CN115607662A - Application of anti-PD-1 antibody in treatment of alveolar soft tissue sarcoma - Google Patents

Application of anti-PD-1 antibody in treatment of alveolar soft tissue sarcoma Download PDF

Info

Publication number
CN115607662A
CN115607662A CN202110783289.9A CN202110783289A CN115607662A CN 115607662 A CN115607662 A CN 115607662A CN 202110783289 A CN202110783289 A CN 202110783289A CN 115607662 A CN115607662 A CN 115607662A
Authority
CN
China
Prior art keywords
homology
antibody
antigen
binding fragment
seq
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
CN202110783289.9A
Other languages
Chinese (zh)
Inventor
谢帆
郭倩
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.)
GENOR BIOPHARMA CO Ltd
Original Assignee
GENOR BIOPHARMA 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 GENOR BIOPHARMA CO Ltd filed Critical GENOR BIOPHARMA CO Ltd
Priority to CN202110783289.9A priority Critical patent/CN115607662A/en
Publication of CN115607662A publication Critical patent/CN115607662A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • 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
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/51Complete heavy chain or Fd fragment, i.e. VH + CH1
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/515Complete light chain, i.e. VL + CL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Abstract

The present disclosure relates to the use of anti-PD-1 antibodies in the treatment of alveolar soft tissue sarcoma. In particular, the disclosure relates to the use of an anti-PD-1 antibody or antigen-binding fragment thereof in the manufacture of a medicament for the treatment or prevention of alveolar soft tissue sarcoma, and further relates to methods of treating or preventing alveolar soft tissue sarcoma using an anti-PD-1 antibody or antigen-binding fragment thereof or a pharmaceutical composition comprising the anti-PD-1 antibody or antigen-binding fragment thereof.

Description

Application of anti-PD-1 antibody in treatment of alveolar soft tissue sarcoma
Technical Field
The present disclosure relates to the field of tumor therapy, more particularly to the use of an anti-PD-1 antibody or antigen-binding fragment thereof in the manufacture of a medicament for the treatment or prevention of alveolar soft tissue sarcoma, and further to methods of treating or preventing alveolar soft tissue sarcoma using an anti-PD-1 antibody or antigen-binding fragment thereof or a pharmaceutical composition comprising the anti-PD-1 antibody or antigen-binding fragment thereof.
Background
Acinar Soft Tissue Sarcoma (ASPS) is a rare and unique subtype, accounting for only 0.5% -1.0% of Soft-Tissue Sarcoma (STS), while STS accounts for approximately 0.8% of all malignant tumor types, with approximately 39,900 new cases in 2014 in china. ASPS can occur in women of all ages, most commonly between 13-35 years old, can often occur in the pelvic cavity and soft tissues of the extremities, and in children also can occur head and neck lesions, often metastasizing to the lungs, bones, and brain. ASPS grow slowly and patients may develop overt clinical symptoms years after initial diagnosis, but most are found to be progressive, with metastasis in the lungs or other organs being the first manifestation of the disease in a significant proportion of cases.
Treatment of alveolar soft tissue sarcoma typically involves surgical resection. Early detection, extensive resection, remains the key to the treatment of alveolar soft tissue sarcoma. Tumor recurrence is less after complete resection, but distant metastasis often occurs, and patients often die from lung, brain and bone metastases. The prognosis is closely related to the position and size of the tumor, whether the surgical resection is thorough, and the like. Multiple relapses, metastasis of vital organs has occurred, or the tumor cannot be completely resected, the prognosis is poor.
Lieberman et al reviewed 102 ASPS with a recurrence rate of 16.7% and a transfer rate of 66.7% (Lieberman PH, brennan MF, kimmel M, et al, alveolar soft-part sarcoma. A. Clinicopathic study of half a center. Cancer, 1989, 63 (1): 1-13); rekhi et al reviewed 47 ASPS with 24% recurrence rate and 72% transfer rate (Rekhi B, ingleA, agarwal M, et al, alvelar soft part partially viewed': clinic ocular review of 47 cases from a specific cancer center, including immunological expression of TFE3 in 22 cases and 21 other organs Pathology, 2012, 44 (1): 11-17. DOI: 10.1097/PAT.0b013e324d7ba4); domestic Liu jun et al summarized 42 cases of alveolar soft tissue sarcoma received treatment in this hospital from 5 to 8 in 1962, and 27 cases (64.3%) had relapses or metastases during the follow-up period, wherein 1 case of local recurrence was simple, 3 cases of local recurrence combined with distant metastasis, and 23 cases of distant metastasis (Liu jun et al, clinical characteristics and treatment efficacy analysis of alveolar soft tissue sarcoma, chinese tumor clinic, 2012,8 461-464.
ASPS are not sensitive to radiation or chemotherapy and lack effective systemic treatment. Since ASPS are mostly vascular and associated with angiogenic pathways, there are currently a few sporadic reports of the application of anti-angiogenic therapy to ASPS, where the oral Vascular Endothelial Growth Factor (VEGF) receptor tyrosine kinase inhibitors Cediranib (AZD 2171), pazopanib (Pazopanib) and Sunitinib (Sunitinib or SU 011248) show primary anti-tumor activity in ASPS. In a recent study of phase II Cediranib, the Objective Remission Rate (ORR) was 19% in 31 patients administered Cediranib (Judson, i., et al, "Cediranib in patients with animal soft-part sarcoma (CASPS): a double-bl, placbo-controlled, randomised, phase 2 trial" The Lancet Oncology 20.7 (2019)). In the chinese clinical oncology society (CSCO) guideline of 2021, angutinib was recommended as a first line treatment for advanced alveolar soft tissue flesh, and in its related ALTER0203 study, angutinib had an ORR of 23.7% (investigator assessment) or 32.4% (independent image assessment) for patients with alveolar soft tissue sarcoma.
In conclusion, early detection, extensive resection, remains the key to treatment of ASPS, and ASPS is not sensitive to radiotherapy or chemotherapy and therefore has a poor prognosis. Thus, there is an urgent need for an effective improved treatment regimen for ASPS, particularly for recurrent, metastatic, or non-surgically resectable ASPS.
Disclosure of Invention
The present inventors have surprisingly found that the anti-PD-1 antibodies of the present disclosure show superior clinical efficacy and improved prognosis as anti-cancer therapeutics, clinically significant, compared to certain known chemotherapeutic agents alone. The present disclosure demonstrates that by administering the anti-PD-1 antibody of the present disclosure to ASPS subjects, the subjects' therapeutic effects are significantly superior to existing commonly used drugs, and the Objective Remission Rate (ORR) is significantly higher than existing commonly used drugs, such as, for example, nilotinib (allotinib) and vascular endothelial growth factor receptor tyrosine kinase inhibitors (VEGFR-TKIs) such as Pazopanib (Pazopanib) or Cediranib (Cediranib), indicating superior clinical efficacy and improved prognosis.
It has also been unexpectedly found that objective remission and improved prognosis can still be obtained in a subject with ASPS who is relapsed, metastatic, or non-surgically resectable by treatment with an anti-PD-1 antibody of the present disclosure. The present disclosure demonstrates that treatment with the anti-PD-1 antibodies of the present disclosure can still achieve significantly improved Objective Remission Rate (ORR) in ASPS (i.e., recurrent ASPS) subjects that have relapsed after previous first-, second-, or third-line and above treatment, or ASPS (i.e., metastatic ASPS) subjects that have metastasized to vital organs (e.g., lung, bone, brain), or non-surgically resectable ASPS subjects, indicating superior clinical efficacy and improved prognosis.
The present inventors have surprisingly found that, using the presently disclosed dosing regimen, the presently disclosed anti-PD-1 antibodies show significant clinical efficacy as anti-cancer therapeutics, while being well tolerated, particularly in ASPS for the treatment of recurrent, metastatic, or non-surgical resection.
It has also been unexpectedly found that, for ASPS subjects, particularly recurrent, metastatic, or non-surgically resectable ASPS subjects, no matter how many lines of anti-tumor therapy the subject has received since, and how positive the rate of PD-L1, etc., may benefit from treatment with the anti-PD-1 antibodies of the present disclosure, indicating superior clinical efficacy and improved prognosis.
Accordingly, in one aspect, the present disclosure provides the use of an anti-PD-1 antibody or antigen-binding fragment thereof in the manufacture of a medicament for treating or preventing alveolar soft tissue sarcoma in a subject in need thereof, wherein the anti-PD-1 antibody or antigen-binding fragment thereof comprises one or more CDRs comprising or consisting of SEQ ID No. 1, SEQ ID No. 2, SEQ ID No. 3, SEQ ID No. 4, SEQ ID No. 5 and SEQ ID No. 6.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a light chain CDR1 (LCDR 1), wherein the light chain LCDR1 sequence has at least 80% homology, at least 81% homology, at least 82% homology, at least 83% homology, at least 84% homology, at least 85% homology, at least 86% homology, at least 87% homology, at least 88% homology, at least 89% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology, or at least 99% homology to the amino acid sequence of SEQ ID No. 1.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a light chain CDR2 (LCDR 2), wherein the light chain LCDR2 sequence has at least 80% homology, at least 81% homology, at least 82% homology, at least 83% homology, at least 84% homology, at least 85% homology, at least 86% homology, at least 87% homology, at least 88% homology, at least 89% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology, or at least 99% homology to the amino acid sequence of SEQ ID No. 2.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a light chain CDR3 (LCDR 3), wherein the light chain LCDR3 sequence has at least 80% homology, at least 81% homology, at least 82% homology, at least 83% homology, at least 84% homology, at least 85% homology, at least 86% homology, at least 87% homology, at least 88% homology, at least 89% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology, or at least 99% homology to the amino acid sequence of SEQ ID No. 3.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a heavy chain CDR1 (HCDR 1), wherein the heavy chain HCDR1 sequence has at least 80% homology, at least 81% homology, at least 82% homology, at least 83% homology, at least 84% homology, at least 85% homology, at least 86% homology, at least 87% homology, at least 88% homology, at least 89% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology, or at least 99% homology to the amino acid sequence of SEQ ID No. 4.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a heavy chain CDR2 (HCDR 2), wherein the heavy chain HCDR2 sequence has at least 80% homology, at least 81% homology, at least 82% homology, at least 83% homology, at least 84% homology, at least 85% homology, at least 86% homology, at least 87% homology, at least 88% homology, at least 89% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology, or at least 99% homology to the amino acid sequence of SEQ ID No. 5.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a heavy chain CDR3 (HCDR 3), wherein the heavy chain HCDR3 sequence has at least 80% homology, at least 81% homology, at least 82% homology, at least 83% homology, at least 84% homology, at least 85% homology, at least 86% homology, at least 87% homology, at least 88% homology, at least 89% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology, or at least 99% homology to the amino acid sequence of SEQ ID No. 6.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises light chain LCDR1, LCDR2, and LCDR3, said light chain LCDR1, LCDR2, and LCDR3 comprising an amino acid sequence having at least 80% homology, at least 85% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology, or at least 99% homology to the amino acid sequences of SEQ ID NOs 1, 2, and 3, respectively; and heavy chains HCDR1, HCDR2 and HCDR3, said heavy chains HCDR1, HCDR2 and HCDR3 comprising an amino acid sequence having at least 80% homology, at least 85% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology or at least 99% homology with the amino acid sequences of SEQ ID NOs 4, 5 and 6, respectively.
In one embodiment, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a light chain comprising LCDR1, LCDR2, and LCDR3 as shown in SEQ ID No. 1, SEQ ID No. 2, and SEQ ID No. 3, respectively; the heavy chain comprises HCDR1, HCDR2 and HCDR3 as shown in SEQ ID NO. 4, SEQ ID NO. 5 and SEQ ID NO. 6, respectively.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a light chain variable region and a heavy chain variable region, wherein the light chain variable region comprises an amino acid sequence having at least 80% homology, at least 85% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology, or at least 99% homology to the amino acid sequence of SEQ ID No. 7; the heavy chain variable region comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% homology to the amino acid sequence of SEQ ID No. 8.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises, consists essentially of, or consists of the amino acid sequence of SEQ ID No. 7; the heavy chain variable region comprises, consists essentially of, or consists of the amino acid sequence of SEQ ID NO 8.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a full-length light chain having an amino acid sequence at least 80% homologous, at least 85% homologous, at least 90% homologous, at least 91% homologous, at least 92% homologous, at least 93% homologous, at least 94% homologous, at least 95% homologous, at least 96% homologous, at least 97% homologous, at least 98% homologous, or at least 99% homologous to the amino acid sequence of SEQ ID No. 9.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a full-length heavy chain having an amino acid sequence that is at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% homologous to the amino acid sequence of SEQ ID No. 10.
In one embodiment, the full length light and heavy chain sequences of the anti-PD-1 antibody or antigen-binding fragment thereof comprise or consist of SEQ ID NO 9 and SEQ ID NO 10, respectively. In one embodiment, the antibody of the present disclosure is designated "G-Ab1" and its light and heavy chain sequences are shown in SEQ ID NO 9 and SEQ ID NO 10, respectively.
In some embodiments, the alveolar soft tissue sarcoma is a recurrent, metastatic, or non-surgically resectable alveolar soft tissue sarcoma.
In some embodiments, there is provided a use of an anti-PD-1 antibody or antigen-binding fragment thereof in the manufacture of a medicament for treating or preventing alveolar soft tissue sarcoma in a subject in need thereof, wherein the medicament is a pharmaceutical composition, and wherein the pharmaceutical composition comprises a therapeutically effective amount of the anti-PD-1 antibody or antigen-binding fragment thereof and a pharmaceutically acceptable excipient or adjuvant.
In some embodiments, the pharmaceutically acceptable excipient or adjuvant comprises glutamic acid, proline, mannitol, sucrose, polysorbate-80, and sodium hydroxide.
In some embodiments, the medicament is administered at a dose of the anti-PD-1 antibody or antigen-binding fragment thereof in the range of about 1 mg/kg to about 10 mg/kg body weight about 1 time every 1, 2, 3, or 4 weeks. In still other embodiments, the medicament is administered at a fixed dose of at least about 200 mg, at least about 220 mg, at least about 240 mg, at least about 260 mg, at least about 280 mg, at least about 300 mg, at least about 320 mg, at least about 340 mg, at least about 360 mg, at least about 380 mg, or at least about 400 mg of the anti-PD-1 antibody or antigen-binding fragment thereof about 1 time every 1, 2, 3, or 4 weeks. In a specific embodiment, the medicament is administered at a fixed dose of about 280 mg of the anti-PD-1 antibody or antigen-binding fragment thereof about 1 time every 3 weeks.
In one embodiment, the medicament is administered at a dose of about 3mg/kg body weight of the anti-PD-1 antibody or antigen-binding fragment thereof about 1 time every 2 weeks.
In some embodiments, the tumor of the subject has at least about 1%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, or at least about 50% PD-L1 positive cells.
In some embodiments, the subject is a subject who has previously relapsed after receiving first, second or third line and more therapy or who has not responded to previous therapy.
In some embodiments, the subject is a subject who has previously experienced a relapse following surgery, ambertinib, VEGFR-TKI treatment (such as pazopanib or cediranib), or other anti-tumor chemotherapy or radiation therapy, or who has not responded to a previous treatment.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof is for administration in combination with one or more additional chemotherapeutic agents, radiotherapeutic agents, cytokines, or other antibodies.
In one aspect, there is provided a method of treating or preventing acinar soft tissue sarcoma in a subject in need thereof, comprising administering to the subject an effective amount of an anti-PD-1 antibody or antigen-binding fragment thereof, wherein the anti-PD-1 antibody or antigen-binding fragment thereof is as defined above.
In some embodiments, the alveolar soft tissue sarcoma is a recurrent, metastatic, or non-surgically resectable alveolar soft tissue sarcoma.
In some embodiments, there is provided a method of treating or preventing acinar soft tissue sarcoma in a subject in need thereof, comprising administering to the subject a pharmaceutical composition comprising one or more anti-PD-1 antibodies or antigen-binding fragments thereof of the present disclosure and a pharmaceutically acceptable excipient or adjuvant.
In some embodiments, the pharmaceutical composition comprises a therapeutically effective amount of the anti-PD-1 antibody or antigen-binding fragment thereof and a pharmaceutically acceptable excipient or adjuvant.
In some embodiments, the pharmaceutically acceptable excipient or adjuvant comprises glutamic acid, proline, mannitol, sucrose, polysorbate-80, and sodium hydroxide.
In some embodiments, there is provided a method of treating or preventing acinar soft tissue sarcoma in a subject in need thereof, comprising administering the pharmaceutical composition to the subject about 1 time every 1, 2, 3, or 4 weeks at a dose of the anti-PD-1 antibody, or antigen-binding fragment thereof, in the range of about 1 mg/kg to about 10 mg/kg body weight. In some further embodiments, there is provided a method of treating or preventing alveolar soft tissue sarcoma in a subject in need thereof, comprising administering the pharmaceutical composition to the subject about 1 time every 1, 2, 3, or 4 weeks at a fixed dose of at least about 200 mg, at least about 220 mg, at least about 240 mg, at least about 260 mg, at least about 280 mg, at least about 300 mg, at least about 320 mg, at least about 340 mg, at least about 360 mg, at least about 380 mg, or at least about 400 mg of the anti-PD-1 antibody or antigen-binding fragment thereof. In a specific embodiment, there is provided a method of treating or preventing alveolar soft tissue sarcoma in a subject in need thereof, comprising administering to the subject the pharmaceutical composition at a fixed dose of about 280 mg of the anti-PD-1 antibody or antigen-binding fragment thereof about 1 time every 3 weeks.
In one embodiment, there is provided a method of treating or preventing acinar soft tissue sarcoma in a subject in need thereof, comprising administering the pharmaceutical composition at a dose of about 3mg/kg body weight of the anti-PD-1 antibody or antigen-binding fragment thereof about 1 time every 2 weeks.
In some embodiments, the tumor of the subject has at least about 1%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, or at least about 50% PD-L1 positive cells.
In some embodiments, the subject is a subject that has relapsed after a previous first, second or third line and more therapy or has not responded to a previous therapy.
In some embodiments, the subject has previously relapsed or not responded to a previous surgical treatment, an ambertinib treatment, a VEGFR-TKI treatment (such as pazopanib or cediranib), or other anti-tumor chemotherapy or radiation therapy.
In some embodiments, there is provided a method of treating or preventing acinar soft tissue sarcoma in a subject in need thereof, comprising administering to the subject an anti-PD-1 antibody or antigen-binding fragment thereof of the present disclosure in combination with one or more additional chemotherapeutic agents, radiotherapeutic agents, cytokines, or other antibodies.
In another aspect, the present disclosure provides an anti-PD-1 antibody or antigen-binding fragment thereof for use in the treatment or prevention of acinar soft tissue sarcoma in a subject in need thereof, wherein the anti-PD-1 antibody or antigen-binding fragment thereof is as defined above.
The following detailed description and examples illustrate embodiments of the invention in detail. It is to be understood that this invention is not limited to the particular embodiments described herein and, thus, may include many variations and modifications. Those skilled in the art will recognize that variations and modifications are within the scope and spirit of the invention.
Drawings
Figure 1 shows that the G-Ab1 antibody specifically binds to PD-1, blocking its binding to PD-L1.
Detailed Description
Unless defined otherwise, all scientific and technical terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. In case of conflict, the present specification, including definitions, will control.
The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. Although only certain exemplary materials and methods are described herein, many methods and materials similar or equivalent to those described herein can be used in the practice of the present disclosure.
As used herein, the singular forms "a", "an" and "the" are intended to include the plural forms as well, unless the context clearly indicates otherwise. Furthermore, the open-ended expressions "comprising" and "comprises" are to be interpreted as meaning that structural elements or method steps not mentioned can also be present, but it is to be noted that the open-ended expressions also cover the case of consisting only of the stated components and method steps (i.e. the closed-ended expression "consisting of 8230; \8230; consists of).
In general, the term "about" is used herein to refer to a change in numerical values above and below the stated value by 5% modification.
As used throughout, ranges are used as shorthand for describing each and every value that is within the range. Any number within a range, such as an integer value, a value that is incremented by one tenth (when the end of the range is one digit after the decimal point), or a value that is incremented by one hundredth (when the end of the range is two digits after the decimal point) may be selected as the end point of the range. For example, a range of 1 to 10 is used to describe all values within that range, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 82300, \ 82309, 9, 9.1, 9.2, 9.3, 9.4, 9.5, 9.6, 9.7, 9.8, 9.9, and 10 (in tenths increments), and includes all subranges such as 1 to 1.5, 2.0 to 3.0, 4.0 to 5.0, 6.0 to 7.0, 8.0 to 9.0, and the like.
The present disclosure has demonstrated that the anti-PD-1 antibodies of the present disclosure show superior clinical efficacy and improved prognosis as anti-cancer therapeutics compared to certain known chemotherapeutic agents alone, with clinically significant significance. More specifically, by administering the anti-PD-1 antibodies of the present disclosure to ASPS subjects, the subjects' Objective Remission Rate (ORR) is significantly higher than current commonly used drugs, e.g., amboinib, VEGFR-TKI such as pazopanib, or cediranib, indicating superior clinical efficacy and improved prognosis.
The present disclosure has demonstrated that objective remission and improved prognosis can still be obtained in subjects with recurrent, metastatic, or non-surgically resectable ASPS on treatment with the anti-PD-1 antibodies of the present disclosure. More specifically, treatment with the anti-PD-1 antibodies of the present disclosure still results in a significantly improved Objective Remission Rate (ORR) in ASPS (i.e., recurrent ASPS) subjects that have relapsed after previous first-, second-, or third-line and beyond treatment, or ASPS (i.e., metastatic ASPS) subjects that have metastasized to vital organs (e.g., lung, bone, brain), or non-surgically resectable ASPS subjects, indicating superior clinical efficacy and improved prognosis.
The present disclosure has demonstrated that, using the dosing regimen of the present disclosure, the anti-PD-1 antibodies of the present disclosure exhibit significant clinical efficacy, improved tolerability, and safety as anti-cancer therapeutics, particularly in ASPS for treatment of recurrent, metastatic, or non-surgical resection.
The disclosure also demonstrates that, for ASPS subjects, particularly recurrent, metastatic, or non-surgically resectable ASPS subjects, no matter how many lines of anti-tumor therapy the subject has received since, and how positive the rate of PD-L1, etc., can benefit from treatment with the anti-PD-1 antibodies of the disclosure, indicating superior clinical efficacy and improved prognosis.
Accordingly, in one aspect, the present disclosure provides the use of an anti-PD-1 antibody or an antigen-binding fragment thereof, wherein the anti-PD-1 antibody or an antigen-binding fragment thereof comprises one or more CDRs selected from the group consisting of SEQ ID NO 1, SEQ ID NO 2, SEQ ID NO 3, SEQ ID NO 4, SEQ ID NO 5 and SEQ ID NO 6, in the manufacture of a medicament for treating or preventing alveolar soft tissue sarcoma in a subject in need thereof.
As used herein, the term "subject" means a mammal, such as a rodent, feline, canine, and primate, and the like. Preferably, the subject of the present disclosure is a human.
The term "treatment" as used herein refers to both therapeutic and prophylactic treatment. Subjects in need of treatment include those already with the disease or condition as well as those likely to develop the disease or condition as well as those targeted to prevent, delay or alleviate the disease or condition.
anti-PD-1 antibodies or antigen-binding fragments thereof
As used herein, the term "antibody" refers to a binding protein having at least one antigen binding domain. The antibody or antigen binding fragment thereof of the present disclosure may be a whole antibody or any fragment thereof. Accordingly, the antibodies or antigen-binding fragments thereof of the present disclosure include monoclonal antibodies or fragments thereof and antibody variants or fragments thereof, as well as immunoconjugates. Examples of antigen-binding fragments include Fab fragments, fab 'fragments, F (ab)' fragments, fv fragments, isolated CDR regions, single chain Fv molecules (scFv), and other antigen-binding fragments known in the art. Antibodies or antigen-binding fragments thereof may also include recombinant polypeptides, fusion proteins, and bispecific antibodies. The anti-PD-1 antibodies or antigen-binding fragments thereof of the present disclosure can have an IgG1, igG2, igG3, or IgG4 isotype. The term "isotype" refers to the class of antibodies encoded by the heavy chain constant region genes. In one embodiment, an anti-PD-1 antibody or antigen-binding fragment thereof of the present disclosure has an IgG1 or IgG4 isotype. The anti-PD-1 antibodies or antigen-binding fragments thereof of the present disclosure can be derived from any species, including, but not limited to, mouse, rat, rabbit, primate, llama, and human. The anti-PD-1 antibody or antigen-binding fragment thereof can be a chimeric antibody, a humanized antibody, or a fully human antibody. In one embodiment, the anti-PD-1 antibody is a murine antibody. In another embodiment, the anti-PD-1 antibody is a chimeric antibody. In another embodiment, the chimeric antibody is a mouse-human chimeric antibody. In another embodiment, the antibody is derived from a mouse and is humanized. In a further embodiment, the antibody is a fully human antibody.
As used herein, the term "antigen-binding fragment" refers to one or more antibody fragments that retain the ability to specifically bind to the antigen to which the intact antibody binds.
A "chimeric antibody" is an antibody having at least a portion of a heavy chain variable region and at least a portion of a light chain variable region derived from one species and at least a portion of a constant region derived from another species. For example, in one embodiment, a chimeric antibody can comprise a murine variable region and a human constant region.
A "humanized antibody" is an antibody comprising Complementarity Determining Regions (CDRs) derived from a non-human antibody, and framework and constant regions derived from a human antibody. For example, an anti-PD-1 antibody provided herein can comprise CDRs derived from one or more murine antibodies as well as human framework and constant regions. Thus, in one embodiment, a humanized antibody provided herein binds to the same epitope on PD-1 as a murine antibody from which the CDRs of the antibody are derived. Exemplary humanized antibodies are provided herein. Additional anti-PD-1 antibodies or variants thereof comprising the light and heavy chain CDRs provided herein can be produced using any human framework sequence and are also encompassed by the invention. In one embodiment, suitable for the invention of the framework sequences include the structure similar to the framework sequences provided herein those framework sequences. Other modifications in the framework regions can be made to improve the properties of the antibodies provided herein. Such other framework modifications may include chemical modifications; point mutations to reduce immunogenicity or to remove T cell epitopes; or back mutated to a residue in the original germline sequence.
The antibodies and antigen binding fragments thereof disclosed herein are specific for PD-1. In one embodiment, the antibodies and fragments thereof are specific for human PD-1. In one embodiment, the antibodies and fragments provided herein bind to human or primate PD-1, but not to PD-1 from any other mammal. In another embodiment, the antibodies and fragments thereof do not bind to mouse PD-1. Having specificity means that the antibody and fragments thereof bind to the PD-1 receptor with greater affinity than to any other target. In one embodiment, the PD-1 antibodies and fragments provided herein are specific for PD-1 and do not cross-react with CTLA4, ICOS, or CD 28.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a light chain LCDR1 sequence having at least 80% homology, at least 81% homology, at least 82% homology, at least 83% homology, at least 84% homology, at least 85% homology, at least 86% homology, at least 87% homology, at least 88% homology, at least 89% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology, or at least 99% homology to the amino acid sequence of SEQ ID No. 1.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a light chain LCDR2 sequence having at least 80% homology, at least 81% homology, at least 82% homology, at least 83% homology, at least 84% homology, at least 85% homology, at least 86% homology, at least 87% homology, at least 88% homology, at least 89% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology, or at least 99% homology to the amino acid sequence of SEQ ID No. 2.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a light chain LCDR3 sequence having at least 80% homology, at least 81% homology, at least 82% homology, at least 83% homology, at least 84% homology, at least 85% homology, at least 86% homology, at least 87% homology, at least 88% homology, at least 89% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology, or at least 99% homology to the amino acid sequence of SEQ ID No. 3.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a heavy chain HCDR1 sequence having at least 80% homology, at least 81% homology, at least 82% homology, at least 83% homology, at least 84% homology, at least 85% homology, at least 86% homology, at least 87% homology, at least 88% homology, at least 89% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology, or at least 99% homology to the amino acid sequence of SEQ ID No. 4.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a heavy chain HCDR2 sequence having at least 80% homology, at least 81% homology, at least 82% homology, at least 83% homology, at least 84% homology, at least 85% homology, at least 86% homology, at least 87% homology, at least 88% homology, at least 89% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology, or at least 99% homology to the amino acid sequence of SEQ ID No. 5.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a heavy chain HCDR3 sequence having at least 80% homology, at least 81% homology, at least 82% homology, at least 83% homology, at least 84% homology, at least 85% homology, at least 86% homology, at least 87% homology, at least 88% homology, at least 89% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology, or at least 99% homology to the amino acid sequence of SEQ ID No. 6.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises light chain LCDR1, LCDR2, and LCDR3, said light chain LCDR1, LCDR2, and LCDR3 comprising an amino acid sequence having at least 80% homology, at least 85% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology, or at least 99% homology to the amino acid sequences of SEQ ID NOs 1, 2, and 3, respectively; and heavy chain HCDR1, HCDR2 and HCDR3, said heavy chain HCDR1, HCDR2 and HCDR3 comprising an amino acid sequence having at least 80% homology, at least 85% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology or at least 99% homology to the amino acid sequences of SEQ ID NOs 4, 5 and 6, respectively.
In one embodiment, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a light chain comprising LCDR1, LCDR2, and LCDR3 as shown in SEQ ID No. 1, SEQ ID No. 2, and SEQ ID No. 3, respectively; the heavy chain comprises HCDR1, HCDR2 and HCDR3 as shown in SEQ ID NO 4, SEQ ID NO 5 and SEQ ID NO 6, respectively.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a light chain variable region and a heavy chain variable region, wherein the light chain variable region comprises an amino acid sequence having at least 80% homology, at least 85% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology, or at least 99% homology to the amino acid sequence of SEQ ID No. 7; the heavy chain variable region comprises an amino acid sequence having at least 80% homology, at least 85% homology, at least 90% homology, at least 91% homology, at least 92% homology, at least 93% homology, at least 94% homology, at least 95% homology, at least 96% homology, at least 97% homology, at least 98% homology, or at least 99% homology to the amino acid sequence of SEQ ID NO. 8.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises, consists essentially of, or consists of the amino acid sequence of SEQ ID No. 7; the heavy chain variable region comprises, consists essentially of, or consists of the amino acid sequence of SEQ ID NO 8.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a full-length light chain having an amino acid sequence at least 80% homologous, at least 85% homologous, at least 90% homologous, at least 91% homologous, at least 92% homologous, at least 93% homologous, at least 94% homologous, at least 95% homologous, at least 96% homologous, at least 97% homologous, at least 98% homologous, or at least 99% homologous to the amino acid sequence of SEQ ID No. 9.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a full-length heavy chain having an amino acid sequence at least 80% homologous, at least 85% homologous, at least 90% homologous, at least 91% homologous, at least 92% homologous, at least 93% homologous, at least 94% homologous, at least 95% homologous, at least 96% homologous, at least 97% homologous, at least 98% homologous, or at least 99% homologous to the amino acid sequence of SEQ ID No. 10.
In one embodiment, the full length light and heavy chain sequences of the anti-PD-1 antibody or antigen-binding fragment thereof comprise or consist of SEQ ID NO 9 and SEQ ID NO 10, respectively.
In one embodiment, the antibodies and fragments provided herein comprise a light chain and a heavy chain, each comprising three CDR regions. Exemplary LCDR1, LCDR2, LCDR3, HCDR1, HCDR2, and HCDR3 of PD-1 antibodies described herein are provided in table 1 below. Exemplary light chain variable region and heavy chain variable region sequences of the PD-1 antibodies described herein are provided in table 2 below. Exemplary full-length light chain and full-length heavy chain sequences of the PD-1 antibodies described herein are provided in table 3 below.
TABLE 1 light and heavy chain CDR sequences
Name (R) Numbering Sequence of
LCDR1 SEQ ID NO: 1 RASESVDNYGYSFMN
LCDR2 SEQ ID NO: 2 RASNLES
LCDR3 SEQ ID NO: 3 QQSNADPT
HCDR1 SEQ ID NO: 4 NFGMN
HCDR2 SEQ ID NO: 5 WISGYTREPTYAADFKG
HCDR3 SEQ ID NO: 6 DVFDY
TABLE 2 light and heavy chain variable region sequences
Figure DEST_PATH_IMAGE002
TABLE 3 full Length light chain and full Length heavy chain sequences
Figure DEST_PATH_IMAGE004
The anti-PD-1 antibodies or antigen-binding fragments thereof described herein, including chimeric, humanized or human monoclonal antibodies, can be prepared and isolated by methods well known in the art.
In some embodiments, the alveolar soft tissue sarcoma is a recurrent, metastatic, or non-surgically resectable alveolar soft tissue sarcoma. In one embodiment, the alveolar soft tissue sarcoma is recurrent alveolar soft tissue sarcoma. In one embodiment, the alveolar soft tissue sarcoma is metastatic alveolar soft tissue sarcoma. In some embodiments, the alveolar soft tissue sarcoma is a non-surgically resectable alveolar soft tissue sarcoma.
Dosing regimens
In some embodiments, there is provided a use of an anti-PD-1 antibody or antigen-binding fragment thereof in the manufacture of a medicament for treating or preventing alveolar soft tissue sarcoma in a subject in need thereof, wherein the medicament is a pharmaceutical composition and comprises one or more anti-PD-1 antibodies or antigen-binding fragments thereof described herein and one or more pharmaceutically acceptable excipients or adjuvants.
As used herein, "pharmaceutically acceptable excipient or adjuvant" refers to physiologically compatible solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, such as salts, buffers, antioxidants, sugars, aqueous or non-aqueous carriers, preservatives, wetting agents, surfactants or emulsifiers, or combinations thereof.
In some embodiments, the pharmaceutically acceptable excipient or adjuvant includes, for example, a buffer, a protectant, an osmotic pressure modifier, a surfactant, and/or a pH modifier.
In some embodiments, the buffer is, for example, glutamic acid, phosphate buffered saline, sodium acetate, sodium citrate, and Tris buffer; such as mannitol, sucrose and proline; surfactants such as polysorbates, for example, polysorbate-80, polysorbate-60, polysorbate-40 and polysorbate-20; the pH adjuster is, for example, sodium hydroxide.
In one embodiment, the pharmaceutically acceptable excipient or adjuvant comprises glutamic acid, proline, mannitol, sucrose, polysorbate-80, and sodium hydroxide.
In some embodiments, the pharmaceutical composition comprises about 1-11 mg/mL, e.g., 1-10 mg/mL, of the anti-PD-1 antibody or antigen-binding fragment thereof and one or more pharmaceutically acceptable excipients or adjuvants selected from the group consisting of glutamic acid, proline, mannitol, sucrose, polysorbate-80, and sodium hydroxide.
By "administering" is meant introducing a composition comprising a therapeutic agent to a subject using any of a variety of methods and delivery systems known to those of skill in the art. Routes of administration of the pharmaceutical compositions of the present disclosure include intravenous, intramuscular, subcutaneous, intraperitoneal, spinal or other parenteral routes of administration, for example by injection or infusion. The phrase "parenteral administration" as used herein refers to modes of administration other than enteral and topical administration, typically by injection, and includes, but is not limited to, intravenous, intramuscular, intraarterial, intrathecal, intralymphatic, intralesional, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural, and intrasternal injection and infusion, and in vivo electroporation. In some embodiments, the composition is administered by a non-parenteral route, and in some embodiments, by oral administration. Other non-parenteral routes include topical, epidermal or mucosal routes of administration, such as intranasal, vaginal, rectal, sublingual or topical. Administration may also be performed, for example, once, multiple times, and/or over one or more extended periods of time.
Dosage regimens can be adjusted as needed to provide the optimal response, e.g., the maximum therapeutic response and/or the minimum adverse effect. In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof of the present disclosure is administered in a weight-based dose. For administration of the anti-PD-1 antibody as a monotherapy or in combination with another anticancer agent at a weight-based dose, the dose can range from about 0.01 mg/kg to about 20 mg/kg, from about 0.1 mg/kg to about 10 mg/kg, from about 0.1 mg/kg to about 5 mg/kg, from about 1 mg/kg to about 5 mg/kg, from about 2 mg/kg to about 5 mg/kg, from about 7.5 mg/kg to about 12.5 mg/kg, or from about 0.1 mg/kg to about 30 mg/kg of the subject's body weight. For example, the dose may be about 0.1 mg/kg, about 0.3 mg/kg, about 1 mg/kg, about 2 mg/kg, about 3mg/kg, about 5 mg/kg or about 10 mg/kg body weight, or about 0.3 mg/kg, about 1 mg/kg, about 2 mg/kg, about 3mg/kg or about 5 mg/kg body weight. Based on the typical pharmacokinetic properties of antibodies, dosing schedules are typically designed to achieve exposures that result in sustained Receptor Occupancy (RO). Exemplary treatment regimens require administration about once per week, about once every 2 weeks, about once every 3 weeks, about once every 4 weeks, about once per month, about once every 3-6 months, or more. In certain embodiments, an anti-PD-1 antibody of the present disclosure is administered to a subject about once every 2 weeks. In other embodiments, the anti-PD-1 antibodies of the present disclosure are administered about once every 3 weeks. The dosage and timing may vary during the course of treatment. For example, a dosing schedule for an anti-PD-1 antibody monotherapy of the present disclosure can include administering an antibody: (i) about every 2 weeks for about 6-weeks; (ii) About every 4 weeks for about 6 doses, then about every 3 months; (iii) about every 3 weeks; (iv) About 3mg/kg to about 10 mg/kg once, followed by about 1 mg/kg every about 2 to 3 weeks.
In some embodiments, the pharmaceutical composition is administered to the subject at a dose of the anti-PD-1 antibody or antigen-binding fragment thereof in the range of about 1 mg/kg to about 10 mg/kg body weight about 1 time every 1, 2, 3, or 4 weeks.
In one embodiment, the pharmaceutical composition is administered at a dose of about 3mg/kg body weight of the anti-PD-1 antibody or antigen-binding fragment thereof about 1 time every 2 weeks.
In some embodiments, a dosage regimen of an anti-PD-1 antibody of the present disclosure comprises administering at least about 0.3 mg/kg to at least about 10 mg/kg body weight, at least about 1 mg/kg to at least about 5 mg/kg body weight, or at least about 1 mg/kg to at least about 3mg/kg body weight intravenously, wherein the antibody is administered every about 14-21 days over a period of up to about 6-weeks or about 12-weeks, until complete response or confirmation of a progressive disease. In certain embodiments, an anti-PD-1 antibody monotherapy of the present disclosure is administered at 3mg/kg every 2 weeks until progressive disease or unacceptable toxicity. In some embodiments, the anti-PD-1 antibody treatment of the present disclosure or any combination treatment disclosed herein lasts for at least about 1 month, at least about 3 months, at least about 6 months, at least about 9 months, at least about 1 year, at least about 18 months, at least about 24 months, at least about 3 years, at least about 5 years, or at least about 10 years.
When used in combination with other cancer agents, the dose of the anti-PD-1 antibodies of the present disclosure can be reduced, e.g., above 0.001 mg/kg and below 3mg/kg, compared to a monotherapy dose. In some embodiments, the dose is from about 0.001 mg/kg to about 1 mg/kg, from about 0.01 mg/kg to about 1 mg/kg, from about 0.1 mg/kg to about 1 mg/kg, or from about 0.001 mg/kg to about 0.1 mg/kg of body weight. In some embodiments, the dose is at least about 0.001 mg/kg, at least about 0.005 mg/kg, at least about 0.01 mg/kg, at least about 0.05 mg/kg, at least about 0.1 mg/kg, at least about 0.5 mg/kg, or at least about 1.0 mg/kg body weight.
In certain embodiments, the dose of the anti-PD-1 antibody of the present disclosure is a fixed dose in a pharmaceutical composition. In embodiments, the fixed dose of the anti-PD-1 antibody or antigen-binding fragment thereof is at least about 100 mg, 120 mg, 140 mg, 160 mg, 180 mg, 200 mg, 220 mg, 240 mg, 260 mg, 280 mg, 300 mg, 400 mg, 420 mg, 440 mg, 460 mg, 480mg, 500 mg, 520 mg, 540 mg, 560 mg, or 600 mg. For example, a fixed dose of an anti-PD-1 antibody of the present disclosure may be about 280 mg. In embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof of the present disclosure is administered at a dose of about 240 mg. In embodiments, an anti-PD-1 antibody or antigen-binding fragment thereof of the present disclosure is administered at a dose of about 360 mg. In embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof of the present disclosure is administered at a dose of about 480 mg. In embodiments, the fixed dose of the anti-PD-1 antibody or antigen-binding fragment thereof of the present disclosure is administered about once every week, every two weeks, every three weeks, every four weeks, every five weeks, or every six weeks. In one embodiment, 360 mg of the anti-PD-1 antibody or antigen-binding fragment is administered once every 3 weeks. In another embodiment, 480mg of the anti-PD-1 antibody or antigen-binding fragment is administered once every 4 weeks.
In some embodiments, the pharmaceutical composition is administered to the subject at a fixed dose of at least about 200 mg, at least about 220 mg, at least about 240 mg, at least about 260 mg, at least about 280 mg, at least about 300 mg, at least about 320 mg, at least about 340 mg, at least about 360 mg, at least about 380 mg, or at least about 400 mg of the anti-PD-1 antibody or antigen-binding fragment thereof about 1 time every 1, 2, 3, or 4 weeks.
In a specific embodiment, the pharmaceutical composition is administered to the subject at a fixed dose of about 280 mg of the anti-PD-1 antibody or antigen-binding fragment thereof about 1 time every 3 weeks.
Baseline characteristics
As described above and demonstrated in the examples of the present application, ASPS subjects, particularly recurrent, metastatic, or non-surgically resectable ASPS subjects, benefit from treatment with the anti-PD-1 antibodies of the present disclosure, regardless of the past few lines of anti-tumor therapy the subject had received, and regardless of the subject's PD-L1 positive rate, and the like, and result in significantly improved Objective Remission Rates (ORR), as well as significantly improved efficacy and prognosis.
The present inventors have surprisingly found that the presently disclosed anti-PD-1 antibodies achieve an ORR of 37.8% and are clinically significant in the treatment of recurrent, metastatic, or non-surgically resectable ASPS, compared to Objective Remission Rates (ORR) of 23.7% (investigator assessment in ALTER0203 study) or 32.4% (independent image assessment in ALTER0203 study), 16.7% -27.6%, 19% for several other agents such as, for example, anitinib, pazopanib, cediranib for treatment of ASPS.
In particular, as demonstrated in the examples herein, the ORR after treatment with the anti-PD-1 antibodies of the present disclosure was 35.7% even in subjects treated with line 0; the ORR following treatment with the anti-PD-1 antibodies of the disclosure can be as high as 39.1% in subjects treated prior to line 1 and beyond. Also as demonstrated in the examples herein, the anti-PD-1 antibodies of the present disclosure have a post-treatment ORR of 33.3% for CPS (combined positive score) ≧ 1; for subjects with CPS <1, the ORR after treatment with an anti-PD-1 antibody of the disclosure is 40.0%.
In one embodiment, the PD-L1 expression level can be measured by methods known in the art. The detection of PD-L1 is based on the detection of the cellular protein level, so that the clinical test is mainly based on the immunohistochemical method. Immunohistochemistry is a classic method for detecting protein expression, and evaluates expression conditions by staining antibodies, preparing digital sections and observing staining depth. In another embodiment, PD-L1 expression levels are measured by automated IHC. A PD-L1 positive tumor can have at least about 1%, at least about 2%, at least about 5%, at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or about 100% PD-L1 expressing tumor cells as measured by automated IHC.
As used herein, the term "CPS (combined positive score)" refers to a combined positive score calculated by (number of tumor cells positive for any intensity PD-L1 membrane staining + number of PD-L1 positive tumor associated immune cells/total number of tumor cells) 100.
In some embodiments, the subject is a subject who has previously relapsed after receiving first, second or third line and more therapy or who has not responded to previous therapy.
In some embodiments, the subject is a subject who has previously experienced a relapse following surgery, ambertinib, VEGFR-TKI treatment (such as pazopanib or cediranib), or other anti-tumor chemotherapy or radiation therapy, or who has not responded to a previous treatment.
In some embodiments, the subject is a subject that has relapsed after a previous first, second or third line and more treatment. In one embodiment, the subject has previously received an aniotinib treatment. In some embodiments, the subject has previously been treated with a VEGFR-TKI. In one embodiment, the subject has received pazopanib. In another embodiment, the subject has received cediranib. In another embodiment, the subject has received sunitinib. In some embodiments, the subject has previously received other anti-tumor chemotherapy or radiation therapy.
Combination drug
In some embodiments, there is provided a method of treating or preventing acinar soft tissue sarcoma in a subject in need thereof, comprising administering to the subject an anti-PD-1 antibody or antigen-binding fragment thereof described herein in combination with one or more additional therapeutic agents selected from a chemotherapeutic agent, a radiotherapeutic agent, a cytokine, or other antibody.
In one embodiment, the anti-PD-1 antibodies and antigen-binding fragments thereof can be administered to a subject before, during, and/or after the additional therapeutic agent is administered to the subject. In one embodiment, the anti-PD-1 antibody and the additional therapeutic agent exhibit therapeutic synergy when administered together (whether simultaneously or sequentially). In one embodiment, the anti-PD-1 antibody and the additional therapeutic agent are administered in separate formulations. In another embodiment, the anti-PD-1 antibody and the additional therapeutic agent are administered in the same formulation. In one embodiment, the anti-PD-1 antibody or antigen-binding fragment thereof provided herein enhances the immunomodulatory effect of the one or more additional therapeutic agents. In another embodiment, the one or more additional therapeutic agents enhance the effect of the anti-PD-1 antibody or antigen-binding fragment thereof.
Method of treatment
In one aspect, the invention provides a method of treating or preventing acinar soft tissue sarcoma in a subject in need thereof, comprising administering to the subject an effective amount of an anti-PD-1 antibody or antigen-binding fragment thereof, wherein the anti-PD-1 antibody or antigen-binding fragment thereof is as defined herein.
As used herein, the term "effective amount" refers to the amount of active agent required to provide a therapeutic and/or prophylactic benefit to a subject.
In some embodiments, the alveolar soft tissue sarcoma is a recurrent, metastatic, or non-surgically resectable alveolar soft tissue sarcoma.
In another aspect, the present disclosure provides an anti-PD-1 antibody or antigen-binding fragment thereof for use in treating or preventing acinar soft tissue sarcoma in a subject in need thereof, wherein the anti-PD-1 antibody or antigen-binding fragment thereof is as defined above.
While various embodiments of the present invention have been described above, it should be understood that they have been presented by way of example only, and not limitation. Many modifications to the disclosed embodiments can be made in accordance with the disclosure herein without departing from the spirit or scope of the invention. Thus, the breadth and scope of the present invention should not be limited by any of the above-described embodiments.
All documents referred to in this disclosure are incorporated herein by reference. All publications and patent documents cited in this application are incorporated by reference for all purposes to the same extent as if each individual publication or patent document were individually indicated to be so incorporated.
Examples
The light and heavy chain sequences of the anti-PD-1 antibody used in the examples are shown in SEQ ID NO 9 and SEQ ID NO 10 and designated G-Ab1.
Example 1 inhibition of PD-1 Signaling by G-Ab1
Experimental methods
PD-1 acts as an important regulatory immune checkpoint molecule, negatively regulating the activation of T-cells. This experiment simulates the regulatory mechanism of the above APC on T-cells by constructing a stably activated NFAT promoter-driven luciferase reporter and a Jurkat T-cell line stably expressing PD-1 (Jurkat/PD-1 cells) to function as T effector cells, and a CHOK cell line stably expressing PD-L1 (CHO/PD-L1 cells) as an artificial APC cell. When Jurkat/PD-1 and CHO/PD-L1 cells were cultured in admixture, PD-L1 and PD-1 bound to inhibit luciferase reporter gene expression induced by the activating NFAT promoter in Jurkat T-cells. After the G-Ab1 is added, the G-Ab1 is combined with PD-1 on the cell surface of Jurkat/PD-1, the combination pathway of PD-1 and PD-L1 is blocked, the inhibition effect of PD-1 on the expression of NFAT-luciferase reporter gene is relieved, the expression of luciferase reporter gene is increased, chemiluminescence is enhanced under the action of a reaction substrate, and the increase of chemiluminescence intensity is in direct proportion to the inhibition activity of G-Ab1 on the combination of PD-1 and PD-L1.
Collecting CHO/PD-L1 cells (Promega Corporation, cat. No. CS187108, lot. No. 0000141847) in logarithmic growth phase, digesting with pancreatin, counting, centrifuging appropriate amount of cell suspension, re-suspending with cell culture medium, and adjusting cell density to 4 × 10 5 Per ml, 100. Mu.L per well was added to a white impermeable cell culture plate (white 96 well plate, costa, cat. No. 3917) for overnight culture. RPMI 1640 (Gibco, cat 22400-089, lot 1687968) +2% FBS (Gibco, cat 11765-054, lot 1674921) medium was prepared as a diluent, and G-Ab1 (concentration: 10.4 mg/ml) and isotype control IgG4 antibody (control IgG4-AB150092 shown in FIG. 1, med. Camycopsis, cat E0849-T1501, lot AB150092, concentration 4.8 mg/ml) were diluted to 50. Mu.g/ml (2X), and 2.5-fold diluted 11 concentration gradients were performed as 2X working solutions. Counting the cell suspension of Jurkat/PD1 (Promega Corporation, cat. No. CS187102, batch No. 0000129197) in logarithmic growth phase, centrifuging, re-suspending with diluent, and adjusting the cell density to 1.25 × 10 6 One per ml. After removing the cell plate inoculated with CHO/PD-L1 and aspirating 95. Mu.L of culture supernatant per well, 40. Mu.L of the working solution and 40. Mu.L of Jurkat/PD1 cell suspension were added, respectively, to set up a negative control well (containing no antibody). Subjecting the loaded cell culture plate to 37 ℃ and 5% CO 2 After 6 h incubation in the incubator, the cells were removed and left at room temperature for 10 min, and 80. Mu.L of Bio-Glo was added to each well TM Substrate solution (Bio-Glo Luciferase assay system, promega Corporation, cat # G7940, lot # 0000165135) was incubated at room temperature for 10 min. The full wavelength chemiluminescence (Luminescence) of each well was read with a multifunctional microplate reader (Molecular Devices, model M5, code A-04-035), with the average value of the negative control wells being RLU no antibody control The ratio induction times Fold of Industtio per well were calculatedn=RLU Ab dilution /RLU no antibody control
Conclusion
The G-Ab1 concentration was logarithmized as an abscissa, and the chemiluminescence value (RLU) per well was determined Ab dilution ) Divided by the mean value of the luminescence (RLU) of the negative control wells (without antibody) no antibody control ) As a vertical coordinate, a non-linear regression analysis method in GraphPad Prism (GraphPad Software Inc., san Diego, california) is adopted, and a Sigmoidal dose-Response (Variable Slope) mode is selected for fitting a curve to be plotted, so that an inhibition curve of G-Ab1 for blocking the combination of PD-1 and PD-L1 is obtained (figure 1). The results show that G-Ab1 inhibits the binding of PD-1 and PD-L1 in a dose-dependent manner, and can reach saturation, and the inhibition curve is in an S shape. Isotype control IgG4 did not show any inhibitory effect, indicating that G-Ab1 is capable of specifically binding to PD-1, blocking its binding to PD-L1.
Example 2 therapeutic efficacy of G-Ab1 in the treatment of recurrent, metastatic, or non-surgically resectable Alveolar Soft Part Sarcoma (ASPS)
Test population: patients with recurrent, metastatic or non-surgically resectable alveolar soft tissue sarcoma (ASPS)
Inclusion criteria
1. Signing an informed consent;
2. age 18 to 75 years old; the nature is not limited;
ECOG score 0-1;
4. the expected survival time is more than 3 months;
5. histologically or cytologically confirmed patients with recurrent, metastatic, or non-surgically resectable alveolar soft tissue sarcoma;
6. at least one measurable lesion is defined as a lesion with a single diameter that can be accurately measured (RECIST 1.1: the longest diameter of the non-lymph node lesion is more than or equal to 10mm, and the short diameter of the lymph node lesion is more than or equal to 15 mm);
7. the time to completion of previous treatment prior to administration must be greater than or equal to 4 weeks or greater than or equal to the half-life (shorter) of 5 previous therapeutic agents (interval of at least 1 week between previous treatment and study groups); the elution period of the nitrosourea, mitomycin C and RANKL inhibitors is at least 6 weeks; the last radiotherapy is completed for at least more than or equal to 4 weeks; the previous treatment with other monoclonal antibody must be completed for more than or equal to 6 weeks;
8. if the patient receives the adriamycin with the cumulative dose of more than 350 mg/m 2 The heart color ultrasonography examination is required, and the Left Ventricular Ejection Fraction (LVEF) is more than or equal to 50%;
9. absolute neutrophil count ≥ 1.5X 10 9 L, platelet is more than or equal to 100 multiplied by 10 9 The hemoglobin is more than or equal to 80 g/L;
10. total bilirubin ≦ 1.5x ULN (Gilbert disease is known to tolerate ≦ 3 x ULN), AST/ALT ≦ 3 x ULN (liver metastasis patients tolerate AST and/or ALT ≦ 5x ULN), ALP ≦ 2.5 x ULN (liver metastasis or bone metastasis patients tolerate ≦ 5x ULN);
11. the creatinine clearance rate is more than or equal to 50 mL/min/1.73 m 2 (calculated by the Cockcroft-Gault formula) or Cr is less than or equal to 1.5xULN;
12. confirmed non-pregnant women within 72 hours prior to dosing; female and male patients in childbearing age have consented to full contraception prior to enrollment, during treatment, and 6 months after the last dose;
13. lactating women consented to cessation of lactation during the trial;
14. providing consent to archive tumor tissue specimens or fresh tissue specimens.
Test pharmaceutical composition:
the PD-1 antibody pharmaceutical composition is prepared by Jia and biopharmaceutical industries, inc., and comprises G-Ab1 at a concentration of 10.0 mg/ml; and 2.94mg/mL glutamic acid, 9.8mg/mL proline, 30mg/mL mannitol, 15mg/mL sucrose, 1.0mg/mL polysorbate-80, 0.63mg/mL sodium hydroxide and water for injection. The specification was 7mL: 70mg.
Design of research
Subjects will receive 3mg/kg of G-Ab, once every 2 weeks (q 2 w), i.v. until the end of the study or withdrawal of notice or development of disease Progression (PD), development of intolerant test drug related adverse events, initiation of subsequent anti-tumor therapy, death or loss of visit. The study employed a multicenter, open, single arm design, with all subjects receiving trial drug therapy. The study included 3 phases: a screening period (up to 28 days), a treatment period (14 days as one administration cycle, 3mg/kg body weight on day 1 of each cycle, q2w administration) and a follow-up period (including safety follow-up, disease progression follow-up (only applicable to subjects who did not continue to take medication but did not progress) and survival follow-up).
Tumor remission is assessed using the solid tumor efficacy evaluation criteria (RECIST version 1.1) and immunotherapy-modified RECIST efficacy evaluation criteria (irrecist). Tumor remission was assessed by Objective Remission Rate (ORR), progression Free Survival (PFS), duration of remission (DOR), disease Control Rate (DCR), overall Survival (OS), and objective remission rate based on ireist (iorrr), progression free survival (iPFS), duration of remission (iDOR), and disease control rate (iDCR) based on RECIST version 1.1. Wherein, the Objective Remission Rate (ORR) is the main clinical efficacy evaluation index. Tumor/efficacy assessments received throughout the study period (assessment range including thoracic, abdominal and pelvic) should be consistent during the screening period and should all be done on a central image. If the subjects quit the visit early after taking part in the study, and the efficacy assessment specified in the last protocol is more than or equal to 6 weeks, and the subsequent anti-tumor treatment is not started, the efficacy assessment should be completed as far as possible before leaving the study. Screening phase assessments should be performed within 28 days prior to the first test drug administration. The first tumor assessment was performed at a distance of 6 weeks from the first dose, followed by 1 assessment every 6 weeks (+ -7 days) until either of the following events occurred: end of study or withdrawal of informed consent or development of disease Progression (PD), development of intolerance of test drug-related adverse events, initiation of subsequent anti-tumor therapy, death or loss of visit. After remission (CR or PR) was recorded at 1 st time, CR or PR should be confirmed at the next tumor efficacy assessment time point (6 weeks ± 7 days). After the end of the treatment, for subjects who had not yet developed PD and had not started subsequent anti-tumor treatment, the procedure was the same as that for the study treatment period until the end of the study or withdrawal of informed consent or development of PD, start of subsequent anti-tumor treatment, death or loss of visit, with 1 efficacy assessment every 6 weeks (± 7 days) followed by 1 efficacy assessment every 12 weeks.
Subjects with known or suspected brain metastases at the time of screening should be examined for head MRI prior to initiation of study treatment. This part of the subjects should be followed up during the study and repeated for MRI assessment, with the same frequency as RECIST assessment. Brain metastases were evaluated as non-target lesions.
If the investigator is unable to determine the progression of the disease, especially with uncertain confidence in non-target lesions (NTL) and new lesions, it is recommended to continue treatment until clinically indicated or to re-evaluate the subject's status at the next scheduled evaluation time. Scan review if disease progression is confirmed, the date of progression should be the date of the first scan.
Following preliminary RECIST judgment of disease Progression (PD) by investigators based on RECIST, subject is allowed to continue to receive treatment if subject is deemed to be clinically benefited and resistant to the trial drug, subject must terminate study treatment after evidence of immune-related confirmed disease progression (icapd) is obtained, according to the revised RECIST efficacy assessment criteria (irest) for immunotherapy.
The safety evaluation was performed by evaluating safety indices such as Adverse Events (AE), immune-related adverse events (irAE), severe Adverse Events (SAE), laboratory examinations, physical examinations, vital signs, and electrocardiograms. The safety of the test drugs is assessed by recording, reporting and analyzing baseline conditions, adverse events, physical examinations, vital signs, electrocardiograms, laboratory examinations and others. During the whole experiment, the subjects were comprehensively evaluated for any adverse reactions starting from the subjects signing informed consent.
Subjects who fail to be evaluated for safety may be replaced by new entry subjects, such as subjects who do not complete the trial on schedule, including early withdrawal, non-compliance with entry criteria, non-scheduled completion of treatment and interview, and the like.
Subjects can quit the trial at any time without reason. If the subject is in poor compliance or violating a regimen, the investigator can also decide on its own whether to take the subject out of the trial.
Clinical results:
subject distribution:
a total of 37 subjects in this study received the test drug.
In the Full Analysis Set (FAS), all 37 subjects were in the han-nationality, the mean age (mean ± standard deviation) was 30.7 ± 8.31 years, and 59.5% of the subjects were female. The ECOG baseline score was between 0-1 for all subjects, with 45.9% of subjects scored ECOG 0 and 54.1% of subjects scored ECOG 1. Prior to enrollment, most subjects had metastases (36/37, 97.3%), with the sites of metastasis being primarily lung (35/37, 94.6%), followed by head (9/37, 24.3%) and bone (5/37, 13.5%). Except for 2 (5.4%) subjects with stage-wise TNM deletion, the remaining 35 (94.6%) subjects were TNM stage IV. Detailed subject baseline characteristics are summarized in table 4.
Table 4 baseline characterization summary of subject disease (full analysis set)
Figure DEST_PATH_IMAGE006
The curative effect results are as follows:
the main efficacy end point:
in FAS, after confirmation of the effect of CR and PR, the independent assessment committee assessed ASPS patients for the following effect on G-Ab1 treatment based on RECIST version 1.1: the Best Overall Response (BOR) was PR for 14 (37.8%) subjects, SD for 18 (48.6%) subjects, and PD for 5 (13.5%) subjects. ORR (95% CI) was 37.8% (22.46%, 55.24%), with a lower limit of 95% CI of 22.46%, greater than the preset efficacy threshold of 10%. Thus, the efficacy of G-Ab1 in a population of patients with recurrent, metastatic, or non-surgically resectable ASPS has significant clinical implications, according to pre-planned analysis. ORR results from investigators evaluated based on RECIST version 1.1 are consistent with results from independent evaluation committee evaluated based on RECIST version 1.1 (see table 5). While the reported ORR for anrotinib (ASPS standard of care drug) was 23.7% (investigator evaluation in ALTER0203 study) or 32.4% (independent image evaluation in ALTER0203 study).
Table 5 summary of tumor overall evaluation (independent evaluation committee and investigator, RECIST1.1 evaluation) (full analysis set)
Figure DEST_PATH_IMAGE008
Note: n = number of subjects in the corresponding analysis pool; n (%) = number of subjects satisfying a particular category and percentage calculation of percentage subject number based on corresponding analysis set; CI = confidence interval, estimated using the method of cloner-pearson.
EXAMPLE 3 subgroup efficacy of G-Ab1 in treating recurrent, metastatic, or non-surgically resectable alveolar soft tissue sarcoma (ASPS)
A subgroup analysis of efficacy endpoints was performed based on the IRC assessment of FAS in example 2.
The research carries out subgroup analysis on main curative effect indexes ORR according to the treatment line number (0 line treatment/1 line treatment and above) of the previous antitumor drugs and the positive rate (CPS < 1/> 1) of PD-L1.
In FAS, after confirmation of the effect of CR and PR, the independent assessment committee assessed ASPS patients for the following effect on G-Ab1 treatment based on RECIST version 1.1: for subjects treated with line 0, ORR (95% CI) after G-Ab1 treatment was 35.7% (12.76%, 64.86%); in the subjects treated with line 1 and above, ORR (95% CI) after G-Ab1 treatment was 39.1% (19.71%, 61.46%) (Table 6).
Table 6 summary of tumor overall evaluation based on several lines of treatment (independent evaluation committee, RECIST1.1 evaluation) (full analysis set)
Figure DEST_PATH_IMAGE010
Note: n = the number of corresponding sub-group subjects in the corresponding analysis set; n (%) = the number of subjects meeting a particular category and the percentage calculated based on the number of corresponding sub-group subjects in the corresponding analysis set; CI = confidence interval, estimated using the method of cloner-pearson.
In FAS, after confirmation of the effect of CR and PR, the independent assessment committee evaluated ASPS patients for the following G-Ab1 treatment based on RECIST version 1.1: ORR (95% CI) after G-Ab1 treatment was 33.3% (9.92%, 65.11%) for CPS (combined positive score) ≧ 1; for subjects with CPS <1, ORR (95% CI) after G-Ab1 treatment was 40.0% (21.13%, 61.33%) (table 7).
Table 7 summary of tumor gross evaluation based on PD-L1 expression (independent evaluation committee, RECIST1.1 evaluation) (full analysis set)
Figure DEST_PATH_IMAGE012
Note: n = the number of corresponding sub-group subjects in the corresponding analysis set; n (%) = number of subjects satisfying a particular category and percentage, calculation of percentage based on number of corresponding subset of subjects in corresponding analysis set; CI = confidence interval, estimated using the capper-pearson method. In FAS, based on confirmed PR/CR, the results of the independent assessment committee based on RECIST version 1.1 assessment are: the ORR (95% CI) was 33.3% (7.49%, 70.07%) in the past subjects who had been treated with antrodia after G-Ab1, and 39.3% (21.50%, 59.42%) in the past subjects who had not been treated with antrodia (table 8).
Table 8 summary of therapeutic efficacy of past use of the erlotinib subgroup (independent assessment committee, RECIST1.1 assessment) (full analysis set)
Figure DEST_PATH_IMAGE014
Note: n = number of corresponding sub-group subjects in the corresponding analysis set; the percentage is calculated based on the number of corresponding sub-group subjects in the corresponding analysis set; ORR and DCR calculated 95% CI using the Clopper-pearson method.
Furthermore, the ORR (95% CI) was 36.8% (n = 7/19) (16.3%, 61.6%) in the subjects who had received VEGFR-TKIs after the G-Ab1 treatment, and the ORR (95% CI) was 38.9% (n = 7/18) (17.3%, 64.3%) in the subjects who had not used VEGFR-TKIs after the G-Ab1 treatment.
In conclusion, the therapeutic efficacy of the anti-PD-1 antibodies described in this disclosure in a population of patients with ASPS that are recurrent, metastatic or non-surgically resectable has significant clinical significance, with an ORR significantly higher than that reported for the known drugs for treatment of ASPS, anitinib and Cediranib (Cediranib). In addition, the PD-1 antibody disclosed by the disclosure has curative effects on ASPS patients with first-line and second-line or third-line and above, and also has curative effects on patients who have previously undergone surgery, radiotherapy and VEGFR-TKI treatment failure, relapse or metastasis, patients with any PD-L1 positive rate (CPS < 1/> or less 1), or ASPS patients who have failed, relapse or metastasis anconii treatment.
While various embodiments of the present invention have been described above, it should be understood that they have been presented by way of example only, and not limitation. The present invention is susceptible to various changes and modifications without departing from the spirit and scope of the present invention, and such changes and modifications are intended to be within the scope of the present invention as claimed. The scope of the invention is defined by the appended claims and equivalents thereof.
<110> Jia and biopharmaceutical industries Ltd
<120> application of anti-PD-1 antibody in treatment of alveolar soft tissue sarcoma
<130> CPCH2161664N
<160> 10
<170> PatentIn version 3.5
<210> 1
<211> 15
<212> PRT
<213> Artificial sequence
<220>
<223> LCDR1
<400> 1
Arg Ala Ser Glu Ser Val Asp Asn Tyr Gly Tyr Ser Phe Met Asn
1 5 10 15
<210> 2
<211> 7
<212> PRT
<213> Artificial sequence
<220>
<223> LCDR2
<400> 2
Arg Ala Ser Asn Leu Glu Ser
1 5
<210> 3
<211> 8
<212> PRT
<213> Artificial sequence
<220>
<223> LCDR3
<400> 3
Gln Gln Ser Asn Ala Asp Pro Thr
1 5
<210> 4
<211> 5
<212> PRT
<213> Artificial sequence
<220>
<223> HCDR1
<400> 4
Asn Phe Gly Met Asn
1 5
<210> 5
<211> 17
<212> PRT
<213> Artificial sequence
<220>
<223> HCDR2
<400> 5
Trp Ile Ser Gly Tyr Thr Arg Glu Pro Thr Tyr Ala Ala Asp Phe Lys
1 5 10 15
Gly
<210> 6
<211> 5
<212> PRT
<213> Artificial sequence
<220>
<223> HCDR3
<400> 6
Asp Val Phe Asp Tyr
1 5
<210> 7
<211> 110
<212> PRT
<213> Artificial sequence
<220>
<223> light chain variable region
<400> 7
Asp Ile Val Leu Thr Gln Ser Pro Ala Ser Leu Ala Val Ser Pro Gly
1 5 10 15
Gln Arg Ala Thr Ile Thr Cys Arg Ala Ser Glu Ser Val Asp Asn Tyr
20 25 30
Gly Tyr Ser Phe Met Asn Trp Phe Gln Gln Lys Pro Gly Gln Pro Pro
35 40 45
Lys Leu Leu Ile Tyr Arg Ala Ser Asn Leu Glu Ser Gly Val Pro Ala
50 55 60
Arg Phe Ser Gly Ser Gly Ser Arg Thr Asp Phe Thr Leu Thr Ile Asn
65 70 75 80
Pro Val Glu Ala Asp Asp Thr Ala Asn Tyr Tyr Cys Gln Gln Ser Asn
85 90 95
Ala Asp Pro Thr Phe Gly Gln Gly Thr Lys Leu Glu Ile Lys
100 105 110
<210> 8
<211> 114
<212> PRT
<213> Artificial sequence
<220>
<223> heavy chain variable region
<400> 8
Gln Ile Gln Leu Val Gln Ser Gly Ser Glu Leu Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Asn Phe
20 25 30
Gly Met Asn Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Lys Trp Met
35 40 45
Gly Trp Ile Ser Gly Tyr Thr Arg Glu Pro Thr Tyr Ala Ala Asp Phe
50 55 60
Lys Gly Arg Phe Val Ile Ser Leu Asp Thr Ser Val Ser Thr Ala Tyr
65 70 75 80
Leu Gln Ile Ser Ser Leu Lys Ala Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Asp Val Phe Asp Tyr Trp Gly Gln Gly Thr Leu Val Thr Val
100 105 110
Ser Ser
<210> 9
<211> 217
<212> PRT
<213> Artificial sequence
<220>
<223> full Length light chain
<400> 9
Asp Ile Val Leu Thr Gln Ser Pro Ala Ser Leu Ala Val Ser Pro Gly
1 5 10 15
Gln Arg Ala Thr Ile Thr Cys Arg Ala Ser Glu Ser Val Asp Asn Tyr
20 25 30
Gly Tyr Ser Phe Met Asn Trp Phe Gln Gln Lys Pro Gly Gln Pro Pro
35 40 45
Lys Leu Leu Ile Tyr Arg Ala Ser Asn Leu Glu Ser Gly Val Pro Ala
50 55 60
Arg Phe Ser Gly Ser Gly Ser Arg Thr Asp Phe Thr Leu Thr Ile Asn
65 70 75 80
Pro Val Glu Ala Asp Asp Thr Ala Asn Tyr Tyr Cys Gln Gln Ser Asn
85 90 95
Ala Asp Pro Thr Phe Gly Gln Gly Thr Lys Leu Glu Ile Lys Arg Thr
100 105 110
Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu Gln Leu
115 120 125
Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr Pro
130 135 140
Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln Ser Gly
145 150 155 160
Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser Thr Tyr
165 170 175
Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys His
180 185 190
Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser Pro Val
195 200 205
Thr Lys Ser Phe Asn Arg Gly Glu Cys
210 215
<210> 10
<211> 440
<212> PRT
<213> Artificial sequence
<220>
<223> full-Length heavy chain
<400> 10
Gln Ile Gln Leu Val Gln Ser Gly Ser Glu Leu Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Asn Phe
20 25 30
Gly Met Asn Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Lys Trp Met
35 40 45
Gly Trp Ile Ser Gly Tyr Thr Arg Glu Pro Thr Tyr Ala Ala Asp Phe
50 55 60
Lys Gly Arg Phe Val Ile Ser Leu Asp Thr Ser Val Ser Thr Ala Tyr
65 70 75 80
Leu Gln Ile Ser Ser Leu Lys Ala Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Asp Val Phe Asp Tyr Trp Gly Gln Gly Thr Leu Val Thr Val
100 105 110
Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe Pro Leu Ala Pro Cys
115 120 125
Ser Arg Ser Thr Ser Glu Ser Thr Ala Ala Leu Gly Cys Leu Val Lys
130 135 140
Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser Gly Ala Leu
145 150 155 160
Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gln Ser Ser Gly Leu
165 170 175
Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser Ser Leu Gly Thr
180 185 190
Lys Thr Tyr Thr Cys Asn Val Asp His Lys Pro Ser Asn Thr Lys Val
195 200 205
Asp Lys Arg Val Glu Ser Lys Tyr Gly Pro Pro Cys Pro Pro Cys Pro
210 215 220
Ala Pro Glu Phe Leu Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys
225 230 235 240
Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val
245 250 255
Val Val Asp Val Ser Gln Glu Asp Pro Glu Val Gln Phe Asn Trp Tyr
260 265 270
Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu
275 280 285
Gln Phe Asn Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His
290 295 300
Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys
305 310 315 320
Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln
325 330 335
Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Gln Glu Glu Met
340 345 350
Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro
355 360 365
Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn
370 375 380
Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu
385 390 395 400
Tyr Ser Arg Leu Thr Val Asp Lys Ser Arg Trp Gln Glu Gly Asn Val
405 410 415
Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gln
420 425 430
Lys Ser Leu Ser Leu Ser Leu Gly
435 440

Claims (11)

1. Use of an anti-PD-1 antibody or antigen-binding fragment thereof in the manufacture of a medicament for treating or preventing alveolar soft tissue sarcoma in a subject in need thereof, wherein the anti-PD-1 antibody or antigen-binding fragment thereof comprises a light chain comprising LCDR1, LCDR2, and LCDR3 as set forth in SEQ ID No. 1, SEQ ID No. 2, and SEQ ID No. 3, respectively, and a heavy chain comprising HCDR1, HCDR2, and HCDR3 as set forth in SEQ ID No. 4, SEQ ID No. 5, and SEQ ID No. 6, respectively.
2. The use of claim 1, wherein the light chain variable region of the anti-PD-1 antibody or antigen-binding fragment thereof comprises an amino acid sequence that is at least 80% homologous to the amino acid sequence of SEQ ID No. 7 and the heavy chain variable region comprises an amino acid sequence that is at least 80% homologous to the amino acid sequence of SEQ ID No. 8.
3. The use according to claim 1 or 2, wherein the light chain and the heavy chain of the anti-PD-1 antibody or an antigen-binding fragment thereof comprise or consist of the sequences shown as SEQ ID NO 9 and 10, respectively.
4. The use of any one of claims 1-3, wherein the alveolar soft tissue sarcoma is a recurrent, metastatic, or non-surgically resectable alveolar soft tissue sarcoma.
5. The use of any one of claims 1-4, wherein the medicament comprises a therapeutically effective amount of the anti-PD-1 antibody or antigen-binding fragment thereof and a pharmaceutically acceptable excipient or adjuvant.
6. The use of claim 5, wherein the medicament comprises glutamic acid, proline, mannitol, sucrose, polysorbate-80, and sodium hydroxide.
7. The use of any one of claims 1-6, wherein (i) the medicament is administered at a dose of the anti-PD-1 antibody or antigen-binding fragment thereof in the range of about 1 mg/kg to about 10 mg/kg body weight about 1 time every 1, 2, 3, or 4 weeks; or (ii) the medicament is administered about 1 time every 1, 2, 3, or 4 weeks at a fixed dose of at least about 200 mg, at least about 220 mg, at least about 240 mg, at least about 260 mg, at least about 280 mg, at least about 300 mg, at least about 320 mg, at least about 340 mg, at least about 360 mg, at least about 380 mg, or at least about 400 mg of the anti-PD-1 antibody or antigen-binding fragment thereof.
8. The use of any one of claims 1-7, wherein the subject is a subject that has relapsed after having received first, second or third and above treatment or has not responded to a previous treatment.
9. The use of claim 8, wherein the subject has previously received surgery, chemotherapy, or radiation therapy followed by a relapse or is unresponsive to the previous therapy.
10. The use of claim 9, wherein the subject has previously received an amboinib therapy, a VEGFR-TKI therapy.
11. The use of any one of claims 1-10, wherein the anti-PD-1 antibody or antigen-binding fragment thereof is for administration in combination with one or more additional chemotherapeutic agents, radiotherapeutic agents, cytokines, or other antibodies.
CN202110783289.9A 2021-07-12 2021-07-12 Application of anti-PD-1 antibody in treatment of alveolar soft tissue sarcoma Pending CN115607662A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110783289.9A CN115607662A (en) 2021-07-12 2021-07-12 Application of anti-PD-1 antibody in treatment of alveolar soft tissue sarcoma

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110783289.9A CN115607662A (en) 2021-07-12 2021-07-12 Application of anti-PD-1 antibody in treatment of alveolar soft tissue sarcoma

Publications (1)

Publication Number Publication Date
CN115607662A true CN115607662A (en) 2023-01-17

Family

ID=84855604

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110783289.9A Pending CN115607662A (en) 2021-07-12 2021-07-12 Application of anti-PD-1 antibody in treatment of alveolar soft tissue sarcoma

Country Status (1)

Country Link
CN (1) CN115607662A (en)

Similar Documents

Publication Publication Date Title
US20220023285A1 (en) Combination of a pd-1 antagonist and a vegfr/fgfr/ret tyrosine kinase inhibitor for treating cancer
US11866509B2 (en) Humanized antibodies against CEACAM1
TWI663983B (en) Combination of a pd-1 antagonist and a vegfr inhibitor for treating cancer
KR102289394B1 (en) Stable formulations of antibodies to human programmed death receptor pd-1 and related treatments
US20220265844A1 (en) Dosing regimens for anti-tf-antibody drug-conjugates
KR20160108568A (en) Combination of a pd-1 antagonist and an ido1 inhibitor for treating cancer
CN113891714A (en) Triple therapy to enhance cancer cell killing in low immunogenic cancers
US20210317214A1 (en) Combination therapy of a pd-1 antagonist and lag3 antagonist for treating patients with non-microsatellite instability-high or proficient mismatch repair colorectal cancer
CN117603360A (en) Multispecific antibodies for treating cancer
KR20200078483A (en) Composition and method for treating liver cancer
US20240010729A1 (en) Combination therapy of a pd-1 antagonist and lag3 antagonist and lenvatinib or a pharmaceutically acceptable salt thereof for treating patients with cancer
CN115607662A (en) Application of anti-PD-1 antibody in treatment of alveolar soft tissue sarcoma
CN115607664A (en) Application of anti-PD-1 antibody in treatment of cervical cancer
US11427647B2 (en) Polynucleotides encoding humanized antibodies against CEACAM1
US20240092934A1 (en) Assessment of ceacam1 expression on tumor infiltrating lymphocytes
CN115607663A (en) Use of anti-PD-1 antibodies in the treatment of primary mediastinal large B-cell lymphoma
CN115611981A (en) Use of anti-PD-1 antibodies in the treatment of peripheral T cell lymphoma
WO2023174278A1 (en) Pharmaceutical composition of anti-tim-3 antibody and hypomethylating agent
US20230365691A1 (en) Use of anti-pd-1 antibody in treatment of nasopharyngeal carcinoma
RU2361614C2 (en) APPLICATION OF ANTIBODIES AGAINST α5β1 FOR INHIBITION OF CANCER CELLS PROLIFERATION
KR102662228B1 (en) Combination of PD-1 antagonists and VEGFR/FGFR/RET tyrosine kinase inhibitors to treat cancer
TW202333783A (en) Methods of treating cancer with a combination of an anti-pd-1 antibody and an anti-cd30 antibody-drug conjugate
CN115697400A (en) anti-CD 30 antibody-drug conjugates and their use for the treatment of non-Hodgkin&#39;s lymphoma
KR20240064733A (en) Combination of a pd-1 antagonist and a vegfr/fgfr/ret tyrosine kinase inhibitor for treating cancer

Legal Events

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