates with the pharmaceutical factory.
Topotecan hydrochloride for injection: the manufacturer: jiangsu Osekang pharmaceutical Co., Ltd.
Mode of administration
BD0801 mab dosing regimen: the specified dose (0.5 mg/kg, 1mg/kg, 1.5mg/kg, 2 mg/kg) was administered once every 2 weeks (14 days).
Paclitaxel: 80mg/m2And the administration is carried out once on the 1 st, 8 th, 15 th and 22 th days in the 28-day administration cycle. Applied as per the instructions.
Topotecan hydrochloride: 4 mg/m2Intravenous infusion was given 1 time each on days 1, 8, and 15 of the 28-day dosing cycle. Applied as per the instructions.
The administration sequence is as follows: the administration was carried out according to the respective administration cycles. If the BD0801 monoclonal antibody and the chemotherapeutic agent are administered on the same day, the BD0801 monoclonal antibody is infused first.
Test grouping
BD0801 mab + paclitaxel: every 2 weeks (14 days) of BD0801 monoclonal antibody is a dosing cycle, and the BD0801 monoclonal antibody is infused first when the BD0801 monoclonal antibody is dosed once on days 1, 8, 15 and 22 in a paclitaxel 28-day dosing cycle, if the BD0801 monoclonal antibody is dosed on the same day.
BD0801 mab + topotecan hydrochloride group: BD0801 monoclonal antibody is administered every 2 weeks (14 days) for one administration cycle, and topotecan hydrochloride is administered 1 time on days 1, 8 and 15 in a 28-day administration cycle, if the two are administered on the same day, BD0801 monoclonal antibody is infused first.
Treatment continues until disease progression or intolerance of adverse effects occurs or other end-of-treatment criteria are met.
Research index and evaluation standard thereof
The safety index is as follows: DLT, adverse events, clinical safety laboratory examinations, electrocardiogram, vital signs, echocardiogram, etc.
The therapeutic end point is as follows: objective Remission Rate (ORR), Disease Control Rate (DCR), Progression Free Survival (PFS), and Overall Survival (OS).
Pharmacokinetic indexes are as follows: cmax, Tmax, AUC(TAU)、Accumulation Ratio(AR)、Ctrough
Statistics and analysis
Analyzing the data set:
total analysis set (FAS): all subjects enrolled and received study medication and had at least one tumor assessment after the baseline period.
Security data set (SS): all subjects were enrolled and received study medication. DLT assessment analysis set: the DLT assessment set included subjects who completed at least one treatment with BD0801 mab during the dose escalation phase and completed the DLT assessment observation period (e.g., 28 days from first dose) or withdrawal from DLT during the observation period.
PK concentration set (PKCS): all subjects were enrolled and received study medication with at least 1 effective blood level data during the trial.
PK parameters analysis set (PKPS): all subjects enrolled and received study medication with at least 1 effective PK parameter during the trial. Those not including PKPS also include: a) those who have entered a serious violation scenario, affect the PK parameter outcome, or cannot estimate the parameters; b) concomitant medication occurred during the course of the trial and had an effect on PK parameters.
And (3) safety analysis:
analysis was performed according to the SS/DLT assessment analysis set. DLT and MTD were determined. Descriptive statistics are used to analyze adverse events, laboratory examination indices, vital signs, electrocardiograms, and the like. Adverse events, adverse reactions, major adverse events, severe adverse events, adverse events leading to discontinuation of medication, and the like were summarized according to dose groups.
Pharmacokinetic analysis:
performing descriptive statistical summary on the plasma concentration of the BD0801 monoclonal antibody according to the dose level and the planning time; PK parameters were analyzed by dose level descriptive statistics. The drug-time curve and the mean drug-time curve for each subject were plotted against the blood concentration-time data for each subject measured in the experiment.
Immunogenicity analysis:
the frequency and rate of immunogenicity positive reactions were summarized and the corresponding titer values were given as tabulated by subject and study date. And the influence of immunogenicity on the pharmacokinetics and safety of the BD0801 monoclonal antibody is discussed.
And (3) analyzing the curative effect:
objective Remission Rate (ORR), Disease Control Rate (DCR) and its 95% confidence interval were calculated from the total analysis set (FAS). For progression-free survival (PFS) and Overall Survival (OS), a Kaplan-Meier method was used to estimate survival curves and plot survival curves. Descriptive statistics were performed for tumor size changes.
Objective Remission Rate (ORR): ORR and its 95% confidence interval (Clopper-Pearson exact method) were calculated.
ORR =(CR+PR)/( CR+PR+SD+PD+NE) ×100%
Confirmed (confirmed) and unconfirmed (unconfirmed) CR and PR results were analyzed simultaneously.
Disease Control Rate (DCR): the DCR and its 95% confidence interval were calculated (Clopper-Pearson exact method).
DCR=(CR+PR+SD)/( CR+PR+SD+PD+NE) ×100%
Both confirmed (confirmed) and unconfirmed (unconfirmed) results were analyzed.
Progression Free Survival (PFS): the time between the start of study medication until the first recorded PD or death date, depending on which occurred first; if no disease progression is observed, the truncation date should be the date of the last tumor measurement prior to receiving a new anti-tumor drug. The PFS was summarized using the Kaplan-Meier test plot, listing the deletion rate, quartile and 95% CI (Brookmeyer-Crowley method), and estimating the progression free survival rate at different times at 95% CI (loglog conversion).
PFS (month) = (first disease progression or death date or last tumor measurement date-first dosing date + 1)/30.4375
Overall Survival (OS): time from study medication initiation to death date (for any reason); if no mortality is observed, the last survival date is known as the truncation date. The OS was summarized using Kaplan-Meier test plots, listing the deletion rate, quartile and 95% CI (Brookmeyer-Crowley method), and estimating the survival rate at different times and 95% CI (loglog conversion).
OS (month) = (death date or last follow-up date-first administration date + 1)/30.4375
Tumor size change: descriptive statistics were performed on the measurable tumor Sum of Longest Diameter (SLD) versus baseline changes. And the maximum percentage of each patient's SLD reduction from the screening period was waterfall. In addition, the change in SLD from baseline at each tumor assessment after each patient dose will also be listed.
Results
1. Population of subjects
A total of 29 subjects participated in the trial and received at least 1 study drug treatment. Of 29 subjects, the mean course was about 28.19 months, most of which were epithelial ovarian cancer (25, 86.2%), primary peritoneal and fallopian tube cancer (2 each (6.9%), pathological diagnosis was mainly serous adenocarcinoma (25, 86.2%), clear cell adenocarcinoma 2 (6.9%), mixed epithelial cancer 1 (3.4%), and others 1 (3.4%). See table 5 for details.
TABLE 5 history of tumor
Figure 940969DEST_PATH_IMAGE007
The 29 subjects all had target lesions with a maximum sum of 195.54mm, a minimum sum of 12mm and an average of 71.342 mm.
The subjects in the group had received chemotherapy, had received no radiation therapy, and 1 subject (3.4%) had received PARP inhibitor treatment and 8 subjects (27.6%) had received additional treatment.
The majority of 29 subjects (26, 89.7%) scored 0 for ECOG and 3 subjects (10.3%) scored 1 for inclusion.
All 29 subjects had a significant surgical history.
The most common concomitant diseases are hypertension (5 cases, 17.2%) and diabetes (5 cases, 17.2%), and the most common concomitant medication is systemic antibacterial (2 cases, 6.9%).
In 29 subjects, 22 (75.9%) were treated with the subsequent chemotherapeutic anti-tumor treatment after the end of the study treatment, 2 in the 0.5mg/kg dose group, 11 in the 1mg/kg dose group, and 9 in the 1.5mg/kg dose group.
2. Safety feature
There were no DLT events in this study, MTD was not achieved, and the recommended dose was 1.5mg/kg Q2W.
By the end of the study, 29 subjects (100%) had adverse events, and the frequency of adverse events was not different between each dose group and the different chemotherapy regimen groups, as hematological toxicity was more common. Grade 3 and above adverse events with incidence > 5% were decreased neutrophil count (51.72%), decreased leukocyte count (41.38%), decreased platelet count (17.24%), anemia (17.24%), increased gamma-glutamyl transferase (10.34%), emesis (10.34%), hypertension (10.34%), hypertriglyceridemia (6.90%).
A total of 27 subjects (93.10%) experienced adverse events associated with BD0801 mab, with similar incidence of drug-related adverse events in the three dose groups, with the highest incidence of drug-related adverse events being decreased white blood cell counts (24.14%) according to PT classification.
A total of 1 death-causing SAE occurred, intracranial bleeding occurring in the 1.5mg/kg dose group (in combination with topotecan hydrochloride), and the investigator considered this SAE to be possibly unrelated to the test drug and to the combined chemotherapeutic drug. In a total of 11 subjects, after 15 treatments, SAE, the highest incidence of SAE was a decrease in neutrophil count (6.9%) according to PT classification.
In total 26 subjects (89.66%), adverse events were observed that resulted in a loss or suspension of BD0801 mab, with the highest incidence of decreased neutrophil count (44.83%); in total, 3 subjects (10.34%) had adverse events leading to permanent withdrawal of BD0801 mAb after administration, 2 in the 1.5mg/kg dose group, decreased platelet count and proteinuria, and 1 in the 1.0mg/kg dose group, respectively, as abdominal pain.
Adverse events of particular interest for this study included: gastrointestinal perforation or fistulae, hypertension, proteinuria, embolism and thrombotic events, various types of bleeding, etc., occur as detailed in fig. 6. According to SOC classification, the highest incidence of AESI was renal and urological disease (11/37.93%), followed by various examinations (9/31.03%), vascular and lymphatic diseases (7/24.14%), and respiratory, thoracic and mediastinal diseases (3/10.34%). According to the PT classification, the highest incidence of AESI was proteinuria (11 cases/37.93%), followed by urine protein detection (7 cases/24.14%), hypertension (6 cases/20.69%).
TABLE 6 analysis of adverse events with specific attention after dosing System (SS)
Figure 676844DEST_PATH_IMAGE009
Grade 3 and above adverse event analysis
In total, 25 subjects (86.21%) had grade 3 adverse events with a post-dose severity of 100%, 91.67% and 78.57% for the 0.5mg/kg, 1.0mg/kg and 1.5mg/kg groups, respectively. Grade 3 and above adverse events with incidence rates greater than or equal to 5% included decreased neutrophil count (51.72%), decreased leukocyte count (41.38%), decreased platelet count (17.24%), anemia (17.24%), increased gamma-glutamyltransferase (10.34%), hypertension (10.34%), decreased neutrophil percentage (6.90%), emesis (10.34%), hypertriglyceridemia (6.90%). See table 7 for details.
TABLE 7 adverse event subsystem analysis (SS) grade no less than 3 after dosing
Figure 141324DEST_PATH_IMAGE011
Grade 3 or more adverse event incidence associated with BD0801 mab was 37.93%, with 57.14% for the 1.5mg/kg group, slightly higher than the 1.0mg/kg group (25.0%), and slightly higher for the taxol subgroup than the topotecan hydrochloride subgroup (45.45% vs 33.33%). Grade 3 adverse events associated with BD0801 monoclonal antibody with an incidence of 5% or more included decreased leukocyte count (10.34%), decreased neutrophil count (10.34%), hypertension (10.34%), increased gamma-glutamyltransferase (6.90%), and decreased platelet count (6.90%). See table 8 for details.
Adverse events associated with BD0801 monoclonal antibody and not associated with chemotherapy at grade 3 or more after no drug administration.
TABLE 8 grade 3 sub-System analysis (SS) of adverse events associated with BD0801 monoclonal antibody after drug administration
Figure 262732DEST_PATH_IMAGE012
Pharmacokinetics/pharmacodynamics
The pharmacokinetic parameters of BD0801 monoclonal antibodies at different doses after administration are detailed in tables 9-1, 9-2 and 9-3. Wherein, after a single administration, the dose range of T is 0.5-1.5 mg/kg1/256.41-93.4 h, CL 0.35-0.38 ml/h/kg, V28.52-48.63 ml/kg; major exposure parameter (C)max、AUC0-t 、AUC0-inf) Increases with increasing dose and exhibits a better linear relationship. After multiple administrations, T is in the dose range of 0.5-1.5 mg/kg1/263.5-101.97 h, 0.35-0.49 ml/h/kg CL and 32.27-61.84 ml/kg V; major exposure parameter (C)max、AUC0-t、AUC0-inf) Increases with increasing dose and exhibits a better linear relationship. AUC and in steady stateThe AUC value of the first administration is approximate, and the BD0801 monoclonal antibody has no obvious accumulation after multiple administrations.
TABLE 9-1 pharmacokinetic parameters (Mean + -SD) after administration of BD0801 monoclonal antibody
Figure 724938DEST_PATH_IMAGE013
Remarking: tmax Described by mean (range); AR, accumulation coefficient, AR = AUC0-t(Steady State)/AUC0-t(first time).
TABLE 9-2 pharmacokinetic parameters (Mean + -SD) after administration of BD0801 monoclonal antibody
Figure 744846DEST_PATH_IMAGE014
Remarking: tmax Described by mean (range); AR, accumulation coefficient, AR = AUC0-t(Steady State)/AUC0-t(first time).
TABLE 9-3 pharmacokinetic parameters (Mean + -SD) of monoclonal antibody BD0801 after administration
Figure 685120DEST_PATH_IMAGE015
Remarking: tmax Described by mean (range); AR, accumulation coefficient, AR = AUC0-t(Steady State)/AUC0-t(first time).
Immunogenicity
BD0801 mab total 29 subjects included immunogenicity analysis in a platinum-resistant recurrent epithelial ovarian cancer, fallopian tube cancer and primary peritoneal carcinoma patient study. Compared with the baseline level, after the BD0801 monoclonal antibody is administered, the sera of 9 patients detect positive ADA (anti-drug antibody), and the positive detection rate of ADA individuals is 31.0 percent (9/29).
Effectiveness of
(1) Analysis (confirmation) of overall best curative effect of tumor focus
The overall Objective Remission Rate (ORR) was 31%, the clinical benefit was found in a total of 20 subjects, the clinical benefit rate (DCR) was 69.0%, and the overall optimal therapeutic effect was CR in a total of 1 (3.4%) subjects, which was a subset of paclitaxel in the 1.0mg/kg dose group. The best efficacy was 8 (27.6%) subjects with PR, with 1 in the 0.5mg/kg dose group (33.3%), 3 in the 1mg/kg dose group (25.0%), and 4 in the 1.5mg/kg dose group (28.6%). The optimal therapeutic effect was SD in a total of 11 subjects (37.9%), 6 in the 1mg/kg dose group (50.0%), and 5 in the 1.5mg/kg dose group (35.7%). A detailed overall best efficacy analysis for each dose group is shown in table 6.
Remission occurred in 7 of 11 subjects with BD0801 mab in combination with paclitaxel subgroup, with an overall objective remission rate of 63.6%, 8 clinical benefits, and a DCR of 72.7%. ORRs of the 0.5mg/kg, 1.0mg/kg, and 1.5mg/kg dose groups were 50.0%, 66.7%, and 66.7%, respectively.
In 18 subjects in the group of BD0801 monoclonal antibody and topotecan hydrochloride, relief appears in 2 subjects, the overall objective relief rate is 11.1%, 12 subjects benefit clinically, and the DCR is 66.7%. ORRs of the 0.5mg/kg dose group, 1.0mg/kg dose group, and 1.5mg/kg dose group were 0, 22.2%, and 0, respectively.
TABLE 10 Total best curative (validation) analysis of tumor Foci (FAS)
Figure 677347DEST_PATH_IMAGE016
Note: optimal efficacy refers to the best efficacy recorded from the start of treatment to before disease progression/recurrence.
Objective Remission Rate (ORR) = N1/N, N1= CR + PR;
clinical benefit rate (DCR) = N2/N, N2= CR + PR + SD;
the disease conditions that could not be classified as CR, PR, SD and PD were classified as NE (not evaluable).
And (3) confirming that: CR or PR patients must be reviewed 4 weeks after initial assessment as CR or PR to confirm efficacy.
(2) Comprehensive analysis of overall best treatment of tumor lesions (not confirmed)
The overall objective remission rate was 37.9% and the clinical benefit rate was 69.0% for the unidentified subjects, and the overall best efficacy was CR for 1 (3.4%) of the subjects, which was the 1.0mg/kg dose group. The overall best effect was found in 10 subjects (34.5%) with PR, with 1 in the 0.5mg/kg dose group (33.3%), 3 in the 1.0mg/kg dose group (25.0%), and 6 in the 1.5mg/kg dose group (42.9%). A detailed overall best-fit analysis of each dose group is shown in table 11.
Objective remission with an ORR of 72.7% was observed in 8 out of 11 subjects treated with BD0801 mab in combination with paclitaxel, with ORRs in the 0.5mg/kg, 1.0mg/kg, and 1.5mg/kg dose groups being 50.0%, 66.7%, and 83.3%, respectively. In total, 8 subjects had clinical benefit, with DCR of 72.7%, DCR of 50.0%, 66.7% and 83.3% in the 0.5mg/k, 1.0mg/kg and 1.5mg/kg dose groups, respectively.
BD0801 mab in combination with topotecan hydrochloride treatment subgroup of 18 subjects showed objective remission with an ORR of 16.7%, with 0.5mg/kg, 1.0mg/kg and 1.5mg/kg dose groups having ORRs of 0, 22.2% and 12.5%, respectively. A total of 12 subjects had clinical benefit with DCR of 66.7%, 0.5mg/kg, 1.0mg/kg and 1.5mg/kg dose groups with DCR of 0, 88.9%, 50.0%, respectively.
TABLE 11 Overall best efficacy (not confirmed) of tumor lesions general analysis (FAS)
Figure 372771DEST_PATH_IMAGE017
Note: optimal efficacy refers to the best efficacy recorded from the start of treatment to before disease progression/recurrence.
Objective Remission Rate (ORR) = N1/N, N1= CR + PR;
clinical benefit rate (DCR) = N2/N, N2= CR + PR + SD;
the disease conditions that could not be classified as CR, PR, SD and PD were classified as NE (not evaluable).
Not confirmed: unconfirmed CR or PR (calculated without consideration of the next tumor assessment confirmation).
(3) Disease progression free survival (monthly) analysis
Of the 29 subjects, 27 (93.1%) subjects developed PFS events and 2 subjects in the 1.5mg/kg dose group were deleted and were a subset of paclitaxel. Median PFS was 5.4 months, with median PFS for the 0.5mg/kg, 1.0mg/kg and 1.5mg/kg groups being 1.9 months, 5.9 months and 4.6 months, respectively. The survival rate of no progress is 65.5% in 3 months, 40.3% in 6 months and 4.0% in 1 year. Detailed progression-free survival analysis is shown in tables 12-13.
PFS events occurred in 9 (81.8%) of 11 subjects of the BD0801 mAb and paclitaxel subgroup with median PFS of 5.3 months, 5.2 months, 5.3 months and 5.7 months in the 0.5mg/kg, 1.0mg/kg and 1.5mg/kg groups, respectively. The survival rate of no progress is 72.7% in 3 months, 43.6% in 6 months and 14.5% in 1 year.
A subset of 18 subjects (100%) with BD0801 mab combined with topotecan hydrochloride had PFS events with a median PFS of 5.5 months, 1.0mg/kg and 1.5mg/kg groups for 1.0 month, 6.1 months and 4.0 months, respectively. The survival rate of 3 months without progress is 61.1%, the survival rate of 6 months without progress is 38.9%, and the survival rate of 1 year without progress is 0.
TABLE 12 disease progression free survival analysis (FAS)
Figure 338365DEST_PATH_IMAGE018
Note: 95% CI was calculated using the Brookmeyer-Crowley method.
TABLE 13 progression free survival (%) at different times (month) (FAS)
Figure 816751DEST_PATH_IMAGE019
Note: the 95% CI was calculated using a loglog transformation.
(4) Total Life time (month) analysis
By 20 days 10 months after 2020, median follow-up time was 12.8 months, 15 (51.7%) OS events were followed, 14 were missed (survived 10, missed 4), median OS was 14.9 months, 0.5mg/kg dose group mOS did not reach, 1.0mg/kg dose group, 1.5mg/kg dose group mean OS was 13.5 months, 18.9 months, respectively. The total survival rate in 6 months is 86.2%, the total survival rate in 1 year is 71.0%, and the total survival rate in 2 years is 31.2%. The detailed overall survival and survival analysis are shown in tables 14-15.
While 4 (36.4%) OS events were co-visited in 11 subjects treated with BD0801 mAb in combination with paclitaxel, 2 subjects in the 0.5mg/kg dose group did not reach OS, and median OS was 14.2 months and 18.9 months in the 1.0mg/kg dose group and 1.5mg/kg dose group, respectively. The 6-month survival rates of the 0.5mg/kg dose group, the 1.0mg/kg dose group, and the 1.5mg/kg dose group were 100%, and 83.3%, respectively, and the 1-year overall survival rates were 100%, 66.7%, and 83.3%, respectively.
BD0801 mab in combination with topotecan hydrochloride treatment group of 18 subjects out of 11 (61.1%) had OS events with a median OS of 12.8 months. The median OS was 3.5 months, 12.8 months and 15.1 months in the 0.5mg/kg, 1.0mg/kg and 1.5mg/kg dose groups, respectively. The total survival rate in 6 months is 83.3%, the total survival rate in 1 year is 65.7%, and the total survival rate in 2 years is 24.6%.
TABLE 14 Total Life time (month) analysis (FAS)
Figure 725801DEST_PATH_IMAGE020
TABLE 15 Total survival (%) at different times (months) (FAS)
Figure 529809DEST_PATH_IMAGE021
(5) Analysis of relationship between exposure and curative effect
In 11 patients with BD0801 mab, a Paclitaxel (PTX) chemotherapy regimen with an ORR of 7/11 (63.6%), the 11 PTX groups of subjects were further analyzed by dose-grouping and dose-response relationships between Objective Remission Rate (ORR) and dose administered for optimal overall efficacy in the subjects were analyzed as shown in fig. 4.
In the BD0801 mab with Paclitaxel (PTX) or topotecan hydrochloride chemotherapy regimen, subjects were ranked from low to high AUC, layered 3 total. The dose-effect relationship between the ORR of the best overall curative effect of the subjects and the drug exposure (AUC) was analyzed in a layered manner, and the ORR and the BD0801 monoclonal antibody exposure showed a positive correlation, compared with the BD0801 monoclonal antibody combined with topotecan hydrochloride (TOPO), which showed no significant correlation between the ORR and the exposure, as shown in FIG. 5.
There was no significant correlation between Progression Free Survival (PFS) and dose at different doses in the overall population, detailed in figure 6.
The percentage of tumor longest diameter (SLD) shrinkage associated with exposure of BD0801 mab in combination with Paclitaxel (PTX) is shown in FIG. 7, where the exposure of BD0801 mab is positively correlated with tumor shrinkage.
Sequence listing
<110> Jiangsu Xiansui pharmaceutical Co., Ltd
Application of anti-human VEGF antibody and chemical drug combination in preparation of drugs for treating ovarian cancer
<160> 17
<170> SIPOSequenceListing 1.0
<210> 1
<211> 122
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 1
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Lys Pro Gly Gly
1 5 10 15
Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Ser Phe Ser Asn Asn
20 25 30
Asp Val Met Cys Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp
35 40 45
Ile Gly Cys Ile Met Thr Thr Asp Val Val Thr Glu Tyr Ala Asn Trp
50 55 60
Ala Lys Ser Arg Phe Thr Val Ser Arg Asp Ser Ala Lys Asn Ser Val
65 70 75 80
Tyr Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Phe
85 90 95
Cys Ala Arg Asp Ser Val Gly Ser Pro Leu Met Ser Phe Asp Leu Trp
100 105 110
Gly Pro Gly Thr Leu Val Thr Val Ser Ser
115 120
<210> 2
<211> 330
<212> PRT
<213> Homo sapiens
<400> 2
Ala Ser Thr Lys Gly Pro Ser Val Phe Pro Leu Ala Pro Ser Ser Lys
1 5 10 15
Ser Thr Ser Gly Gly Thr Ala Ala Leu Gly Cys Leu Val Lys Asp Tyr
20 25 30
Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser Gly Ala Leu Thr Ser
35 40 45
Gly Val His Thr Phe Pro Ala Val Leu Gln Ser Ser Gly Leu Tyr Ser
50 55 60
Leu Ser Ser Val Val Thr Val Pro Ser Ser Ser Leu Gly Thr Gln Thr
65 70 75 80
Tyr Ile Cys Asn Val Asn His Lys Pro Ser Asn Thr Lys Val Asp Lys
85 90 95
Lys Val Glu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys
100 105 110
Pro Ala Pro Glu Leu Leu Gly Gly Pro Ser Val Phe Leu Phe Pro Pro
115 120 125
Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys
130 135 140
Val Val Val Asp Val Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp
145 150 155 160
Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu
165 170 175
Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu
180 185 190
His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn
195 200 205
Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly
210 215 220
Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg Asp Glu
225 230 235 240
Leu Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr
245 250 255
Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn
260 265 270
Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe
275 280 285
Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn
290 295 300
Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn His Tyr Thr
305 310 315 320
Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys
325 330
<210> 3
<211> 112
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 3
Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Leu Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Asn Cys Gln Ala Ser Gln Ser Ile Tyr Asn Asn
20 25 30
Asn Glu Leu Ser Trp Tyr Gln Gln Lys Pro Gly Lys Pro Pro Lys Leu
35 40 45
Leu Ile Tyr Arg Ala Ser Thr Leu Ala Ser Gly Val Pro Ser Arg Phe
50 55 60
Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu
65 70 75 80
Gln Pro Glu Asp Val Ala Thr Tyr Tyr Cys Gly Gly Tyr Lys Ser Tyr
85 90 95
Ser Asn Asp Gly Asn Gly Phe Gly Gly Gly Thr Lys Val Glu Ile Lys
100 105 110
<210> 4
<211> 107
<212> PRT
<213> Homo sapiens
<400> 4
Arg Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu
1 5 10 15
Gln Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe
20 25 30
Tyr Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln
35 40 45
Ser Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser
50 55 60
Thr Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu
65 70 75 80
Lys His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser
85 90 95
Pro Val Thr Lys Ser Phe Asn Arg Gly Glu Cys
100 105
<210> 5
<211> 6
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 5
Asn Asn Asp Val Met Cys
1 5
<210> 6
<211> 8
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 6
Gly Phe Ser Phe Ser Asn Asn Asp
1 5
<210> 7
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 7
Gly Phe Ser Phe Ser Asn Asn Asp Val
1 5
<210> 8
<211> 17
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 8
Cys Ile Met Thr Thr Asp Val Val Thr Glu Tyr Ala Asn Trp Ala Lys
1 5 10 15
Ser
<210> 9
<211> 6
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 9
Met Thr Thr Asp Val Val
1 5
<210> 10
<211> 8
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 10
Ile Met Thr Thr Asp Val Val Thr
1 5
<210> 11
<211> 12
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 11
Asp Ser Val Gly Ser Pro Leu Met Ser Phe Asp Leu
1 5 10
<210> 12
<211> 14
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 12
Ala Arg Asp Ser Val Gly Ser Pro Leu Met Ser Phe Asp Leu
1 5 10
<210> 13
<211> 13
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 13
Gln Ala Ser Gln Ser Ile Tyr Asn Asn Asn Glu Leu Ser
1 5 10
<210> 14
<211> 8
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 14
Gln Ser Ile Tyr Asn Asn Asn Glu
1 5
<210> 15
<211> 7
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 15
Arg Ala Ser Thr Leu Ala Ser
1 5
<210> 16
<211> 3
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 16
Arg Ala Ser
1
<210> 17
<211> 12
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 17
Gly Gly Tyr Lys Ser Tyr Ser Asn Asp Gly Asn Gly
1 5 10

Claims (13)

1. Use of an anti-human VEGF antibody in combination with a chemotherapeutic agent for the manufacture of a medicament for the treatment of ovarian cancer, wherein the chemotherapeutic agent is selected from the group consisting of taxanes or camptothecins, and wherein the antibody comprises a heavy chain variable region comprising HCDR1, HCDR2 and HCDR3 and a light chain variable region comprising LCDR1, LCDR2 and LCDR3,
the sequence of the HCDR1 is shown as SEQ ID NO. 5;
the sequence of the HCDR2 is shown as SEQ ID NO. 8;
the sequence of the HCDR3 is shown as SEQ ID NO. 11;
the sequence of the LCDR1 is shown as SEQ ID NO. 13;
the sequence of the LCDR2 is shown as SEQ ID NO. 15; and the combination of (a) and (b),
the sequence of LCDR3 is shown in SEQ ID NO. 17.
2. The use of claim 1, wherein the heavy chain variable region has the sequence shown in SEQ ID NO. 1 and the light chain variable region has the sequence shown in SEQ ID NO. 3.
3. The use of claim 2, wherein the antibody further comprises a heavy chain constant region having the sequence set forth in SEQ ID NO. 2, and a light chain constant region having the sequence set forth in SEQ ID NO. 4.
4. The use according to any one of claims 1 to 3, wherein the ovarian cancer is epithelial ovarian cancer, fallopian tube cancer or primary peritoneal cancer.
5. The use of claim 4, wherein the ovarian cancer is platinum-resistant recurrent ovarian cancer.
6. The use according to claim 4, wherein the pathological type of ovarian cancer is serous adenocarcinoma, clear cell adenocarcinoma or mixed epithelial carcinoma.
7. The use according to any one of claims 1 to 3, wherein the taxane compound is paclitaxel and the camptothecin compound is topotecan hydrochloride.
8. The use according to any one of claims 1 to 3, wherein the effective amount of the anti-human VEGF antibody administered is 0.5 to 2 mg/kg.
9. The use of claim 8, wherein said effective amount of anti-human VEGF antibody administered is 0.5mg/kg, 1mg/kg, 1.5mg/kg or 2 mg/kg.
10. The use of claim 8, wherein said anti-human VEGF antibody is administered in a single dose, each dose containing an amount of antibody effective for administration to a patient.
11. The use of claim 10, wherein said single dose dosage form contains 25-150 mg of said anti-human VEGF antibody.
12. The use of claim 11, wherein said single dose dosage form contains 25mg, 50mg, 80mg, 100mg or 150mg of said anti-human VEGF antibody.
13. The use of claim 7, wherein the effective amount of paclitaxel administered is 80mg/m2(ii) a The hydrochloric acid topoietAn effective amount of kang is 4 mg/m2
CN202110742169.4A 2021-07-01 2021-07-01 Application of anti-human VEGF antibody and chemical drug combination in preparation of drugs for treating ovarian cancer Pending CN113244388A (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202211290780.9A CN115814076A (en) 2021-07-01 2021-07-01 Application of anti-human VEGF antibody and chemical drug combination in preparation of drugs for treating ovarian cancer
CN202110742169.4A CN113244388A (en) 2021-07-01 2021-07-01 Application of anti-human VEGF antibody and chemical drug combination in preparation of drugs for treating ovarian cancer

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110742169.4A CN113244388A (en) 2021-07-01 2021-07-01 Application of anti-human VEGF antibody and chemical drug combination in preparation of drugs for treating ovarian cancer

Related Child Applications (1)

Application Number Title Priority Date Filing Date
CN202211290780.9A Division CN115814076A (en) 2021-07-01 2021-07-01 Application of anti-human VEGF antibody and chemical drug combination in preparation of drugs for treating ovarian cancer

Publications (1)

Publication Number Publication Date
CN113244388A true CN113244388A (en) 2021-08-13

Family

ID=77190302

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202211290780.9A Pending CN115814076A (en) 2021-07-01 2021-07-01 Application of anti-human VEGF antibody and chemical drug combination in preparation of drugs for treating ovarian cancer
CN202110742169.4A Pending CN113244388A (en) 2021-07-01 2021-07-01 Application of anti-human VEGF antibody and chemical drug combination in preparation of drugs for treating ovarian cancer

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN202211290780.9A Pending CN115814076A (en) 2021-07-01 2021-07-01 Application of anti-human VEGF antibody and chemical drug combination in preparation of drugs for treating ovarian cancer

Country Status (1)

Country Link
CN (2) CN115814076A (en)

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1419452A (en) * 2000-03-27 2003-05-21 布里斯托尔-迈尔斯斯奎布公司 Synergistic methods and compositions for treating cancer
CN1829741A (en) * 2003-05-30 2006-09-06 健泰科生物技术公司 Treatment with anti-VEGF antibodies
CN101080227A (en) * 2004-12-15 2007-11-28 诺瓦提斯公司 Combinations of therapeutic agents for treating cancer
CN101918579A (en) * 2007-10-22 2010-12-15 先灵公司 Fully human anti-VEGF antibodies and using method
WO2011106300A2 (en) * 2010-02-23 2011-09-01 Genentech, Inc. Anti-angiogenesis therapy for the treatment of ovarian cancer
CN103417965A (en) * 2012-05-17 2013-12-04 江苏先声药物研究有限公司 Anti-VEGF antibody containing pharmaceutical composition
CN104334577A (en) * 2012-03-13 2015-02-04 霍夫曼-拉罗奇有限公司 Combination therapy for the treatment of ovarian cancer
CN110227159A (en) * 2018-03-05 2019-09-13 义慧科技(深圳)有限公司 The drug and CPT1a inhibitor for improving entity tumor confrontation blood vessel drug eluting drug resistance are in application wherein
AU2020202054A1 (en) * 2003-05-30 2020-04-09 Genentech, Inc. Treatment with anti-VEGF antibodies

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1419452A (en) * 2000-03-27 2003-05-21 布里斯托尔-迈尔斯斯奎布公司 Synergistic methods and compositions for treating cancer
CN1829741A (en) * 2003-05-30 2006-09-06 健泰科生物技术公司 Treatment with anti-VEGF antibodies
AU2020202054A1 (en) * 2003-05-30 2020-04-09 Genentech, Inc. Treatment with anti-VEGF antibodies
CN101080227A (en) * 2004-12-15 2007-11-28 诺瓦提斯公司 Combinations of therapeutic agents for treating cancer
CN101918579A (en) * 2007-10-22 2010-12-15 先灵公司 Fully human anti-VEGF antibodies and using method
WO2011106300A2 (en) * 2010-02-23 2011-09-01 Genentech, Inc. Anti-angiogenesis therapy for the treatment of ovarian cancer
CN103237810A (en) * 2010-02-23 2013-08-07 霍夫曼-拉罗奇有限公司 Anti-angiogenesis therapy for the treatment of ovarian cancer
CN104334577A (en) * 2012-03-13 2015-02-04 霍夫曼-拉罗奇有限公司 Combination therapy for the treatment of ovarian cancer
CN103417965A (en) * 2012-05-17 2013-12-04 江苏先声药物研究有限公司 Anti-VEGF antibody containing pharmaceutical composition
CN110227159A (en) * 2018-03-05 2019-09-13 义慧科技(深圳)有限公司 The drug and CPT1a inhibitor for improving entity tumor confrontation blood vessel drug eluting drug resistance are in application wherein

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
JIANGSU SIMCERE PHARMACEUTICAL CO., LTD.: "A Phase 1b Study of Sevacizumab in Combination With Chemotherapy in Chinese Patients With Platinum-Resistant Recurrent Ovarian Cancer.", 《CLINICALTRIALS.GOV》 *
PUJADE-LAURAINE 等: "Bevacizumab Combined With Chemotherapy for Platinum-Resistant Recurrent Ovarian Cancer: The AURELIA Open-Label Randomized Phase III Trial", 《JOURNAL OF CLINICAL ONCOLOGY》 *
郑双双 等: "铂耐药复发性卵巢癌的治疗进展", 《天津医科大学学报》 *
鲁巍 江苏先声药业有限公司: "注射用BD0801联合化疗对比安慰剂联合化疗在含铂化疗方案治疗失败的复发性上皮卵巢癌、输卵管癌和原发性腹膜癌患者中的研究", 《药智数据》 *

Also Published As

Publication number Publication date
CN115814076A (en) 2023-03-21

Similar Documents

Publication Publication Date Title
US20220162332A1 (en) Activatable anti-pdl1 antibodies, and methods of use thereof
CN113347996B (en) Combined pharmaceutical composition for treating tumors
US20230279096A1 (en) Combination therapy with anti-il-8 antibodies and anti-pd-1 antibodies for treating cancer
IL178042A (en) Pharmaceutical compositions containing anti-alpha5beta1 antibodies for inhibiting cancer cell proliferation
CN113939315B (en) Combined pharmaceutical composition for treating melanoma
KR20210076025A (en) Cancer Combination Therapy
TWI728400B (en) Cd226 agonist antibodies
WO2021254481A1 (en) Anti-claudin18.2 antibody and use thereof
US20230058779A1 (en) Combination cancer treatment using a pd-1 antagonist, an ilt4 antagonist, and lenvatinib or salts thereof
WO2019089973A1 (en) Anti-tissue factor antibody-drug conjugates and their use in the treatment of cancer
CN116406288A (en) Methods for treating cancer or von-hippel-lindau disease using a combination of a PD-1 antagonist, a HIF-2 alpha inhibitor, and lenvatinib or a pharmaceutically acceptable salt thereof
KR20220149740A (en) A method of treating cancer using a combination of a PD-1 antagonist, a CTLA4 antagonist and lenvatinib or a pharmaceutically acceptable salt thereof
WO2017070475A1 (en) Methods of treating cancer by administering a mek inhibitor and a combination of anti-egfr antibodies
US20230250182A1 (en) Methods for treating cancer or von-hippel lindau disease using a combination of a pd-1 antagonist, a hif-2 alpha inhibitor, and lenvatinib or a pharmaceutically acceptable salt thereof
WO2022068894A1 (en) Bifunctional molecule simultaneously targeting pd-l1 and vegf and medical use thereof
JPWO2021180205A5 (en)
CN113244388A (en) Application of anti-human VEGF antibody and chemical drug combination in preparation of drugs for treating ovarian cancer
CN112175087A (en) Bispecific antibody for resisting CD4 and TGF beta, pharmaceutical composition and application thereof
CN115531531A (en) Application of anti-human VEGF antibody and chemotherapy in preparation of medicine for treating colorectal cancer
TWI835885B (en) Combination therapy for cancer
CN113164599B (en) Use of anti-PD-L1 monoclonal antibodies for the treatment of cancer
WO2023134787A2 (en) Use of combination of anti-pd-1 antibody and anti-vegf antibody in treatment of hepatocellular carcinoma
US20230140694A1 (en) Combination treatment for cancer involving anti-icos and anti-pd1 antibodies, optionally further involving anti-tim3 antibodies
CN115531530A (en) Application of anti-human VEGF antibody in preparation of drugs for treating solid tumors
TW202333786A (en) Methods to reverse treml1-induced immune suppression

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
TA01 Transfer of patent application right

Effective date of registration: 20230626

Address after: 264006 No.1 Heilongjiang Road, Yantai Economic and Technological Development Zone, Yantai City, Shandong Province

Applicant after: SHANDONG SIMCERE BIO-PHARMACEUTICAL Co.,Ltd.

Address before: 210042 699 Xuanwu Road, Xuanwu District, Nanjing, Jiangsu -18

Applicant before: JIANGSU SIMCERE PHARMACEUTICAL Co.,Ltd.

TA01 Transfer of patent application right