CN113365659A - Use of anti-PD-L1 antibodies for the treatment of head and neck cancer - Google Patents

Use of anti-PD-L1 antibodies for the treatment of head and neck cancer Download PDF

Info

Publication number
CN113365659A
CN113365659A CN202080011333.8A CN202080011333A CN113365659A CN 113365659 A CN113365659 A CN 113365659A CN 202080011333 A CN202080011333 A CN 202080011333A CN 113365659 A CN113365659 A CN 113365659A
Authority
CN
China
Prior art keywords
seq
antibody
amino acid
chemotherapy
acid sequence
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.)
Granted
Application number
CN202080011333.8A
Other languages
Chinese (zh)
Other versions
CN113365659B (en
Inventor
张喜全
王训强
陈杰
赵伟
陈德培
朱漫杰
魏娇
陈飞洋
周雅倩
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.)
Chia Tai Tianqing Pharmaceutical Group Co Ltd
Nanjing Shunxin Pharmaceutical Co Ltd
Original Assignee
Chia Tai Tianqing Pharmaceutical Group Co Ltd
Nanjing Shunxin Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Chia Tai Tianqing Pharmaceutical Group Co Ltd, Nanjing Shunxin Pharmaceutical Co Ltd filed Critical Chia Tai Tianqing Pharmaceutical Group Co Ltd
Publication of CN113365659A publication Critical patent/CN113365659A/en
Application granted granted Critical
Publication of CN113365659B publication Critical patent/CN113365659B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants

Abstract

The present application provides the use of an anti-PD-L1 antibody for the treatment of head and neck cancer, comprising administering to the subject a therapeutically effective amount of an inhibitor of the interaction between the PD-1 receptor and its ligand PD-L1, wherein the inhibitor is an anti-PD-L1 antibody.

Description

Use of anti-PD-L1 antibodies for the treatment of head and neck cancer Technical Field
The present application provides a method of treating cancer in a subject comprising administering to the subject a therapeutically effective amount of an inhibitor of the interaction between the PD-1 receptor and its ligand PD-L1.
Background
The innate immune system, which contains T lymphocytes, has a powerful anti-cancer capacity with a wide range of abilities and precise specificity to respond to a variety of tumor antigens. Emerging cancer immunotherapy enhances anti-tumor immune responses by adoptive transfer of activated effector cells, immunization against relevant antigens, or provision of non-specific immunostimulants. Over the last decade, researchers have struggled to develop specific immune checkpoint inhibitors and expect to provide new immunotherapeutic protocols for the treatment of cancer, including the development of antibodies (antibodies) Ipilimumab (Ipilimumab) (a) that bind to and inhibit CTLA-4
Figure PCTCN2020074083-APPB-000001
) For treating patients with advanced melanoma (Hodi et al (2010) N Engl J Med 363:711-23), and development of antibodies such as Nivolumab (Nivolumab) (r)
Figure PCTCN2020074083-APPB-000002
) And Pabolizumab (Pembrolizumab) (Pembrolizumab)
Figure PCTCN2020074083-APPB-000003
) They specifically bind to programmed death receptor-1 (PD-1) and block the inhibitory PD-1/PD-1 ligand pathway (Topalian et al (2012a) N Engl J Med 366: 2443-54). Among them, PD-1(programmed death-1) is a key immune checkpoint receptor expressed by activated T lymphocytes and B lymphocytes and mediates immunosuppression, and its ligands include at least PD-L1 and PD-L2.
PD-L1(Programmed death-ligand 1), also known as CD274 and B7-H1, is a 40kDa type 1 transmembrane protein encoded by the CD274 gene and is a ligand for PD-1. Both PD-L1 and PD-1 belong to the immunoglobulin superfamily and both consist of two extracellular Ig domains, an N-terminal V domain and a C-terminal constant domain. The binding interface of PD-L1 to programmed death receptor-1 (PD-1) and B7-1(CD80) is on an IgV-like domain (Lin et al (2008) PNAS105: 3011-3016). While PD-L1 contains a conserved short intracellular tail region (about 30 amino acids), PD-1 contains two cytoplasmic tyrosine-based signaling motifs, namely an immunoreceptor tyrosine-based inhibitory motif (ITIM) and an immunoreceptor tyrosine-based switching motif (ITSM). Following T cell stimulation, PD-1 recruits the tyrosine phosphatase SHP-2 to the ITSM motif in its cytoplasmic tail, resulting in dephosphorylation of effector molecules involved in the CD3T cell signaling cascade, such as CD3 ζ, PKC θ and ZAP70 (Freeman et al (2000) J Exp Med 192: 1027-34; Latchman et al (2001) Nat Immunol 2: 261-8; Carter et al (2002) Eur J Immunol 32: 634-43). PD-L1 is widely distributed not only on leukocytes and nonhematopoietic cells in lymphoid and non-lymphoid tissues, but also in various cancer cells, is highly expressed on the surface of various tumor cells, and the degree of malignancy and poor prognosis of tumors are closely related to the expression level of PD-L1. There are clinical data indicating that high tumor expression of PD-L1 is associated with increased tumor invasiveness and poor prognosis. The PD-1/PD-L1 complex forms a transmission inhibition signal and negatively regulates T cell immune response; it inhibits TCR-mediated T cell activation, cytokine production and T cell proliferation (Fife et al (2011) Nature Immunology 10: 1185-1193); induction of depletion or anergy in cognate antigen-specific T cells (Hofmeyer et al (2011) Journal of Biomedicine and Biotechnology 2011: 1-9); promote the differentiation of Th1 cells into Foxp3+ regulatory T cells (Armanath et al (2011) Science TransMed 3: 1-13; Francisco et al (2009) J. exp. Med.206: 3015-; and inducing apoptosis of effector T cells. Disruption of the PD-L1 gene resulted in an upregulated T cell response and the generation of autoreactive T cells (Latchman et al (2004) PNAS 101: 10691-10696). Antibody blockade of PD-1 or PD-L1 resulted in increased anti-tumor immunity (Iwai et al (2002) PNAS99: 12293-12297).
The anti-PD-L1 antibody can block the interaction of PD-L1 with PD-1 and CD80, so that the related negative regulation signals can not be started and conducted, thereby preventing the activity of effector T cells in a tumor microenvironment from being inhibited, and enabling the T cells to play the functions of killing and inhibiting the tumor cells. The anti-PD-L1 antibody can directly act on tumor tissues, so that the antibody has high specificity and safety. The major anti-PD-L1 monoclonal antibody drug products currently include Atezolizumab, Roche, Durvalumab, Asricon, and Avelumab, Merck & Hurride, among others. Patent WO2016022630 also discloses an anti-PD-L1 antibody, which has high affinity for PD-L1, can significantly inhibit the interaction between PD-L1 and PD-1 on the cell surface, and significantly promote the secretion of IL-2 and IFN-gamma by T cells.
Head and neck cancers include oral, nasopharyngeal, oropharyngeal, and laryngeal cancers, accounting for around 5% of global cancers (excluding non-melanoma skin cancers) (Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer invasion and mortality world wide: sources, methods and major Patterns in GLOBOCAN 2012. J Cancer 2015; 136: E359-E386). In 2012, about 68 ten thousand head and neck cancers are newly diagnosed, and 37 ten thousand patients die of the disease. Of these new cases reported, nearly 14 million cases occur in europe, with over 90% of cases histologically classified as squamous cell carcinoma. In the united states, an estimated 6.1 million people were diagnosed with head and neck cancer in 2016, and over 1.3 million people would die from the cancer. Head and neck cancer is mainly squamous carcinoma. Squamous Cell Carcinoma of Head and Neck (SCCHN) affecting the oral cavity, larynx, oropharynx and hypopharynx account for 75% of all SCCHNs and is closely associated with alcohol and tobacco use. Oropharyngeal SCCHN is also closely associated with Human Papilloma Virus (HPV) type 16 infection. Over the past 20 years, HPV-positive oropharyngeal cancers have increased dramatically, suggesting an important role for HPV infection in oropharyngeal cancer. Despite the decreased incidence of HPV-negative SCCHN (following the trend of decreased use of tobacco products worldwide), the cure of HPV-negative tumors has not been optimal. Of newly diagnosed SCCHN patients, about 60% of patients are locally or regionally advanced disease. The failure rate of regional treatment is between 35% and 65% depending on the tumor site, stage and resectability. The median PFS (progression-free survival) for this disease was 1.9 years, with a 3-year PFS rate of 61.2% reported. Most of the treated patients eventually recur locally, with distant metastases occurring in 10-30% of patients. Current treatment options for regional and distant recurrence are limited. The prognosis for both relapses is poor, and only a few patients are eligible to receive a potentially curative treatment for regional disease recurrence, but the incidence is high. According to the CSCO head and neck tumor guidelines (2018.V1), first line treatment of recurrent metastatic head and neck squamous carcinoma recommends (1A) a chemotherapy regimen comprising cisplatin/carboplatin +5-FU with an average OS (overall survival) of 7.4-8.7 months. At 5/2016 and 1/12, the PD-1 inhibitors Keytruda (Pembrolizumab, morsandon) and Opdivo (Nivolumab, bosch-mai-gui) were FDA approved for treatment of recurrent or metastatic treatment SCCHN, respectively, and are currently used for second line therapy.
Disclosure of Invention
Summary of the present application
The present application provides a method of treating, ameliorating or ameliorating cancer in a subject comprising administering to the subject a therapeutically effective amount of an inhibitor of the interaction between a PD-1 receptor and its ligand PD-L1, wherein the inhibitor is a PD-L1 antibody.
The present application also provides a use of an anti-PD-L1 antibody for treating, ameliorating, or ameliorating cancer, comprising administering to the subject a therapeutically effective amount of an inhibitor of the interaction between the PD-1 receptor and its ligand PD-L1, wherein the inhibitor is an anti-PD-L1 antibody.
The application also provides use of an anti-PD-L1 antibody in the manufacture of a medicament for treating, ameliorating, or ameliorating cancer.
The present application also provides anti-PD-L1 antibodies for use in treating, ameliorating, or ameliorating cancer in a subject.
In some embodiments, the anti-PD-L1 antibody comprises the amino acid sequence: a heavy chain CDR1 region having at least 80% homology to the amino acid sequence set forth in SEQ ID NO.1 or SEQ ID NO. 4; a heavy chain CDR2 region having at least 80% homology to the amino acid sequence set forth in SEQ ID NO. 2 or SEQ ID NO. 5; a heavy chain CDR3 region having at least 80% homology to the amino acid sequence set forth in SEQ ID NO. 3 or SEQ ID NO. 6; a light chain CDR1 region having at least 80% homology to the amino acid sequence set forth in SEQ ID NO. 7 or SEQ ID NO. 10; a light chain CDR2 region having at least 80% homology to the amino acid sequence set forth in SEQ ID NO. 8 or SEQ ID NO. 11; a light chain CDR3 region having at least 80% homology to the amino acid sequence set forth in SEQ ID NO. 9 or SEQ ID NO. 12.
In some embodiments, the anti-PD-L1 antibody is administered at a dose of 1mg/kg, 2mg/kg, 3mg/kg, 5mg/kg, 6mg/kg, 9mg/kg, 10mg/kg, 15mg/kg, 20mg/kg, 30mg/kg body weight for continuous dosing.
In some embodiments, the anti-PD-L1 antibody is administered in one or more unitary doses that are effective to treat the cancer. In some embodiments, wherein the unitized dose is in the range of about 20mg to about 2000mg of anti-PD-L1 antibody. In some embodiments, wherein the unitized dose is selected from about 300mg, about 600mg, about 900mg, about 1000mg, about 1200mg, about 1500mg, about 1800mg, about 2100mg, or about 2400mg of the anti-PD-L1 antibody. In some embodiments, the unitized dose is selected from about 1200mg of anti-PD-L1 antibody.
In some embodiments, the anti-PD-L1 antibody is administered about every week (q1w), about every 2 weeks (q2w), about every 3 weeks (q3w), or about every 4 weeks (q4 w). In some embodiments, the patient is administered a uniform dose of anti-PD-L1 antibody about every 3 weeks. In some embodiments, the anti-PD-L1 antibody is administered at a dose of 1200mg per patient, approximately once every 3 weeks, for a sustained period.
In some embodiments, the anti-PD-L1 antibody is administered as an intravenous infusion. In some embodiments, the anti-PD-L1 antibody is administered as an intravenous infusion over about 1-2 hours, preferably about 1 hour.
In some embodiments, the anti-PD-L1 antibody is a naked antibody, an intact antibody, or an antibody fragment comprising an antigen binding region.
In some embodiments, the method results in an objective response, preferably a complete response or a partial response.
In some embodiments, the subject has not previously received systemic chemotherapy. In some embodiments, the subject has previously received surgical treatment, radiation therapy, induction chemotherapy, concurrent chemotherapy, and/or adjuvant chemotherapy. In some embodiments, the subject has not previously received systemic chemotherapy, but has received surgical treatment, radiation therapy, induction chemotherapy, concurrent chemotherapy, and/or adjuvant chemotherapy. In some embodiments, the subject has complete remission following surgical treatment, radiation therapy, induction chemotherapy, concurrent chemotherapy, and/or adjuvant chemotherapy before disease progression occurs again. In some embodiments, the subject has failed to complete remission or failed to partial remission following surgical treatment, radiation therapy, induction chemotherapy, concurrent chemotherapy, and/or adjuvant chemotherapy. In some embodiments, the subject undergoes metastasis following surgical treatment, radiation treatment, induction chemotherapy, concurrent chemotherapy, and/or adjuvant chemotherapy.
In some embodiments, the cancer is squamous cell carcinoma of the head and neck (SCCHN). In some embodiments, the primary site of head and neck squamous cell carcinoma is the oropharynx, oral cavity, hypopharynx, and/or larynx.
In some embodiments, the cancer is recurrent and/or metastatic head and neck squamous cell carcinoma.
In some embodiments, the cancer is recurrent or metastatic head and neck squamous cell carcinoma. In some embodiments, the recurrent or metastatic head and neck squamous cell carcinoma is a recurrent or metastatic head and neck squamous cell carcinoma with no indication of a local radical therapy.
In some embodiments, the head and neck squamous cell carcinoma is recurrent.
In some embodiments, the head and neck squamous cell carcinoma is metastatic.
In some embodiments, the head and neck squamous cell carcinoma is refractory.
In some embodiments, the cancer treatment is a first line treatment for recurrent or metastatic head and neck squamous cell carcinoma.
In some embodiments, the treatment is a first line treatment for recurrent or metastatic head and neck squamous cell carcinoma in combination with standard chemotherapy. In some embodiments, the standard chemotherapy comprises administering a chemotherapeutic agent to the patient. In some embodiments, the standard chemotherapy comprises administering cisplatin and 5-fluorouracil to the patient. In some embodiments, the standard chemotherapy comprises administering carboplatin and 5-fluorouracil to the patient. In some embodiments, the anti-PD-L1 antibody and the chemotherapeutic agent are each in the form of a pharmaceutical composition that can be administered simultaneously, sequentially, or at intervals.
In some embodiments, the treatment is given intravenously at 1200mg of anti-PD-L1 on the first day (D1) of each cycle in a cycle of 3 weeks (21 days), and standard chemotherapy is given after the anti-PD-L1 antibody infusion is complete. In some embodiments, the standard chemotherapy regimen is the first day (D1) intravenous infusion of cisplatin at 75mg/m2Or carboplatin AUC5 by intravenous infusion and 5-fluorouracil 750mg/m by daily intravenous infusion on days one to five (D1-D5)2. In some embodiments, the standard chemotherapy regimen is the first day (D1) intravenous infusion of 75mg/m cisplatin2And 5-Fluorouracil was administered at 750mg/m by daily intravenous infusion on days one to five (D1-D5)2. In some embodiments, the standard chemotherapy regimen is carboplatin AUC5 given by intravenous infusion on day one (D1) and 5-fluorouracil 750mg/m by intravenous infusion daily on days one to five (D1-D5)2
The present application also provides a combination pharmaceutical composition for treating recurrent or metastatic head and neck squamous cell carcinoma comprising an anti-PD-L1 antibody and a chemotherapeutic agent.
In some embodiments, the combination pharmaceutical composition comprises a pharmaceutical composition comprising an anti-PD-L1 antibody and a pharmaceutical composition comprising a chemotherapeutic agent. In some embodiments, the chemotherapeutic agent in the combination pharmaceutical composition may be cisplatin, carboplatin, and/or 5-fluorouracil. In some embodiments, the pharmaceutical composition comprising the anti-PD-L1 antibody is included in a combination pharmaceutical composition, in particular, the anti-PD-L1 antibody is stored in a container in a fixed dose selected from the group consisting of about 300mg, about 600mg, about 900mg, about 1000mg, about 1200mg, about 1500mg, about 1800mg, about 2100mg, and about 2400mg of the anti-PD-L1 antibody.
The present application also provides a kit for treating recurrent or metastatic head and neck squamous cell carcinoma, the kit comprising a pharmaceutical composition of an anti-PD-L1 antibody and a chemotherapeutic agent, and instructions for the use of an anti-PD-L1 antibody in combination with a chemotherapeutic agent for treating recurrent or metastatic head and neck squamous cell carcinoma.
The present application provides an article of manufacture comprising a container containing a fixed dose of an anti-PD-L1 antibody. The present application also provides for the use of an anti-PD-L1 antibody in the manufacture of an article of manufacture for the treatment of cancer comprising a container comprising a fixed dose of an anti-PD-L1 antibody. In some embodiments, the container is a vial. The fixed dose is selected from about 300mg, about 600mg, about 900mg, about 1000mg, about 1200mg, about 1500mg, about 1800mg, about 2100mg, and about 2400mg of the anti-PD-L1 antibody. In some embodiments, the article of manufacture further comprises a package insert or package insert instructing the user to administer the fixed dose to a cancer patient. In some embodiments, the article of manufacture comprises 1 or more than 1 vial containing about 300mg or 600mg of anti-PD-L1 antibody. In some embodiments, the article of manufacture comprises 1 vial containing about 300mg of the anti-PD-L1 antibody. In some embodiments, the article of manufacture comprises 1 vial containing about 600mg of anti-PD-L1 antibody.
This applicationDetailed description and preferred embodiments of the invention
The present application provides a method for treating, ameliorating or improving a subject having a cancer or tumor comprising administering to the subject a therapeutically effective amount of an inhibitor of the interaction between the PD-1 receptor and its ligand PD-L1.
The present application also provides a method for treating, ameliorating or improving a subject having a cancer or tumor, the method comprising administering to the subject a therapeutically effective amount of: an antibody or antigen-binding portion thereof that binds programmed death ligand 1(PD-L1) and/or inhibits PD-L1 activity.
The present application also provides a monotherapy or a combination therapy for treating, ameliorating or ameliorating a subject having a cancer or tumor, the method comprising administering to the subject a therapeutically effective amount of: an antibody or antigen-binding portion thereof that binds programmed death ligand 1(PD-L1) and/or inhibits PD-L1 activity.
The present application also provides a method of treating, ameliorating or improving a subject having a cancer or tumor that is squamous cell carcinoma of the head and neck (SCCHN), comprising: (i) measuring the level of PD-L1 in a sample from the subject, wherein the subject is PD-L1 positive, and (ii) administering to the subject a therapeutically effective amount of an anti-PD-L1 antibody or antigen-binding portion thereof.
The present application provides a method for treating, ameliorating or improving a subject having a cancer or tumor. In certain embodiments, the subject is a patient that has been histopathologically diagnosed as head and neck Squamous Cell Carcinoma (SCCHN). For patients with cancer as described herein, the primary site of squamous cell carcinoma of the head and neck may be oropharynx, oral cavity, hypopharynx, or larynx. The patient's cancer may be refractory, recurrent or metastatic head and neck squamous cell carcinoma, e.g., in certain patients, whose head and neck squamous cell carcinoma is recurrent; in certain patients, the head and neck squamous cell carcinoma is metastatic; in certain patients, squamous cell carcinoma of the head and neck is refractory. In some embodiments, the cancer is recurrent and/or metastatic head and neck squamous cell carcinoma. In some embodiments, the squamous cell carcinoma of the head and neck is recurrent SCCHN with no indication of a curative partial therapy. In some embodiments, the squamous cell carcinoma of the head and neck is metastatic SCCHN with no indication of a curative partial therapy.
In some embodiments, the subject has previously received surgery, chemotherapy, and/or radiation therapy. In some embodiments, the subject has re-developed disease progression after achieving complete remission following surgery, chemotherapy, and/or radiation therapy. In some embodiments, the subject has failed to complete remission or failed to partial remission following surgery, chemotherapy, and/or radiation therapy.
In some aspects of the present application, the head and neck squamous cell carcinoma patient may be a patient who has not previously received systemic chemotherapy. In some versions of the present application, a patient with head and neck squamous cell carcinoma has previously received surgery, radiation therapy, induction chemotherapy, concurrent chemotherapy, and/or adjuvant chemotherapy. In some embodiments, the head and neck squamous cell carcinoma patient is a patient who has not previously received systemic chemotherapy, but who has received surgical treatment, radiation therapy, induction chemotherapy, concurrent chemotherapy, and/or adjuvant chemotherapy. In some embodiments of the present application, the head and neck squamous cell carcinoma patient is a patient who experiences disease progression again after complete remission following surgical treatment, radiation therapy, induced chemotherapy, concurrent chemotherapy, and/or adjuvant chemotherapy. In some embodiments of the present application, the head and neck squamous cell carcinoma patient is a patient who has failed to complete remission or to partial remission following surgical treatment, radiation therapy, induced chemotherapy, concurrent chemotherapy, and/or adjuvant chemotherapy. In some embodiments of the present application, the cancer of the patient has metastasized.
In some embodiments of the present application, the treatment is a first line treatment for recurrent or metastatic head and neck squamous cell carcinoma. In some embodiments of the present application, the treatment is a first line treatment for recurrent or metastatic head and neck squamous cell carcinoma in combination with standard chemotherapy. The standard chemotherapy comprises administering a chemotherapeutic agent to the patient. In some embodiments, the chemotherapeutic agent comprises a platinum-containing chemotherapeutic drug.In some embodiments, the standard chemotherapy comprises administering cisplatin and 5-fluorouracil to a patient, or administering carboplatin and 5-fluorouracil to a patient. In some embodiments, the standard chemotherapy comprises platinum-containing duplex chemotherapy of the patient. In some embodiments, the anti-PD-L1 antibody and the chemotherapeutic agent are each in the form of a pharmaceutical composition that can be administered simultaneously, sequentially, or at intervals. In some embodiments, the treatment is administered to the patient in a 3 week (21 day) cycle, with each cycle giving a uniform dose of about 300mg, about 600mg, about 900mg, about 1000mg, about 1200mg, about 1500mg, about 1800mg, about 2100mg, or about 2400mg of the anti-PD-L1 antibody. In some specific embodiments, the treatment is administered in a cycle of 3 weeks (21 days), 300mg, about 600mg, about 900mg, about 1000mg, about 1200mg, about 1500mg, about 1800mg, about 2100mg, or about 2400mg of the anti-PD-L1 antibody intravenously the first day (D1) of each cycle, followed by infusion of the anti-PD-L1 antibody by standard chemotherapy regimen. In other specific embodiments, the treatment is performed in a cycle of 3 weeks (21 days), with approximately 1200mg of the anti-PD-L1 antibody administered intravenously the first day (D1) of each cycle, followed by infusion of the anti-PD-L1 antibody by a standard chemotherapy regimen. In some embodiments, the standard chemotherapeutic regimen comprises: the first day (D1) cisplatin was administered by intravenous infusion at 75-100mg/m2Or carboplatin AUC5 was administered by intravenous infusion and 5-fluorouracil 750mg/m 1000mg/m was administered by intravenous infusion daily on days one to five (D1-D5)2. In some embodiments, the standard chemotherapeutic regimen comprises: the first day (D1) cisplatin was administered by intravenous infusion at 75-100mg/m2And 5-fluorouracil 750-1000mg/m was administered by intravenous infusion daily on the first to fifth days (D1-D5)2. In some embodiments, the standard chemotherapeutic regimen comprises: carboplatin AUC5 was given by intravenous infusion on day one (D1), and 5-fluorouracil 750-1000mg/m was given by intravenous infusion daily on day one to day five (D1-D5)2. Wherein the intravenous infusion administration of cisplatin is 75-100mg/m2Optionally intravenous infusion of 75mg/m2、80mg/m 2、85mg/m 2、90mg/m 2Or 100mg/m2Preferably 75mg/m by intravenous infusion2Or by intravenous infusion at 100mg/m2(ii) a The intravenous infusion was given 5-fluorouracil 750-1000mg/m2Optionally, intravenous infusion of 750mg/m2、800mg/m 2、850mg/m 2、900mg/m 2Or 1000mg/m2Preferably 750mg/m for intravenous infusion2Or by intravenous infusion at 1000mg/m2
The present application also provides combination therapy for first-line treatment of subjects with recurrent or metastatic head and neck squamous cell carcinoma, wherein the patient receives an anti-PD-L1 antibody in combination with a standard chemotherapy regimen, 1 cycle for 3 weeks (21 days), 1200mg of anti-PD-L1 antibody is administered intravenously the first day (D1) of each cycle, and after completion of the infusion of anti-PD-L1 antibody, administration of the standard chemotherapy regimen is initiated: the first day of each cycle (D1) cisplatin was infused intravenously at 75mg/m25-Fluorouracil 750mg/m intravenous infusion on days one to five (D1-D5) of each cycle2/d。
The present application also provides combination therapy for first-line treatment of subjects with recurrent or metastatic head and neck squamous cell carcinoma, wherein the patient receives an anti-PD-L1 antibody in combination with a standard chemotherapy regimen, 1 cycle for 3 weeks (21 days), 1200mg of anti-PD-L1 antibody is administered intravenously the first day (D1) of each cycle, and after completion of the infusion of anti-PD-L1 antibody, administration of the standard chemotherapy regimen is initiated: carboplatin AUC5 was intravenously infused on the first day of each cycle (D1), and 5-fluorouracil 750mg/m was intravenously infused from the first to fifth days of each cycle (D1-D5)2/d。
The present application also provides a combination pharmaceutical composition for treating recurrent or metastatic head and neck squamous cell carcinoma comprising an anti-PD-L1 antibody and a chemotherapeutic agent. In some embodiments, the combination pharmaceutical composition comprises a pharmaceutical composition comprising an anti-PD-L1 antibody and a pharmaceutical composition comprising a chemotherapeutic agent. In some embodiments, the chemotherapeutic agent comprises a platinum-containing chemotherapeutic drug. In some embodiments, the chemotherapeutic agent in the combination pharmaceutical composition can be cisplatin, carboplatin, and/or 5-fluorouracil. In some embodiments, the pharmaceutical composition comprising the anti-PD-L1 antibody is included in a combination pharmaceutical composition, in particular, the anti-PD-L1 antibody is stored in a container in a fixed dose selected from the group consisting of about 300mg, about 600mg, about 900mg, about 1000mg, about 1200mg, about 1500mg, about 1800mg, about 2100mg, and about 2400mg of the anti-PD-L1 antibody. In some embodiments, the combination pharmaceutical composition comprises about 300mg, about 600mg, about 900mg, or about 1200mg of the anti-PD-L1 antibody, and a therapeutically effective amount of cisplatin and 5-fluorouracil. In some embodiments, the combination pharmaceutical composition comprises about 300mg, about 600mg, or about 1200mg of the anti-PD-L1 antibody, and a therapeutically effective amount of carboplatin and 5-fluorouracil.
The present application also provides a method for identifying a subject having head and neck squamous cell carcinoma who is suitable for anti-PD-L1 antibody therapy, the method comprising measuring the level of PD-L1 in a sample from the subject, and wherein a therapeutically effective amount of an anti-PD-L1 antibody, or antigen-binding portion thereof, is administered to the subject.
The present application also provides a kit for treating a subject having head and neck squamous cell carcinoma, the kit comprising: (a) an anti-PD-L1 antibody or antigen-binding portion thereof; (b) instructions for treating the tumor.
The present application provides methods of treating cancer using one or more immune checkpoint inhibitors (e.g., an anti-PD-L1 antibody or antigen-binding portion thereof, or an anti-PD-1 antibody or antigen-binding portion thereof). In one embodiment, the cancer is a primary cancer. In one embodiment, the cancer is metastatic or recurrent cancer. In one embodiment, the cancer is a relapsed or refractory cancer. In one embodiment, the cancer is head and neck squamous cell carcinoma. In one embodiment, the cancer is recurrent or metastatic head and neck squamous cell carcinoma.
The present application also provides for the use of an anti-PD-L1 antibody in the first line treatment of squamous cell carcinoma of the head and neck. The present application also provides the use of an anti-PD-L1 antibody in a second line treatment of squamous cell carcinoma of the head and neck. The application also provides the use of an anti-PD-L1 antibody in the three-line treatment of squamous cell carcinoma of the head and neck. In some embodiments, the anti-PD-L1 antibody is used for first line treatment of recurrent or metastatic head and neck squamous cell carcinoma.
In certain embodiments, the subject is a human patient. In certain embodiments, the subject has received another cancer treatment (e.g., surgical treatment, radiation treatment, induction chemotherapy, concurrent chemotherapy, and/or adjuvant chemotherapy), but is resistant or refractory to such another cancer treatment. In certain embodiments, the subject is a human patient. In certain embodiments, the subject has received another cancer treatment (e.g., surgical treatment, radiation treatment, induction chemotherapy, concurrent chemotherapy, and/or adjuvant chemotherapy), but is post-relapsed, or metastatic, to such another cancer treatment.
In certain embodiments, the present application provides a method for treating a subject having a tumor, the method comprising administering to the subject a therapeutically effective amount of an immune checkpoint inhibitor, e.g., an anti-PD-L1 antibody. In certain embodiments, the present application relates to a method of treating a subject having a tumor that is a recurrent or metastatic squamous cell carcinoma of the head and neck, comprising: (i) measuring the level of PD-L1 in a sample of the subject, wherein the subject expresses PD-L1, and (ii) administering to the subject a therapeutically effective amount of an immune checkpoint inhibitor, e.g., an anti-PD-L1 antibody or an antigen-binding portion thereof. In certain embodiments, the anti-PD-L1 antibody is 13C5, 5G11, ch13C5-hIgG1, ch13C5-hIgG4, ch5G11-hIgG1, ch5G11-hIgG4, hu13C5-hIgG1, hu13C5-hIgG4, hu5G11-hIgG1, or hu5G11-hIgG4 mab. In other embodiments, the anti-PD-L1 antibody competes for binding with 5G11 mab.
In some embodiments, the cancer or tumor expresses PD-L1. The PD-L1 expression level of a tumor in a subject can be measured prior to administration of any composition or using any method disclosed herein. In one embodiment, the tumor has a PD-L1 expression level of at least about 0.5%, at least about 0.6%, at least about 0.7%, at least about 0.8%, at least about 0.9%, at least about 1%, at least about 2%, at least about 3%, at least about 4%, at least about 5%, at least about 6%, at least about 7%, at least about 8%, at least about 9%, at least about 10%, at least about 11%, at least about 12%, at least about 13%, at least about 14%, at least about 15%, at least about 20%, or greater than at least about 20%. In another embodiment, the tumor has a PD-L1 expression level of at least about 1%. In other embodiments, the subject has a PD-L1 expression level of at least about 5%. In a particular embodiment, the tumor has a PD-L1 expression level of at least about 10%. Measurement of PD-L1 expression levels can be performed using methods such as antibodies, in situ mRNA hybridization, automated IHC, and the like.
The present application provides a method of treating, ameliorating, or ameliorating a subject having a cancer or tumor, the method comprising administering to the subject a therapeutically effective amount of an anti-PD-L1 antibody, or antigen-binding portion thereof, wherein a tumor sample obtained from the patient has been determined to have a detectable expression level of PD-L1 in about 1% or more (e.g., about 1%, about 2%, about 3%, or about 4% or more) of the cells. In some embodiments, the tumor sample obtained from the patient has been determined to have a detectable expression level of PD-L1 in cells comprising about 1% to about 65% or more (e.g., about 1% to about 5%, about 5% to about 10%, about 10% to about 20%, about 20% to about 30%, about 30% to about 40%, about 40% to about 50%, or about 50% to about 65%). The present application also provides a method of determining whether a patient suffering from head and neck squamous cell carcinoma is likely to respond to treatment with a therapeutic agent comprising an anti-PD-L1 antibody, the method comprising determining the expression level of PD-L1 in tumor cells in a tumor sample obtained from the patient, wherein a detectable expression level of PD-L1 in cells comprising about 1% or more of the tumor sample indicates that the patient is likely to respond to treatment with a therapeutic agent comprising an anti-PD-L1 antibody. The present application also provides a method for predicting responsiveness of a patient suffering from head and neck squamous cell carcinoma to treatment with a therapeutic agent comprising an anti-PD-L1 antibody, the method comprising determining expression levels of PD-L1 in cells in a tumor sample obtained from the patient, wherein a detectable expression level of PD-L1 in tumor cells comprising about 1% or more of the tumor sample indicates that the patient is likely to respond to treatment with a therapeutic agent comprising an anti-PD-L1 antibody. The present application also provides a method for selecting a therapy for a patient suffering from head and neck squamous cell carcinoma, the method comprising determining the expression level of PD-L1 in tumor cells in a tumor sample obtained from the patient, and selecting a therapy for the patient comprising a therapeutic agent that is an anti-PD-L1 antibody based on the detectable expression level of PD-L1 in tumor cells comprising about 1% or more of the tumor sample. In some embodiments, the tumor sample obtained from the patient has been determined to have a detectable expression level of PD-L1 in tumor cells comprising about 5% or more of the tumor sample. In some embodiments, the tumor sample obtained from the patient has been determined to have a detectable expression level of PD-L1 in tumor cells comprising at least about 10% of the tumor sample. The present application also provides a method for determining whether a patient suffering from head and neck squamous cell carcinoma is likely to respond to treatment with a therapeutic agent comprising an anti-PD-L1 antibody or antigen-binding portion thereof, the method comprising determining a subtype of a tumor obtained from the patient, wherein a relapsed and/or metastatic head and neck squamous cell carcinoma indicates that the patient is likely to respond to treatment with a therapeutic agent comprising an anti-PD-L1 antibody. The present application provides a method for predicting responsiveness of a patient suffering from head and neck squamous cell carcinoma to treatment with a therapeutic agent comprising an anti-PD-L1 antibody, the method comprising determining a subtype of a tumor obtained from the patient, wherein a relapsed and/or metastatic head and neck squamous cell carcinoma indicates that the patient is likely to respond to treatment with a therapeutic agent comprising an anti-PD-L1 antibody. The present application provides a method for selecting a therapy for a patient suffering from head and neck squamous cell carcinoma, the method comprising determining a subtype of a tumor obtained from the patient from the tumor sample, and selecting a therapy for the patient comprising a therapeutic agent for an anti-PD-L1 antibody based on the tumor determining that the head and neck squamous cell carcinoma is recurrent and/or metastatic. In some embodiments, the method further comprises administering to the patient a therapeutically effective amount of a therapeutic agent that is an anti-PD-L1 antibody based on the expression level of PD-L1 in tumor cells in the tumor sample.
In certain embodiments, the expression level of the molecular marker in a tumor sample or blood tissue sample obtained from the patient has been determined to be altered, e.g., increased, relative to a reference level of the at least one gene. In other embodiments, the expression level of microrna in the tumor sample obtained from the patient has been determined to be altered relative to a reference level of the microrna, in some embodiments, the alteration is a decrease in the expression level. In some embodiments, the reference level is a non-tumor sample or a blood tissue sample of the non-diseased subject as a reference. In some embodiments, the reference level is a non-tumor sample of the patient as a reference.
In certain embodiments, the level of genetic mutation, level of genetic modification, level of transcription, and/or level of expression of at least one of PD-L1, TMB, MSI, 9p24.1 in a tumor sample or blood tissue sample obtained from the patient has been determined to be altered relative to a reference level of the at least one gene. In some embodiments, the reference level is a non-tumor sample or a blood tissue sample of the non-diseased subject as a reference. In some embodiments, the reference level is a non-tumor sample of the patient as a reference.
In certain embodiments, the objective response rate of a subject administered a therapeutically effective amount of an anti-PD-L1 antibody is between about 10% to about 40% (e.g., about 10% to about 20%, about 20% to about 30%, about 30% to about 40%). In yet another embodiment, the objective response rate of a patient administered a therapeutically effective amount of an anti-PD-L1 antibody, or antigen-binding portion thereof, is between about 15% to about 25%. In other embodiments, the objective response rate of a patient administered a therapeutically effective amount of a therapeutic agent against a PD-L1 antibody is at least about 15%. In other embodiments, the objective response rate of a patient administered a therapeutically effective amount of a therapeutic agent against a PD-L1 antibody is at least about 20%.
In certain embodiments, the therapies of the present application (e.g., anti-PD-L1 antibodies) are effective to increase the duration of survival of a subject. In certain embodiments, the anti-PD-L1 antibody therapy of the present application increases the duration of survival of a subject compared to standard of care therapy. In certain embodiments, the therapies of the present application increase the overall survival of the subject. In certain embodiments, the subject exhibits an overall survival of at least about 6 months, 7 months, 8 months, 9 months, 10 months, at least about 11 months, at least about 12 months, at least about 13 months, at least about 14 months, at least about 15 months, at least about 16 months, at least about 17 months, at least about 18 months, at least about 19 months, at least about 20 months, at least about 21 months, at least about 22 months, at least about 23 months, at least about 2 years, at least about 3 years, at least about 4 years, or at least about 5 years after administration. In certain embodiments, the survival or duration of total survival of a subject is increased by 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 40%, at least about 50%, or at least about 75% when compared to another subject treated with standard of care therapy alone. In other embodiments, the survival or duration of total survival of a subject is increased by at least about 1 month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 6 months, at least about 1 year, at least about 18 months, at least about 2 years, at least about 3 years, at least about 4 years, or at least about 5 years when compared to another subject treated with standard of care therapy alone.
In certain embodiments, the therapies of the present application are effective to increase the duration of progression-free survival of a subject. For example, progression free survival of a subject is increased by at least about 2 weeks, at least about 1 month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 6 months, or at least about 1 year when compared to another subject treated with standard of care therapy alone. In certain embodiments, after administration of the anti-PD-L1 antibody therapy, the subject exhibits a total response rate that is at least about 30%, 35%, 36%, 37%, 39%, 40%, 45%, or 50% higher than the response rate after administration of the standard of care therapy.
Immune checkpoint inhibitors suitable for use in the disclosed methods include anti-PD-L1 antibodies that bind to PD-L1 with high specificity and affinity, block the binding of PD-L1, and inhibit the immunosuppressive effects of the PD-1 signaling pathway. In any of the methods of treatment disclosed herein, an anti-PD-1 or anti-PD-L1 "antibody" includes an antigen-binding moiety that binds to the PD-1 or PD-L1 receptor, respectively, and exhibits functional properties in inhibiting ligand binding and upregulating the immune system similar to the functional properties of an intact antibody. In some embodiments, the anti-PD-1 antibody, anti-PD-L1 antibody, or antigen-binding portion thereof is a chimeric, humanized, or human monoclonal antibody, or portion thereof. In certain embodiments for treating a human subject, the antibody is a humanized antibody. In other embodiments for treating a human subject, the antibody is a human antibody. Antibodies of the IgG1, IgG2, IgG3 or IgG4 isotype may be used.
In certain embodiments, the anti-PD-1 antibody, anti-PD-L1 antibody, or antigen-binding portion thereof, comprises a heavy chain constant region of human IgG1 or IgG4 isotype. In certain other embodiments, the sequence of the IgG4 heavy chain constant region of the anti-PD-1 antibody, anti-PD-L1 antibody, or antigen-binding portion thereof, contains the S228P mutation that replaces a serine residue in the hinge region with a proline residue typically found at the corresponding position of an IgG1 isotype antibody. This mutation present in the monoclonal antibody prevents Fab arm exchange with endogenous IgG4 antibody while retaining low affinity for activation of the Fc receptor associated with wild-type IgG4 antibody (Wang et al, 2014Cancer Immunol res.2(9): 846-56). In other embodiments, the antibody comprises a light chain constant region that is a human kappa or lambda constant region. In other embodiments, the anti-PD-1 antibody, anti-PD-L1 antibody, or antigen-binding portion thereof is a mAb or antigen-binding portion thereof.
In WO2016022630, it has been disclosed that an anti-PD-L1 antibody has high affinity for PD-L1, can significantly inhibit the interaction of PD-L1 and PD-1 on the cell surface, and significantly promote the secretion of IL-2 and IFN-gamma by T cells.
In certain embodiments, the anti-PD-L1 antibody or fragment thereof cross-competes with 5G11 or 13C5 mab. In other embodiments, the anti-PD-L1 antibody or fragment thereof binds to the same or a similar epitope as the 5G11 or 13C5 mab. In certain embodiments, the anti-PD-L1 antibody has the same CDRs as the 5G11 or 13C5 mab.
In certain embodiments, the anti-PD-L1 antibody or fragment thereof cross-competes with 5G11 or 13C5 mab for binding to the same epitope region of human PD-L1. For administration to a human subject, these cross-competitive antibodies are chimeric or humanized or human antibodies. Such chimeric, humanized or human mabs may be prepared and isolated by methods well known in the art.
anti-PD-L1 antibodies useful in the disclosed inventive methods also include antigen-binding portions of the above antibodies. It is well established that the antigen binding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed by the term "antigen-binding portion" of an antibody include: (i) fab fragments, i.e., monovalent fragments consisting of the VL, VH, CL and CH1 domains; (ii) a F (ab')2 fragment, i.e. a bivalent fragment comprising 2 Fab fragments linked by a disulfide bond at the hinge region; (iii) an Fd fragment consisting of the VH and CH1 domains; and (iv) Fv fragments (including, e.g., scFv) consisting of VL and VH domains of a single arm of an antibody.
An anti-PD-L1 antibody suitable for use in the disclosed compositions is an antibody that binds to PD-L1 with high specificity and affinity, blocks the binding of PD-1, and inhibits the immunosuppressive effects of the PD-L1/PD-1 signaling pathway. In any of the compositions or methods disclosed herein, an anti-PD-L1 "antibody" includes an antigen-binding portion or fragment that binds to a PD-L1 ligand and exhibits functional properties similar to those of an intact antibody in inhibiting receptor binding and upregulating the immune system. In certain embodiments, the anti-PD-L1 antibody or antigen-binding portion thereof cross-competes with 5G11 or 13C5 mab for binding to human PD-L1. In other embodiments, the anti-PD-L1 antibody or antigen-binding portion thereof is a chimeric, humanized, or human monoclonal antibody or portion thereof. In certain embodiments, the antibody is a humanized antibody. In other embodiments, the antibody is a human antibody. Antibodies of the IgG1, IgG2, IgG3 or IgG4 isotype may be used.
In certain embodiments, the anti-PD-L1 antibody used in the methods may be replaced with another anti-PD-1 antagonist or an anti-PD-L1 antagonist. For example, because anti-PD-1 antibodies prevent the interaction between PD-1 and PD-L1, thereby exerting a similar effect on the signaling pathway of PD-1, anti-PD-1 antibodies may replace the use of anti-PD-L1 antibodies in the methods disclosed herein. Thus, in one embodiment, the present application relates to a method for treating a subject having a tumor that is recurrent or metastatic squamous cell carcinoma of the head and neck, comprising administering to the subject a therapeutically effective amount of an anti-PD-L1 antibody.
In certain embodiments, the anti-PD-L1 antibody or antigen-binding fragment thereof is a 13C5, 5G11, ch13C5-hIgG1, ch13C5-hIgG4, ch5G11-hIgG1, ch5G11-hIgG4, hu13C5-hIgG1, hu13C5-hIgG4, hu5G11-hIgG1, or hu5G11-hIgG4 monoclonal antibody or antigen-binding fragment thereof (see WO2016022630 or CN 107001463A).
The present application provides antigen binding polypeptides or pharmaceutical compositions comprising antigen binding polypeptides for the treatment of cancer or tumors. In some embodiments, the antigen binding polypeptide is an antibody or antigen binding portion thereof that binds programmed death ligand 1(PD-L1) and/or inhibits PD-L1 activity. In some embodiments, the antigen binding polypeptide is an anti-PD-L1 antibody.
The present application provides an isolated antibody or fragment thereof that binds PD-L1, wherein the antibody can be produced by a hybridoma selected from the group consisting of the hybridomas designated herein as 13C5, 5G 11. Accordingly, the present application also includes hybridomas 13C5, 5G11, and any hybridoma that produces an antibody disclosed herein. The present application also provides isolated polynucleotides encoding the antibodies and fragments thereof provided herein. The present application also includes expression vectors comprising the isolated polynucleotides, and host cells comprising the expression vectors.
The present application provides an anti-PD-L1 antibody comprising heavy chain Complementarity Determining Regions (CDRs) selected from the group consisting of the 13C5 or 5G11 antibodies, and light chain complementarity determining regions selected from the group consisting of the 13C5 or 5G11 antibodies. In one embodiment, the application provides an anti-PD-L1 antibody comprising a variable heavy chain selected from a ch5G11-hIgG1, ch5G11-hIgG4, ch13C5-hIgG1, ch13C5-hIgG4 chimeric antibody, and a variable light chain selected from a ch5G11-hIgG1, ch5G11-hIgG4, ch13C5-hIgG1, ch13C5-hIgG4 chimeric antibody. In one embodiment, the present application provides an anti-PD-L1 antibody comprising a variable heavy chain selected from a hu13C5-hIgG1, hu13C5-hIgG4, hu5G11-hIgG1, or hu5G11-hIgG4 humanized antibody, and a variable light chain selected from a hu13C5-hIgG1, hu13C5-hIgG4, hu5G11-hIgG1, or hu5G11-hIgG4 humanized antibody. Reference may be made to the description of patent documents WO2016022630 or CN 107001463A: 13C5, ch13C5-hIgG1, ch13C5-hIgG4, hu13C5-hIgG1, or hu13C5-hIgG4 has the HCDR1 sequence SYGMS (SEQ ID NO:4), the HCDR2 sequence SISSGGSTYYPDSVKG (SEQ ID NO:5), the HCDR3 sequence GYDSGFAY (SEQ ID NO:6), the LCDR1 sequence ASQSVSTSSSSFMH (SEQ ID NO:10), the LCDR2 sequence YASNLES (SEQ ID NO:11), and the LCDR3 sequence QHSWEIPYT (SEQ ID NO: 12); the sequence of HCDR1 of 5G11, ch5G11-hIgG1, ch5G11-hIgG4, hu5G11-hIgG1 or hu5G11-hIgG4 is TYGVH (SEQ ID NO:1), the sequence of HCDR2 is VIWRGVTTDYNAAFMS (SEQ ID NO:2), the sequence of HCDR3 is LGFYAMDY (SEQ ID NO:3), the sequence of LCDR1 is KASQSVSNDVA (SEQ ID NO:7), the sequence of LCDR2 is YAANRYT (SEQ ID NO:8), and the sequence of LCDR3 is QQDYTSPYT (SEQ ID NO: 9).
In certain embodiments, an isolated anti-PD-L1 antibody described herein comprises: a heavy chain CDR1 region having the amino acid sequence shown in SEQ ID NO.1, a heavy chain CDR2 region having the amino acid sequence shown in SEQ ID NO. 2, a heavy chain CDR3 region having the amino acid sequence shown in SEQ ID NO. 3; and a light chain CDR1 region having the amino acid sequence shown in SEQ ID NO. 7, a light chain CDR2 region having the amino acid sequence shown in SEQ ID NO. 8, and a light chain CDR3 region having the amino acid sequence shown in SEQ ID NO. 9.
In certain embodiments, an immune checkpoint inhibitor (e.g., an anti-PD-L1 antagonist) for use herein is a PD-L1Fc fusion protein.
In certain embodiments, the application administers to a subject a therapeutically effective amount of an anti-PD-L1 antibody, wherein the anti-PD-L1 antibody is administered alone. In some embodiments, the separate administration refers to that the anti-PD-L1 antibody may not be used in combination with other anti-cancer drugs, and/or not be administered simultaneously with other anti-cancer drugs. In some embodiments, the separate administration refers to the anti-PD-L1 antibody may be administered without a chemotherapeutic agent in combination with, and/or concurrently with, the chemotherapeutic agent. In some embodiments, the separate administration refers to that the anti-PD-L1 antibody may not be used in combination with, and/or not administered simultaneously with, other targeted drugs. In some embodiments, the separate administration refers to the anti-PD-L1 antibody may not be administered in combination with, and/or concurrently with, other anti-cancer antibodies. In some embodiments, the separate administration refers to that the anti-PD-L1 antibody may not be administered in combination with, and/or concurrently with, radiation therapy.
In certain embodiments, the application administers to a subject a therapeutically effective amount of an anti-PD-L1 antibody, wherein the anti-PD-L1 antibody is co-administered.
In certain embodiments, the immune checkpoint inhibitor (e.g., an anti-PD-1 antibody or an anti-PD-L1 antibody) is administered in combination with one or more other anti-cancer drugs. In certain embodiments, the one or more anti-cancer agents have been administered to the subject prior to administration of the anti-PD-1 antibody or anti-PD-L1 antibody or prior to combination with the anti-PD-1 antibody or anti-PD-L1 antibody. In certain embodiments, the one or more anti-cancer agents are not effective in treating the cancer. In certain embodiments, the additional anti-cancer agent is any anti-cancer agent described herein or known in the art.
In certain embodiments, an anti-PD-1 antibody or an anti-PD-L1 antibody can be combined with another immunotherapy. In certain embodiments, immunotherapy involving the blockade of immune checkpoints is administered as monotherapy. In other embodiments, immunotherapy involving the blockade of immune checkpoints is administered in combination with other therapies.
The therapeutic agents of the present application can be formulated in a composition, for example, a pharmaceutical composition comprising an antibody and a pharmaceutically acceptable carrier. As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, which are physiologically compatible. In one embodiment, the carrier for the antibody-containing composition is suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal, or epidermal administration (e.g., by injection or infusion), while the carrier for the TKI (tyrosine kinase inhibitor) -containing composition is suitable for non-parenteral (e.g., oral) administration. The pharmaceutical compositions of the present application may include one or more pharmaceutically acceptable salts, antioxidants, aqueous and non-aqueous carriers, and/or adjuvants such as preservatives, wetting agents, emulsifying agents, and dispersing agents.
Dosage regimens are adjusted to provide the most desirable response, e.g., the maximum therapeutic response and/or the minimum adverse effects. In certain embodiments, the methods of the present application can be used with a uniform dose or a weight-based dose. In other embodiments, the anti-PD-1 antibody, anti-PD-L1 antibody, or antigen-binding portion thereof, is administered as a unitary dose. In other embodiments, the anti-PD-1 antibody, anti-PD-L1 antibody, or antigen-binding portion thereof, is administered as a weight-based dose. For administration of the anti-PD-L1 antibody (as monotherapy or in combination with another anti-cancer agent), the dose may be in the following range: from about 0.01 to about 40mg/kg, from about 0.1 to about 30mg/kg, from about 0.1 to about 20mg/kg, from about 0.1 to about 15mg/kg, from about 0.1 to about 10mg/kg, from about 1 to about 15mg/kg, from about 1 to about 20mg/kg, from about 1 to about 3mg/kg, from about 3 to about 10mg/kg, from about 3 to about 15mg/kg, from about 3 to about 20mg/kg, from about 3 to about 30mg/kg, from about 10 to about 20mg/kg, or from about 15 to about 20mg/kg of the body weight, or from about 60mg to at least about 2400mg, from about 90mg to at least about 1800mg, from about 120mg to at least about 1500mg, from about 300mg to at least about 9000mg, from about 600mg to at least about 900mg, from about 300mg to at least about 1200mg, from about 600mg to at least 1200mg, or from about 900mg to at least 1200 mg. For example, the dose may be about 0.1, about 1, about 2, about 3, about 5, about 6, about 9, about 10, about 15, about 20, or about 30mg/kg body weight; or about 30mg, about 60mg, about 120mg, about 150mg, about 180mg, about 300mg, about 600mg, about 900mg, about 1200mg, about 1800mg, about 2100mg, or about 2400 mg. Dosing schedules are generally designed to achieve exposures that result in sustained Receptor Occupancy (RO) (based on the typical pharmacokinetic properties of antibodies). One exemplary treatment regimen entails administration about once per week (q1w), about once every 2 weeks (q2w), about once every 3 weeks (q3w), about once every 4 weeks (q4w), about once every 1 month (q1m), about once every 3-6 months or longer. In certain embodiments, an anti-PD-L1 antibody, such as 13C5, ch13C5-hIgG1, ch13C5-hIgG4, hu13C5-hIgG1, hu13C5-hIgG4, 5G11, ch5G11-hIgG1, ch5G11-hIgG4, hu5G11-hIgG1, or hu5G11-hIgG4 monoclonal antibody, is administered to the subject about once every 2 weeks. In other embodiments, the antibody is administered about once every 3 weeks. The dosage and schedule may vary during the course of treatment. Given that IgG4 antibodies typically have a half-life of 2-3 weeks, the dosage regimen of the anti-PD-L1 antibodies of the present application comprises at least about 1 to at least about 30mg/kg body weight, at least about 3 to at least about 20mg/kg body weight, at least about 10 to at least about 15mg/kg body weight, or at least about 300 to at least about 1200mg via intravenous administration of the antibody every about 14-21 days in up to about 6 weeks or about 12 weeks of the cycle until complete response or confirmation of progressive disease. In certain embodiments, anti-PD-L1 monotherapy is administered at 3mg/kg every 2 weeks until progressive disease or unacceptable toxicity. In other embodiments, anti-PD-L1 monotherapy is administered at 1200mg every 3 weeks until progressive disease or unacceptable toxicity. In certain embodiments, the antibody treatment 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-L1 antibody can be reduced relative to a monotherapy dose. The dose of 13C5, ch13C5-hIgG1, ch13C5-hIgG4, hu13C5-hIgG1, hu13C5-hIgG4, 5G11, ch5G11-hIgG1, ch5G11-hIgG4, hu5G11-hIgG1 or hu5G11-hIgG4 monoclonal antibodies lower than the typical 20mg/kg but not less than 0.001mg/kg is a sub-therapeutic dose. Sub-therapeutic doses of anti-PD-L1 antibody used in the methods herein are above 0.001mg/kg and below 20 mg/kg. In certain embodiments, the subtherapeutic dose is from about 0.001mg/kg to about 3mg/kg, from about 0.01mg/kg to about 3mg/kg, from about 0.001mg/kg to about 10mg/kg, or from about 0.01mg/kg to about 10mg/kg of body weight. In certain embodiments, the subtherapeutic dose is at least about 0.001mg/kg, at least about 0.005mg/kg, at least about 0.01mg/kg, at least about 0.05mg/kg, at least about 0.1mg/kg, at least about 0.5mg/kg, at least about 1.0mg/kg body weight, or at least about 3.0mg/kg body weight. In certain embodiments, the sub-therapeutic unitized dose is less than about 600mg every 3 weeks, e.g., about 300mg or about 120mg every 3 weeks. In certain embodiments, 3mg/kg administration may allow sufficient exposure to result in maximal biological activity.
In certain embodiments, the dose of the anti-PD-L1 antibody or anti-PD-1 antibody is a fixed dose in a pharmaceutical composition. In other embodiments, the methods of the present application can be used in a uniform dose (a dose administered to a patient without regard to the patient's weight). For example, a uniform dose of 13C5, ch13C5-hIgG1, ch13C5-hIgG4, hu13C5-hIgG1, hu13C5-hIgG4, 5G11, ch5G11-hIgG1, ch5G11-hIgG4, hu5G11-hIgG1, or hu5G11-hIgG4 mAb may be about 1200 mg. In certain embodiments, the anti-PD-L1 antibody, or antigen-binding portion thereof, is administered at a dose of about 1200 mg. In certain embodiments, the anti-PD-L1 antibody, or antigen-binding portion thereof, is administered at a dose of about 900 mg. In certain embodiments, the anti-PD-L1 antibody, or antigen-binding portion thereof, is administered at a dose of about 600 mg. In one embodiment, 900mg of the anti-PD-L1 antibody or antigen-binding fragment is administered once every 3 weeks. In another embodiment, 1200mg of the anti-PD-L1 antibody or antigen-binding fragment is administered once every 4 weeks.
For administration of the anti-PD-L1 antibody (as monotherapy or in combination with another anti-cancer agent), the dose may be in the following range: from about 0.01 to about 20mg/kg, from about 0.1 to about 10mg/kg, from about 0.1 to about 5mg/kg, from about 3 to about 10mg/kg, from about 3 to about 15mg/kg, or from about 0.1 to about 30mg/kg of body weight or from about 80mg to at least about 800mg, from about 80mg to at least about 700mg, from about 80mg to at least about 600mg, from about 80mg to at least about 500mg, from about 80mg to at least about 400mg, from about 80mg to at least about 300mg, from about 100mg to at least about 300mg, or from about 200mg to about 300 mg. For example, the dose may be about 0.1, about 0.3, about 1, about 2, about 3, about 5, or about 10mg/kg body weight, or about 0.3, about 1, about 2, about 3, or about 5mg/kg body weight; or about 80mg, about 100mg, about 160mg, about 200mg, about 240mg, about 300mg, about 320mg, about 400mg, about 500mg, about 600mg, about 700mg, or about 800 mg. Dosing schedules are generally designed to achieve exposures that result in sustained Receptor Occupancy (RO) (based on the typical pharmacokinetic properties of antibodies). An exemplary treatment regimen entails administration about once per week, about once every 2 weeks, about once every 3 weeks, about once every 4 weeks, about 1 time per month, about once every 3-6 months, or longer.
In certain embodiments, a dose of about 3mg/kg to about 30mg/kg of body weight of the subject is administered intravenously on day 1 of the cycle (D1) for about 21 days for 1 cycle until efficacy is assessed as disease progression and an intolerable toxic response is present. In certain embodiments, a body weight dose of about 3mg/kg to about 20mg/kg is administered intravenously on day 1 of the cycle (D1) for 1 cycle of about 21 days until efficacy is assessed as disease progression with intolerable toxicity. In certain embodiments, a 3mg/kg dose is received by intravenous drip on day 1 of the cycle (D1), for about 21 days for 1 cycle, until efficacy is assessed as disease progression, with intolerable toxic reactions. In certain embodiments, a dose of about 10mg/kg body weight is administered by intravenous drip on day 1 of the cycle (D1) for about 21 days for 1 cycle until therapeutic efficacy is assessed as disease progression and an intolerable toxic response occurs. In certain embodiments, a dose of about 15mg/kg body weight is administered by intravenous drip on day 1 of the cycle (D1) for about 21 days for 1 cycle until therapeutic efficacy is assessed as disease progression and an intolerable toxic response occurs. In certain embodiments, a dose of about 20mg/kg body weight is administered intravenously on day 1 of the cycle (D1) for 1 cycle for 21 days until efficacy is assessed as disease progression and an intolerable toxic response is present. In certain embodiments, a dose of about 30mg/kg body weight is administered intravenously on day 1 of the cycle (D1) for 1 cycle for 21 days until efficacy is assessed as disease progression and an intolerable toxic response is present. In certain embodiments, a body weight dose of about 3mg/kg, 10mg/kg, 15mg/kg, 20mg/kg or 30mg/kg is administered by intravenous drip on day 1 of each cycle (D1) for 1 cycle of about 2 weeks, about 3 weeks or about 4 weeks until the therapeutic effect is assessed as disease progression with intolerable toxic reactions. In certain embodiments, a single dose intravenous drip of about 1200mg is received on day 1 of the cycle (D1) for 1 cycle of about 2 weeks, about 3 weeks, or about 4 weeks, until efficacy is assessed as disease progression, with intolerable toxic reactions.
In certain embodiments, a uniform dose of about 600mg to about 1200mg is received as an intravenous drip on day 1 of the cycle (D1), for about 1 cycle for about 21 days, until efficacy is assessed as disease progression, with intolerable toxicity. In certain embodiments, a uniform dose of about 600mg to about 900mg per cycle on day 1 (D1) is administered by intravenous drip for about 21 days for 1 cycle until efficacy is assessed as disease progression and an intolerable toxic response is present. In certain embodiments, a 1200mg bolus intravenous drip is received on day 1 of the cycle (D1) for 1 cycle of about 21 days until efficacy is assessed as disease progression with intolerable toxic response. In certain embodiments, a uniform dose of about 900mg to about 1200mg per cycle on day 1 (D1) is administered by intravenous drip for about 21 days for 1 cycle until efficacy is assessed as disease progression and an intolerable toxic response is present. In certain embodiments, a 600mg bolus intravenous drip is received on day 1 of the cycle (D1) for 1 cycle for 21 days until efficacy is assessed as disease progression and an intolerable toxic response occurs. In certain embodiments, a 900mg bolus intravenous drip is received on day 1 of the cycle (D1) for 1 cycle of 21 days until efficacy is assessed as disease progression with intolerable toxic reactions. In certain embodiments, a 1200mg bolus intravenous drip is received on day 1 of the cycle (D1) for 1 cycle for 21 days until efficacy is assessed as disease progression and an intolerable toxic response occurs. In certain embodiments, a single dose intravenous drip of about 900mg is received on day 1 of the cycle (D1) for 1 cycle of about 2 weeks, about 3 weeks, or about 4 weeks, until efficacy is assessed as disease progression, with intolerable toxic reactions. In certain embodiments, a single dose intravenous drip of about 1200mg is received on day 1 of the cycle (D1) for 1 cycle of about 2 weeks, about 3 weeks, or about 4 weeks, until efficacy is assessed as disease progression, with intolerable toxic reactions.
In certain embodiments, the dose of the anti-PD-L1 antibody or anti-PD-1 antibody is a fixed dose in a pharmaceutical composition.
In certain embodiments, the dose of the anti-PD-L1 antibody or anti-PD-1 antibody is a fixed dose in a pharmaceutical composition containing a second anti-cancer agent.
The actual dosage level of one or more of the active ingredients in the pharmaceutical compositions of the present application can be varied so as to obtain an amount of the active ingredient that is effective to achieve a desired therapeutic response for a particular patient, composition, and mode of administration, without undue toxicity to the patient. The selected dosage level will depend upon a variety of pharmacokinetic factors including the activity of the particular composition of the application employed, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in conjunction with the particular composition employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts. The compositions of the present application can be administered via one or more routes of administration using one or more of a variety of methods well known in the art. The skilled artisan will appreciate that the route and/or mode of administration will vary with the desired result.
The present application is also directed to a pharmaceutical composition comprising an antibody and at least one or more of a buffer, an isotonicity adjusting agent, a stabilizer and/or a surfactant. In particular, the pharmaceutical composition comprises 1-150mg/mL of an anti-PD-L1 humanized monoclonal antibody (mab), 3-50mM buffer, 2-150mg/mL of an isotonicity adjusting/stabilizing agent and 0.01-0.8mg/mL of a surfactant, and has a pH of about 4.5-6.8.
In some embodiments, the anti-PD-L1 humanized mab concentration is about 5-150mg/mL, calculated as w/v; preferably about 10-60 mg/mL; more preferably about 10-30 mg/mL. In some embodiments, the anti-PD-L1 humanized monoclonal antibody has a mass-volume concentration of about 10mg/mL, about 20mg/mL, about 30mg/mL, about 40mg/mL, about 50mg/mL, about 60mg/mL, about 70mg/mL, about 80mg/mL, about 90mg/mL, about 100mg/mL, about 110mg/mL, or about 120mg/mL, preferably about 10mg/mL, about 20mg/mL, about 30mg/mL, about 40mg/mL, about 50mg/mL, or about 60mg/mL, more preferably about 10mg/mL, about 20mg/mL, or about 30 mg/mL. In some embodiments, the anti-PD-L1 humanized mab mass volume concentration is about 10 mg/mL. In other embodiments, the anti-PD-L1 humanized monoclonal antibody mass volume concentration is about 30 mg/mL. In other embodiments, the anti-PD-L1 humanized monoclonal antibody mass volume concentration is about 60 mg/mL.
In some embodiments, the buffer is a histidine salt buffer. The histidine salt buffer is present at a concentration of about 5 to about 30mM, preferably about 10 to about 25mM, more preferably about 10 to about 20mM, and most preferably about 10 to about 15 mM. In some embodiments, the histidine salt buffer is about 5mM, about 10mM, about 15mM, about 20mM, about 25mM, or about 30 mM. In some embodiments, the histidine salt buffer is about 10 mM. In other embodiments, the histidine salt buffer is about 15 mM. In other embodiments, the histidine salt buffer is about 20 mM. Wherein the histidine salt buffer comprises histidine and hydrochloric acid.
In some embodiments, the isotonicity adjusting/stabilizing agent is about 20-150mg/mL sucrose, preferably about 40-100mg/mL sucrose, more preferably about 60-80mg/mL sucrose, calculated as w/v. In some embodiments, the sucrose is at a concentration of about 40mg/mL, 50mg/mL, 60mg/mL, 70mg/mL, 80mg/mL, 90mg/mL, or 100 mg/mL. In some embodiments, the sucrose concentration is about 60 mg/mL. In some embodiments, the sucrose concentration is about 70 mg/mL. In some embodiments, the sucrose concentration is about 80 mg/mL. In some embodiments, the sucrose concentration is about 90 mg/mL.
In some embodiments, the surfactant is selected from polysorbate 80, polysorbate 20, poloxamer 188; preferably polysorbate 80 or polysorbate 20; more preferably polysorbate 80. In some embodiments, the surfactant is present at a concentration of about 0.05 to about 0.6mg/mL, preferably about 0.1 to about 0.4mg/mL, and more preferably about 0.2 to about 0.3mg/mL, w/v.
In some embodiments, the surfactant is about 0.01-0.8mg/mL polysorbate 80 or polysorbate 20, calculated as w/v. In some embodiments, the surfactant is about 0.05-0.6mg/mL polysorbate 80, preferably about 0.1-0.4mg/mL polysorbate 80, more preferably about 0.2-0.3mg/mL polysorbate 80, and most preferably about 0.2mg/mL polysorbate 80. In some embodiments, polysorbate 80 is present in the pharmaceutical composition at about 0.1mg/mL, 0.2mg/mL, 0.3mg/mL, 0.4mg/mL, 0.5mg/mL, or 0.6 mg/mL; preferably, the pharmaceutical composition has polysorbate 80 at a level of about 0.2mg/mL, 0.3mg/mL, 0.4mg/mL, or 0.5 mg/mL; more preferably, the pharmaceutical composition comprises polysorbate 80 at a level of about 0.2mg/mL, 0.3mg/mL, or 0.4 mg/mL; most preferably, the pharmaceutical composition has polysorbate 80 at a level of about 0.2 mg/mL. In some embodiments, the polysorbate 80 content of the pharmaceutical composition is about 0.1 mg/mL. In other embodiments, the pharmaceutical composition comprises polysorbate 80 at about 0.2 mg/mL. In some embodiments, the polysorbate 80 content of the pharmaceutical composition is about 0.3 mg/mL. In other embodiments, the pharmaceutical composition comprises polysorbate 80 at a level of about 0.4 mg/mL. In some embodiments, the polysorbate 80 content of the pharmaceutical composition is about 0.5 mg/mL.
In some embodiments, the pH of the aqueous solution of the pharmaceutical composition is selected from 4.0-6.8; preferably 4.5 to 6.5; more preferably 5.5 to 6.0; most preferably 5.5. In some embodiments, the pH of the aqueous pharmaceutical composition solution is about 4.5, about 4.8, about 5.0, about 5.2, about 5.4, about 5.5, about 5.6, about 5.8, or about 6.0, preferably about 5.0, about 5.2, about 5.4, about 5.5, or about 5.6, more preferably about 5.5. In some embodiments, the pH of the aqueous pharmaceutical composition solution is about 5.0. In some embodiments, the pH of the aqueous pharmaceutical composition solution is about 5.2. In some embodiments, the pH of the aqueous pharmaceutical composition solution is about 5.4. In some embodiments, the pH of the aqueous pharmaceutical composition solution is about 5.5. In some embodiments, the pH of the aqueous pharmaceutical composition solution is about 5.6. In some embodiments, the pH of the aqueous pharmaceutical composition solution is about 5.8. In some embodiments, the pH of the aqueous pharmaceutical composition solution is about 6.0.
The present application provides an isolated antibody or fragment thereof that binds PD-L1. The anti-PD-L1 humanized monoclonal antibody provided by the application comprises the following amino acid sequence: a heavy chain CDR1 region having at least 80% (e.g., 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%) homology to the amino acid sequence set forth in SEQ ID NO 1 or SEQ ID NO 4; a heavy chain CDR2 region having at least 80% (e.g., 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%) homology to the amino acid sequence set forth in SEQ ID NO 2 or SEQ ID NO 5; a heavy chain CDR3 region having at least 80% (e.g., 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%) homology to the amino acid sequence set forth in SEQ ID NO 3 or SEQ ID NO 6; a light chain CDR1 region having at least 80% (e.g., 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%) homology to the amino acid sequence set forth in SEQ ID NO 7 or SEQ ID NO 10; a light chain CDR2 region having at least 80% (e.g., 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%) homology to the amino acid sequence set forth in SEQ ID NO 8 or SEQ ID NO 11; a light chain CDR3 region having at least 80% (e.g., 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%) homology to the amino acid sequence set forth in SEQ ID NO 9 or SEQ ID NO 12.
In a specific embodiment, the anti-PD-L1 humanized monoclonal antibodies provided herein comprise the amino acid sequence: selected from the heavy chain CDR1 regions shown as SEQ ID NO.1 or SEQ ID NO. 4; selected from the heavy chain CDR2 regions shown as SEQ ID NO. 2 or SEQ ID NO. 5; selected from the heavy chain CDR3 regions shown as SEQ ID NO. 3 or SEQ ID NO. 6; selected from the light chain CDR1 regions shown as SEQ ID NO. 7 or SEQ ID NO. 10; selected from the light chain CDR2 regions shown as SEQ ID NO 8 or SEQ ID NO 11; selected from the light chain CDR3 regions shown as SEQ ID NO 9 or SEQ ID NO 12.
Preferably, the anti-PD-L1 humanized mab provided herein comprises the amino acid sequence: a heavy chain variable region having at least 80% (e.g., 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%) homology to the amino acid sequence set forth in SEQ ID NO 13 or SEQ ID NO 14; a light chain variable region that is at least 80% (e.g., 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%) homologous to the amino acid sequence set forth in SEQ ID NO 15 or SEQ ID NO 16.
In a specific embodiment, the anti-PD-L1 humanized monoclonal antibody provided herein comprises the amino acid sequence shown as the heavy chain variable region of SEQ ID NO: 13; the light chain variable region shown as SEQ ID NO. 15.
In another embodiment, the anti-PD-L1 humanized monoclonal antibody provided herein comprises the amino acid sequence shown as the heavy chain variable region of SEQ ID NO: 14; the variable region of the light chain as shown in SEQ ID NO 16.
Each CDR region described herein and the various variants thereof described above are capable of specifically recognizing and binding to PD-L1, thereby effectively blocking signaling between PD-L1 and PD-1.
In a specific embodiment, the anti-PD-L1 humanized monoclonal antibody provided herein comprises a heavy chain amino acid sequence as set forth in SEQ ID NO.17 and a light chain amino acid sequence as set forth in SEQ ID NO. 18.
In another specific embodiment, the anti-PD-L1 humanized monoclonal antibody provided herein comprises a heavy chain amino acid sequence as set forth in SEQ ID NO. 19 and a light chain amino acid sequence as set forth in SEQ ID NO. 20.
In another specific embodiment, the anti-PD-L1 humanized monoclonal antibody provided herein comprises a heavy chain amino acid sequence as set forth in SEQ ID NO:21 and a light chain amino acid sequence as set forth in SEQ ID NO: 18.
In a specific embodiment, the anti-PD-L1 humanized monoclonal antibodies provided herein comprise conservative substitution variants selected from one or more of SEQ ID NO 1, SEQ ID NO 2, SEQ ID NO 3, SEQ ID NO 4, SEQ ID NO 5, SEQ ID NO 6, SEQ ID NO 7, SEQ ID NO 8, SEQ ID NO 9, SEQ ID NO 10, SEQ ID NO 11, SEQ ID NO 12, SEQ ID NO 13, SEQ ID NO 14, SEQ ID NO 15, SEQ ID NO 16, SEQ ID NO 17, SEQ ID NO 18, SEQ ID NO 19, SEQ ID NO 20, SEQ ID NO 21. An anti-PD-L1 humanized mab comprising the conservative substitution variants retains the ability to specifically recognize and bind to PD-L1.
The anti-PD-L1 humanized monoclonal antibody provided by the application can be an IgG1 or IgG4 antibody, preferably, the anti-PD-L1 humanized monoclonal antibody is an IgG1 antibody, and more preferably is a glycosylated IgG1 antibody.
In a specific embodiment of the present application, the pharmaceutical composition comprises: (a) an anti-PD-L1 humanized mab at a mass volume concentration of about 20mg/mL, (b) sucrose at a mass volume concentration of about 70mg/mL, (c) polysorbate 80 at a mass volume concentration of about 0.1mg/mL, (d) histidine at a molar concentration of about 20mM, (e) optionally, a suitable amount of hydrochloric acid to adjust the pH of the composition to about 5.0.
In another specific embodiment of the present application, the pharmaceutical composition comprises: (a) an anti-PD-L1 humanized mab at a concentration by mass volume of about 10mg/mL, (b) sucrose at a concentration by mass volume of about 80mg/mL, (c) polysorbate 80 at a concentration by mass volume of about 0.2mg/mL, (d) histidine at a molarity of about 10mM, (e) optionally, a suitable amount of hydrochloric acid to adjust the pH of the composition to about 5.5.
In yet another specific embodiment of the present application, the pharmaceutical composition comprises: (a) an anti-PD-L1 humanized mab at a concentration by mass volume of about 50mg/mL, (b) sucrose at a concentration by mass volume of about 80mg/mL, (c) polysorbate 80 at a concentration by mass volume of about 0.3mg/mL, (d) histidine at a molarity of about 10mM, (e) optionally, a suitable amount of hydrochloric acid to adjust the pH of the composition to about 5.5.
In yet another more specific embodiment of the present application, the pharmaceutical composition comprises: (a) an anti-PD-L1 humanized mab at a concentration by mass volume of about 100mg/mL, (b) sucrose at a concentration by mass volume of about 80mg/mL, (c) polysorbate 80 at a concentration by mass volume of about 0.5mg/mL, (d) histidine at a molarity of about 10mM, (e) optionally, a suitable amount of hydrochloric acid to adjust the pH of the composition to about 5.5.
In yet another specific embodiment of the present application, the pharmaceutical composition comprises: (a) an anti-PD-L1 humanized mab at a concentration by mass volume of about 30mg/mL, (b) sucrose at a concentration by mass volume of about 80mg/mL, (c) polysorbate 80 at a concentration by mass volume of about 0.2mg/mL, (d) histidine at a molarity of about 10mM, (e) optionally, a suitable amount of hydrochloric acid to adjust the pH of the composition to about 5.5.
In yet another specific embodiment of the present application, the pharmaceutical composition comprises: (a) an anti-PD-L1 humanized mab at a concentration by mass volume of about 60mg/mL, (b) sucrose at a concentration by mass volume of about 80mg/mL, (c) polysorbate 80 at a concentration by mass volume of about 0.2mg/mL, (d) histidine at a molarity of about 10mM, (e) optionally, a suitable amount of hydrochloric acid to adjust the pH of the composition to about 5.5.
In yet another specific embodiment of the present application, the pharmaceutical composition comprises: (a) an anti-PD-L1 humanized mab at a mass volume concentration of about 10mg/mL, (b) sucrose at a mass volume concentration of about 70mg/mL, (c) polysorbate 80 at a mass volume concentration of about 0.4mg/mL, (d) histidine at a molar concentration of about 20mM, (e) optionally, a suitable amount of acetic acid, adjusting the pH of the composition to about 6.5.
In yet another specific embodiment of the present application, the pharmaceutical composition comprises: (a) an anti-PD-L1 humanized mab at a mass volume concentration of about 10mg/mL, (b) sucrose at a mass volume concentration of about 80mg/mL, (c) polysorbate 80 at a mass volume concentration of about 0.2mg/mL, (d) histidine at a molar concentration of about 20mM, (e) optionally, a suitable amount of hydrochloric acid to adjust the pH of the composition to about 5.5.
In some embodiments, the pharmaceutical composition is an aqueous injection solution, including but not limited to an aqueous formulation that is not lyophilized or an aqueous formulation reconstituted from a lyophilized powder. In other embodiments, the pharmaceutical composition is a lyophilized formulation. The lyophilized formulation refers to a formulation prepared by subjecting an aqueous solution to a lyophilization process, which is a stabilization process in which the substance is first frozen and then the amount of solvent is reduced by sublimation (primary drying process) and then by desorption (secondary drying process) until the amount of solvent is a value that no longer supports a biological activity or chemical reaction. The lyophilized formulations of the present application can also be dried by other methods known in the art, such as spray drying and bubble drying.
The present application provides formulations which do not exceed 1.1%, preferably not exceed 0.9%, more preferably not exceed 0.5% of the polymer when stored at 2-8 ℃ or 25 ℃ for at least 6 months.
The present application also provides methods of preparing the aforementioned pharmaceutical compositions comprising admixing the anti-PD-L1 humanized mab with other agents, such as one or more of a buffer, an isotonicity adjusting/stabilizing agent, and/or a surfactant.
The present application also provides a method for treating, ameliorating or ameliorating a neoplasia condition in a subject comprising administering to the subject the aforementioned pharmaceutical composition.
The present application provides an article of manufacture comprising a container containing a fixed dose of an anti-PD-L1 antibody. The present application also provides for the use of an anti-PD-L1 antibody in the manufacture of an article of manufacture for the treatment of cancer comprising a container comprising a fixed dose of an anti-PD-L1 antibody. In some embodiments, the container is a vial. The fixed dose is selected from about 300mg, about 600mg, about 900mg, about 1000mg, about 1200mg, about 1500mg, about 1800mg, about 2100mg, and about 2400mg of the anti-PD-L1 antibody. In some embodiments, the article of manufacture further comprises a package insert or package insert instructing the user to administer the fixed dose to a cancer patient. In some embodiments, the article of manufacture comprises 1 or more than 1 vial containing about 300mg or 600mg of anti-PD-L1 antibody. In some embodiments, the article of manufacture comprises 1 vial containing about 300mg of the anti-PD-L1 antibody. In some embodiments, the article of manufacture comprises 1 or more than 1 vial, each of which contains about 10mL of a pharmaceutical composition comprising an anti-PD-L1 antibody. In some embodiments, the article of manufacture comprises 1 or more than 1 vial, each of which contains about 20mL of the anti-PD-L1 antibody-containing pharmaceutical composition. In some embodiments, the pharmaceutical composition comprises 1-150mg/mL anti-PD-L1 humanized monoclonal antibody (mab), 3-50mM buffer, 2-150mg/mL isotonic adjusting/stabilizing agent, and 0.01-0.8mg/mL surfactant, and has a pH of about 4.5-6.8. In some embodiments, the article of manufacture comprises 1 or more than 1 vial, each of which contains about 10mL of a pharmaceutical composition comprising an anti-PD-L1 antibody, wherein the pharmaceutical composition comprises 30mg/mL of the anti-PD-L1 humanized monoclonal antibody. In some embodiments, the article of manufacture comprises 1 or more than 1 vial, each of which contains about 20mL of a pharmaceutical composition comprising an anti-PD-L1 antibody, wherein the pharmaceutical composition comprises 30mg/mL of an anti-PD-L1 humanized monoclonal antibody. In some embodiments, the pharmaceutical composition is any one of the pharmaceutical compositions provided herein.
In some embodiments, 300mg of the anti-PD-L1 antibody can be placed in 1 container and 4 such containers made up an article of manufacture, or 600mg of the anti-PD-L1 antibody can be placed in 1 container and 2 such containers made up an article of manufacture, optionally further comprising a therapeutically effective amount of cisplatin and/or 5-fluorouracil, or alternatively comprising a therapeutically effective amount of carboplatin and/or 5-fluorouracil. The substance of the article may be conveniently administered to a patient.
Definitions and explanations of terms
In order that the present application may be more readily understood, certain terms are defined. As used in this application, each of the following terms shall have the following meaning, except where otherwise explicitly provided herein. Additional definitions are set forth in the present application.
As used herein, the term "antibody" refers to a binding protein having at least one antigen binding domain. The antibodies and fragments thereof of the present application can be whole antibodies or any fragment thereof. Thus, the antibodies and fragments of the present application include monoclonal antibodies or fragments thereof and antibody variants or fragments thereof, as well as immunoconjugates. Examples of antibody fragments include Fab fragments, Fab 'fragments, F (ab')2 fragments, Fv fragments, isolated CDR regions, single chain Fv molecules (scFv), Fd fragments, and other antibody fragments known in the art. Antibodies and fragments thereof can also include recombinant polypeptides, fusion proteins, and bispecific antibodies. The anti-PD-L1 antibodies and fragments thereof disclosed herein may be of the IgG1, IgG2, IgG3, or IgG4 isotype. The term "isotype" refers to the class of antibodies encoded by the heavy chain constant region gene. In one embodiment, the anti-PD-L1 antibodies and fragments thereof disclosed herein are of the IgG1 or IgG4 isotype. The PD-L1 antibodies and fragments thereof of the present application may be derived from any species, including but not limited to mouse, rat, rabbit, primate, llama, and human. The PD-L1 antibody and fragments thereof can be chimeric, humanized or fully human. In one embodiment, the anti-PD-L1 antibody is an antibody produced by a mouse-derived hybridoma cell line. Thus, in one embodiment, the anti-PD-L1 antibody is a murine antibody. In another embodiment, the anti-PD-L1 antibody is a chimeric antibody. In another embodiment, the chimeric antibody is a mouse-human chimeric antibody. In another embodiment, the antibody is a humanized antibody. In another embodiment, the antibody is derived from a murine antibody and is humanized.
"humanized antibodies" are the following antibodies: the antibody comprises Complementarity Determining Regions (CDRs) derived from a non-human antibody; and framework regions and constant regions derived from human antibodies. For example, an anti-PD-L1 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-L1 as a murine antibody from which the CDRs of the antibody are derived. Provided herein are exemplary humanized antibodies. Additional anti-PD-L1 antibodies or variants thereof comprising the heavy and light chain CDRs provided herein can be produced using any human framework sequence and are also included in the present application. In one embodiment, framework sequences suitable for use in the present application include those similar in structure to the framework sequences provided herein. Additional modifications may be made in the framework regions to improve the properties of the antibodies provided herein. Such additional framework modifications may include chemical modifications; point mutations to reduce immunogenicity or to remove T cell epitopes; or reverting the mutation to a residue in the original germline sequence. In some embodiments, such modifications include those corresponding to the mutations exemplified herein, including back mutations to germline sequences. For example, in one embodiment, one or more amino acids in the human framework regions of the VH and/or VL of the humanized antibodies provided herein are back mutated to the corresponding amino acids in the parent murine antibody. For example, for the VH and VL of humanized 5G11 and humanized 13C5, several sites of the framework amino acids of the template human antibody described above were back mutated to the corresponding amino acid sequences in the mouse 5G11 and 13C5 antibodies. In one embodiment, the amino acids at positions 53 and/or 60 and/or 67 of the light chain variable region are back mutated to the corresponding amino acids found at said positions in the mouse 5G11 or 13C5 light chain variable region. In another embodiment, the amino acid at position 24 and/or 28 and/or 30 and/or 49 and/or 73 and/or 83 and/or 94 of the heavy chain variable region is back mutated to the corresponding amino acid found at said position in the mouse 5G11 or 13C5 heavy chain variable region. In one embodiment, the humanized 5G11 antibody comprises a light chain variable region wherein the amino acid at position 60 is mutated from ser(s) to asp (d) and the amino acid at position 67 is mutated from ser(s) to tyr (y); and a heavy chain variable region wherein the amino acid at position 24 is mutated from phe (f) to val (v), the amino acid at position 49 is mutated from ala (a) to gly (g), the amino acid at position 73 is mutated from thr (t) to asn (n), and the amino acid at position 83 is mutated from thr (t) to asn (n). In one embodiment, the humanized 13C5 antibody comprises a light chain variable region wherein the amino acid at position 53 is mutated from tyr (y) to lys (k); and a heavy chain variable region wherein the amino acid at position 28 is mutated from Thr (T) to Ile (I), the amino acid at position 30 is mutated from Ser (S) to Arg (R), the amino acid at position 49 is mutated from Ser (S) to Ala (A), and the amino acid at position 94 is mutated from Tyr (Y) to Asp (D). Additional or alternative back mutations can be made in the framework regions of the humanized antibodies provided herein to improve the properties of the antibody. The present application also includes humanized antibodies that bind PD-L1 and comprise framework modifications corresponding to the exemplary modifications described herein relative to any suitable framework sequence, as well as other framework modifications that otherwise improve the properties of the antibody.
By "isolated antibody" is meant an antibody that: it is substantially free of other antibodies having different antigen specificities (e.g., an isolated antibody that specifically binds PD-1 is substantially free of antibodies that specifically bind antigens other than PD-1). However, an isolated antibody that specifically binds PD-1 may have cross-reactivity with other antigens (such as PD-1 molecules from different species). Furthermore, the isolated antibody may be substantially free of other cellular material and/or chemicals.
The term "monoclonal antibody" ("mAb") refers to a non-naturally occurring preparation of antibody molecules of single molecular composition (i.e., antibody molecules whose base sequences are substantially identical and which exhibit a single binding specificity and affinity for a particular epitope). mabs are an example of an isolated antibody. Mabs can be produced by hybridoma techniques, recombinant techniques, transgenic techniques, or other techniques known to those of skill in the art.
An "antigen-binding portion" (also referred to as an "antigen-binding fragment") of an antibody refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen bound by an intact antibody.
As used herein, the term "derived" when used in reference to a molecule or polypeptide relative to a reference antibody or other binding protein means a molecule or polypeptide that is capable of specifically binding the same epitope as the reference antibody or other binding protein.
The antibodies and antigen binding fragments thereof disclosed herein are specific for PD-L1. In one embodiment, the antibody or fragment thereof is specific for PD-L1. In one embodiment, the antibodies and fragments provided herein bind to human or primate PD-L1, but not PD-L1 from any other mammal. In another embodiment, the antibody or and fragments thereof does not bind to mouse PD-L1. The terms "human PD-L1", "hPD-L1", and "huPD-L1", and the like, are used interchangeably herein, and refer to variants or isoforms of human PD-L1 and human PD-L1. By "specific" is meant that the antibody and fragments thereof bind PD-L1 with greater affinity than any other target.
As used herein, the term "EC 50" refers to the effective concentration, 50% of the maximal response of an antibody. As used herein, the term "IC 50" refers to the inhibitory concentration, 50% of the maximal response of an antibody. Both EC50 and IC50 may be measured by ELISA or FACS analysis or any other method known in the art.
As used herein, the term "treatment" refers to both therapeutic treatment as well as preventative or prophylactic measures. Subjects in need of treatment include those already with the disease or condition, as well as those who may have the disease or condition and whose purpose is to prevent, delay or attenuate the disease or condition.
As used herein, the term "systemic treatment" refers to treatment in which a drug substance is transported through the bloodstream to reach and affect cells throughout the body.
As used herein, the term "systemic chemotherapy" refers to systemic chemotherapy that does not include chemotherapy for locally advanced disease as one of the links of multimodal treatment, wherein chemotherapy for locally advanced disease includes induction chemotherapy, concurrent chemotherapy with radiotherapy, and adjuvant chemotherapy.
As used herein, the term "subject" means a mammal, such as a rodent, feline, canine, and primate. Preferably, the subject according to the present application is a human.
By "administering" is meant physically introducing a composition comprising a therapeutic agent to a subject using any of a variety of methods and delivery systems known to those skilled in the art. Routes of administration of immune checkpoint inhibitors (e.g., anti-PD-1 antibodies or anti-PD-L1 antibodies) include intravenous, intramuscular, subcutaneous, intraperitoneal, spinal, or other parenteral routes of administration, e.g., by injection or infusion. The phrase "parenteral administration" as used herein refers to modes of administration other than enteral and topical administration, typically by injection, and includes, but is not limited to, intravenous, intramuscular, intraarterial, intrathecal, intralymphatic, intralesional, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion, and in vivo electroporation. In certain embodiments, the immune checkpoint inhibitor (e.g., an anti-PD-1 antibody or an anti-PD-L1 antibody) is administered by a non-parenteral route, and in certain embodiments, orally. Other non-parenteral routes include topical, epidermal or mucosal routes of administration, e.g., intranasally, vaginally, rectally, sublingually or topically. Administration may also be performed, for example, once, multiple times, and/or over one or more extended periods of time.
As used herein, an "adverse effect" (AE) is any adverse and often unintended or undesirable sign (including abnormal laboratory findings), symptom or disease associated with the use of medical treatment. For example, an adverse event can be associated with activation of the immune system or expansion of cells of the immune system (e.g., T cells) in response to a treatment. The medical treatment may have one or more related AEs, and each AE may have the same or different severity level. Reference to a method capable of "altering an adverse event" refers to a treatment regimen that reduces the incidence and/or severity of one or more AEs associated with the use of a different treatment regimen.
As used herein, "dosing interval" refers to the amount of time that elapses between multiple doses of a formulation disclosed herein administered to a subject. The dosing interval may thus be indicated as a range.
The term "dosing frequency" as used herein means the frequency of doses administered of a formulation disclosed herein over a given time. The frequency of administration may be indicated as the number of doses per given time, e.g. 1 time per week or 1 time per 2 weeks.
The use of the term "flat dose" refers to a dose that is administered to a patient without regard to the weight or Body Surface Area (BSA) of the patient. Thus, a uniform dose is provided as an absolute amount of the agent (e.g., anti-PD-L1 antibody) rather than as a mg/kg dose. For example, a 60kg human and a 100kg human will receive the same dose of antibody (e.g., 240mg anti-PD-L1 antibody).
The use of the term "fixed dose" in reference to a composition of the present application means that two or more different antibodies in a single composition are present in the composition in a specific (fixed) ratio to each other. In certain embodiments, the fixed dose is based on the weight of the antibody (e.g., mg). In certain embodiments, the fixed dose is based on the concentration of the antibody (e.g., mg/mL). In certain embodiments, the ratio of mg of the first antibody to mg of the second antibody is at least about 1:1, about 1:2, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:15, about 1:20, about 1:30, about 1:40, about 1:50, about 1:60, about 1:70, about 1:80, about 1:90, about 1:100, about 1:120, about 1:140, about 1:160, about 1:180, about 1:200, about 200:1, about 180:1, about 160:1, about 140:1, about 120:1, about 100:1, about 90:1, about 80:1, about 70:1, about 60:1, about 50:1, about 40:1, about 30:1, about 20:1, about 15:1, about 10:1, about 1: 8:1, about 1:1, about 5: 7, about 1:1, about 1: 6:1, about 1:1, about 1:1, about 1:1, about 1:1, about 1:1, about 1:1, about 1:1, about 1:1, about 1:1, about 6, about 1:1, about 1, about 4:1, about 3:1, or about 2: 1. For example, a 3:1 ratio of primary and secondary antibodies may mean that the vial may contain about 240mg of primary and 80mg of secondary antibody, or about 3mg/ml of primary and 1mg/ml of secondary antibody.
The term "weight-based dose" as referred to herein refers to a dose administered to a patient that is calculated based on the weight of the patient. For example, when a patient with a weight of 60kg requires 3mg/kg of anti-PD-1 antibody and 1mg/kg of anti-CTLA-4 antibody, one can extract appropriate amounts of anti-PD-1 antibody (i.e., 180mg) and anti-CTLA-4 antibody (i.e., 60mg) at a time from a 3:1 ratio fixed dose formulation of anti-PD-1 antibody and anti-CTLA-4 antibody.
The term "immunotherapy" means the treatment of a subject having a disease or at risk of infection or of suffering from a relapse of a disease by a method that includes inducing, enhancing, suppressing or otherwise altering an immune response. By "treatment" or "therapy" of a subject is meant any type of intervention or process performed on the subject, or the administration of an active agent to a subject, with the purpose of reversing, alleviating, ameliorating, inhibiting, slowing, or preventing the onset, progression, severity, or recurrence of a symptom, complication, or condition, or biochemical indicator associated with the disease.
As used herein, "PD-L1 positive" may be used interchangeably with "at least about 1% of PD-L1 expression". In one embodiment, PD-L1 expression may be used by any method known in the art. In another embodiment, PD-L1 expression is measured by automated IHC. A PD-L1 positive tumor may thus have at least about 1%, at least about 2%, at least about 5%, at least about 10%, or at least about 20% PD-L1 expressing tumor cells as measured by automated IHC. In certain embodiments, "PD-L1 positive" means that there are at least 100 cells expressing PD-L1 on the cell surface.
"programmed death receptor-1 (PD-1)" means an immunosuppressive receptor belonging to the CD28 family. PD-1 is expressed predominantly on previously activated T cells in vivo and binds to both ligands PD-L1 and PD-L2. The term "PD-1" as used herein includes variants, homologs, and species homologs of human PD-1(hPD-1), hPD-1, and analogs having at least one common epitope with hPD-1.
"programmed death ligand-1 (PD-L1)" is one of two cell surface glycoprotein ligands for PD-1 (the other being PD-L2) that down-regulates T cell activation and cytokine secretion upon binding to PD-1.
"subject" includes any human or non-human animal. The term "non-human animal" includes, but is not limited to, vertebrates such as non-human primates, sheep, dogs, and rodents such as mice, rats, and guinea pigs. In certain embodiments, the subject is a human. The terms "subject" and "patient" are used interchangeably herein in certain contexts.
A "therapeutically effective amount" or "therapeutically effective dose" of a drug or therapeutic agent is any amount of drug that, when used alone or in combination with another therapeutic agent, protects a subject from the onset of a disease or promotes disease regression as evidenced by a reduction in the severity of disease symptoms, an increase in the frequency and duration of disease symptom-free stages, or prevention of injury or disability caused by the affliction of the disease. The ability of a therapeutic agent to promote disease regression can be evaluated using a variety of methods known to skilled practitioners, such as in human subjects during clinical trials, in animal model systems predicting efficacy for humans, or by determining the activity of the agent in an in vitro assay.
As used herein, a "sub-therapeutic dose" refers to a dose of a therapeutic compound (e.g., an antibody) that is lower than the usual or typical dose of the therapeutic compound when administered alone for the treatment of a hyperproliferative disease (e.g., cancer).
As an example, an "anti-cancer drug" promotes cancer regression in a subject or prevents further tumor growth. In certain embodiments, the therapeutically effective amount of the drug promotes cancer regression to the point of eliminating the cancer. By "promoting cancer regression" is meant that an effective amount of a drug, administered alone or in combination with an anti-neoplastic agent, results in the reduction of tumor growth or size, necrosis of the tumor, a reduction in the severity of at least one disease symptom, an increase in the frequency and duration of disease symptom-free stages, or the prevention of injury or disability resulting from the affliction of the disease. Furthermore, the terms "effective" and "effectiveness" with respect to treatment include pharmacological effectiveness and physiological safety. Pharmacological efficacy refers to the ability of a drug to promote cancer regression in a patient. Physiological safety means the level of toxicity or other adverse physiological effects (adverse effects) at the cellular, organ and/or organism level resulting from drug administration.
As an example for treating a tumor, a therapeutically effective amount of an anti-cancer agent can inhibit cell growth or tumor growth by at least about 10%, at least about 20%, at least about 40%, at least about 60%, or at least about 80% relative to an untreated subject, or, in certain embodiments, relative to a patient treated with standard of care therapy. In other embodiments of the present application, tumor regression may be observed for a period of at least about 20 days, at least about 40 days, or at least about 60 days. Despite these final measures of therapeutic effectiveness, the evaluation of immunotherapeutic drugs must also take into account "immune-related" response patterns.
By "immune-related" response pattern is meant the clinical response pattern often observed in cancer patients treated with immunotherapeutic agents that produce an anti-tumor effect by inducing a cancer-specific immune response or by altering the innate immune process. This response pattern is characterized by beneficial therapeutic effects following an initial increase in tumor burden or the appearance of new lesions, which would be classified as disease progression and would be synonymous with drug failure in the evaluation of traditional chemotherapeutic agents. Thus, proper evaluation of immunotherapeutic agents may require long-term monitoring of the effect of these agents on the target disease.
A therapeutically effective amount of a drug includes a "prophylactically effective amount," which is any amount of drug that inhibits the occurrence or recurrence of cancer when administered, alone or in combination with an anti-neoplastic agent, to a subject at risk of developing cancer (e.g., a subject with a premalignant condition) or a subject at risk of cancer recurrence. In certain embodiments, the prophylactically effective amount completely prevents the occurrence or recurrence of cancer. By "inhibiting" the occurrence or recurrence of cancer is meant reducing the likelihood of occurrence or recurrence of cancer, or completely preventing the occurrence or recurrence of cancer.
A "recurrent" cancer is one that regenerates at the primary site or a distant site in response to an initial treatment (e.g., surgery). A "locally recurrent" cancer is one that occurs at the same location after treatment as a previously treated cancer.
A "non-resectable" cancer is one that cannot be removed by surgery.
"metastatic" cancer refers to cancer that spreads from one part of the body (e.g., the lungs) to another part of the body.
The use of alternatives (e.g., "or") should be understood to refer to either, both, or any combination of alternatives. The indefinite articles "a" or "an" as used herein shall be understood to mean "one or more" of any listed or enumerated component.
The terms "about," about, "or" consisting essentially of mean a value or composition within an acceptable error range for the particular value or composition as determined by one of ordinary skill in the art, which will depend in part on how the value or composition is measured or determined, i.e., the limitations of the measurement system. For example, "about" or "consisting essentially of can mean within 1 or more than 1 standard deviation, as practiced in the art. Alternatively, "about" or "consisting essentially of may refer to a range of up to 10% or 20% (i.e., ± 10% or ± 20%), for example, fluctuating within ± 5% of the particular numerical range given, preferably fluctuating within ± 2%, more preferably fluctuating within ± 1%. For example, about 3mg may include any number between 2.7mg to 3.3mg (for 10%) or between 2.4mg to 3.6mg (for 20%). For example, a pH of about 5.5 means a pH of 5.5. + -. 5%, preferably a pH of 5.5. + -. 2%, more preferably a pH of 5.5. + -. 1%. Furthermore, particularly with respect to biological systems or processes, the term may refer to up to an order of magnitude or up to at most 5 times the numerical value. Where a particular value or composition is provided in the application and claims, unless otherwise stated, the meaning of "about" or "consisting essentially of" should be assumed to be within an acceptable error range for that particular value or composition.
As used herein, the terms "about once per week", "about once per two weeks" or any other similar dosing interval term refer to approximations. "about once per week" may include every 7 days ± 1 day, i.e., every 6 days to every 8 days. "about once every two weeks" may include every 14 days ± 3 days, i.e., every 11 days to every 17 days. Similar approximations apply, for example, about once every 3 weeks, about once every 4 weeks, about once every 5 weeks, about once every 6 weeks, and about once every 12 weeks. In certain embodiments, a dosing interval of about once every 6 weeks or about once every 12 weeks means that a first dose may be administered on any day of the first week, and then a second dose may be administered on any day of the sixth or twelfth week, respectively. In other embodiments, a dosing interval of about once every 6 weeks or about once every 12 weeks refers to administration of a first dose on a particular day of the first week (e.g., monday) followed by administration of a second dose on the same day of the sixth or twelfth week (i.e., monday), respectively. Similar principles apply to phrases including, but not limited to, "about 1 time every 2 weeks," "about 1 time per month," and the like.
As used herein, any concentration range, percentage range, ratio range, or integer range should be understood to include the value of any integer within the recited range, and when appropriate, to include fractions thereof (such as tenths and hundredths of integers), unless otherwise indicated.
In this document, unless otherwise indicated, the terms "comprises, comprising and including" or equivalents thereof, are open-ended and mean that elements, components and steps other than those listed may be included.
All patents, patent applications, and other established publications are herein expressly incorporated by reference for the purpose of description and disclosure. These publications are provided solely for their disclosure prior to the filing date of the present application. All statements as to the date of these documents or representation as to the contents of these documents is based on the information available to the applicant and does not constitute any admission as to the correctness of the dates of these documents or the contents of these documents. Moreover, any reference to such publications in this specification does not constitute an admission that the publications form part of the common general knowledge in the art in any country.
Detailed Description
The present application is further described below with reference to specific examples, which, however, are only for illustration and do not limit the scope of the present application. Likewise, the present application is not limited to any particular preferred embodiment described herein. It will be understood by those skilled in the art that equivalents may be made to the features of the present application or modifications may be made thereto without departing from the scope of the invention. The reagents used in the following examples are commercially available products, and the solutions can be prepared by techniques conventional in the art, except where otherwise specified. In the examples, the anti-PD-L1 humanized monoclonal antibody was prepared as described in WO2016022630, and after affinity chromatography, an eluate containing the antibody was obtained by a conventional antibody purification method.
TABLE 1 abbreviation table
Figure PCTCN2020074083-APPB-000004
Figure PCTCN2020074083-APPB-000005
Example 1 preclinical pharmacodynamic test
This example discloses the results of a pharmacodynamic study of an anti-PD-L1 antibody in vitro and in vivo in mice.
hu5G11-hIgG1 (heavy chain amino acid sequence is shown as SEQ ID NO.17, light chain amino acid sequence is shown as SEQ ID NO. 18) combined with human PD-L1 protein, and its EC50 was 21.3 ng/mL; the hu5G11-hIgG1 and human PD-L1 remarkably induce the secretion of IFN-gamma in a DC cell MLR, the effect of the IFN-gamma is obvious dose-dependent, and the EC50 is 35.0 +/-11.3 ng/mL, so that the hu5G11-hIgG1 and DC-expressed PD-L1 are combined to inhibit a PD-L1/PD-1 signal channel, thereby stimulating the secretion of the IFN-gamma in a CD4+ T cell.
The hu5G11-hIgG1(15mg/kg, IP, Q2D X11) had a 91.7% tumor suppression rate against MC-38/H-11 mice subcutaneously transplanted tumors (all calculated based on median tumor volume) and significantly prolonged survival of mice with intraperitoneal inoculation of mouse MC-38/H-11 cells, with median survival >98 days, to 80% survival at the end of the experiment (D98) (p <0.01, compared to the human IgG 15mg/kg group).
Therefore, in vivo and in vitro pharmacodynamic tests prove that hu5G11-hIgG1 can prevent PD-L1 from being combined with PD-1 and B7.1 receptors on the surface of T cells, so that the T cells recover the activity, the immune response is enhanced, and the anti-tumor effect is exerted.
Example 2 preclinical toxicology testing
This example discloses the results of acute toxicity testing and long-term toxicity testing of anti-PD-L1 antibodies in animals.
The cynomolgus monkeys were used 6, and divided into 2 groups of 3, each group had both male and female. The pharmaceutical compositions of the present application containing hu5G11-hIgG1 at a concentration of 10mg/mL were administered at doses of hu5G11-hIgG 1200 mg/kg, 400mg/kg, respectively. The day of administration was taken as day 1 of the test. General conditions of each group of cynomolgus monkeys were observed for 14 consecutive days after administration; body weight was measured before dosing and on days 4, 9, and 14 of the experiment; measuring the food intake on the 2 nd to 3 th, 8 th to 9 th and 12 th to 13 th days of the test; measuring body temperature, II-lead electrocardiogram and blood pressure before administration, about 0-1 hour after administration and 14 days of experiment; performing hematology and blood biochemical detection on the 4 th and 14 th days of the test; all cynomolgus monkeys of each group were anesthetized on day 15 of the experiment and euthanized for anatomical observation. The test results show that: the cynomolgus monkey is injected with 200mg/kg and 400mg/kg of the product by single intravenous injection, and no obvious abnormality is found in general observation, weight, ingestion, body temperature, electrocardiogram, blood pressure, hematology, hematobiochemistry, urine routine and general anatomy, and the Maximum Tolerated Dose (MTD) is 400 mg/kg.
The long-term toxicity test results showed that cynomolgus monkeys were intravenously injected with the pharmaceutical composition of the present application containing hu5G11-hIgG1 for 4 weeks, and were recovered from withdrawal for 4 weeks with no toxic reactive agent (NOAEL) of 200 mg/kg.
Example 3 clinical phase I protocol and safety, tolerability results
This example discloses pharmacokinetic and tolerability preliminary efficacy results for a single-center, open, dose escalation clinical phase I trial.
Clinical phase I tolerance and pharmacokinetic studies patients with advanced malignancies who were diagnosed unequivocally, failed standard therapy or lacked standard therapy were enrolled, the safety and tolerance using pharmaceutical compositions containing hu5G11-hIgG1 were observed, and the Maximum Tolerated Dose (MTD) and dose-limiting toxicity (DLT) were determined.
Primary end point: DLT; and (4) MTD.
Secondary endpoint: evaluating pharmacokinetics; the antitumor treatment effect is preliminarily examined.
Safety and tolerability results show that 1mg/kg, 3mg/kg, 10mg/kg, 20mg/kg, 30mg/kg of pharmaceutical composition containing hu5G11-hIgG1 was administered to the patients in the group. The administration was continued every 21 days for one cycle. The results show that patients currently in the group are well tolerated during continuous dosing and that the adverse effects associated with the drug do not exceed class II. The dosage can be tolerated at present; detecting the cytokines in the group with the dosage of 1-10mg/kg, and detecting the cytokines without a cytokine storm; serious adverse reactions associated with immunity were also not found for a while.
Preliminary pharmacokinetic evaluation shows that after the test drugs are injected into the patients at different doses (1mg/kg, 3mg/kg and 10mg/kg) through intravenous infusion for the first time, the serum drug exposure level of the patients who have been injected into the patients presents obvious dose dependence and shows linear pharmacokinetic characteristics.
Example 4 clinical phase II trial for treatment of recurrent or metastatic squamous cell carcinoma of head and neck (R/M SCCHN)
The main purpose is as follows: overall Survival (OS) of subjects treated with the pharmaceutical composition comprising hu5G11-hIgG1 compared to placebo was evaluated in recurrent or metastatic head and neck squamous cell carcinoma first-line subjects receiving standard chemotherapy (cisplatin + 5-fluorouracil or carboplatin + 5-fluorouracil) of investigator choice.
The secondary purpose is as follows: in a first line subject of recurrent or metastatic head and neck squamous cell carcinoma who received standard chemotherapy (cisplatin + 5-fluorouracil or carboplatin + 5-fluorouracil) of investigator choice: evaluating duration of remission (DOR) in subjects treated with the pharmaceutical composition comprising hu5G11-hIgG1 as compared to placebo; evaluating Progression Free Survival (PFS) of subjects treated with the pharmaceutical composition comprising hu5G11-hIgG1 as compared to placebo; evaluating the Objective Remission Rate (ORR) of a subject treated with the pharmaceutical composition comprising hu5G11-hIgG1 compared to placebo; evaluating the Disease Control Rate (DCR) of subjects receiving treatment with the pharmaceutical composition comprising hu5G11-hIgG1 compared to placebo; evaluating the safety and immunogenicity of the treatment with the pharmaceutical composition comprising hu5G11-hIgG 1; the quality of life of subjects treated with the pharmaceutical composition comprising hu5G11-hIgG1 was evaluated.
Primary end point: overall Survival (OS): from random grouping to the time of death due to various causes.
Secondary endpoint: (1) duration of remission (DOR): for patients with optimal remission as Complete Remission (CR) or Partial Remission (PR), defined as the time from the first appearance of CR or PR to disease relapse or progression (assessed on independent images) or death from various causes, as assessed by the investigator; for patients who reached remission, had no disease progression before analysis, or tumor recurrence or death, DOR was terminated by the date of last disease assessment. (2) Progression Free Survival (PFS): refers to the time from random grouping to the first occurrence of progression or relapse or death due to various causes. (3) Objective Remission Rate (ORR): after receiving study drug treatment, Objective Remission Rate (ORR) was the percentage of Complete Remission (CR) + Partial Remission (PR) as evaluated according to RECIST1.1 criteria. (4) Disease Control Rate (DCR): is the percentage of Complete Remission (CR) + Partial Remission (PR) + Stable Disease (SD).
Progression Free Survival (PFS): defined as the time from the first dose until objective tumor progression or death.
Overall Survival (OS): defined as the time from the start of the first dose to death due to any cause. In days, the time of the last follow-up is usually calculated as the time of death for an unsuccessfully visited subject.
Duration of remission (DOR): defined as the time from the first evaluation as CR or PR to the first evaluation as PD or death.
Time To Remission (TTR): the subject is dosed for the first time beginning to the time of first evaluation as CR or PR.
Safety evaluation index: (1) the incidence, relevance to the test drug and severity of adverse events and severe adverse events in the treatment of pharmaceutical compositions comprising hu5G11-hIgG 1. (2) Changes in vital signs, physical examination results and laboratory results before, during and after treatment were studied. (3) The immunogenicity (ADA) of the treatment with the pharmaceutical composition containing hu5G11-hIgG1 was evaluated.
Exploratory study evaluation: (1) discovery of biomarkers related to efficacy, mechanism of action, safety, and/or pathological mechanism. For the determination of biomarkers, including but not limited to PD-L1 expression, tumor specimens were collected during the screening period. Tumor tissue biomarkers, including cellular, deoxyribonucleic acid (DNA), ribonucleic acid (RNA), or protein levels, that may be associated with anti-tumor immune response and/or disease progression during treatment with pharmaceutical compositions comprising hu5G11-hIgG1 or in combination with standard chemotherapy. (2) The Pharmacokinetic (PK) profile of hu5G11-hIgG1 following co-administration of cisplatin/carboplatin, 5-fluorouracil and pharmaceutical compositions containing hu5G11-hIgG1 is described.
The experimental study was conducted to evaluate the combination of pharmaceutical compositions containing hu5G11-hIgG1Combination of standard chemotherapy (cisplatin + 5-fluorouracil or carboplatin + 5-fluorouracil) versus standard chemotherapy was used in a multicenter, randomized, double-blind clinical study of first-line treatment of patients with recurrent or metastatic squamous cell carcinoma of the head and neck (R/M SCCHN). The experimental group received the hu5G11-hIgG1 pharmaceutical composition in combination with standard chemotherapy (cisplatin + 5-fluorouracil or carboplatin + 5-fluorouracil) regimen and the control group received placebo in combination with chemotherapy (cisplatin + 5-fluorouracil or carboplatin + 5-fluorouracil) regimen for 1 cycle at 3 weeks (21 days) for a total of 6 cycles. The first day of each cycle (D1) was given intravenously the hu5G11-hIgG1 pharmaceutical composition 1200 mg/placebo, and after completion of hu5G11-hIgG1 pharmaceutical composition/placebo infusion, standard chemotherapy regimens were started: cisplatin 75mg/m2Intravenous infusion (D1), or carboplatin AUC5 intravenous infusion (D1); 5-Fluorouracil 750mg/m2Intravenous infusion (D1-D5), subjects will receive up to 6 cycles of hu5G11-hIgG1 pharmaceutical composition/placebo combined standard chemotherapy, followed by continued hu5G11-hIgG1 pharmaceutical composition/placebo maintenance therapy for 3 weeks (21 days) for 1 cycle, hu5G11-hIgG1 pharmaceutical composition/placebo dosing time of no more than 96 weeks, until disease progression, intolerable toxicity or investigator/subject decision to withdraw from study.
The study population is as follows: recurrent/metastatic squamous cell carcinoma of head and neck (R/M SCCHN) patients.
Inclusion criteria (subjects must meet all of the following criteria to be able to be included): 1) histologically or cytologically confirmed Squamous Cell Carcinoma of Head and Neck (SCCHN) patients with oropharyngeal, oral, hypopharynx or laryngeal primary sites; 2) recurrent/metastatic SCCHN without indications of a partial radical therapy; 3) at least one measurable lesion (assessed according to RECIST 1.1); 4) tumor tissue samples can be provided for PD-L1 expression detection, fresh biopsy tissues are preferentially selected for detection, and archived tissue wax blocks can be provided for detection after slicing if the biopsy tissues can not be provided for detection; 5) the tumor expresses PD-L1, and the Tumor Proportion Score (TPS) is more than or equal to 1 percent; 6) the age is more than or equal to 18 years when signing informed consent; 7) eastern american tumor cooperative group (ECOG) performance status: 0-1 min; 8) the expected life span exceeds 3 months; 9) the major organs function normally.
Exclusion criteria (subjects who meet one or more of the following criteria will be excluded): 1) receiving systemic chemotherapy, but not chemotherapy for locally advanced disease as part of a multimodal treatment; chemotherapy for locally advanced disease includes: inducing chemotherapy, radiotherapy synchronous chemotherapy and adjuvant chemotherapy; 2) treatment for locally advanced SCCHN completes disease progression within 6 months; 3) previous immunotherapy received anti-PD-1, anti-PD-L1, anti-PD-L2, anti-CD 137, or anti-CTLA-4 antibodies or any other antibody or drug specifically targeted to T cell co-stimulation or immune checkpoint pathways; 4) receive cetuximab therapy within 6 months prior to randomization; 5) researchers believe patients who are not eligible for inclusion in the group for other reasons.
Key abort test criteria: the sponsor has the right to terminate the study at any time, but must inform the investigator, ethics committee and national food and drug administration in advance and clarify the reason. Reasons for terminating the study include, but are not limited to, the following: 1. the number and severity of adverse events occurring in this study suggest that continuing the study will cause significant harm to the subject; 2. the group entering speed of the research center is too slow, and the group entering plan cannot be completed within a limited time; 3. the quality of the existing clinical data is not good, which is not beneficial to the continuous development of follow-up research; 4. the study was terminated due to changes in the national drug administration regulatory policies.
Test medication: the pharmaceutical composition containing hu5G11-hIgG1 is 100mg/10mL, is stored at 2-8 ℃ in a dark place, and has a temporary validity period of 24 months, and is prepared by the following steps: the Ningdanqing pharmaceutical industry group, Inc.
Control drugs: placebo, size 10 mL.
hu5G11-hIgG1 pharmaceutical composition/placebo formulation method: the aseptic operation principle should be strictly observed in the configuration process. The required dose of hu5G11-hIgG1 pharmaceutical composition/placebo was drawn from a glass vial under sterile conditions and diluted with sterile pyrogen-free physiological saline to a 250mL polyvinyl chloride (PVC), Polyethylene (PE) or Polyolefin (PO) infusion bag. Mix the diluted solution gently inverted without shaking. As a medicine for injection, the presence or absence of particulate matter and discoloration should be visually checked before administration. The medicine is clear to microemulsion light, and is colorless to light yellow liquid. If cloudiness, discoloration, or the presence of visible particles is observed in the solution, it should be discarded. The vial was not shaken. The product contains no antiseptic. Once prepared, administration is immediate.
The preparation method of the chemotherapeutic drug comprises the following steps: in standard chemotherapy regimens, "cisplatin" or "carboplatin" and "5-fluorouracil" are formulated with reference to the respective package insert.
Test drug management: according to the GCP requirement, research medicines are kept, issued, configured, used and recycled by special persons in hospitals. Used and partially used drug containers, residual drug solutions, empty vials, infusion bags and syringes may be destroyed locally according to the guidelines and procedures established by research centers, local agencies, and the drug packages are recycled and returned to the sponsors. For unused medications, return to the sponsor is required unless there is a significant safety issue with the drug contents that requires immediate destruction in accordance with local regulations. The dispensing, configuration, use and recovery of test drugs require complete records. The use and record condition of the medicine is checked regularly by an inspector, and the recovery condition is inspected at any time.
Study treatment
Study treatment should begin on the day of randomization or as close as possible to the date the treatment was dispensed.
Dispensing therapy
Subjects will receive treatment with one of the following two groups (test and control) (table 2):
TABLE 2 Allocation therapy
Figure PCTCN2020074083-APPB-000006
Note: & hu5G11-hIgG 1/placebo was administered prior to standard chemotherapy for no more than 96 weeks; platinum drugs (cisplatin/carboplatin) can be selected by researchers according to the condition of the subjects, and the dose of the carboplatin cannot exceed 800 mg; # end point event: disease progression, intolerable toxicity or investigator/subject decision to withdraw from study.
Administration of drugs
Dose selection
hu5G11-hIgG 1/placebo: the first day of each cycle (D1) used hu5G11-hIgG 1/placebo, 1200 mg/placebo, D1, intravenous drip. 3 weeks (21 days) is a treatment cycle with dosing times not exceeding 96 weeks until disease progression, intolerable toxicity or the investigator/subject decision to withdraw from the study.
Standard chemotherapeutic drugs: after completion of hu5G11-hIgG 1/placebo infusion, standard chemotherapy regimens were given on days 1 (D1) to 5 (D5).
Standard chemotherapy regimens are as follows:
cisplatin/carboplatin: cisplatin 75mg/m2Carboplatin AUC5, D1, intravenous infusion;
5-fluorouracil: 750mg/m2D1-D5, intravenous infusion;
cis-platinum
The dose of cisplatin should be calculated as the Body Surface Area (BSA) of the subject, calculated before each dose. For BSA > 2.0m2According to 2.0m2The dose is calculated.
At least 1 hour was left between the end of the hu5G11-hIgG 1/placebo infusion and the start of cisplatin infusion. Intravenous pre-hydration can be performed simultaneously with hu5G11-hIgG 1/placebo infusion, but a separate infusion line must be used. Because cisplatin is highly emetogenic, it must be prevented from vomiting to prevent acute or delayed nausea and vomiting.
Carboplatin
The dose of carboplatin was calculated based on the renal function (creatinine clearance) of the subject using the formula AUC5, maximum dose of 800 mg.
At least 1 hour was left between the end of the hu5G11-hIgG 1/placebo infusion and the beginning of the carboplatin infusion.
5-Fluorouracil
5-Fluorouracil was calculated from the Body Surface Area (BSA) of the subjects, which was calculated prior to each dose. 5-Fluorouracil should be administered after cisplatin/carboplatin administration is complete and the same infusion line can be used.
Dose adjustment
If appropriate, investigators attribute each toxicity event to cisplatin/carboplatin alone, 5-fluorouracil or hu5G11-hIgG 1/placebo, or combination therapy, as well as to gradual dose reduction as possible.
The subject may stop chemotherapy and continue hu5G11-hIgG 1/placebo treatment alone. Likewise, subjects may stop hu5G11-hIgG 1/placebo and continue individual chemotherapy (as appropriate) for the first 6 cycles.
Time of administration
The subjects will receive the blinded hu5G11-hIgG 11200 mg (D1) or placebo (D1) in combination with cisplatin 75mg/m2Carboplatin AUC 5(D1) + 5-fluorouracil 750mg/m2(D1-D5) Q3W for 6 cycles, then received blinded hu5G11-hIgG 11200 mg (D1) or placebo (D1) Q3W until disease progression/treatment was complete. hu5G11-hIgG 1/placebo will be administered prior to chemotherapy. Trial treatments should be administered on day 1 of each cycle, after all procedures and assessments are completed. The test treatment may be administered on day 1 +3 of the target per cycle, with cycle 1 being administered only on day 1 +3 of the target.
hu5G11-hIgG 1/placebo dosing time: the administration is carried out by using an infusion apparatus which is provided with an aseptic, pyrogen-free and low-protein adsorption online filter (the pore diameter is 0.2-0.22 mu m), and the infusion is completed within 60 minutes. If the first infusion is tolerated by the subject, subsequent administrations will complete the infusion in 30 minutes. The infusion set is not required to be shared with other medicines. The study center should be as complete as possible to bring the hu5G11-hIgG1 containing pharmaceutical composition/placebo infusion time as close to 60 minutes as possible. However, a-10 minute to +20 minute time window (i.e., infusion time 50-80 minutes) is allowed to account for variations in infusion pumps between different centers.
Cisplatin/carboplatin dosing time: according to local practice and specification requirements, cisplatin is 75mg/m2Q3W will be administered by 60-120 min intravenous infusion for 6 cycles. Carboplatin AUC5mg/mL/min60 min was administered by intravenous infusion of Q3W for 6 cycles.
5-Fluorouracil dosing time: according to local practice and specification requirements, 5-fluorouracil 750mg/m224 is smallContinuous infusion was given as intravenous infusion of Q3W for 6 cycles.
Evaluation of effectiveness
The main efficacy end point: overall Survival (OS): time to death from enrollment to various causes.
Secondary efficacy endpoints: duration of remission (DOR): for patients with optimal remission as Complete Remission (CR) or Partial Remission (PR), defined as the time from the first appearance of CR or PR to relapse or progression of the disease (based on independent assessment) or death from various causes, as assessed by the investigator; for patients who reached remission, had no disease progression before analysis, or tumor recurrence or death, DOR was terminated by the date of last disease assessment.
Progression Free Survival (PFS): refers to the time from enrollment until the first appearance of progression or relapse or death from various causes.
Objective Remission Rate (ORR): after receiving study drug treatment, Objective Remission Rate (ORR) was the percentage of Complete Remission (CR) + Partial Remission (PR) as evaluated according to RECIST1.1 criteria.
Disease Control Rate (DCR): is the percentage of Complete Remission (CR) + Partial Remission (PR) + Stable Disease (SD).
Imaging examination
The imaging examination of this study requires a multi-row spiral ct (multiple detector ct) enhanced scan. The examined part is the whole body, including the neck, chest, abdomen and pelvic cavity. To reduce the detection error between scanners, the test requires the same consistent inspection method to be used for inspection. In the case of patients with sensitivity to CT contrast agents, Magnetic Resonance (MR) imaging can be used as an alternative to CT imaging. Otherwise, please select the same examination means as the baseline. During the study, outside of the conventional examination points and sites, if a new lesion is suspected, the investigator decides to add another corresponding examination. All experimental imaging data and associated additional imaging examination data are required to be archived for retrospective confirmation.
Evaluation of therapeutic Effect
The study mainly uses RECIST1.1 standard for efficacy evaluation.
irRECIST is an adapted version of RECIST1.1, the purpose of which is to explain the unique tumor remission of immunotherapeutic drugs. irRECIST will be used in imaging evaluation by researchers at the study center to assess tumor remission and progression and to make treatment decisions. Subjects should not discontinue treatment if feasible until imaging evaluation by the investigator at the study center confirms disease progression. Given that some subjects may develop a transient tumor outbreak within the first few months after starting immunotherapy and then have disease remission, subjects are allowed to continue to receive therapy after the first appearance of imaging Progression (PD). For subjects judged to be clinically unstable, PD need not be confirmed by repeated tumor imaging examinations.
A tumor outbreak includes any of the following: 1) worsening of the original target lesion; 2) worsening of the original non-target lesion; 3) appearance of new lesions for subjects who were evaluated for preliminary evidence of imaging PD according to RECIST1.1, the primary investigator could decide on his own whether to continue to administer study treatment to the subject until imaging examination results are obtained again. The clinical judgment made by the investigator at the study center is based on the overall clinical status of the subject, including physical performance, clinical symptoms, and laboratory test results. Subjects may continue to receive study treatment and re-receive tumor assessments after 4 weeks or more for study centers to confirm PD according to irRECIST.
Clinical stability is defined as follows: 1) clinically significant symptoms and signs (including worsening laboratory test values) that suggest disease progression did not appear. 2ECOG performance status score did not decrease. 3) No rapid disease progression occurred. 4) The critical anatomical sites do not present progressive tumors (e.g., spinal cord compression) that require other urgent medical intervention.
Subjects judged to be clinically unstable should discontinue trial therapy starting with first-evaluated radiographic evidence of PD and not require confirmation of PD by repeated radiographic examinations.
In determining whether tumor burden increases or decreases according to irRECIST, researchers at the research center should consider all target and non-target lesions, as well as any newly emerging lesions.
According to irRECIST, PD cannot be confirmed if, in repeated imaging examinations, all of the following occur: 1) the sum of the diameters of the target lesions increased < 20% above the lowest value, or the absolute value increased <5 mm. 2) Resulting in stabilization or improvement of the non-target focal condition of the initial PD. 3) Resulting in stabilization or improvement of the new focal condition of the original PD. 4) New lesions did not increase since the last evaluation. 5) New non-target lesions did not progress since the last evaluation.
Repeated imaging examinations by investigators at the study center, as assessed by irRECIST, did not confirm PD, and the subject's clinical condition remained stable, the study treatment continued and the imaging examinations performed on a normal schedule.
According to irRECIST, PD can be confirmed if, in repeated imaging examinations, any of the following occurs: 1) the sum of the diameters of the target lesions is increased by more than or equal to 20 percent compared with the minimum value, and the absolute value is increased by at least 5 mm. 2) Leading to worsening of the non-target focal condition of the initial PD. 3) Leading to worsening of the new focal condition of the original PD. 4) New lesions appeared since the last evaluation. 5) The new non-target lesion progressed again since the last evaluation and if any of the above events occurred in repeated imaging examinations, resulting in confirmation of PD, the subject would be discontinued from study treatment.
Statistical analysis
Sample size
The test is a multi-center, random, double-blind study, the main endpoint index is overall survival time (OS), and Lorrank difference test is adopted. Assuming that subjects received 8.5 months of smos treated with cisplatin/carboplatin + 5-fluorouracil, the mOS treated with the test drug hu5G11-hIgG1 in combination with cisplatin/carboplatin + 5-fluorouracil was expected to be 12 months, i.e., 3.5 months (HR 0.70) improvement over the standard chemotherapy regimen of the control group, as bilateral α 0.05, with 80% confidence (1- β), 18 months for the group, 18 months for the follow-up, no more than 15% for the test control shedding rate, sample size calculated using PASS11 at a 1:1 group ratio, at least 265 events were obtained in this study, and 334 samples were obtained in the group (167 in the test group and the control group, respectively).
Statistical analysis set
Full assay set (FAS, Full analysis set): all subjects enrolled in the group and administered at least one trial dose.
Protocol analysis Set (PPS, Per Protocol Set): in the total analysis set, subjects who have completed medication according to the protocol, have not used contraband drugs, have not seriously violated the protocol, and subjects who quit the test due to the occurrence of an endpoint event will be included in the PP set.
Safety data Set (SS, Safety Set): the cases with the study drug used at least once after the group entry and with safety evaluation data.
Analysis of efficacy
Efficacy assays will be performed simultaneously in FAS and PPS.
In the main efficacy index overall survival time (OS) of the study, the median survival time and 95% CI thereof were calculated by the Kaplan-Meier method, and a survival curve graph was drawn.
In Objective Remission Rate (ORR) analysis, the ORR of patients and their 95% CI were calculated.
The analysis indexes of the survival period comprise: duration of reaction (DOR), Progression Free Survival (PFS), median survival and 95% CI were calculated using the Kaplan-Meier method and plotted as a survival graph. In Objective Remission Rate (ORR) and Disease Control Rate (DCR) analysis, the ORR and DCR of patients and their 95% CI were calculated.
Security analysis
The safety analysis population, including all patients who received any dose of study drug, will be analyzed according to the treatment the subject received.
Adverse events were described by the corresponding terms in the MeDRA dictionary and rated according to NCI CTCAE v 5.0. In addition, all serious adverse events will also be summarized.
Cases of death reported during study treatment and during follow-up after completion of treatment or termination of treatment will be summarized by treatment group.
Adverse events leading to treatment termination and early withdrawal from the study will be summarized by treatment group and reason for withdrawal.
In addition, study exposure will be summarized by treatment group, including number of cycles received and cumulative dose.
Laboratory data outside the normal range will be identified. Additional selected laboratory data were summarized by treatment group and NCI CTCAE v5.0 grading. Clinically significant abnormal laboratory data will be reported as adverse events and summarized in the adverse event table.
The positive rate of immunogenicity (ADA) in the subject is summarized.
Example 5 clinical phase II trial results for treatment of recurrent or metastatic squamous cell carcinoma of head and neck (R/M SCCHN)
1. The test subject in 2018 has a confirmed histopathological diagnosis of squamous cell carcinoma of throat in 2018, the pathological tissue acquisition way is surgery and lymph node metastasis occurs, and cisplatin chemotherapy is performed after the surgery in 8 months in 2018 to 10 months in 2018 to synchronously perform radiotherapy of laryngeal and lymph node regions. Relapse occurred in 2019 in 8 months. Cycle 1 treatment started on day 1 of month 10 2019 (hu5G11-hIgG1 injection 1200 mg; carboplatin 425 mg; 5-FU1080mg/24 h continuous instillation for 5 days); cycle 2 therapy started at 23.10.2019 (hu5G11-hIgG1 injection 1200 mg; carboplatin 478 mg; 5-FU1035mg/24 h continuous instillation for 5 days);
in terms of efficacy assessment, the subjects received hu5G11-hIgG1 injection/placebo in combination with carboplatin and fluorouracil for 1 cycle at 21 days, and continued dosing, with a 39.4% reduction in target lesions from baseline for primary tumor assessment at 11 months and 11 days in 2019, and efficacy assessment of PR.
Focal condition:
and (3) screening period: 65mm of target focus
After 6 weeks: a39.4% reduction in the target lesion at 39.4mm resulted in PR.
2.② test subject, pathological diagnosis is (hypopharyngeal) mesodifferentiation squamous cell carcinoma in 2018, 4 and 23 days, and radiotherapy is carried out in 2018, 5 and 14 days, 2018, 7 and 10 days. Liver metastases appeared in the 6 th month review in 2019. Cycle 1 therapy started 7/9/2019 (hu5G11-hIgG1 injection 1200 mg; carboplatin 442.1 mg; 5-FU5625mg/24 h continuous instillation for 5 days); cycle 2 therapy started on 31/7/2019 (hu5G11-hIgG1 injection 1200 mg; carboplatin 354.2 mg; 5-FU5601mg/24 h continuous instillation for 5 days); cycle 3 therapy started on day 21 of 8/2019 (hu5G11-hIgG1 injection 1200 mg; carboplatin 401.7 mg; 5-FU5577mg/24 h continuous instillation for 5 days); cycle 4 therapy started on 12.9.2019 (hu5G11-hIgG1 injection 1200 mg; carboplatin 385.5G; 5-FU5601mg/24 h continuous instillation for 5 days); cycle 5 therapy started on 30.9/2019 (hu5G11-hIgG1 injection 1200 mg; carboplatin 394.2 mg; 5-FU5601mg/24 h continuous instillation for 5 days); cycle 6 therapy started on day 23 of month 10 2019 (hu5G11-hIgG1 injection 1200 mg; carboplatin 388.9 mg; 5-FU5553mg/24 h continuous instillation for 5 days); cycle 7 treatment starting on day 3 of 12 months in 2019 (hu5G11-hIgG1 injectable 1200 mg);
in terms of efficacy assessment, the subjects received hu5G11-hIgG1 injection/placebo in combination with carboplatin and fluorouracil for 1 cycle at 21 days, and continued dosing, with a 38.4% reduction in baseline and PR efficacy assessment of the target lesion at the initial tumor assessment day 8 and 19 in 2019.
Focal condition:
and (3) screening period: target lesion 151.5mm
After 6 weeks: a93.3 mm reduction in target lesion of 38.4% results in PR
After 12 cycles: a46.9% reduction in 80.5mm of the target lesion resulted in PR
After 18 cycles: a46.9% reduction of 80.4mm in the target lesion resulted in PR.
3.③ test subject, 6.2019, 25.8.6, laryngoscope biopsy is pathologically diagnosed as pharyngeal squamous cell carcinoma, and the pathological tissue acquisition way is laryngoscope biopsy, and lymph node metastasis occurs with mediastinal lymph node metastasis. Cycle 1 therapy started on day 11, 7/2019 (hu5G11-hIgG1 injection: 1200 mg; carboplatin: 800 mg; 5-FU: 6558.75mg/24 h continuous instillation for 5 days); cycle 2 therapy started at month 8, 1, 2019 (hu5G11-hIgG1 injection 1200 mg; carboplatin 800 mg; 5-FU 6558.75mg/24 h continuous instillation for 5 days);
in terms of efficacy assessment, the subjects received hu5G11-hIgG1 injection/placebo in combination with carboplatin and fluorouracil for 1 cycle for 21 days, and continued dosing, with a 17% reduction in target lesions from baseline for primary tumor assessment on day 21 of 08 month 2019, an SD for efficacy assessment, and a 64.3% reduction in target lesions from baseline for secondary efficacy assessment on day 29 of 9 month 2019, a PR for efficacy assessment.
Focal condition:
and (3) screening period: target focus 28mm
After 6 weeks: the 23mm reduction in the target lesion by 17% resulted in SD
After 12 weeks: a10 mm reduction in the target lesion by 64.3% resulted in PR.
The clinical experiment results show that the hu5G11-hIgG1 injection can effectively inhibit and treat head and neck tumors and reduce target lesions.
While the compositions and methods of this application have been described in terms of preferred embodiments in light of the present disclosure, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the application.
The disclosures of all documents cited herein are incorporated by reference herein, to the extent that they provide exemplary, procedural and other details supplementary to those set forth herein.

Claims (42)

  1. A method of treating, ameliorating or improving cancer in a subject comprising administering to the subject a therapeutically effective amount of an anti-PD-L1 antibody that is an inhibitor of the interaction between a PD-1 receptor and its ligand PD-L1, wherein the cancer is Squamous Cell Carcinoma of Head and Neck (SCCHN), and wherein the anti-PD-L1 antibody comprises the amino acid sequence: a heavy chain CDR1 region having at least 80% homology to the amino acid sequence set forth in SEQ ID NO.1 or SEQ ID NO. 4; a heavy chain CDR2 region having at least 80% homology to the amino acid sequence set forth in SEQ ID NO. 2 or SEQ ID NO. 5; a heavy chain CDR3 region having at least 80% homology to the amino acid sequence set forth in SEQ ID NO. 3 or SEQ ID NO. 6; a light chain CDR1 region having at least 80% homology to the amino acid sequence set forth in SEQ ID NO. 7 or SEQ ID NO. 10; a light chain CDR2 region having at least 80% homology to the amino acid sequence set forth in SEQ ID NO. 8 or SEQ ID NO. 11; a light chain CDR3 region having at least 80% homology to the amino acid sequence set forth in SEQ ID NO. 9 or SEQ ID NO. 12.
  2. The method of claim 1, wherein the anti-PD-L1 antibody comprises a conservative substitution variant selected from one or more of SEQ ID NO 1, SEQ ID NO 2, SEQ ID NO 3, SEQ ID NO 4, SEQ ID NO 5, SEQ ID NO 6, SEQ ID NO 7, SEQ ID NO 8, SEQ ID NO 9, SEQ ID NO 10, SEQ ID NO 11, SEQ ID NO 12, SEQ ID NO 13, SEQ ID NO 14, SEQ ID NO 15, SEQ ID NO 16, SEQ ID NO 17, SEQ ID NO 18, SEQ ID NO 19, SEQ ID NO 20, and SEQ ID NO 21 and the anti-PD-L1 antibody retains the ability to specifically recognize and bind PD-L1.
  3. The method of claim 1 or 2, wherein the anti-PD-L1 antibody comprises: a heavy chain CDR1 region having the amino acid sequence shown in SEQ ID NO.1, a heavy chain CDR2 region having the amino acid sequence shown in SEQ ID NO. 2, a heavy chain CDR3 region having the amino acid sequence shown in SEQ ID NO. 3; and a light chain CDR1 region having the amino acid sequence shown in SEQ ID NO. 7, a light chain CDR2 region having the amino acid sequence shown in SEQ ID NO. 8, and a light chain CDR3 region having the amino acid sequence shown in SEQ ID NO. 9.
  4. The method according to any one of the preceding claims, wherein the anti-PD-L1 antibody is administered about every week (q1w), about every 2 weeks (q2w), about every 3 weeks (q3w), or about every 4 weeks (q4 w).
  5. The method according to any one of the preceding claims, wherein the anti-PD-L1 antibody is administered in one or more uniform doses effective to treat the cancer.
  6. The method according to any one of the preceding claims, wherein the patient is administered a uniform dose of the anti-PD-L1 antibody about every 3 weeks.
  7. The method of any one of claims 5-6, wherein the unitized dose is in the range of about 20mg to about 2400mg of anti-PD-L1 antibody.
  8. The method according to any one of claims 5-7, wherein the unitized dose is selected from about 300mg, about 600mg, about 900mg, about 1000mg, about 1200mg, about 1500mg, about 1800mg, about 2100mg, or about 2400mg of anti-PD-L1 antibody.
  9. The method according to any one of claims 5-8, wherein the unitized dose is selected from about 1200mg of anti-PD-L1 antibody.
  10. The method according to any one of claims 5-9, wherein the anti-PD-L1 antibody is administered at a dose of about 1200mg per patient, about once every 3 weeks, for a sustained period.
  11. The method according to any one of claims 1-4, wherein the anti-PD-L1 antibody is administered at a dose of 1mg/kg, 3mg/kg, 10mg/kg, 15mg/kg, 20mg/kg, 25mg/kg, 30mg/kg body weight for continuous dosing.
  12. The method of any one of the preceding claims, wherein the anti-PD-L1 antibody is administered as an intravenous infusion.
  13. The method of any one of the preceding claims, wherein the anti-PD-L1 antibody is administered as a 1-2 hour intravenous infusion, preferably a 1 hour intravenous infusion.
  14. The method of any one of the preceding claims, wherein the anti-PD-L1 antibody is a naked antibody, an intact antibody, or an antibody fragment comprising an antigen binding region.
  15. The method according to any of the preceding claims, wherein the method results in an objective response, preferably a complete response or a partial response.
  16. The method of any one of the preceding claims, wherein the subject has not previously received systemic chemotherapy.
  17. The method of any one of the preceding claims, wherein the subject has previously received surgical treatment, radiation therapy, induction chemotherapy, concurrent chemotherapy, and/or adjuvant chemotherapy.
  18. The method of any one of the preceding claims, wherein the subject has not previously received systemic chemotherapy, but has received surgical treatment, radiation therapy, induction chemotherapy, concurrent chemotherapy, and/or adjuvant chemotherapy.
  19. The method of any of the preceding claims, wherein the subject has achieved complete remission following surgical treatment, radiation therapy, induced chemotherapy, concurrent chemotherapy and/or adjuvant chemotherapy before disease progression occurs again.
  20. The method of any one of the preceding claims, wherein the subject has failed to complete remission or partial remission following surgical treatment, radiation treatment, induced chemotherapy, concurrent chemotherapy, and/or adjuvant chemotherapy.
  21. The method of any one of the preceding claims, wherein the cancer of the subject has metastasized.
  22. The method according to any preceding claim, wherein the primary site of head and neck squamous cell carcinoma is oropharynx, oral cavity, hypopharynx, and/or larynx.
  23. The method of any one of the preceding claims, wherein the cancer is recurrent and/or metastatic head and neck squamous cell carcinoma.
  24. The method according to any preceding claim, wherein the recurrent or metastatic head and neck squamous cell carcinoma is that without indication of a partial radical treatment.
  25. The method according to any preceding claim, wherein the squamous cell carcinoma of the head and neck is refractory.
  26. The method of any preceding claim, wherein the treatment of cancer is a first line treatment of recurrent or metastatic head and neck squamous cell carcinoma.
  27. The method according to any of the preceding claims, wherein the treatment is a first line treatment of recurrent or metastatic head and neck squamous cell carcinoma in combination with standard chemotherapy.
  28. The method of claim 27, wherein the standard chemotherapy comprises administering a chemotherapeutic agent to the patient.
  29. The method of any one of claims 27-28, wherein the standard chemotherapy comprises administering cisplatin and 5-fluorouracil to the patient, or carboplatin and 5-fluorouracil to the patient.
  30. The method of claim 29, wherein the anti-PD-L1 antibody and the chemotherapeutic agent are each in the form of a pharmaceutical composition that can be administered simultaneously, sequentially, or at intervals.
  31. The method of any one of claims 27-30, wherein the treatment is for 3 weeks (21 days) as a cycle, 1200mg of anti-PD-L1 antibody is administered intravenously the first day (D1) of each cycle, and a standard chemotherapy regimen is administered after the anti-PD-L1 antibody infusion.
  32. The method of claim 31, wherein the standard chemotherapeutic regimen comprises: the first day (D1) cisplatin was administered at 75mg/m by intravenous infusion2Or carboplatin AUC5 by intravenous infusion and 5-fluorouracil 750mg/m by intravenous infusion on day one to day five (D1-D5)2
  33. The method of any one of claims 31-32, wherein the standard chemotherapeutic regimen comprises: the first day (D1) cisplatin was administered at 75mg/m by intravenous infusion2And 5-Fluorouracil was administered by intravenous infusion on day one to day five (D1-D5) at 750mg/m2
  34. The method of any one of claims 31-32, wherein the standard chemotherapeutic regimen comprises: carboplatin AUC5 was given by intravenous infusion or intravenous infusion on day one (D1), and 5-fluorouracil 750mg/m was given by intravenous infusion on day one to day five (D1-D5)2
  35. A combination pharmaceutical composition for treating recurrent or metastatic head and neck squamous cell carcinoma comprising an anti-PD-L1 antibody and a chemotherapeutic agent, wherein the anti-PD-L1 antibody comprises the amino acid sequence: a heavy chain CDR1 region having at least 80% homology to the amino acid sequence set forth in SEQ ID NO.1 or SEQ ID NO. 4; a heavy chain CDR2 region having at least 80% homology to the amino acid sequence set forth in SEQ ID NO. 2 or SEQ ID NO. 5; a heavy chain CDR3 region having at least 80% homology to the amino acid sequence set forth in SEQ ID NO. 3 or SEQ ID NO. 6; a light chain CDR1 region having at least 80% homology to the amino acid sequence set forth in SEQ ID NO. 7 or SEQ ID NO. 10; a light chain CDR2 region having at least 80% homology to the amino acid sequence set forth in SEQ ID NO. 8 or SEQ ID NO. 11; a light chain CDR3 region having at least 80% homology to the amino acid sequence set forth in SEQ ID NO. 9 or SEQ ID NO. 12.
  36. The combination pharmaceutical composition of claim 35, wherein the combination pharmaceutical composition comprises a pharmaceutical composition comprising an anti-PD-L1 antibody and a pharmaceutical composition comprising a chemotherapeutic agent.
  37. The combination pharmaceutical composition according to any one of claims 35-36, wherein the anti-PD-L1 antibody comprises a conservative substitution variant selected from one or more of SEQ ID No.1, SEQ ID No. 2, SEQ ID No. 3, SEQ ID No. 4, SEQ ID No. 5, SEQ ID No. 6, SEQ ID No. 7, SEQ ID No. 8, SEQ ID No. 9, SEQ ID No. 10, SEQ ID No. 11, SEQ ID No. 12, SEQ ID No. 13, SEQ ID No. 14, SEQ ID No. 15, SEQ ID No. 16, SEQ ID No.17, SEQ ID No.18, SEQ ID No. 19, SEQ ID No. 20 and SEQ ID No. 21, and the anti-PD-L1 antibody retains the ability to specifically recognize and bind PD-L1.
  38. The combination pharmaceutical composition according to any one of claims 35-37 wherein the chemotherapeutic agent is selected from cisplatin, carboplatin and/or 5-fluorouracil.
  39. The pharmaceutical combination composition of any one of claims 36-38, wherein the pharmaceutical composition comprising the anti-PD-L1 antibody is an article of manufacture stored in a container comprising a fixed dose of the anti-PD-L1 antibody selected from the group consisting of about 300mg, about 600mg, about 900mg, about 1000mg, about 1200mg, about 1500mg, about 1800mg, about 2100mg, and about 2400mg of the anti-PD-L1 antibody.
  40. The combination pharmaceutical composition of claim 39, wherein the preparation comprises 1 or more than 1 vial containing about 300mg or 600mg of anti-PD-L1 antibody.
  41. The pharmaceutical combination composition of any one of claims 39-40, wherein the article of manufacture further comprises a package insert or a package insert instructing the user to administer the fixed dose to a cancer patient.
  42. A kit for treating recurrent or metastatic head and neck squamous cell carcinoma, the kit comprising the anti-PD-L1 antibody-containing pharmaceutical composition of any one of claims 36-41 and a chemotherapeutic agent, and instructions for the use of the anti-PD-L1 antibody and chemotherapeutic agent in combination for the treatment of recurrent or metastatic head and neck squamous cell carcinoma.
CN202080011333.8A 2019-01-31 2020-01-31 Use of anti-PD-L1 antibodies for the treatment of head and neck cancer Active CN113365659B (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN2019100957345 2019-01-31
CN201910095734 2019-01-31
PCT/CN2020/074083 WO2020156500A1 (en) 2019-01-31 2020-01-31 Use of anti-pd-l1 antibody in treatment of head and neck cancer

Publications (2)

Publication Number Publication Date
CN113365659A true CN113365659A (en) 2021-09-07
CN113365659B CN113365659B (en) 2023-08-18

Family

ID=71839951

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202080011333.8A Active CN113365659B (en) 2019-01-31 2020-01-31 Use of anti-PD-L1 antibodies for the treatment of head and neck cancer

Country Status (2)

Country Link
CN (1) CN113365659B (en)
WO (1) WO2020156500A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023098798A1 (en) * 2021-12-01 2023-06-08 正大天晴药业集团股份有限公司 Drug combination for treating non-small cell lung cancer

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103987405A (en) * 2011-11-28 2014-08-13 默克专利股份公司 Anti-PD-L1 antibodies and uses thereof
WO2016022630A1 (en) * 2014-08-05 2016-02-11 Jiping Zha Anti-pd-l1 antibodies
WO2017011666A1 (en) * 2015-07-14 2017-01-19 Bristol-Myers Squibb Company Method of treating cancer using immune checkpoint inhibitor
CN107460221A (en) * 2016-06-02 2017-12-12 正大天晴药业集团股份有限公司 A kind of cell culture processes for reducing protein polymer in anti-PD L1 antibody
WO2018178040A1 (en) * 2017-03-30 2018-10-04 Merck Patent Gmbh Combination of an anti-pd-l1 antibody and a dna-pk inhibitor for the treatment of cancer
CN109153723A (en) * 2016-05-13 2019-01-04 瑞泽恩制药公司 The treatment of cancer with combinations of anti-PD-1 antibody and radiation

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2015335029B2 (en) * 2014-10-24 2021-09-23 Astrazeneca Ab Combination
CN116327955A (en) * 2015-11-04 2023-06-27 杜克大学 Combination therapy of immunotoxins and checkpoint inhibitors

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103987405A (en) * 2011-11-28 2014-08-13 默克专利股份公司 Anti-PD-L1 antibodies and uses thereof
WO2016022630A1 (en) * 2014-08-05 2016-02-11 Jiping Zha Anti-pd-l1 antibodies
CN107001463A (en) * 2014-08-05 2017-08-01 中美冠科生物技术(太仓)有限公司 Anti- PD L1 antibody
WO2017011666A1 (en) * 2015-07-14 2017-01-19 Bristol-Myers Squibb Company Method of treating cancer using immune checkpoint inhibitor
CN107922502A (en) * 2015-07-14 2018-04-17 百时美施贵宝公司 Use the method for immunity inspection point inhibitor for treating cancer
CN109153723A (en) * 2016-05-13 2019-01-04 瑞泽恩制药公司 The treatment of cancer with combinations of anti-PD-1 antibody and radiation
CN107460221A (en) * 2016-06-02 2017-12-12 正大天晴药业集团股份有限公司 A kind of cell culture processes for reducing protein polymer in anti-PD L1 antibody
WO2018178040A1 (en) * 2017-03-30 2018-10-04 Merck Patent Gmbh Combination of an anti-pd-l1 antibody and a dna-pk inhibitor for the treatment of cancer

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
ALFIERI等: "Immunotherapy for recurrent/metastatic head and neck cancer", 《CURRENT OPINION IN OTOLARYNGOLOGY & HEAD AND NECK SURGERY》, vol. 26, no. 2, pages 152 - 156 *
BAUML等: "Pembrolizumab for Platinum- and Cetuximab-Refractory Head and Neck Cancer: Results From a Single-Arm, Phase II Study", 《JOURNAL OF CLINICAL ONCOLOGY:OFFICIAL JOURNAL OF THE AMERICAN SOCIETY OF CLINICAL ONCOLOGY》, vol. 35, no. 14, pages 1542 - 1549 *
CABEZAS-CAMARERO等: "Durable response to first-line nivolumab in a patient with oligometastatic PD-L1 positive nasopharyngeal cancer", 《ORAL ONCOLOGY》, vol. 87, pages 204 - 206, XP085559748, DOI: 10.1016/j.oraloncology.2018.10.019 *
SHETH等: "Pembrolizumab and its use in the treatment of recurrent or metastatic head and neck cancer", 《FUTURE ONCOLOGY》, vol. 14, no. 16, pages 11547 - 1558 *
毛璐等: "程序性细胞死亡受体-1与其配体信号通路的调控及其在头颈鳞状细胞癌治疗中的研究进展", 《国际口腔医学杂志》, vol. 45, no. 5, pages 560 - 565 *

Also Published As

Publication number Publication date
WO2020156500A1 (en) 2020-08-06
CN113365659B (en) 2023-08-18

Similar Documents

Publication Publication Date Title
CN107743401B (en) Compositions comprising a combination of an anti-PD-1 antibody and an additional antibody
JP2023039448A (en) Combination of pd-1 antagonist and vegfr/fgfr/ret tyrosine kinase inhibitor for treating cancer
US20200262922A1 (en) Stable formulations of anti-ctla4 antibodies alone and in combination with programmed death receptor 1 (pd-1) antibodies and methods of use thereof
CA2955184A1 (en) Combination of an ox40 agonist and a 4-1bb agonist for treating cancer
US20160031990A1 (en) Antagonists of pdl-1 and pd-1 for the treatment of hpv-negative cancers
KR20160108568A (en) Combination of a pd-1 antagonist and an ido1 inhibitor for treating cancer
CN111973739B (en) Use of anti-PD-L1 monoclonal antibodies for the treatment of cancer
CN113365659B (en) Use of anti-PD-L1 antibodies for the treatment of head and neck cancer
US20240050564A1 (en) Combination therapy using an anti-fucosyl-gm1 antibody
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
WO2021155840A1 (en) Use of anti-pd-1 antibody in treatment of malignant tumors
CN113117072A (en) Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody
CN113164599B (en) Use of anti-PD-L1 monoclonal antibodies for the treatment of cancer
EP3668894A1 (en) Treatment of ck8 positive cancers in relation with k-ras gene status
WO2023208001A1 (en) Combination of anti-pd-1 antibody and anti-egfr antibody, and use thereof in treatment of head and neck squamous cell carcinoma
WO2023134787A2 (en) Use of combination of anti-pd-1 antibody and anti-vegf antibody in treatment of hepatocellular carcinoma
WO2024050429A2 (en) Methods for treating cancer using anti-ctla4 antibodies
KR20230087451A (en) Combination therapy of a PD-1 antagonist and an antagonist to VEGFR-2 for treating cancer patients
CN116806226A (en) Combination therapy of PD-1 antagonists and antagonists of VEGFR-2 for treating cancer patients

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant