专利摘要:
The invention generally relates to activable antibodies that specifically bind to PDL1 and methods for making and using these activable anti-PDL1 antibodies in a variety of therapeutic, diagnostic and prophylactic indications.
公开号:BR112019025188A2
申请号:R112019025188-0
申请日:2018-05-31
公开日:2020-06-23
发明作者:Humphrey Rachel;Rachel Humphrey;Carman Lori;Lori Carman;Will Matthias;Matthias Will;Zheng Beiyao;Beiyao Zheng;Balinski Kathe;Kathe Balinski
申请人:Cytomx Therapeutics, Inc.;
IPC主号:
专利说明:

[001] [001] This application claims the benefit of US Provisional Application No. 62 / 513,937, filed on June 1, 2017, US Provisional Application No. 62 / 534,950, filed on July 20, 2017, Provisional Application No. 62 / 555,598, filed on September 7, 2017, and US Provisional Application No. 62 / 657,567, filed on April 13, 2018, the content of each of which is incorporated herein by reference in its entirety. Description of the Electronically Sent Text File
[002] [002] The content of the text file sent electronically is incorporated here by reference in its entirety: A computer-readable copy of the Sequence Listing (file name: CYTM_052_001US_SeqList_ST25, registered date: July 24, 2018, file size 80 kilobytes file). Field of the Invention This invention generally relates to specific dosage regimens for administering activable anti-PDL1 antibodies for the treatment of cancer. Background of the Invention
[003] [003] Antibody-based therapies have proven effective treatments for various diseases, but in some cases, toxicities due to the wide expression of the target have limited its therapeutic effectiveness. In addition, antibody-based therapies exhibited other limitations, such as rapid clearance of circulation after administration.
[004] [004] In the field of small molecule therapies, strategies have been developed to provide prodrugs for an active chemical entity. Such prodrugs are administered in a relatively inactive (or significantly less active) form. Once administered, the prodrug is metabolized in vivo to the active compound. Such prodrug strategies can provide greater selectivity of the drug for its intended target and reduce adverse effects.
[005] [005] Therefore, there is a continuing need in the field of antibody-based therapies for antibodies that mimic the desirable characteristics of the small molecule prodrug. Summary of the Invention
[006] [006] In several respects, the invention provides methods for treating, alleviating a symptom of, or delaying the progression of cancer in an individual, administering intravenously at a dose of about 0.3 mg / kg to 30 mg / kg of an anti-PDL1 antibody activable to the individual. The activable antibody has an antibody (AB) that specifically binds to human PDL1. AB has a heavy chain variable region having a complementarity determining region 1 (CDRH1) having the amino acid sequence of SEQ ID NO: 212, a complementarity determining region 2 (CDRH2) having the amino acid sequence of SEQ ID NO: 246, and a complementarity determining region 3 (CDRH3) having the amino acid sequence or SEQ ID NO: 235; and a light chain variable region having a light chain complementarity determining region 1 (CDRL1) having the amino acid sequence of SEQ ID NO: 209, a light chain complementarity determining region 2, (CDRL2) having the amino acid sequence of SEQ ID NO: 215, a light chain complementarity determining region 3 (CDRL3) having the amino acid sequence of SEQ ID NO: 228; a cleavable (CM) portion bound to AB, where CM is a polypeptide that functions as a substrate for a protease; and a masking portion (MM) attached to the CM.
[007] [007] Also included in the invention are methods of treating, alleviating a symptom of, or delaying the progression of cancer in an individual, by administering intravenously in a fixed dose of about 24 to 2400 mg of an anti-PDL1 antibody activable to individual, in which the activable antibody has an antibody (AB) that specifically binds to human PDL1, in which AB comprises a variable heavy chain region having a complementarity determining region 1 (CDRH1) having the sequence of amino acid of SEQ ID NO: 212, a complementarity determining region 2 (CDRH2) having the amino acid sequence of SEQ ID NO: 246, and a complementarity determining region 3 (CDRH3) having the amino acid sequence or SEQ ID NO: 235 ; and a light chain variable region having a light chain complementarity determining region 1 (CDRL1) having the amino acid sequence of SEQ ID NO: 209, a light chain complementarity determining region 2, (CDRL2) having the sequence amino acid of SEQ ID NO: 215, a region that determines light chain complementarity 3 (CDRL3) having the amino acid sequence of SEQ ID NO: 228; a cleavable (CM) portion linked to AB, where CM is a polypeptide that functions as a substrate for a protease; and a masking portion (MM) connected to the AB.
[008] [008] In another aspect, the invention provides an anti-PDL1 antibody activable with an antibody (AB) that specifically binds to human PDL1. AB has a heavy chain variable region a complementarity determining region 1 (CDRH1) having the amino acid sequence of SEQ ID NO: 212, a complementarity determining region 2 (CDRH2) having the amino acid sequence of SEQ ID NO: : 246, and a complementarity-determining region 3 (CDRH3) having the amino acid sequence or SEQ ID NO: 235; and a light chain variable region having a light chain complementarity determining region 1 (CDRL1) having the amino acid sequence of SEQ ID NO: 209, a light chain complementary determining region 2, (CDRL2) having the sequence amine acid of SEQ ID NO: 215, a region that determines light chain complementarity 3 (CDRL3) having the amino acid sequence of SEQ ID NO: 228; a cleavable portion (CM) linked to AB, where CM is a polypeptide that functions as a substrate for a protease; and a masking portion (MM) attached to the CM, for use in the treatment, relief of a symptom or delay in the progression of a cancer in an individual, and in which the activable antibody is administered intravenously at a dose of about from 0.3 mg / kg to 30 mg / kg.
[009] [009] In another aspect, the invention provides an activable anti-PDL1 antibody having an antibody (AB) that specifically binds to human PDL1. AB has a heavy chain variable region a complementarity determining region 1 (CDRH1) having the amino acid sequence of SEQ ID NO: 212, a complementarity determining region 2 (CDRH2) having the amino acid sequence of SEQ ID NO: 246, and a complementarity determining region 3 (CDRH3) having the amino acid sequence or SEQ ID NO: 235; and a light chain variable region having a light chain complementarity determining region 1 (CDRL1) having the amino acid sequence of SEQ ID NO: 209, a light chain complementarity determining region 2, (CDRL2) having the amino acid sequence of SEQ ID NO: 215, a region that determines complementary light chain 3 (CDRL3) having the amino acid sequence of SEQ ID NO: 228, a cleavable (CM) portion linked to AB, in that CM is a polypeptide that functions as a substrate for a protease; and a masking portion (MM) linked to CM, for use in treatment, relief of a symptom or delay in the progression of cancer in an individual, and in which the activable antibody is administered.
[0010] [0010] MM inhibits the binding of AB to human PDL1 when the activable antibody is in an uncleaved state. In some respects, MM had the amino acid sequence of SEQ ID NO: 63.
[0011] [0011] In some respects, CM has the amino acid sequence of SEQ ID NO: 377.
[0012] [0012] AB has a heavy chain variable region (VH) having the amino acid sequence of SEQ ID NO: 46 and a light chain variable (VL) having the amino acid sequence of SEQ ID NO: 58 or SEQ ID NO: 137.
[0013] [0013] In other respects, the activatable antibody has a light chain having the amino acid sequence of SEQ ID NO: 1008 and a heavy chain having the amino acid sequence of SEQ ID NO: 432.
[0014] Alternatively, the activatable antibody having a light chain having the amino acid sequence of SEQ ID NO: 428 and a heavy chain having the amino acid sequence of SEQ ID NO: 432.
[0015] [0015] The dose is approximately between 3 mg / kg to 10 mg / kg. The dose is approximately between 3 mg / kg to 15 mg / kg. The dose is 0.3 mg / kg, 1 mg / kg, 3 mg / kg, 6 mg / kg, 10 mg / kg, 15 mg / kg, or 30 mg / kg.
[0016] [0016] The fixed dose is 240 mg, 480 mg, 800 mg, 1200 mg, 2400 mg.
[0017] [0017] The activable antibody is administered on a dose schedule every 7-28 days. For example, the activable antibody is administered on a dose schedule every 14 days or 21 days.
[0018] [0018] The activable antibody is administered as a monotherapy. Alternatively, the activable antibody is administered as a component of a combination therapy. Combination therapy includes the administration of a dose of an anti-CTLA-4 antibody or a B-RAF inhibitor
[0019] [0019] The anti-CTLA-4 antibody is, for example, ipilimumab. The anti-CTLA-4 antibody is administered intravenously. The anti-CTLA-4 antibody is administered at a dose of 3 mg / kg, 6 mg / kg or 10 mg / kg. Alternatively, the anti-CTLA-4 antibody is administered in a fixed dose of 240 mg, 480 mg or 800 mg.
[0020] [0020] The B-RAF inhibitor is vemurafenib. The B-RAF inhibitor is administered orally. The B-RAF inhibitor is administered at a dose of 960 mg or at a dose of 875 mg. The activable antibody and the B-RAF inhibitor are administered over the same period of time.
[0021] [0021] In some aspects, the dose of the B-RAF inhibitor is administered twice a day. In other respects, at least 4 doses of each of the activable antibody and the B-RAF inhibitor are administered.
[0022] [0022] In some respects, multiple doses of the activatable antibody and the anti-CTLA-4 antibody are administered over a first period of time, followed by administration of multiple doses of the activable antibody as a monotherapy over a second period of time. time.
[0023] [0023] In other respects, the activable antibody and one dose of the anti-CTLA-4 antibody are administered concomitantly as a combination therapy every 21 days for 4 doses, followed by administration of one dose of the activable antibody as a monotherapy. every 14 days.
[0024] [0024] In yet another aspect, multiple doses of the antibody activable as a monotherapy are administered during a first period of time, followed by the concomitant administration of multiple doses of the activable antibody and the anti-CTLA-4 antibody as combination therapy is administered over a second period of time.
[0025] [0025] In yet another aspect, multiple doses of the activatable antibody are administered as a monotherapy for a first period of time, multiple doses of the activable antibody and the anti-CTLA-4 antibody are subsequently administered as a therapy. combination over a second period of time and multiple doses of the antibody activable as a monotherapy are administered over a third period of time.
[0026] [0026] In other respects, the activable antibody is administered as a monotherapy every 14 days for 4 doses, followed by administration of a dose of activable antibody and a dose of anti-CTLA-4 antibody as a combination therapy every 21 days for 4 doses, followed by administration of a dose of the activable antibody as a monotherapy every 14 days.
[0027] [0027] Cancer is an advanced, unresectable solid tumor or lymphoma. For example, the advanced, unresectable tumor is a type of tumor responsive to PDL1.
[0028] [0028] Cancer is a carcinoma, just like squamous cell carcinoma.
[0029] [0029] Cancer is, for example, anal squamous cell carcinoma, basal cell carcinoma, bladder cancer, bone cancer, intestinal carcinoma, breast cancer, carcinoid, castration-resistant prostate cancer (CRPC), cervical carcinoma , colorectal cancer (CRC), colon cancer, cutaneous squamous cell carcinoma, endometrial cancer, esophageal cancer, gastric carcinoma, gastroesophageal junction cancer, glioblastoma / mixed glioma, glioma, head and neck cancer, hepatocellular carcinoma, malignancy hematogenous, liver cancer, lung cancer, melanoma, Merkel cell carcinoma, multiple myeloma, nasopharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, peritoneal carcinoma, undifferentiated pleomorphic sarcoma, prostate cancer, rectal carcinoma, kidney cancer, sarcoma, salivary gland carcinoma, squamous cell carcinoma, stomach cancer, testicular cancer, thymic carcinoma, thymic epithelial tumor, thymoma, it cancer rheoid, urogenital cancer, urothelial cancer, uterine carcinoma or uterine sarcoma.
[0030] [0030] Cancer is a High Tumor Mutational Load (hTMB) cancer.
[0031] [0031] Breast cancer is triple negative breast cancer or breast cancer positive for estrogen receptors.
[0032] [0032] Haematological malignancy is lymphoma or leukemia. Lymphoma is, for example, B-cell lymphoma, T-cell lymphoma, Hodgkin's lymphoma or EBV lymphoma, primary B-cell medial lymphoma.
[0033] [0033] Cancer is melanoma.
[0034] [0034] Carcinoma of the intestine is, for example, carcinoma of the small intestine or adenocarcinoma of the small intestine. Colon cancer is colon adenocarcinoma.
[0035] [0035] Lung cancer is, for example, non-small cell lung cancer (NSCLC) or small cell lung cancer. The NSCLC is either a non-scaly NSCLC or a scaly NSCLC.
[0036] [0036] Prostate cancer is a neuroendocrine prostate cancer of small cells.
[0037] [0037] Renal cancer is renal cell carcinoma or renal sarcoma.
[0038] [0038] Preferably, the cancer is undifferentiated pleomorphic sarcoma, small bowel adenocarcinoma, Merkel cell carcinoma, thymic carcinoma, anal squamous cell carcinoma, cutaneous squamous cell carcinoma or triple negative breast cancer.
[0039] [0039] The individual exhibits one or more of the following characteristics
[0040] [0040] Unless otherwise defined, all the technical and scientific terms used herein have the same meaning as commonly understood by someone versed in the technique to which this invention belongs. In the specification, singular forms also include the plural, unless the context clearly indicates otherwise. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. In the event of a conflict, this specification, including definitions, will prevail. In addition, the materials, methods and examples are illustrative only and are not intended to be limiting.
[0041] [0041] Other features and advantages of the invention will be evident from the detailed description and claims that follow. Brief Description of Drawings
[0042] [0042] Figure 1A is a schematic representation of the study design for a study in Example 2, where "AA" represents the anti-PDL1 activable antibody referred to herein as PL07-2001-C5H9v2, which comprises the heavy chain sequence of SEQ ID NO: 432 and the light chain sequence of SEQ ID NO: 428.
[0043] [0043] Figure 1B is a schematic representation of a study project for the study in Example 2, where "AA" represents the activable anti-PDL1 antibody referred to herein as PL07-2001-C5H9v2, which comprises the sequence of heavy chain of SEQ ID NO: 432 and the light chain sequence of SEQ ID NO: 428. In comparison to Figure 1A, this schematic representation includes an additional optional Part A2 in the study design.
[0044] [0044] Figure 2 shows a series of graphs showing the detection of cleaved and intact anti-PD-L1 activable antibodies in tumor and plasma samples determined using the WES system (ProteinSimple, San Jose, CA) under similar conditions those described in the WES instrumentation manual.
[0045] [0045] Figures 3A and 3B are a series of graphs showing the screening of anti-idiotypic (anti-id) clones PL07-2001-C5H9v2 against 37% of activable antibody activated by an arm at 0.11, 0.33 and 1 µg / ml in human plasma at 1: 100. Figure 3A is an electropherogram showing 17G1 detection of the decreasing concentration of PL07-2001-C5H9v2 activated by an arm (1, 0.33 and 0.11 ug / ml, referred to in the Figure as AA MIXTURE). Figure 3B shows the relative activation percentage for the 6 main clones of an activable antibody activated by an arm. The relative activation rate is preserved at different concentrations. Clones 21H10 and 27C1 have less affinity, resulting in no data for the concentration of 0.11 µg / ml.
[0046] [0046] Figures 4A, 4B, 4C and 4D are a series of graphs that show that the antibody referred to here as 17G1 has high specificity for the activable antibody (AA) PL07-2001-C5H9v2. 17G1 was evaluated in Wes for specificity by reinforcing 160 ng / ml of PL07-2001-C5H9v2 activated by an arm (activated AA) in human plasma (Figure 4C) or lung tumor lysates (Figure 4D).
[0047] [0047] Figures 5A and 5B are a series of graphs that represent the specific detection of activable antibody therapies
[0048] [0048] Figure 6A and Figure 6B are a series of graphs that represent the preferential activation of activable antibody (AA) therapies in the tumor versus plasma detected in the xenograft tumor model. MDA-MB-231 xenograft mice were treated with 1 mg / ml of the activable anti-PDL1 antibody referred to herein as PL07-2001-C5H9v2. The tumor and plasma samples were collected on day 4. Figures 6A and 6B demonstrate the analysis of tumor and plasma homogenate samples by a Wes-based capillary electrophoresis immunoassay method (ProteinSimple) of the description.
[0049] [0049] Figures 7A and 7B are a series of graphs that represent the preferential activation of antibody therapies activable in the tumor versus plasma detected in another xenograft tumor model. SAS xenograft mice were treated with 0.1 mg / kg of the activable anti-PDL1 antibody referred to herein as PL07- 2001-C5H9v2. Figures 7A and 7B demonstrate the analysis of tumor and plasma homogenate samples by a Wes-based capillary electrophoresis immunoassay method (ProteinSimple) of the description.
[0050] [0050] Figures 8A and 8B are graphs of the mean plasma concentration of PL07-2001-C5H9v2 (nM) intact (not cleaved) and total (ie, intact and cleaved), respectively, versus time (day) after administration of up to 30 mg / kg of q2W for Cohorts A and A2 Cycle 1 Dose
[0051] [0051] Figure 9A represents the best percentage change from baseline in target lesions after the administration of PL07-2001- C5H9v2. Figure 9B is a radar graph that represents the change in the target lesion (%) vs. time after the administration of PL07-2001- C5H9v2. Abbreviations: CR, resistant to castration; ER + BC, estrogen receptor positive breast cancer; HNSCC, head and neck squamous cell carcinoma; PD, progressive disease; PR, partial response; RECIST, Response Assessment Criteria in Solid Tumors; DS, stable disease; TNBC, triple negative breast cancer.
[0052] [0052] Figure 10A represents the best percentage change from baseline in the target lesions after the administration of the PL07-2001-C5H9v2 + ipilimumab combination. FIG 10B is a radar graph that represents the change in the target lesion (%) vs. time after the administration of PL07-2001-C5H9v2. Abbreviations: CR, complete answer; ER + BC, estrogen receptor positive breast cancer; HNSCC, squamous cell carcinoma of the head and neck; PD, progressive disease; PR, partial response, RECIST, Response Assessment Criteria in Solid Tumors; SCC, squamous cell carcinoma; SCLC, small cell lung cancer; SD, stable disease; TNBC, triple negative breast cancer. Detailed Description of the Invention
[0053] [0053] The present description provides methods of treating cancer by administering an activable anti-PDL1 antibody. Specifically, the invention is based on the results of the first human safety and efficacy study of an activable antibody. A dose increase study was conducted to evaluate the safety and efficacy of PL07-2001-C5H9v2 as a monotherapy or in combination with ipilimumab PL07-2001-C5H9v2 is a protease-activated anti-PDL1 antibody. PL07-2001-C5H9v2 is activated by tumor-associated proteases and has been shown to be inactive in the circulation.
[0054] [0054] Patients with solid, metastatic, or advanced unresectable tumors or lymphoma were administered intravenously with 0.03 mg / kg -30 mg / kg of PL07-2001-C5H9v2 every three weeks. Among patients with evaluable data (n = 19), the target lesions decreased from baseline in 8 patients (42%). Target lesions decreased from baseline by dose levels ≥3 mg / kg in 6/10 patients (60%).
[0055] [0055] The disease control rate was 45% for patients in all patients dosed between 0.03 to 30 mg / kg of PL07-2001- C5H9v2. For patients dosed with at least 10 mg / kg, disease control rates were greater than 66%. Surprisingly, pharmacokinetic analysis (PK) has shown that PL07-2001-C5H9v2 circulates in the plasma mainly in the non-activated form and PK is only minimally reduced by the targeted mediated drug disposition.
[0056] [0056] The activable anti-PDL1 antibodies described herein overcome a limitation of antibody therapies, particularly antibody therapies which are known to be toxic to at least some degree in vivo. Target-mediated toxicity is a major limitation for the development of therapeutic antibodies. The activable anti-PDL1 antibodies provided here are designed to address the toxicity associated with target inhibition in normal tissues by traditional therapeutic antibodies. It is important to note that these activable anti-PDL1 antibodies remain masked until they are proteolytically activated at the disease site. ACTIVABLE ANTI-PDL1 ANTIBODIES
[0057] [0057] The activable antibodies used in the compositions and methods of the description were generated and characterized using the methods described in PCT Publication No. WO 2016/149201, whose contents are incorporated by reference here in their entirety.
[0058] [0058] Activable anti-PDL1 antibodies include an antibody that specifically binds to PDL1 coupled to a masking portion (MM), so that coupling the MM reduces the antibody's capacity or its binding fragment antigen to bind to PDL1. MM is coupled via a sequence that includes a substrate for a protease, for example, a protease that is co-located with PDL1 at a treatment site in an individual.
[0059] [0059] In some embodiments, the activable antibodies include an antibody (AB) that is modified by an MM and also includes one or more cleavable moieties (CM). Such activable antibodies exhibit activable / switchable binding to the AB target. Activable antibodies generally include an antibody or antibody fragment (AB), modified by or coupled to a masking portion (MM) and a modifiable or cleavable portion (CM). In some embodiments, CM contains an amino acid sequence that serves as a substrate for at least one protease. In the modalities, the AB has two heavy chains and two light chains.
[0060] [0060] The elements of the activable antibodies are arranged so that the MM and the CM are positioned so that, in a cleaved (or relatively active) state and in the presence of a target, the AB binds to a target while the activable antibody is in an uncleaved (or relatively inactive) state and in the presence of the target, the specific binding of AB to its target is reduced or inhibited. The specific binding of AB to its target may be reduced due to inhibition or masking of the AB's ability to specifically bind to its target by MM.
[0061] [0061] Activable antibodies can be provided in a variety of structural configurations. Exemplary formulas for activable antibodies are provided below. It is specifically contemplated that the N- to C-terminal order of AB, MM and CM can be reversed within an activable antibody. It should be noted that, although MM and CM are indicated as separate components in the formulas below, in all the exemplary modalities (including formulas) described here, it is contemplated that the amino acid sequences of MM and CM may overlap, for example, so that the CM is totally or partially contained in the MM.
[0062] [0062] For example, activable antibodies can be represented by the following formula (in order from an amino (N) terminal region to a carboxyl (C) terminal region: (MM) - (CM) - (AB) (AB) - (CM) - (MM) where MM is a masking moiety, CM is a cleavable moiety and AB is an antibody In addition, the formulas above provide additional amino acid sequences that can be positioned N-terminal or CC- terminal to the elements of activable antibodies.
[0063] [0063] In many embodiments, it may be desirable to insert one or more ligands, for example, flexible ligands, into the construction of activable antibody, in order to provide flexibility in one or more of the MM-CM junctions, the CM junction -AB, or both. For example, AB, MM and / or CM may not contain enough residues (for example, Gly, Ser, Asp, Asn, especially Gly and Ser, particularly Gly) to provide the desired flexibility. As such, the switchable phenotype of such activable antibody constructs can benefit from the introduction of one or more amino acids to provide a flexible linker. In addition, as described below, where the activatable antibody is provided as a conformationally restricted construct, a flexible linker can be operationally inserted to facilitate the formation and maintenance of a cyclic structure in the uncleaved activable antibody.
[0064] [0064] For example, in certain embodiments, an activable antibody comprises one of the following formulas (where the formula below represents an amino acid sequence in the N- to C-terminal direction or C- to N-terminal direction): (MM) -L1- (CM) - (AB) (MM) - (CM) -L2- (AB) (MM) -L1- (CM) -L2- (AB) where MM, CM and AB are as defined above; where L1 and L2 are each independently and optionally present or absent, are the same or different flexible ligands that include at least 1 flexible amino acid (for example, Gly). In addition, the formulas above provide additional sequences of amino acids that can be positioned at the N-terminal or at the C-terminal to the activable antibody elements. Examples include, but are not limited to, targeting portions (eg, a ligand for a cell receptor in a target tissue) and portions that extend the serum half-life (eg, polypeptides that bind serum proteins, such as immunoglobulin (eg, IgG) or serum albumin (eg, human serum albumin (SAH)).
[0065] [0065] When the AB is modified with an MM and is in the presence of the specific binding of the AB target to its target it is reduced or inhibited, compared to the specific binding of the unmodified AB with an MM or the binding parental AB to the target. In comparison to binding the unmodified AB with an MM or binding the parental AB to the target, the AB's ability to bind the target when modified with an MM can be reduced by at least 50%, 60%, 70 %, 80%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% and up to 100% for at least 2, 4, 6, 8, 12, 28, 24, 30, 36, 48, 60, 72, 84 or 96 hours, or 5, 10, 15, 30, 45, 60, 90, 120, 150 or 180 days, or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 months or more when measured in vivo or in an in vitro assay.
[0066] [0066] On the other hand, the binding affinity of the modified AB with an MM and a CM for the target is at least 5, 10, 25, 50, 100, 250, 500, 1,000, 2,500, 5,000, 10,000, 50,000, 100,000, 500,000,
[0067] [0067] When used herein, the term cleaved state refers to the condition of antibodies that can be activated after modification of the CM by at least one protease. The term non-cleaved state, when used herein, refers to the condition of antibodies activable in the absence of CM cleavage by a protease. As discussed above, the term "activable antibodies" is used here to refer to an activable antibody in both its uncleaved (native) and cleaved states. It will be evident to the ordinarily versed specialist that, in some modalities, an activated cleaved antibody may not have an MM due to the protease cleavage of the CM, resulting in the release of at least the MM.
[0068] [0068] By activable or switchable means that the activable antibody exhibits a first level of binding to a target when the activable antibody is in an inhibited, masked or uncleaved state (ie, a first conformation) and a second level of binding to the target in the uninhibited, unmasked and / or cleaved state (ie, a second con-
[0069] [0069] The CM and AB of the activable antibodies are selected so that the AB represents a binding portion for a given target, and the CM represents a substrate for a protease. In some embodiments, the protease is colocalized with the target at a treatment site in an individual. When used here, colocalized refers to being in the same or relatively close proximity. In some embodiments, a protease cleaves a CM to produce an activated antibody that binds to a target located near the cleavage site. The activable antibodies described here find particular use where, for example, a protease capable of cleaving a site in the CM, that is, a protease, is present at relatively higher levels in or near the tissue containing the target of a site treatment or diagnostic site than in the tissue of untreated sites (for example, in healthy tissues). In some modalities, a CM in the description is also cleaved by one or more other proteases. In some modalities, it is the one or more other proteases that are colocalized with the target and that are responsible for the cleavage of CM in vivo.
[0070] [0070] In some embodiments, the activable antibodies provide reduced toxicity and / or adverse side effects that could otherwise result from the binding of AB at non-treatment sites if the AB were not masked or otherwise inhibited by binding to the target.
[0071] [0071] In general, an activable antibody can be designed by selecting an AB of interest and constructing the rest of the activable antibody, so that, when conformationally restricted, the MM provides masking of the AB or reducing the binding of the AB to its target. Structural design criteria can be taken into account to provide this functional feature.
[0072] [0072] For specific cleavage by an enzyme, the contact between the enzyme and the CM is made. When the activable antibody comprising an AB coupled to an MM and a CM is in the presence of sufficient target and enzymatic activity, the CM can be cleaved. Sufficient enzyme activity can refer to the enzyme's ability to make contact with CM and effect cleavage. It can be easily predicted that an enzyme may be in the vicinity of CM, but unable to cleave because of other cellular factors or protein modification of the enzyme.
[0073] [0073] In some embodiments, the serum half-life of the activable antibody is longer than that of the corresponding antibody; for example, the pK of the activatable antibody is greater than that of the corresponding antibody. In some embodiments, the serum half-life of the activable antibody is at least 15 days when administered to an organism. In some embodiments, the serum half-life of the activable antibody is at least 12 days when administered to an organism. In some embodiments, the serum half-life of the activable antibody is at least 11 days when administered to an organism. In some modalities, the serum half-life of the activable antibody is at least 10 days when administered to an organism.
[0074] [0074] An exemplary activable antibody includes an antibody (AB) that specifically binds to human PDL1, having a variable region
[0075] An exemplary activable anti-PDL1 antibody includes an antibody (AB) that specifically binds to human PDL1. AB includes two antibody heavy chains each having a heavy chain variable region having a complementarity determining region 1 (CDRH1) having the amino acid sequence of SEQ ID NO: 212, a complementarity determining region 2 ( CDRH2) having the amino acid sequence of SEQ ID NO: 246, and a complementarity-determining region 3 (CDRH3) having the amino acid sequence for SEQ ID NO: 235; and two antibody light chains, each having a light chain variable region, a light chain complementarity-determining region 1 (CDRL1) having the amino acid sequence of SEQ ID NO: 209, a region determining complementarity light chain 2 (CDRL2) having the amino acid sequence of SEQ ID NO: 215, a complementary determining region of light chain 3 (CDRL3) having the amino acid sequence of SEQ ID NO: 228; two masking portion peptides (MM1s); and two cleavable portion peptides (CM1s), each CM1 being a substrate for a protease. Each MM1 is linked in an N- to C-terminal direction to a CM1, to form two MM1-CM1 peptides where the carboxyl terminus of each MM1-CM1 peptide is linked to the amino terminus of each AB light chain. The CM is, for example, ISSGLLSGRSDNH (SEQ ID NO: 377). The MM is, for example, GIALCPSHFCQLPQT (SEQ ID NO: 63).
[0076] [0076] In some embodiments, the activatable anti-PDL1 antibody has a heavy chain variable region of SEQ ID NO: 46 and a light chain variable region of SEQ ID NO: 137 that includes a CM of SEQ ID NO: 377, an MM of SEQ ID NO: 63 and a VL of SEQ ID NO: 58.
[0077] [0077] In other embodiments, the activable anti-PDL1 antibody has a heavy chain of SEQ ID NO: 432 and a light chain of SEQ ID NO: 428 that includes a CM of SEQ ID NO: 377, an MM of SEQ ID NO : 63, a VL of SEQ ID NO: 58 and a constant domain Capa. .
[0078] Preferred activable anti-PDL1 antibodies useful in the methods of the invention include PL07-2001-C5H9v2, which include a heavy chain variable region of SEQ ID NO: 46 and a light chain variable region of SEQ ID NO: 137 PL07-2001-C5H9v2 full size heavy and light chain includes SEQ ID NO; 432 and SEQ ID NO: 428, respectively.
[0079] [0079] An additional activable anti-PDL1 antibody useful in the methods of the invention includes PL07-2001-C5H9v2-WO which includes a heavy chain variable region of SEQ ID NO: 46 and a light chain variable region of SEQ ID NO : 58. The PL07-2001-C5H9v2-WO natural-sized light and heavy chain includes SEQ ID NO: 432 and SEQ ID
[0080] [0080] PL07-2001- C5H9v2 Heavy Chain Variable Sequence EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKG
[0081] [0081] PL07-2001-C5H9v2 Light Chain Variable Sequence QGQSGSGIALCPSHFCQLPQTGGGSSGGSGGSGGISSGLLSGRSD NHGGSDIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKPG
[0082] [0082] PL07-2001- C5H9v2-W0 Light Chain Variable Sequence GIALCPSHFCQLPQTGGGSSGGSGGSGGISSGLLSGRSDNHGGSDI QMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKPGKAPKLLIY AASSLQSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQDNGY PSTFGGGTKVEIKR (SEQ ID NO:)
[0083] [0083] PL07-2001-C5H9v2 Heavy Chain Sequence EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKG LEWVSSIWRNGIVTVYADSVKGRFTISRDNSKNTLYLQMNSLRAED TAVYYCAKWSAAFDYWGQGTLVTVSSASTKGPSVFPLAPCSRSTSE STAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSS VVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEF LGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVD GVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSN KGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKG
[0084] [0084] PL07-2001-C5H9v2 Light Chain Sequence QGQSGSGIALCPSHFCQLPQTGGGSSGGSGGSGGISSGLLSGRS DNHGGSDIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQK PGKAPKLLIYAASSLQSGVPSRFSGSGSGTDFTLTISSLQPEDFATY YCQQDNGYPSTFGGGTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVV
[0085] [0085] PL07-2001-C5H9v2-WO Light Chain Sequence (without ligand) GIALCPSHFCQLPQTGGGSSGGSGGSGGISSGLLSGRSDNHGGSDI QMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKPGKAPKLLIY AASSLQSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQDNGY PSTFGGGTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYP
[0086] [0086] In some embodiments, activable anti-PDL1 antibody includes an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to an amino acid sequence of SEQ ID NOs: 46, 137, 138, 432, 428 and 1008. Anti-PDL1 Antibodies
[0087] Exemplary anti-PDL1 antibodies useful in the construction of an activable anti-PDL1 antibody described herein include antibodies C5H9 v2, C5H9, C5B10, C5E10, and G12H9. The VH and VL CDRs of C5H9 v2, C5H9, C5B10, C5E10, and G12H9 are shown below in a single row in Table 1
[0088] [0088] Variable heavy and light chain amino acid sequences for anti-PDL1 antibodies C5H9 v2, C5H9, C5B10, C5E10, and G12H9 are shown below.
[0089] [0089] C5H9v2 Anti-PDL1 Light Chain Variable Sequence DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKPGKAPKL
[0090] [0090] C5H9 and C5H9v2 Anti-PDL1 Heavy Chain Variable Sequence EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVR
[0091] [0091] C5H9, C5B10, C5E10 and G12H9 Anti-PDL1 Light Chain Variable Sequence DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKPGKAPKLL
[0092] [0092] C5B10 Anti-PDL1 Heavy Chain Variable Sequence EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGL
[0093] [0093] C5E10 Anti-PDL1 Heavy Chain Variable Sequence EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLE
[0094] [0094] G12H9 Variable Heavy Chain EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMetSWVRQAPGK GLEWVSSIWYQGLVTVYADSVKGRFTISRDNSKNTLYLQMetN SLRAEDTAVYYWKSAQDQYQQQQTQYQ
[0095] [0095] In some embodiments, the anti-PDL1 antibody includes an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to an amino acid sequence of SEQ ID NOs: 58, 46, 12, 48, 50 and 52. Masking Portions
[0096] [0096] The activable anti-PDL1 antibodies provided herein include a masking portion (MM). In some embodiments, the masking moiety is an amino acid sequence (i.e., peptide) that is coupled or otherwise linked to the anti-PDL1 antibody and is positioned within the activable anti-PDL1 antibody construct, so that the masking portion reduces the ability of the anti-PDL1 antibody to specifically bind to PDL1. Suitable masking portions are identified using any of a variety of known techniques. For example, the peptide masking moieties are identified using the methods described in PCT Publication No. WO 2009/025846 by Daugherty et al., The contents of which are incorporated herein by reference in their entirety.
[0097] [0097] MM is a polypeptide of about 2 to 40 amino acids in length. Preferably, MM is a polypeptide of up to about 40 amino acids in length.
[0098] [0098] In some embodiments, the MM polypeptide sequence is different from that of PDL1. In some embodiments, the MM polypeptide sequence is not more than 50% identical to any PDL1. In some modalities, the MM polypeptide sequence is not greater than 40%, 30%, 25%, 20%, 15% or 10% identical to PDL1.
[0099] [0099] MM copies include: YCEVSELFVLPWCMG (SEQ ID NO: 208), SCLMHPHYAHDYCYV (SEQ ID NO: 426), LCEVL MLLQHPWCMG (SEQ ID NO: 59), IACRHFMEQLPFCHH (SEQ ID NO: 60), FGPRCGE 61), ILYCDSWGAGCL-TRP (SEQ ID NO: 62), GIALCPSHFCQLPQT (SEQ ID NO: 63), DGPRCFVSGECSPIG (SEQ ID NO: 64), LCYKLDYDDRSYCHI (SEQ ID NO: 65), PCHPHPYDARPYCNV (SEQ, 66) PCYWHPF- FAYRYCNT (SEQ ID NO: 67), VCYYMDWLGRNWCSS (SEQ ID NO: 68), LCDLFKLREFPYCMG (SEQ ID NO: 69), YLPCHFVPIGACNNK (SEQ ID NO: 70), IFCHMGVVYQA NO (71), IFCHMGVVVPPNC (SE) SEQ ID NO: 72), PCHPAPYDARPYCNV (SEQ ID NO: 73), PCHPHAYDARPYCNV (SEQ ID NO: 74), PCHPHPA- DARPYCNV (SEQ ID NO: 75), PCHPHPYAARPYCNV (SEQ ID NO: 76), PCHPHNVY : 77), PCHPHPYDARPACNV (SEQ ID NO: 78), PCHPHPYDARPYCAV (SEQ ID NO: 79), PCHA HPYDARPYCNV (SEQ ID NO: 80), and PCHPHPYDARAYCNV (SEQ ID NO: 81).
[00100] [00100] A preferred MM includes GIALCPSHFCQLPQT (SEQ ID NO: 63).).
[00101] [00101] In some modalities, MM comprises an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%,
[00102] [00102] The activable anti-PDL1 antibodies provided herein include a cleavable portion (CM). In some embodiments, the cleavable portion includes an amino acid sequence that is a substrate for a protease, usually an extracellular protease. Suitable substrates are identified using any of a variety of known techniques. For example, peptide substrates are identified using the methods described in U.S. Patent No. 7,666,817 to Daugherty et al .; in U.S. Patent No. 8,563,269 to Stagliano et al .; and PCT Publication No. WO 2014/026136 by La Porte et al., the contents of which are incorporated by reference in their entirety. (See also, Boulware et al. "Evolutionary optimization of peptide substrates for proteases that exhibit rapid hydrolysis kinetics." Biotechnol Bioeng. 106.3 (2010): 339-46).
[00103] [00103] In some embodiments, the protease that cleaves CM is active, for example, over-regulated, in diseased tissue, and the protease cleaves CM in the activable antibody when the activable antibody is exposed to the protease.
[00104] [00104] In some embodiments, the protease is colocalized with PDL1 in a tissue, and the protease cleaves CM to the activable antibody when the activable antibody is exposed to the protease.
[00105] [00105] In some embodiments, protease is present at relatively higher levels or in the vicinity of the tissue that contains the target of a treatment site or diagnostic site than in the tissue of non-treatment sites (for example , in healthy tissue) and the protease cleaves CM to the activable antibody when the activable antibody is exposed to the protease.
[00106] [00106] Exemplary CMs include: LSGRSDNH, (SEQ ID NO: 341),
[00107] [00107] A preferred CM includes ISSGLLSGRSDNH, (SEQ ID NO: 377). Spacers and Binders
[00108] [00108] The binders suitable for use in the compositions described here are generally those that provide flexibility of the modified AB or the activable antibodies to facilitate the inhibition of AB binding to the target. Such binders are generally referred to as flexible binders. Suitable ligands can be easily selected and can be of any of different lengths, such as 1 amino acid (eg, Gly) to 20 amino acids, 2 amino acids to 15 amino acids, 3 amino acids to 12 amino acids, including 4 amino acids to 10 amino acids, 5 amino acids to 9 amino acids, 6 amino acids to 8 amino acids or 7 amino acids to 8 amino acids and can be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acids in length.
[00109] Exemplary flexible binders include glycine (G) n polymers, glycine-serine polymers, including, for example, (GS) n, (GSGGS) n (SEQ ID NO: 191) and (GGGS) n (SEQ ID NO: 192), where n is an integer of at least one, Gly-Gly-Ser-Gly (SEQ ID NO: 193), Gly-Gly-Ser-Gly-Gly (SEQ ID NO: 194), Gly- Ser-Gly-Ser-Gly (SEQ ID NO: 195), Gly-Ser-Gly-Gly-Gly (SEQ ID NO: 196), Gly-Gly- Gly-Ser-Gly (SEQ ID NO: 197), Gly -Be-Be-Be-Gly (SEQ ID NO: 198), and the like. Glycine-alanine polymers, alanine polymers
[00110] [00110] In some embodiments, at least one of L1 or L2 comprises an amino acid sequence selected from the group consisting of (GS) n, (GGS) n, (GSGGS) n (SEQ ID NO: 191) and ( GGGS) n (SEQ ID NO: 192), where n is an integer of at least one.
[00111] [00111] In some embodiments, at least one of L1 or L2 comprises an amino acid sequence selected from the group consisting of GGSG (SEQ ID NO: 193), GGSGG (SEQ ID NO: 194), GSGSG (SEQ ID NO : 195), GSGGG (SEQ ID NO: 196), GGGSG (SEQ ID NO: 197), and GSSSG (SEQ ID NO: 198).
[00112] [00112] In some embodiments, L1 comprises the amino acid sequence GSSGGSGGSGGSG (SEQ ID NO: 199), GSSGGSG GSGG (SEQ ID NO: 200), GSSGGSGGSGGS (SEQ ID NO: 201), GSSGGSGGSGGSGGGS (SEQ ID NO: 201) GSSGGSGGSG (SEQ ID NO: 203), or GSSGGSGGSGS (SEQ ID NO: 204).
[00113] [00113] In some embodiments, L2 comprises the amino acid sequence GSS, GGS, GGGS (SEQ ID NO: 205), GSSGT (SEQ ID NO: 206) or GSSG (SEQ ID NO: 207).
[00114] [00114] In some embodiments, the activable antibody also includes a signal peptide. In some embodiments, the signal peptide is conjugated to the activable antibody using a spacer. In some embodiments, the spacer is coupled to the activable antibody in the absence of a signal peptide. In some embodiments, the spacer is attached directly to the MM of the activable antibody. In some modes, the spacer is attached directly to the MM of the antibody active in the structural arrangement of the N-terminal to the C-terminal of the spacer-MM-CM-AB. An example of a spacer attached directly to the MM N-terminus of the activable antibody is, for example, QGQSGS (SEQ ID NO: 923); GQSGS (SEQ ID NO: 1192); QSGS (SEQ ID NO: 1193); SGS (SEQ ID NO: 1194); GS (SEQ ID NO: 1195); S; QGQSGQG (SEQ ID NO: 924); GQSGQG (SEQ ID NO: 395); QSGQG (SEQ ID NO: 925); SGQG (SEQ ID NO: 926); GQG (SEQ ID NO: 927); HQ (SEQ ID NO: 928); G; QGQSGQ (SEQ ID NO: 1196); GQSGQ (SEQ ID NO: 1197); QSGQ (SEQ ID NO: 1198); SGQ (SEQ ID NO: 1198); GQ (SEQ ID NO: 1199); and Q.
[00115] [00115] A preferred spacer includes QGQSGS (SEQ ID NO: 923).
[00116] [00116] In some embodiments, the activatable antibody does not include a spacer sequence. TREATMENT METHOD
[00117] [00117] The invention provides methods to prevent, delay progression, treat, alleviate a symptom or ameliorate a PDL1-mediated disease in an individual, by administering a therapeutically effective amount of activable anti-PDL1 antibody described here to an individual in need of them. The invention provides the use of the activable anti-PDL1 antibody described herein to slow progression, treat, alleviate a symptom of, or otherwise improve a PDL1-mediated disease in an individual, by administering a therapeutically effective amount of anti-PDL1 antibody. PDL1 activable. An amount
[00118] [00118] PDL1 is known to be expressed in a variety of cancers (See, for example, Chen et al., "Molecular Pathways: Next-Generation Immunotherapy - Inhibiting Programmed Death-Ligand 1 and Programmed Death-1," Clin Can. Res., Vol. 18: 6580-6587 (2012), whose contents are incorporated by reference in their entirety).
[00119] [00119] Cancers suitable for delaying the progression, treatment and alleviation of a symptom according to the methods of the invention include, for example, however, they are not limited to, they are anal squamous cell carcinoma, pancellular carcinoma, bladder cancer, bone cancer, intestinal carcinoma, breast cancer, carcinoid, castration-resistant prostate cancer (CRPC), cervical carcinoma, colorectal cancer (CRC), colon cancer, cutaneous squamous cell carcinoma, endometrial cancer , esophageal cancer, gastric carcinoma, gastroesophageal junction cancer, glioblastoma / mixed glioma, glioma, head and neck cancer, hepatocellular carcinoma, hematological malignancy, liver cancer, lung cancer, melanoma, cell carcinoma Merkel, multiple myeloma, nasopharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, peritoneal carcinoma, undifferentiated pleomorphic sarcoma, prostate cancer, rectal carcinoma, kidney cancer, sarcoma, carcinom the salivary gland, squamous cell carcinoma, stomach cancer, testicular cancer, thymic carcinoma, thymic epithelial tumor, thymoma, thyroid cancer, urogenital cancer, urothelial cancer, uterine carcinoma or uterine sarcoma.
[00120] [00120] In some modalities, cancer is a High Tumor Mutational Charge Cancer (hTMB).
[00121] [00121] In other modalities, breast cancer is triple negative breast cancer or breast cancer positive for estrogen receptor. Hematological malignancy is lymphoma, leukemia or multiple myeloma. Lymphoma includes B-cell lymphoma, T-cell lymphoma, Hodgkin's lymphoma or EBV lymphoma, primary B-cell mediatal lymphoma. In some embodiments, Hodgkin's lymphoma is post-allo-HSCT.
[00122] [00122] Carcinoma of the intestine is, for example, carcinoma of the small intestine or adenocarcinoma of the small intestine.
[00123] [00123] Head and neck cancer includes, for example, a squamous cell carcinoma of the head and neck. Esophageal cancer is, for example, esophageal carcinoma.
[00124] [00124] Colon cancer is, for example, colon adenocarcinoma.
[00125] [00125] Lung cancer is, for example, non-small cell lung cancer (NSCLC) or small cell lung cancer.
[00126] [00126] The NSCLC is a non-scaly NSCLC or a scaly NSCLC.
[00127] [00127] Prostate cancer is, for example, small cell neuroendocrine prostate cancer.
[00128] [00128] In some modalities, cancer is a carcinoma, such as, for example, squamous cell carcinoma.
[00129] [00129] In other modalities, cancer is renal cancer, such as renal cell carcinoma or renal sarcoma
[00130] [00130] Cancers particularly suitable in the practice of the methods and uses of the invention include non-differentiated pleomorphic sarcoma, small intestine adenocarcinoma, Merkel cell carcinoma, thymic carcinoma, anal squamous cell carcinoma, cell carcinoma scaly skin and triple negative breast cancer.
[00131] [00131] In some modalities, the cancer is gastric cancer or cancer of the gastroesophageal junction.
[00132] [00132] In some modalities, gastric or gastroesophageal cancer is an advanced, unresectable cancer with a Siewert classification of II / III for those with a significant esophageal component.
[00133] [00133] In some modalities, cancer is a thymoma or thymic cancer. Thymic cancer is a thymicepithelial tumor.
[00134] [00134] In some modalities, cancer is a melanoma. In some modalities, cancer is an eye melanoma.
[00135] [00135] B-cell lymphoma, T-cell lymphoma, Hodgkin's lymphoma / primary mediastinal B-cell lymphoma and chronic myelogenous leukemia.
[00136] [00136] In some modalities, the cancer is due to a tumor that expresses PDL1.
[00137] [00137] Cancer is an advanced, unresectable tumor or solid lymphoma. The advanced, unresectable tumor is a type of tumor responsive to PDL1.
[00138] [00138] In some modalities, the individual has an unresectable solid tumor with no additional standard of care available. In some modalities, the individual has lymphoma without an additional standard of care available. In some modalities, the individual is naive to immunotherapy. In some embodiments, PDL1 / PD1 inhibitory therapy is not approved for the individual's cancer.
[00139] [00139] In some embodiments, the PDL1 status of the individual and / or tumor is unknown. In some modalities, the individual and / or tumor is positive for PDL1 (PDL1 +), for example, the individual has a tumor proportion score score of at least 1% of membranous staining.
[00140] [00140] An activable anti-PDL1 antibody used in any of the modalities of these methods and uses can be administered at any stage of the disease. For example, such an activable anti-PDL1 antibody can be administered to a patient suffering from cancer of any stage, from the beginning to the metastatic one. In some modalities, cancer comprises advanced or recurrent solid tumors or lymphomas. In some embodiments, the individual has a solid, unresectable tumor.
[00141] [00141] The invention also provides methods of treating cancer patients with an autoimmune or inflammatory disease, delivering a therapeutically effective amount of an activable anti-PDL1 antibody described herein to an individual in need thereof. In some embodiments, the autoimmune disease is colitis, RA, pancreatitis, diabetes or pneumonitis.
[00142] [00142] In some modalities, the individual is a mammal, such as a human, non-human primate, companion animal (for example, cat, dog, horse), farm animal, work animal or zoo animal. In some modalities, the individual is human. In some modalities, the individual is a companion animal. In some modalities, the individual is an animal in the care of a veterinarian. Preferably, the individual is human.
[00143] [00143] In various modalities, individuals exhibit one or more of the following characteristics: they are naive to the PD-1 / PDL1 inhibitor, they are naive to the CTLA-4 inhibitor, they are positive to the BRAFV600E mutation, they are naive to the inhibitor of BRAF is either naive to immunotherapy, is positive for PDL1, is unknown for PDL1 or has been previously treated with a PD1 / PDL1 inhibitor.
[00144] [00144] In some modalities, the individual does not have an additional standard of care available.
[00145] [00145] In other modalities, the individual was previously treated with a PD-1 / PDL1 inhibitor, and treatment with the PD-1 inhibitor
[00146] [00146] The method according to any of the previous claims, in which the individual is naive to immunotherapy.
[00147] [00147] The activable anti-PDL1 antibody and its therapeutic formulations is administered to an individual who suffers or is susceptible to a disease or disorder associated with the expression and / or activity of aberrant PDL1. An individual who suffers or is susceptible to a disease or disorder associated with aberrant PDL1 expression and / or activity is identified using any of a variety of methods known in the art. For example, individuals suffering from cancer or another neoplastic condition are identified using a variety of clinical and / or laboratory tests, such as physical examination and blood, urine and / or stool tests to assess their health status. For example, individuals suffering from inflammation and / or inflammatory disorder are identified using any of a variety of clinical and / or laboratory tests such as physical examination and / or analysis of body fluids, for example, blood, urine analysis and / or feces, to assess health status.
[00148] [00148] Administration of an activable anti-PDL1 antibody to a patient suffering from a disease or disorder associated with aberrant PDL1 expression and / or activity is considered successful if any of several clinical or laboratory objectives are achieved. For example, administration of an activable anti-PDL1 antibody to a patient suffering from a disease or disorder associated with aberrant PDL1 expression and / or activity is considered successful if one or more of the symptoms associated with the disease or disorder are relieved. - reduced, inhibited or not progressing to another state, that is, worse. Administration of an activable anti-PDL1 antibody to a patient suffering from a disease or disorder associated with aberrant PDL1 expression and / or activity is considered successful if the disease or disorder goes into remission or does not progress to another state. , that is, worse. DOSAGE AND ADMINISTRATION
[00149] [00149] The cancer therapy provided here, containing an activable anti-PDL1 antibody, is administered in an amount sufficient to exert a therapeutically useful effect. Typically, the active agents are administered in an amount that does not result in undesirable side effects from the patient being treated or that minimizes or reduces the side effects observed.
[00150] [00150] It is within the level of someone skilled in the art to determine the precise amounts of active agents, including activable anti-PDL1 antibodies to be administered to an individual. For example, such agents and uses for the treatment of solid tumors and lymphomas are well known in the art. Thus, the dosages of such agents can be chosen based on standard dosing regimens for this agent under a given administration routine.
[00151] [00151] It is understood that the precise dosage and duration of treatment is a function of the tissue or tumor being treated and can be determined empirically using known test protocols or by extrapolating test data in vivo or in vitro and / or can be determined from known dosage regimens of the particular agent. It should be noted that concentrations and dosage values may also vary with the age of the individual treated, the individual's weight, the administration routine and / or the extent or severity of the disease and other factors that are within the level qualified physician to consider. Generally, dosage regimens are chosen to limit toxicity. It should be noted that the attending physician would know how and when to finish, stop or adjust therapy to reduce the dosage due to toxicity, or bone marrow, liver or kidney or other tissue disorders. On the other hand, the attending physician would also know how and when to adjust treatment to higher levels if the clinical response is not adequate (excluding toxic side effects). It should also be understood that, for any individual in particular, specific dosage regimens must be adjusted over time according to the individual need and the professional diagnosis of the person who administers or supervises the administration of the formulations, and that the concentration bands established here are only exemplary and are not intended to limit your scope.
[00152] [00152] For example, activable anti-PDL1 antibodies are administered in a therapeutically effective amount to decrease the tumor volume.
[00153] [00153] The amount of activable anti-PDL1 antibodies is administered for the treatment of a disease or condition, it can be determined by standard clinical techniques. In addition, in vitro assays and animal models can be employed to help identify ideal dosing ranges. The precise dosage, which can be determined empirically, may depend on the administration routine, the type of disease to be treated and the severity of the disease.
[00154] [00154] The activable anti-PDL1 antibodies provided here are administered intravenously. For intravenous administration, the conjugate can be administered by pressure or bolus, by infusion or by a combination thereof. The infusion time can be about 1 minute to three hours, such as about 1 minute to about two hours, or about 1 minute to about 60 minutes, or at least 10 minutes, 40 minutes or 60 minutes.
[00155] [00155] The dosage amount is between 0.03 mg / kg and 30 mg / kg. In other embodiments, the dosage amount is between 0.3 mg / kg and 30 mg / kg. In other modalities, the dosage amount is between 3 mg / kg and 30 mg / kg; 3 mg / kg and 20 mg / kg; 3 mg / kg and 15 mg / kg, or 3 mg / kg and 10 mg / kg. In some modalities, the dosage amount is between 5 mg / kg and 30 mg / kg; 5 mg / kg and 30 mg / kg; 5 mg / kg and 20 mg / kg; 5 mg / kg and 15 mg / kg; or 5 mg / kg and 10 mg / kg. In other embodiments, the dosage amount is between 10 mg / kg and 30 mg / kg; 10 mg / kg and 20 mg / kg; or 10 mg / kg and 15 mg / kg.
[00156] [00156] For example, the dosage amount is 0.03 mg / kg, 0.10 mg / kg, 0.3 mg / kg, 1.0 mg / kg, 3.0 mg / kg, 10.0 mg / kg or 30.0 mg / kg. The dosage amount is 1 mg / kg, 3 mg / kg, 6 mg / kg or 15.0 mg / kg. Preferably, the dosage amount is 10 mg / kg.
[00157] [00157] The activable anti-PDL1 antibodies provided here are administered in a fixed dose. A fixed dosage is based, for example, on a 65 kg human, a 70 kg human, a 75 kg human, a 75 kg human or an 80 kg human and the mg / kg dosages described here . For example, when the fixed dose is based on an 80 kg human and the desired mg / kg doses are 10 mg / kg, then the fixed dose is 800 mg.
[00158] [00158] A fixed dosage is between 240 mg and 2400 mg. Exemplary fixed doses include 240 mg, 480 mg, 800 mg, 1200 mg and 2400 mg.
[00159] [00159] The frequency and timing of administration, and dosage amounts, can be administered periodically during an administration cycle to maintain a continuous and / or long-term effect of the active agents for a desired period of time. Compositions provided of activable anti-PDL1 antibodies can be administered hourly, daily, weekly, biweekly, monthly, annually or once. The length of the administration cycle can be determined empirically and depends on the disease to be treated, the severity of the disease, the patient in particular and other considerations within the level of experience of the attending physician. The length of time of treatment with a combination therapy provided here can be one week, two weeks, one month, several months, one year, several years or more.
[00160] [00160] The frequency of administration of activable anti-PDL1 antibodies is between once a day and every 28 days; between once a day and once a month, between once a week and once a month; between once a week and once every two months.
[00161] [00161] For example, the frequency of administration of activable anti-PDL1 antibodies is once a day, every other day, twice a week, once a week, once every 2 weeks, once every 3 weeks , once every 4 weeks, once every 5 weeks, once every six weeks, once every seven weeks, once every eight weeks. In other words, the frequency of administration of the activable anti-PDL1 antibodies is once a day, every two days, twice a week, once every 7 days, once every 14 days, once every 21 days , once every 28 days, once every 35 days, once every 42 days, once every 49 days, once every 56 days. Dosages can be divided into a plurality of administration cycles during the course of treatment. For example, activable anti-PDL1 antibodies can be administered in frequency over a period of about a month, 2 months, 3 months, 4 months, 5 months, 6 months, a year or more. The frequency of administration can be the same throughout the cycle or it can be different. For example, an exemplary dosing frequency is twice a week, at least for the first week of an administration cycle. After the first week, the frequency can continue twice a week, it can increase to more than twice a week, or it can be reduced to no more than once a week. It is within the level of an educated person to determine the dosage frequency and administration cycle based on the specific dosage to be administered, the disease or condition to be treated, the severity of the disease or condition, the age of the individual and other similar factors.
[00162] [00162] If the symptoms of the disease persist in the absence of interrupted treatment, treatment can be continued for an additional period of time. Throughout the course of treatment, evidence of disease and / or toxicity or side effects related to treatment can be monitored.
[00163] [00163] The administration cycle of activable anti-PDL1 antibodies can be adapted to add periods of interrupted treatment in order to provide a period of rest from exposure to agents. The time period for discontinuing treatment can be for a predetermined time or it can be determined empirically, depending on how the patient is responding or depending on the observed side effects. For example, treatment can be stopped for a week, two weeks, three weeks, a month or several months. Usually, the interrupted treatment period is incorporated into a dosing cycle for a patient.
[00164] [00164] An exemplary dosage regimen is a 14-day treatment cycle or administration cycle. The active anti-PDL1 antibodies described here are administered on day 1, followed by 13 days without dosing. It is within the level of someone skilled in the art to determine the precise cycle of administration and dosing schedule.
[00165] [00165] As noted above, the administration cycle can be for any desired period of time. Therefore, the 14-day administration cycle can be repeated for any period of time. It is within the skill level of the attending physician to adopt an administration cycle and dosage regimen that meets the patient's needs depending on personal considerations specific to the patient and the disease to be treated.
[00166] [00166] In some embodiments, the activable anti-PDL1 antibodies described here are used as unique active agents, that is, monotherapy. Alternatively, the activable anti-PDL1 antibodies described herein are used in conjunction with one or more additional agents or a combination of additional agents, i.e., combination therapy or co-therapy. Suitable additional agents include current pharmaceutical and / or surgical therapies for an intended application, such as, for example, cancer. For example, activable anti-PDL1 antibodies can be used in conjunction with a chemotherapeutic antineoplastic agent or additional radiation.
[00167] [00167] In some embodiments, the activable anti-PDL1 antibody is administered before and / or during and / or after treatment in combination with one or more additional agent (s) (for example, combination therapy) .
[00168] [00168] Non-limiting examples of additional agents include a chemotherapeutic agent, radiation, a checkpoint inhibitor, a kinase inhibitor, an anti-inflammatory agent, an immunosuppressive agent, a T cell agonist, a cell agonist NK, an agent directed to inhibitors in the tumor microenvironment agent affects the depletion of regulatory T cells, an antigenic agent, agent directed to inhibitors in the tumor microenvironment, a proteasome inhibitor, an antimetabolite, an antimicrobial agent, a topoisomerase inhibitor, a vaccine, an oncovirus, a DC activating agent, a cytotoxic antibiotic and / or any other agent harmful to nucleic acid.
[00169] [00169] In some embodiments, the additional agent (s) is (are) tumor-directed antibody designed to kill the tumor via ADCC or by direct conjugation to a toxin (for example, a drug antibody conjugate ( In some embodiments, the additional agent (s) stimulates co-stimulating molecules In some embodiments, the additional agent (s) is a cell therapeutic agent Adoptive T that affects the transfer of adoptive T cells
[00170] [00170] In some embodiments, the agent inhibits adenosine A2aR. In some embodiments, the agent inhibits arginase. In some embodiments, the agent inhibits CD39. In some embodiments, the agent inhibits CD73. In some embodiments, the agent inhibits CD47.
[00171] [00171] In some modalities, the additional agent is chemotherapeutic agent. Chemotherapeutic agents include, for example, alkylating agents, and taxanes. Alkylating agents include, for example, platinum-based chemotherapy, such as carboplatin, cisplatin, and oxaliplatin.
[00172] [00172] Taxanes include, for example, docetaxel, paclitaxel, Abraxane® (i.e., albumin-conjugated paclitaxel). Other chemotherapeutic agents include doxorubicin, irinotecan, gemcitabine and any chemotherapeutic agents known to those skilled in the art.
[00173] [00173] A tumor microenvironment inhibitor includes, for example, an IDO inhibitor, an α-CSF1R inhibitor, an α-CCR4 inhibitor, TGF-beta block, a myeloid-derived suppressor cell or a regulatory T cell .
[00174] [00174] In some modalities, the agonist is selected from the group consisting of Ox40, GITR, CD137, ICOS, CD27 and HVEM.
[00175] [00175] In some embodiments, the DC activating agent includes, as a non-limiting example, a bell-fold receptor (TLR) and / or α-CD40 agonist.
[00176] [00176] A checkpoint inhibitor inhibits (for example, blocks) immune checkpoint proteins. Immune checkpoints include, for example, CTLA-4, LAG-3, PD1 (also referred to as PD-1), PDL1, TIGIT, TIM-3, B7H4 and Vista.
[00177] [00177] Kinase inhibitors inhibit kinases such as B-RAFi, MEKi and Btk.
[00178] [00178] Exemplary kinase inhibitors include pazopanib, osimertinib, crizotinib, sorafenib or erlotinib
[00179] [00179] A B-RA inhibitor includes, for example, vemurafenib. A Btk inhibitor includes, for example, ibrutinib. MEKi kinase inhibitors include, for example, trametinib, cobimetinib or selumethinbe.
[00180] [00180] In some embodiments, the additional agent is an immunomodulatory agent, such as lenolidominde or IL-2.
[00181] [00181] In some embodiments, the additional agent is a proteasome inhibitor, such as bortezomib or carfilzomib.
[00182] [00182] In some modalities, the additional agent is an agent considered a standard of care by those skilled in the art
[00183] [00183] In some embodiments, the additional agent is a targeted agent, such as another antibody, for example, a monoclonal antibody (for example, ipilimumab or bevacizumab), a bispecific antibody or a multispecific antibody.
[00184] [00184] Additional agents are administered simultaneously or at different times during a treatment regimen. For example, the activable anti-PDL1 antibody is administered concomitantly with the addition agent, prior to the administration of the additional agent, or subsequent to the administration of the additional agent, or alternately. The additional agent is administered in a single dose or in multiple doses.
[00185] [00185] In some embodiments, the additional agent is a targeted agent, such as another antibody, for example, a monoclonal antibody (for example, bevacizumab), a bispecific antibody or a multispecific antibody. In some embodiments, the additional agent is a proteasome inhibitor, such as bortezomib or carfil-
[00186] [00186] In some embodiments, the additional agent is another antibody or its antigen-binding fragment, another conjugated antibody or its antigen-binding fragment, another activable antibody or its antigen-binding fragment and / or another conjugated activable antibody or its antigen-binding fragment. In some embodiments, the additional agent is another antibody or its antigen-binding fragment, another conjugated antibody or its antigen-binding fragment, another activable antibody or its antigen-binding fragment and / or another conjugated activable antibody or its antigen-binding fragment against the same target as the first antibody or its antigen-binding fragment, the first conjugated antibody or its antigen-binding fragment, activable antibody or its antigen-binding fragment and / or an activable antibody conjugate or its antigen-binding fragment, for example, against PDL1. In some embodiments, the additional agent is another antibody or its antigen-binding fragment, another conjugated antibody or its antigen-binding fragment, another activable antibody or its antigen-binding fragment and / or another antibody activatable conjugate or its antigen-binding fragment against a target other than the target of the first antibody or its antigen-binding fragment, the first conjugated antibody or its antigen-binding fragment, activable antibody or its antigen-binding fragment antigen and / or a conjugated activable antibody or its
[00187] [00187] In some embodiments, this additional agent is ipilimumab, a CTLA-binding fragment of ipilimumab and / or an antibody activated by ipilimumab. CD51 CYR61 hGH
[00188] [00188] As a non-limiting example, the additional agent is or is derived from an antibody listed in Table 23. Table 22: Exemplary sources for Additional Agents Antibody Trade Name (antibody name) Target Avastin ™ (bevacizumab) VEGF Lucentis ™ (ranibizumab) VEGF Erbitux ™ (cetuximab) EGFR Vectibix ™ (panitumumab) EGFR Remicade ™ (infliximab) TNFα Humira ™ (adalimumab) TNFα Tysabri ™ (natalizumabe) Integrina α4 Simulect ™ (basiliximabe) IL2R Solirisuma5 (efalizumab) CD11a Bexxar ™ (tositumomab) CD20 Zevalin ™ (ibritumomab tiuxetan) CD20 Rituxan ™ (rituximab) CD20 (Ocrelizumab) CD20 Arzerra ™ (ofatumumab) CD20 Gazyva ™ (Obinutuzumabe) CD20 Zenapaxcabisztabiszaczabetabiszczaczaczabetzaczabetzaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczaczzxzzxzxzxzxzxzxzxzxzxzxzxzxzxzxzxzxzxzxzxqxqxzxzxzxxlxlxlxlxlxlxlxl ) CD30 Myelotarg ™ (gentuzumab) CD33 Mylotarg ™ (gentuzumab ozogamycin) CD33 Campath ™ (alemtuzumab) CD52 ReoPro ™ (abiciximab) Glycoprotein IIb / IIIa Xolair ™ receptor (omalizumab) IgE Herceptin ™ (trastuzumabbe) and trastuzumabbe (Herastinumabbe) Her2 ntansin) Her2 Synagis ™ (palivizumab) RSV F protein (ipilimumab) CTLA-4 (tremelimumab) CTLA-4 Hu5c8 CD40L (pertuzumab) Her2-neu (ertumaxomab) CD3 / Her2-neu Orencia ™ (abataceptte) CTLA-4 (tanezbe ) NGF (bavituximab) Phosphatidylserine (zalutumumab) EGFR (mapatumumab) EGFR (matuzumab) EGFR
[00189] [00189] Additional agents are administered simultaneously or at different times during a treatment regimen. For example, the activable anti-PDL1 antibody is administered concomitantly with the addition agent, prior to the administration of the additional agent, or subsequent to the administration of the additional agent, or alternately. The additional agent is administered in a single dose or in multiple doses.
[00190] [00190] In some embodiments, the activatable anti-PDL1 antibody of the description is used in combination with a CTLA-4 inhibitor. In some embodiments, the activatable anti-PDL1 antibody of the description is used in combination with an anti-CTLA-4 antibody, such as, for example, ipilimumab.
[00191] [00191] The CTLA-4 inhibitor, such as ipilimumab, is administered at a dose between 1 mg / kg to 20 mg / kg, between 3 mg / kg to 15 mg / kg, between 3 mg / kg to 10 mg / kg. For example, the CTLA-4 inhibitor such as ipilimumab is administered at a dosage of 1 mg / kg, 2 mg / kg, 3 mg / kg, 4 mg / kg, 5 mg / kg, 6 mg / kg, 7 mg / kg, 8 mg / kg, 9 mg / kg or 10 mg / kg.
[00192] [00192] In several embodiments, the anti-CTLA-4 antibody, for example, ipilimumab is administered in a fixed dose. A fixed dose is based, for example, on a 65 kg human, a 70 kg human, a 75 kg human or an 80 kg human and the mg / kg dosages described herein. For example, when the fixed dose is based on an 80 kg human and the desired mg / kg dose is 10 mg / kg, then the fixed dose will be 800 mg. If the desired dose in mg / kg is 6 mg / kg, then the fixed dose will be 480 mg. If the desired mg / kg dose is 3 mg / kg, then the fixed dose will be 240 mg. A fixed dosage of the anti-CTLA-4 antibody, for example, ipilimumab is between 140 mg and 1000 mg. Exemplary fixed dosages include 240 mg, 480 mg and 800 mg,
[00193] [00193] In some modalities, ipilimumab is administered at a higher dose than its maximum tolerated dose for a given indication. Alternatively, ipilimumab is administered at a lower dose than its maximum tolerated dose for a given indication.
[00194] [00194] In other modalities, ipilimumab is administered at a higher dose than its recommended dose for a given indication. Alternatively, ipilimumab is administered at a dose lower than the recommended dose for a given indication.
[00195] [00195] In some embodiments, the activable anti-PDL1 antibody and the CTLA-4 inhibitor, for example, an anti-CTLA-4 antibody, for example, ipilimumab, are administered intravenously (IV).
[00196] [00196] The frequency of administration of anti-CTLA-4 antibody, for example, ipilimumab is between once a day and every 28 days; between once a day and once a month, between once a week and once a month; between once a week and once every two months. For example, the frequency of administration of the anti-CTLA-4 antibody, for example, ipilimumab is once a day, every other day, twice a week, once a week, once every 2 weeks, once once every 3 weeks, once every 4 weeks, once every 5 weeks, once every six weeks, once every seven weeks, once every eight weeks. In other words, the frequency of administration of the activable anti-CTLA-4 antibody, for example, ipilimumab is once a day, every other day, twice a week, once every 7 days, once every 14 days, once every 21 days, once every 28 days, once every 35 days, once every 42 days, once every 49 days, once every 56 days.
[00197] [00197] The activable anti-PDL1 antibody and the CTLA-4 inhibitor, for example, an anti-CTLA-4 antibody, for example, ipilimumab, are administered IV at a regular interval. The active anti-PDL1 antibody and the CTLA-4 inhibitor, for example, an anti-CTLA-4 antibody, for example, ipilimumab, are administered IV at the same regular interval. Alternatively, the activable anti-PDL1 antibody and the CTLA-4 inhibitor, for example, an anti-CTLA-4 antibody, for example, iplimimumab, are administered IV at different regular intervals.
[00198] [00198] In some modalities, the frequency of administration of activable anti-PDL1 antibodies is between once a day and every 28 days; between once a day and once a month, between once a week and once a month; between once a week and once every two months and the frequency of administration of the anti-CTLA-4 antibody, for example, ipilimumab is every 7 days, every 14 days or every 28 days.
[00199] [00199] For example, the frequency of administration of activable anti-PDL1 antibodies is once a day, every day, twice a week, once a week, once every 2 weeks, once every 3 weeks, once every 4 weeks, once every 5 weeks, once every six weeks, once every seven weeks, once every eight weeks, the frequency of administration of the anti-CTLA-4 antibody. , for example, ipilimumab is every 7 days, every 14 days or every 28 days.
[00200] [00200] Alternatively, the frequency of administration of the activable anti-PDL1 antibodies is once a day, every two days, twice a week, once every 7 days, once every 14 days, once at every 21 days, once every 28 days, once every 35 days, once every 42 days, once every 49 days, once every 56 days. The frequency of administration of the anti-CTLA-4 antibody, for example, ipilimumab is every 7 days, every 14 days or every 28 days.
[00201] [00201] For example, in some embodiments, the activable anti-PDL1 antibody and the CTLA-4 inhibitor, for example, an anti-CTLA-4 antibody, for example, ipilimumab, are administered IV every 21 days for multiple doses .
[00202] [00202] For example, in some embodiments, the activable anti-PDL1 antibody and the CTLA-4 inhibitor, for example, an anti-CTLA-4 antibody, for example, ipilimumab, are administered IV every 14 days for multiple doses .
[00203] [00203] In some embodiments, activable anti-PDL1 antibody and the CTLA-4 inhibitor, for example, an anti-CTLA-4 antibody, for example, ipilimumab, is administered IV every 21 days for at least
[00204] [00204] In some embodiments, the anti-PDL1 antibody, conjugated anti-PDL1 antibody, activable anti-PDL1 antibody and / or conjugated activable anti-PDL1 antibody of the description and the CTLA-4 inhibitor, for example, an anti- CTLA-4, for example, ipilimumab, is administered IV every 21 days for at least two or more doses, for example, at least four doses, followed by administration of anti-PDL1 antibody activable as a monotherapy for a desired period of time
[00205] [00205] In some embodiments, the anti-PDL1 antibody, conjugated anti-PDL1 antibody, activable anti-PDL1 antibody and / or conjugated activable anti-PDL1 antibody of the description are administered IV in a dosage of 0.3 mg / kg, 1 , 0 mg / kg, 3.0 mg / kg, 10.0 mg / kg and 30.0 mg / kg and the CTLA-4 inhibitor, for example, an anti-CTLA-4 antibody, for example, ipilimumab, IV is administered at a dose of 3 mg / kg. In some embodiments, the activable anti-PDL1 antibody is administered IV at a dose of 10.0 mg / kg, and, 6 mg / kg, or 10 mg / kg or, 6 mg / kg or 10 mg / kg. In some embodiments, the activable anti-PDL1 antibody and the inhibition of CTLA-4, for example, an anti-CTLA-4 antibody, for example, ipilimumab, are administered according to the dosage and / or administration schedule. shown in Figure 1, Part B1 or Part B2 and described in Example 2. In any of these embodiments described here, an activable anti-PDL1 antibody from the description is used. In an exemplary embodiment, the activable anti-PDL1 antibody is PL07-2001-C5H9v2.
[00206] [00206] For example, multiple doses of the activable antibody and the
[00207] [00207] For example, one dose of the activable antibody and one dose of the anti-CTLA-4 antibody are administered concomitantly as a combination therapy every 21 days for 4 doses, followed by administration of one dose of the activable anti-PDL1 antibody as monotherapy every 14 days.
[00208] [00208] In some embodiments, multiple doses of the anti-PDL1 antibody activable as a monotherapy are administered during a first period of time, followed by the concomitant administration of multiple doses of the activable anti-PDL1 antibody and the anti-CTLA-4 antibody as a combination therapy for a second period of time.
[00209] [00209] For example, multiple doses of the activatable antibody are administered as a monotherapy over a first period of time and subsequently multiple doses of the activatable antibody and anti-CTLA-4 antibody as a combination therapy are administered for a second period of time, followed by the administration of multiple doses of the activable antibody as a monotherapy for a third period of time.
[00210] [00210] In some embodiments, the activable antibody is administered as a monotherapy every 14 days for 4 doses, followed by administration of a dose of activable antibody and a dose of anti-CTLA-4 antibody as a combination therapy a every 21 days for 4 doses, followed by administration of a dose of an activable antibody as a monotherapy every 14 days.
[00211] [00211] In some embodiments, the activable anti-PDL1 antibody is used in combination with a B-RAF inhibitor. In some fashion-
[00212] [00212] In some embodiments, the activable anti-PDL1 antibody is administered intravenously (IV) and the B-RAF inhibitor, for example, vemurafenib, is administered orally (PO). In some modalities, the activable anti-PDL1 antibody is administered IV, and multiple doses, for example, two or more doses, of the B-RAF inhibitor, for example, vemurafenib, are administered PO daily. In some embodiments, the activable anti-PDL1 antibody is administered IV and two doses of the B-RAF inhibitor, for example, vemurafenib, are administered PO daily. In some embodiments, the activable anti-PDL1 antibody is administered IV every 14 days, and two doses of the B-RAF inhibitor, for example, vemurafenib, are administered PO daily.
[00213] [00213] In some modalities, the B-RAF inhibitor, for example, vemurafenib, is administered PO in a dose of 960 mg. In some modalities, the B-RAF inhibitor, for example, vemurafenibere, is administered twice daily PO, in a dose of 960 mg.
[00214] [00214] In some embodiments, the BB-RAF inhibitor, for example, vemurafenib, is administered PO in a dose of 875 mg. In some modalities, the B-RAF inhibitor, for example, vemura-fenib, is administered twice daily PO in a dose of 875 mg.
[00215] [00215] In some embodiments, the activable anti-PDL1 antibody is administered IV in a dosage of 1.0 mg / kg, 3.0 mg / kg, 10.0 mg / kg and 30.0 mg / kg and the inhibitor of B-RAF, for example, vemurafenib, PO is administered at a dose of 960 mg. In other embodiments, the activable anti-PDL1 antibody is administered IV at a dosage of 1.0 mg / kg, 3.0 mg / kg, 10.0 mg / kg and 30.0 mg / kg and the BB- RAF, for example, vemurafenib, is administered PO in a dose of 875 mg.
[00216] [00216] In some embodiments, the activable anti-PDL1 antibody is administered IV at a dosage of 10.0 mg / kg, and the B-RAF inhibitor, for example, vemurafenib, is administered PO at a dose of 960 mg.
[00217] [00217] In other embodiments, the activable anti-PDL1 antibody is administered IV at a dosage of 10.0 mg / kg and the B-RAF inhibitor, for example, vemurafenib, is administered PO at a dose of 875 mg.
[00218] [00218] In other embodiments, the activable anti-PDL-1 antibody is administered IV at a dosage of 10.0 mg / kg and the B-RAF inhibitor, for example, vemurafenib administered PO at a dose of 875 mg.
[00219] [00219] In some embodiments, the activable anti-PDL1 antibody and the B-RAF inhibitor, for example, vemurafenib, are administered according to the dosing and / or administration schedule shown in Figure 1, Part C and described in Example 1. ACTIVABLE ANTIBODY-PHARMACEUTICAL CONJUGATES
[00220] [00220] The compositions and methods provided herein allow the binding of one or more agents to one or more cysteine residues in the AB without compromising the activity (for example, the masking, activation or binding activity) of the anti antibody -PDL1 activable. In some embodiments, the compositions and methods provided herein permit the attachment of one or more agents to one or more cysteine residues in the AB without reducing or otherwise disturbing one or more disulfide bonds in the MM. The compositions and methods provided herein produce an activable anti-PDL1 antibody that is conjugated to one or more agents, for example, any of a variety of therapeutic, diagnostic and / or prophylactic agents, for example, in some modalities , without any (any) of the agent (s) being conjugated to the MM of the activable anti-PDL1 antibody. The compositions and methods provided herein produce conjugated activable anti-PDL1 antibodies in which MM maintains the ability to effectively and efficiently mask the AB of the activatable antibody in an uncleaved state. The compositions and methods provided herein produce conjugated activable anti-PDL1 antibodies in which the activable antibody is still activated, that is, cleaved, in the presence of a protease that can cleave CM.
[00221] [00221] In some embodiments, the activable antibodies described here also include an agent conjugated to the activatable antibody. In some embodiments, the conjugate agent is a therapeutic agent, such as an anti-inflammatory and / or antineoplastic agent. In such modalities, the agent is conjugated to an active carbohydrate portion of the antibody, for example, in some embodiments, where the carbohydrate portion is located outside the antigen-binding region of the antibody or antigen-binding fragment in the activable antibody. In some embodiments, the agent is conjugated to a sulfhydryl group of the antibody or antigen-binding fragment in the activable antibody.
[00222] [00222] In some embodiments, the agent is a cytotoxic agent, such as a toxin (for example, an enzymatically active toxin of bacterial, fungal, plant or animal origin or fragments thereof) or a radioactive isotope (that is, a radioconjugate) .
[00223] [00223] In some embodiments, the agent is a detectable portion, such as, for example, a label or other marker. For example, the agent is or includes a radiolabeled amino acid, one or more biotinyl moieties that can be detected by the labeled avidin (for example, streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or calorific methods) (rimometric), one or more radioisotopes or radionuclides, one or more fluorescent labels, one or more enzyme labels and / or one or more chemiluminescent agents. In some embodiments, the detectable portions are linked by spacer molecules.
[00224] [00224] The description also refers to immunoconjugates comprising an antibody conjugated to a cytotoxic agent, such as a toxin (for example, an enzymatically active toxin of bacterial, fungal, plant or animal origin, or fragments thereof) or an isotope radioactive (that is, a radioconjugate). Suitable cytotoxic agents include, for example, dolastatin and its derivatives (for example, auristatin E, AFP, MMAF, MMAE, MMAD, DMAF, DMAE). For example, the agent is monomethyl auristatin E (MMAE) or monomethyl auristatin D (MMAD). In some embodiments, the agent is an agent selected from the group listed in Table 11. In some embodiments, the agent is a dolastatin. In some modalities, the agent is an auristatin or its derivative. In some modalities, the agent is auristatin E or its derivative. In some modalities, the agent is monomethyl auristatin E (MMAE). In some modalities, the agent is monomethyl auristatin D (MMAD). In some embodiments, the agent is a maytansinoid or maytansinoid derivative. In some embodiments, the agent is DM1 or DM4. In some embodiments, the agent is a duocarmycin or its derivative. In some modalities, the agent is a calicheamicin or its derivative. In some embodiments, the agent is a pyrrolobenzodiazepine. In some embodiments, the agent is a pyrrolobenzodiazepine dimer.
[00225] [00225] In some embodiments, the agent is bound to the AB using a caproil-valine-citrulline maleimide ligand or a PEG-valine-citrulline maleimide ligand. In some embodiments, the agent is bound to AB using a caproil-valine-citrulline maleimide linker. In some embodiments, the agent is bound to AB using a PEG-valine-citrulline maleimide linker. In some embodiments, the agent is monomethyl auristatin D (MMAD) bound to AB using a PEG-valine-citrulline-to-aminobenzyloxycarbonyl maleimide linker, and this linker payload construction is referred to here as "vc-MMAD". In some embodiments, the agent is monomethyl auristatin E (MMAE) bound to AB using a PEG-valine-citrulline-to-aminobenzyloxycarbonyl maleimide linker, and this linker payload construction is referred to herein as "vc-MMAE". The vc-MMAD and vc-MMAE structures are shown below: vc-MMAD: vc-MMAE:
[00226] [00226] The description also provides conjugated activable antibodies that include an activatable antibody bound to the monomethyl auristatin D (MMAD) payload, wherein the activable antibody includes an antibody or its binding antigen (AB) fragment that binds specifically to a target, a masking portion (MM) that inhibits the binding of the AB of the activable antibody in a non-cleaved state to the target, and a cleavable portion (CM) coupled to the AB, and the CM is a polypeptide that functions as a substrate for at least one MMP protease.
[00227] [00227] In some embodiments, the MMAD-conjugated activable antibody can be conjugated using any of several methods to bind agents to ABs: (a) binding to the carbohydrate moieties
[00228] [00228] In some embodiments, the polyethylene glycol (PEG) component of a binder of the present description is formed of 2 ethylene glycol monomers, 3 ethylene glycol monomers, 4 ethylene glycol monomers, 5 ethylene glycol monomers, 6 ethylene glycol monomers, 7 ethylene glycol monomers, 8 ethylene glycol monomers, 9 ethylene glycol monomers or at least 10 ethylene glycol monomers. In some embodiments of the present description, the PEG component is a branched polymer. In some embodiments of the present description, the PEG component is an unbranched polymer. In some embodiments, the PEG polymer component is functionalized with an amino group or its derivative, a carboxyl group or its derivative, or both, an amino group or its derivative and a carboxyl group or its derivative.
[00229] [00229] In some embodiments, the PEG component of a linker of the present description is an amino-tetra-ethylene-glycol-carboxyl group or its derivative. In some embodiments, the PEG component of a linker of the present description is an amino-tri-ethylene glycol-carboxyl group or its derivative. In some embodiments, the PEG component of a linker of the present description is an amino-ethylene glycol-carboxyl group or its derivative. In some embodiments, an amino derivative is the formation of an amide bond between the amino group and a carboxyl group to which it is conjugated. In some modalities, a carboxyl derivative is the formation of an amide bond between the carboxyl group and an amino group to which it is conjugated. In some embodiments, a carboxyl derivative is the formation of an ester bond between the carboxyl group and a hydroxyl group to which it is conjugated.
[00230] [00230] Enzymatically active toxins and fragments thereof that can be used include diphtheria A chain, unbound active fragments of diphtheria toxin, exotoxin A chain (from Pseudo-monas aeruginosa), ricin A chain, abrin chain A, modecin A chain, alpha-sarcin, Aleurites fordii proteins, diantin proteins, American Phytolaca proteins (PAPI, PAPII and PAP-S), momordica charantia inhibitor, curcine, crotin, saponary officinalis inhibitor, gelonin, mitogelin, restrictocin, phenomycin, enomycin and trichothecenes. A variety of radionuclides are available for the production of radioconjugated antibodies. Examples include Bi, 131 I, 131 In, 90 Y, and 186 Re. In some embodiments, the isotope is zirconium.
[00231] [00231] Those skilled in the art will recognize that a wide variety of possible portions can be coupled to the activable antibodies resulting from the description. (See, for example, "Conjugate Vaccines", Contributions to Microbiology and Immunology, JM Cruse and RE Lewis, Jr (eds), Carger Press, New York, (1989), whose entire contents are hereby incorporated by reference ). PHARMACEUTICAL COMPOSITIONS
[00232] [00232] The antibodies, conjugated antibodies, activable antibodies and / or conjugated activable antibodies of the description (also referred to herein as "active compounds") and derivatives, fragments, analogs and homologues thereof, can be incorporated into pharmaceutical compositions suitable for administration. Such compositions typically comprise the antibody, the conjugated antibody, the activatable antibody and / or the conjugated activable antibody and a pharmaceutically acceptable carrier. When used herein, the term "pharmaceutically acceptable carrier" is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption retarding agents, and the like, compatible with pharmaceutical administration. Suitable vehicles are described in the most recent edition of Remington's Pharmaceu-
[00233] [00233] A pharmaceutical composition of the description is formulated to be compatible with the intended administration routine. Examples of administration routines include parenteral administration, for example, intravenous, intradermal, subcutaneous, oral (for example, inhalation), transdermal (ie, topical), transmucosal and rectal. Solutions or suspensions used for parenteral, intradermal or subcutaneous application may include the following components: a sterile diluent, such as water for injection, saline, fixed oils, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents waistband; antibacterial agents such as benzyl alcohol or methyl parabenos; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylene diaminetetraacetic acid (EDTA); buffers such as acetates, citrates or phosphates and agents for adjusting tonicity such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. Parenteral preparation can be included in ampoules, disposable syringes or multi-dose vials made of glass or plastic.
[00234] [00234] Pharmaceutical compositions suitable for injection
[00235] [00235] Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients listed above, when required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound in a sterile vehicle containing a basic dispersion medium and the other necessary ingredients from those listed above. In the case of sterile powders for the preparation of sterile injectable solutions, the preparation methods are vacuum drying and lyophilization which produce a powder of the active ingredient plus any additional desired ingredient from a previously sterile filtered solution.
[00236] [00236] Oral compositions generally include an inert diluent or an edible carrier. They can be closed in gelatin capsules or compressed into tablets. For purposes of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches or capsules. Oral compositions can likewise be prepared using a fluid carrier for use as a mouthwash, in which the compound in the fluid carrier is applied orally and agitated and expectorated or swallowed. Pharmaceutically compatible binding agents and / or adjuvant materials can be included as part of the composition. Tablets, pills, capsules, troches and the like can contain any of the following ingredients or compounds of a similar nature: a binder such as microcrystalline cellulose, tragacanth gum or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent, such as peppermint vegetable, methyl salicylate or orange flavoring.
[00237] [00237] For administration by inhalation, the compounds are released in the form of an aerosol spray from a pressurized container or dispenser that contains a suitable propellant, for example, a gas such as carbon dioxide or a nebulizer.
[00238] [00238] Systemic administration can likewise be by transmucosal or transdermal means. For transmuted administration
[00239] [00239] The compounds can likewise be prepared in the form of suppositories (for example, with conventional suppository bases, such as cocoa butter and other glycerides) or retention enemas for rectal release.
[00240] [00240] In one embodiment, the active compounds are prepared with vehicles that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including microencapsulated implants and delivery systems. Biodegradable and biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorectants and polylactic acid. The methods for preparing such formulations will be apparent to those skilled in the art. The materials can likewise be commercially obtained from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to cells infected with monoclonal antibodies to viral antigens) can likewise be used as pharmaceutically acceptable vehicles. . These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811.
[00241] [00241] It is especially advantageous to formulate oral or parenteral compositions in the form of a dosage unit to facilitate administration and uniformity of dosage. The dosage unit form when used here refers to physically discrete units suitable as unitary dosages for the individual to be treated; each unit containing a predetermined amount of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the description is dictated and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be obtained, and the limitations inherent in the technique of composing such an active compound. for the treatment of individuals.
[00242] [00242] The pharmaceutical compositions can be included in a container, package or distributor together with instructions for administration. DEFINITIONS:
[00243] [00243] Unless otherwise defined, the scientific and technical terms used in connection with the present description will have the meanings that are commonly understood by those skilled in the art. The term "one (a)" entity or "one (an)" entity refers to one or more of these entities. For example, a compound refers to one or more compounds. As such, the terms "one", "one", "one or more" and "at least one" can be used interchangeably. In addition, unless otherwise required by the context, singular terms must include pluralities and plural terms must include the singular. Generally, the nomenclatures used in connection with the techniques of cell and tissue culture, molecular and chemical biology and hybridization of proteins and oligo or polynucleotides described here are those well known and commonly used in the art. Standard techniques are used for recombinant DNA, oligonucleotide synthesis and tissue culture and transformation (eg, electroporation, lipofection). Enzymatic reactions and purification techniques
[00244] [00244] When used in accordance with this description, the following terms, unless otherwise indicated, should be understood to have the following meanings:
[00245] [00245] When used herein, the term "antibody" refers to immunoglobulin molecules and immunologically active portions of immunoglobulin (Ig) molecules, that is, molecules that contain an antigen-binding site that binds specifically (immunoreacts with) an antigen. By "specific binding" or "immunoreacts with" or "immunospecific binding" means that the antibody reacts with one or more antigenic determinants of the desired antigen and does not react with other polypeptides or binds with much less affinity (Kd> 10 -6). Antibodies include, however, are not limited to, polyclonal, monoclonal, chimeric, domain, single chain antibodies, Fab and F (ab ') 2 fragments, scFvs and a Fab expression library.
[00246] [00246] The structural unit of the basic antibody is known to comprise a tetramer. Each tetramer is composed of two parts
[00247] [00247] The term "monoclonal antibody" (mAb) or "monoclonal antibody composition", when used herein, refers to a population of antibody molecules that contain only one molecular species of antibody molecule consisting of a genetic product exclusive light chain and a single heavy chain gene product. In particular, the complementarity-determining regions (CDRs) of the monoclonal antibody are identical across all molecules in the population. MAbs contain an antigen binding site capable of immunoreacting with a particular epitope of the antigen characterized by a unique binding affinity for it.
[00248] [00248] The term "antigen binding site" or "binding moiety" refers to the part of the immunoglobulin molecule that participates in antigen binding. The antigen binding site is formed by amino acid residues from the variable regions of the N ("V") terminal of the heavy ("H") and light ("L") chains. Three highly divergent stretches in the V regions of the heavy and light chains, called "hypervariable regions", are interposed between more conserved flanking stretches, known as "structural regions" or "FRs". Thus,
[00249] [00249] When used herein, the term "epitope" includes any protein determinant capable of specific binding to an immunoglobulin, an scFv or a T cell receptor. The term "epitope" includes any protein determinant capable of specific binding to an immunoglobulin or T-cell receptor. Epitopic determinants generally consist of chemically active surface clusters of molecules such as amino acids or sugar side chains and generally have specific three-dimensional structural characteristics as well as specific load characteristics. . For example, antibodies can be raised against N-terminal or C-terminal peptides of a polypeptide. An antibody is said to specifically bind to an antigen when the dissociation constant is ≤ 1 µM; in some modalities, ≤ 100 nM and in some modalities, ≤ 10 nM.
[00250] [00250] When used herein, the terms "specific binding", "immunological binding" and "immunological binding properties" refer to non-covalent interactions of the type that occur between an immunoglobulin molecule and an antigen for which the immunoglobulin is specific. - ca. The resistance or affinity of immunological binding interactions can be expressed in terms of the dissociation constant (Kd) of the interaction, where a smaller Kd represents a greater affinity. The immunological binding properties of the selected polypeptides can be quantified using methods well known in the art. One of these methods involves measuring the rates of formation and dissociation of the antigen-binding site / antigen complex, where these rates depend on the concentrations of the complex partners, the affinity of the interaction and the geometric parameters that also influence the rate at both directions. In this way, both the "constant on rate" (Kon) and the "constant on off rate" (Koff) can be determined by calculating the actual concentrations and rates of association and decoupling. (See Nature 361: 186-87 (1993)). The Koff / Kon relationship allows the cancellation of all parameters not related to affinity and is equal to the dissociation constant Kd. (See, generally, Davies et al., (1990) Annual Rev Biochem 59: 439-473). An antibody of the present description is said to specifically bind to the target, when the binding constant (Kd) is <1 μM, in some modalities <100 nM, in some modalities <10 nM and in some modalities <100 pM at about 1 pM, as measured by tests, such as radioligand binding assays or similar assays, known to those skilled in the art.
[00251] [00251] The term "isolated polynucleotide", when used here, will mean a polynucleotide of genomic, cDNA or synthetic origin or some combination of them that, by virtue of its origin, the "isolated polynucleotide" (1) is not associated with all or a portion of a polynucleotide in which the "isolated polynucleotide" is found
[00252] [00252] The term "isolated protein" referred to herein means a cDNA protein, recombinant RNA or synthetic origin or some combination thereof that, by virtue of its origin or source of derivation, the "isolated protein" (1) it is not associated with proteins found in nature, (2) it is free of other proteins from the same source, for example, free of murine proteins, (3) it is expressed by a cell of a different species or (4) it does not occur in nature.
[00253] [00253] The term "polypeptide" is used here as a generic term to refer to proteins, fragments or analogues native to a polypeptide sequence. Therefore, fragments of native proteins and analogues are species of the polypeptide genus. Polypeptides according to the description comprise the heavy chain immunoglobulin molecules shown here and the light chain immunoglobulin molecules shown here, as well as the antibody molecules formed by combinations comprising the heavy chain immunoglobulin molecules. with light chain immunoglobulin molecules, such as the light chain immunoglobulin molecules cape and vice versa, as well as their fragments and analogues.
[00254] [00254] The term "natural occurrence", when used here, applied to an object, refers to the fact that an object can be found in nature. For example, a polypeptide or polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a natural source and that was not intentionally-
[00255] [00255] The term "operationally linked", when used here, refers to the positions of the components thus described, in a relationship that allows them to function as intended. A control sequence "operationally linked" to a coding sequence is linked in such a way that the expression of the coding sequence is achieved under conditions compatible with the control sequences.
[00256] [00256] The term "control sequence", when used here, refers to polynucleotide sequences necessary to perform the expression and processing of coding sequences to which they are linked. The nature of such control sequences differs depending on the host organism in the prokaryotes, such control sequences generally include promoter, ribosomal binding site and transcription termination sequence in eukaryotes, generally, these control sequences include promoters and transcription termination sequence. The term "control sequences" is intended to include, at a minimum, all components whose presence is essential for expression and processing and may likewise include additional components whose presence is advantageous, for example, leader strings and partner strings fusion The term "polynucleotide", when referred to herein, means nucleotides at least 10 bases in length, ribonucleotides or deoxynucleotides or a modified form of any type of nucleotide. The term includes single and double stranded forms of DNA.
[00257] [00257] The term oligonucleotide referred to herein includes naturally occurring and modified nucleotides linked together by naturally occurring and non-naturally occurring oligonucleotide bonds. Oligonucleotides are a subset of polynucleotides that generally
[00258] [00258] The term "naturally occurring nucleotides" referred to herein includes deoxyribonucleotides and ribonucleotides. The term "modified nucleotides" referred to herein includes nucleotides with modified or substituted sugar groups and the like. The term "oligonucleotide bonds" referred to herein includes oligonucleotide bonds, such as phosphorothioate, phosphorodithioate, phosphoroselerloate, phosphorodiselenoate, phosphorananothioate, phosphoraniladate, phosphoronmidate and the like. See, for example, LaPlanche et al., Nucl. Acids Res. 14: 9081 (1986); Stec et al., J. Am. Chem. Soc. 106: 6077 (1984), Stein et al., Nucl. Acs ids. 16: 3209 (1988), Zon et al., Anti Cancer Drug Design 6: 539 (1991); Zon et al., Oligonucleotides and Analogues: A Practical Approach, pp. 87-108 (F. Eckstein, Ed., Oxford University Press, Oxford England (1991)); Stec et al., U.S. Patent No. 5,151,510; Uhlmann and Peyman Chemical Reviews 90: 543 (1990). An oligonucleotide can include a marker for detection, if desired.
[00259] [00259] When used here, the twenty conventional amino acids and their abbreviations follow conventional use. See Immunology - A Synthesis (2nd Edition, E. S. Golub and D. R. Green, Eds., Sinauer Associates, Sunderland, Mass. (1991)). Stereoisomers (for example, D amino acids) of the twenty conventional amino acids, unnatural amino acids such as α-, α-disubstituted amino acids, N-alkyl amino acids, lactic acid and other non-conventional amino acids
[00260] [00260] Likewise, unless otherwise specified, the left end of the single-stranded polynucleotide sequences is the 5 'end; the left direction of the double stranded polynucleotide sequences is referred to as the 5 'direction. The direction of the 5 'to 3' addition of the nascent RNA transcripts is referred to as the transcription direction, the sequence regions on the DNA strand having the same sequence as the RNA and which are 5 'to the 5' end of the transcription of RNAs are referred to as "upstream sequences", sequence regions on the DNA strand having the same sequence as RNA and which are 3 'to the 3' end of the RNA transcript are referred to as "downstream sequences".
[00261] [00261] As applied to polypeptides, the term "substantial identity" means that two sequences of peptides, when perfectly aligned, as in GAP or BESTFIT programs using standard range weights, share at least 80 percent identity in some embodiments, at least 90 percent sequence identity, in some embodiments, at least 95 percent sequence identity and, in some modalities, at least 99 percent sequence identity.
[00262] [00262] In some embodiments, the positions of residues that are not identical differ by conservative amino acid substitutions.
[00263] [00263] As discussed here, small variations in the amino acid sequences of antibodies or immunoglobulin molecules are contemplated when covered by the present description, since variations in the amino acid sequence maintain at least 75%, in some embodiments, at least 80% , 90%, 95% and, in some modalities, 99%. In particular, conservative amino acid substitutions are contemplated. Conservative substitutions are those that occur within a family of related amino acids in their side chains. Genetically encoded amino acids are generally divided into families: (1) acidic amino acids are aspartate, glutamate; (2) basic amino acids are lysine, arginine, histidine; (3) non-polar amino acids are alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan and (4) uncharged polar amino acids are glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine. Hydrophilic amino acids include arginine, asparagine, aspartate, glutamine, glutamate, histidine, lysine, serine and threonine. Hydrophobic amino acids include alanine, cysteine, isoleucine, leucine, methionine, phenylalanine, proline, tryptophan, tyrosine and valine. Other families of amino acids include (i) serine and threonine, which are the aliphatic-hydroxy family; (ii) asparagine and glutamine, which are the family containing amide; (iii) alanine, valine, leucine and isoleucine, which are the aliphatic family; and (iv) phenylalanine, tryptophan and tyrosine, which are the aromatic family. For example, it is reasonable to expect that an isolated substitution of a leucine for an isoleucine or valine, an aspartate for a glutamate, a threonine for a serine or a similar substitution of an amino acid for a structurally related amino acid will not have an effect important in the binding or properties of the resulting molecule, especially if the substitution does not involve an amino acid within a site of the structure. Whether an amino acid change results in a functional peptide can be easily determined by assessing the specific activity of the polypeptide derivative. The tests are described in detail here. Fragments or analogues of antibodies or immunoglobulin molecules can easily be prepared by those skilled in the art. The appropriate amino and carboxy terminals of fragments or analogues occur close to the limits of the functional domains. The structural and functional domains can be identified by comparing the nucleotide and / or amino acid sequence data with public or proprietary sequence databases. In some modalities, computerized comparison methods are used to identify motifs of sequence or predicted domains of protein conformation that occur in other proteins of known structure and / or function. Methods for identifying protein sequences that fold into a known three-dimensional structure are known. Bowie et al., Science 253: 164 (1991). Thus, the previous examples demonstrate that those skilled in the art can recognize motifs of sequence and structural conformations that can be used to define structural and functional domains according to the description.
[00264] [00264] Suitable amino acid substitutions are those that: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter the binding affinity for the formation of protein complexes, (4) they alter binding affinities and (5) confer or modify other physicochemical or functional properties of these analogs. Analogs can include several muteins of a sequence other than the naturally occurring peptide sequence. For example, single or multiple amino acid substitutions (for example, conservative amino acid substitutions) can be made in the naturally occurring sequence (for example, in the portion of polypeptide outside the domain (s), forming contacts The amino acid substitution should not substantially alter the structural characteristics of the origin sequence (for example, a substitution amino acid should not tend to break a helix that occurs in the origin sequence or interrupt other types of secondary structure that characterize the origin sequence.) Examples of secondary and tertiary polypeptide structures recognized in the art are described in Proteins, Structures and Molecular Principles (Creighton, Ed., WH Freeman and Company, New York (1984)); Introduction to Protein Structure (C. Branden and J. Tooze, eds., Garland Publishing, New York, NY (1991)) and Thornton et al., Nature 354: 105 (1991).
[00265] [00265] The term "polypeptide fragment", when used herein, refers to a polypeptide that has an amino and / or carboxy terminal deletion and / or one or more internal deletion (s), rém, where the remaining amino acid sequence is identical to the corresponding positions in the naturally occurring sequence minus, for example, a full-size cDNA sequence. Fragments typically have at least 5, 6, 8 or 10 amino acids in length, in some embodiments, at least 14 amino acids in length, in some embodiments, at least 20 amino acids in length, generally at least 50 amino acids in length and in some modalities, at least 70 amino acids in length. The term "analog", when used herein, refers to polypeptides that are composed of a segment of at least 25 amino acids that have substantial identity with a portion of a deduced sequence of amino acids and that have specific binding to the target, under suitable binding conditions. Typically, polypeptide analogs comprise a conservative amino acid substitution (or addition or deletion) in relation to the naturally occurring sequence. Analogs typically are at least 20 amino acids long, in some embodiments, at least 50 amino acids long.
[00266] [00266] The term "agent" is used here to denote a chemical compound, a mixture of chemical compounds, a biological macromolecule or an extract made from biological materials.
[00267] [00267] When used herein, the terms "marker" or "marked" refer to the incorporation of a detectable marker, for example, by incorporating a radiolabelled amino acid or by connecting to a polypeptide portions of biotinyl that can be detected by avidin labeled (for example, streptavidin containing a fluorescent marker or enzyme activity that can be detected by optical or calorimetric methods). In certain situations, the label or marker may likewise be therapeutic. Various methods of marking polypeptides and glycoproteins are known in the art and can be used. Examples of polypeptide clips include, but are not limited to, radioisotopes or radionuclides (eg, 3H, 14 15 35 35 90 99 111 125 131 C, N, N, S, Y, Tc, In, I, I), tags fluorescent (eg, FITC, rhodamine, lanthanide matches), enzyme labels (eg, horseradish peroxidase, p-galactosidase, luciferase, alkaline phosphatase), chemiluminescent groups, biotinyl, recognized predetermined polypeptide epitopes by a secondary reporter (for example, sequences of leucine zipper pairs, binding sites for secondary antibodies, metal binding domains, epitope labels). In some embodiments, the labels are connected by spacer arms of various lengths to reduce potential steric impediment. The term "pharmaceutical agent or drug", when used herein, refers to a compound or chemical composition capable of inducing a desired therapeutic effect when properly administered to a patient.
[00268] [00268] Other chemical terms used here are used according to conventional usage in the art, as exemplified by The McGraw-Hill Dictionary of Chemical Terms (Parker, S., Ed., McGraw-Hill, San Francisco (1985)).
[00269] [00269] When used here, "substantially pure" means that an object species is the predominant species present (that is, on a molar basis it is more abundant than any other individual species in the composition) and, in some modalities, a substantially purified fraction is a composition in which the object species comprise at least about 50% (on a molar basis) of all macromolecular species present.
[00270] [00270] Generally, a substantially pure composition will comprise more than 80% of all macromolecular species present in the composition, in some modalities, more than 85%, 90%, 95% and 99%. In some embodiments, the object species is purified for essential homogeneity (contaminating species cannot be detected in the composition by conventional detection methods), in which the composition consists essentially of a single macromolecular species.
[00271] [00271] The term patient includes human and veterinary individuals.
[00272] [00272] The terms individual and patient are used interchangeably here.
[00273] [00273] The antibodies and / or activable antibodies of the description bind specifically to a specific target, for example, a human target protein, such as human PDL1. Likewise, the antibodies and / or activable antibodies that bind to the same epitope as the antibodies and / or activable antibodies described here are included in the description. Also included in the description are antibodies and / or antibodies activable by antibodies that compete with an anti-PDL1 antibody and / or an activable anti-PDL1 antibody described herein for binding to PDL1, for example, human PDL1. Likewise included
[00274] [00274] Those skilled in the art will recognize that it is possible to determine, without undue experimentation, whether a monoclonal antibody (for example, a murine or humanized monoclonal antibody) has the same specificity as a monoclonal antibody used in the methods described here to verify if the former prevents the latter from connecting to the target. If the monoclonal antibody to be tested competes with the monoclonal antibody of the description, as shown by a decrease in the binding of the monoclonal antibody of the description, the two monoclonal antibodies bind to the same, or to a closely related epitope. An alternative method for determining whether a monoclonal antibody has the specificity of a monoclonal antibody of the description is to preincubate the monoclonal antibody of the description with the target and then add the monoclonal antibody being tested to determine whether the monoclonal antibody being tested being tested is inhibited in its ability to bind the target. If the monoclonal antibody under test is likely to be inhibited, it has the same epitopic specificity, or functionally equivalent, as the monoclonal antibody of the description.
[00275] [00275] The invention will be further described in the following examples, which do not limit the scope of the invention described in the claims.
[00276] [00276] This example assesses safety, tolerability, pharmacokinetics (PK), pharmacodynamics (PD) and preliminary antitumor activity of one of more doses, for example, a single dose or multiple domains.
[00277] [00277] This example used the activatable anti-PDL1 antibody referred to here as activable anti-PDL1 antibody PL07-2001-C5H9v2, which comprises the following sequences of variable regions of the heavy and light chain: Variable Heavy Chain of PL07-2001-C5H9v2 EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGL
[00278] [00278] Activable anti-PDL1 antibody PL07-2001-C5H9v2 comprises the following heavy and light chain sequences: PL07-2001-C5H9v2 heavy chain sequence EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGL EWVSSIWRNGIVTVYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTA VYYCAKWSAAFDYWGQGTLVTVSSASTKGPSVFPLAPCSRSTSEST AALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSV VTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEF LGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVD GVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVS NKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVK GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSR
[00279] [00279] It is known that tumors can evade host immunity by expression of programmed death ligand 1 (PD-L1), a ligand that negatively regulates programmed cell death 1 (PD-1), an inhibitory receptor expressed in cells Activated T's (Herbst RS et al., Nature. Vol. 515: 563-67 (2014)). Antibodies to PD-L1 have shown activity against a variety of cancers and are being tested in combination with other immunotherapies in an effort to improve response rates and response durability (Iwai Y et al., J Biomed Sci., Vol. 24:26 (2017) However, significant toxicities related to life-threatening immunity (irAEs) are known toxins of antibodies that block the PD1 / PDL1 axis, especially when used in a wide variety of combinations with other immunotherapies , including with ipilimumab (Wolchok JD et al., N Engl J Med., Vol. 369: 122-33 (2013); Larkin J et al., N Engl J Med., Vol. 373: 23-34 (2015) , vemurafenib, cobimetinib, vemurafenib / cobimetinib, pazopanib or osimertinib (Ahn MJ et al., Expert Opin Drug Drug, vol. 16: 465-469 (2017); Hwu P et al., Ann Online, vol. 27: 379-400 (2016)).
[00280] [00280] More than 90% of tumor samples from 200 patients with a variety of malignancies demonstrated activation of the activable antibody PL07-2001-C5H9v2 in in situ studies (see, PCT International Publication Number WO / 2014 / 107599, published on July 10, 2014, by Vasiljeva O, et al., For representative testing techniques), a finding that corroborates the presence of proteases from the tumor microenvironment in the overwhelming majority of tumors necessary to ensure the activation of the activable antibody in vivo. In addition, preclinical results demonstrated equivalent efficacy for a mouse substitute of the activable antibody PL07- 2001-C5H9v2 compared to the mouse substitute parent antibody, while minimizing the induction of systemic autoimmunity in non-obese diabetic mice susceptible to dia- betes (Wong C. et al., Presented at CRI-CIMT-EATI-AACR; September 16, 2015; New York, NY). The mouse substitute for the activable antibody PL07-2001-C5H9v2 likewise exhibited reduced peripheral binding to circulating T cells in tumor-bearing mice compared to the parental antibody (Wong C. et al., Ibid.).
[00281] [00281] The study described here is a phase 1/2, open label, multicenter study with dose increase, performed in several parts, as shown in Figures 1A and 1B, where "AA" represents the antibody activable anti-PDL1 PL07-2001-C5H9v2.
[00282] [00282] The study includes dose-increasing groups that receive activable antibodies: monotherapy groups (parts A and A2), a combination therapy with the ipilimumab group, however, two different regimens (Parts B1 and B2), one combination therapy with the vemurafenib group (Part C), and a monotherapy group in a dose expansion phase (Figure 1A does not include Part A2; Figure 1B includes Part A2). Within each Party, the dose increase followed a 3 + 3 project. Not all individuals in the study were necessarily enrolled in Part A2, but for those who, in some modalities, enrollment in Part A2 requires completion successful monotherapy dose level in Part A. For those enrolling in Part A2, this Part A2 refines the selection of the BAT / maximum dose obtained by assessing the dose / exposure relationship safely and effectively and with the levels of PL07-2001-C5H9v2 activated in the tumor microenvironment and in plasma in patients with PD-L1 + tumors. Starting cohort enrollment in Parts B1, B2 and C requires successful completion of the subsequent monotherapy dose level tested in at least Part A. Enrollment in Part D, the expansion phase, begins after the dose increase in Part A is complete and the maximum tolerated dose (BAT) was determined. Treatment continues for up to 2 years or until the progression of the disease is confirmed or the toxicity becomes unacceptable.
[00283] [00283] In Part A, monotherapy with activable antibody PL07- 2001-C5H9v2 was administered at the indicated dose (ie 0.03, 0.1, 0.3, 1, 3, 10, 30 mg / kg) IV every 14 days. For individuals enrolled in Part A2, Part A2, monotherapy with activable antibody PL07-2001-C5H9v2 is administered at the indicated dose IV every 14 days to study biomarkers and efficacy in PD-L1 + tumors. In Part B1, the concomitant schedule was an active antibody PL07-2001- C5H9v2 at the indicated dose plus ipilimumab at 3 mg / kg, administered IV every 21 days for 4 doses, followed by monotherapy with an activated antibody PL07-2001-C5H9v2 IV every 14 days. In Part B2, the phased scheme is monotherapy with activable antibody PL07-2001-C5H9v2 administered IV every 14 days for 4 doses, followed by the activable antibody PL07-2001-C5H9v2 at the indicated dose plus IV administered by ipilimumab each 21 days for 4 doses, followed by PL07-2001-C5H9v2, monotherapy with active antibodies administered IV every 14 days. In part C, the activable antibody PL07-2001-C5H9v2 at the indicated dose was administered IV every 14 days plus vemurafenib at 960 mg / kg PO administered twice daily. In part D, the PL07-2001-C5H9v2 activable antibody is administered
[00284] [00284] In Figures 1A and 1B, IV represents intravenous administration, PO represents oral administration and BAT refers to the maximum level of tolerated dose.
[00285] [00285] The activable antibody PL07-2001-C5H9v2 in Part A was dosed as follows: a first cohort was administered 0.03 mg / kg, a second cohort was administered 0.10 mg / kg, a third cohort was administered 0.3 mg / kg, the fourth cohort was administered 1.0 mg / kg, a fifth cohort was administered 3.0 mg / kg, a sixth cohort was administered at 10.0 mg / kg and a seventh cohort was administered in 30.0 mg / kg.
[00286] [00286] For individuals who enroll in Part A2, the activable antibody PL07-2001-C5H9v2 in Part A2 is dosed as follows: a first cohort is administered 0.3 mg / kg, a second cohort is administered 1 , 0 mg / kg, a third cohort is administered 3.0 mg / kg, and a fourth cohort is administered 10.0 mg / kg.
[00287] [00287] Part B1 subjects were dosed as follows: ipilimumab is administered 3 mg / kg IV and the activable antibody PL07-2001-C5H9v2 was dosed so that a first cohort is administered at 0.3 mg / kg , a second cohort is administered at 1.0 mg / kg, a third cohort is administered at 3.0 mg / kg, a fourth cohort is administered at 10.0 mg / kg and a fifth cohort is administered at 30.0 mg / kg.
[00288] [00288] Individuals in Part B2 are dosed as follows: ipilimumab is administered 3 mg / kg IV and the activable antibody PL07- 2001-C5H9v2 is dosed so that a first cohort is administered at 3.0 mg / kg, a second cohort is administered at 10.0 mg / kg, and a third cohort is administered at 30.0 mg / kg, a fourth cohort is administered at 10.0 mg / kg and a fifth cohort is administered at 30 , 0 mg / kg.
[00289] [00289] Part C individuals are dosed as follows: vermurafenib is released 960 mg / kg PO and the activable antibody PL07- 2001-C5H9v2 is dosed so that a first cohort is administered 1.0 mg / kg , a second cohort is administered at 3.0 mg / kg, and a third cohort is administered at 10.0 mg / kg, and a fourth cohort is administered at 30.0 mg / kg.
[00290] [00290] Part D individuals are dosed as follows: the activable antibody PL07-2001-C5H9v2 is administered in BAT.
[00291] [00291] Within each part of the study, the dose increase of the administered anti-PDL1 activable antibody follows a 3 + 3 project, which is a rules-based project in which the lowest dose level is allocated to the first cohort, the dose is adaptively increased progressively or not progressively increased based on the observed dose limiting toxicities (DLTs), and the adaptive increase or non-increase is repeated until the maximum tolerated dose (BAT) is reached. In Part A, an individual is each enrolled in the dosage cohorts of 0.03, 0.1 and 0.3 mg / kg, and subsequent dose levels will follow the 3 + 3 project.
[00292] [00292] In this study, enrollment in Part A2, as depicted in Figure 1B requires successful completion of the monotherapy dose level in Part A. Part A2 will register at least six more patients with PD-L1 + cancer in each indicated dose, including a minimum of 2 individuals per cohort with thymoma, thymic carcinoma or a thymic epithelial tumor. Part A2 will refine the BAT / maximum achieved dose (MAD), to assess the relationship between dose / exposure and safety, efficacy and pharmacodynamic biomarkers, and the levels of activated antibody in the tumor microenvironment and in the plasma.
[00293] [00293] In this study, starting cohort enrollment in Parts B1, B2 and C of Figures 1A or 1B requires successful completion of the subsequent monotherapy dose level tested in at least Part A. Part D is started after the dose increase is complete for Part A and the maximum tolerated dose is determined. Treatment is continued for up to 2 years or until confirmed disease progression or unacceptable toxicity.
[00294] [00294] In this study, when Part A2 is included, up to 175 patients are enrolled in the dose increase cohorts (1-6 patients per dose cohort in Part A, approximately 6 patients per dose cohort in Part A2, and 3-6 patients per dose cohort in Parts B1, B2 and C). Approximately 20 patients are enrolled in the dose expansion cohort (Part D). If Part A2 is not included, up to 150 patients will be enrolled as established immediately above, omitting Part A2. The main eligibility criteria for enrolled patients are shown in Table A below. Table A. Main eligibility criteria. All parts • age ≥ 18 years • ECOG performance status 0-1 Part A • Advanced, unresectable solid tumor or lymphoma with no additional standard of care available • Naive to immunotherapy (including therapy with PD-1 / inhibitors / PD-L1 and CTLA-4) • Immunotherapy unavailable for the patient's disease Part A2 (Opci- • The same requirements as in Part A, and must be PD-L1 + (onal score) of the tumor proportion in at least 1% of staining remember) • You must agree to participate in the biomarker analysis and have a safe tumor site for biopsy
[00295] [00295] In some modalities, when a cohort assignment occurs, patients with a known PD-L1 status are assigned to Part A, however, the PD-L1 status is not an inclusion criterion
[00296] [00296] The main objectives of this study are: (i) safety and tolerability of the activable antibody PL07-2001-C5H9v2 alone or in combination with ipilimumab or vemurafenib and / or (ii) maximum tolerated dose and dose-limiting toxicities of the activable antibody PL07 - 2001-C5H9v2 alone or in combination with ipilimumab or vemurafenib.
[00297] [00297] The secondary objectives of this study may include any of the following, or any combination of them: objective response according to the Response Assessment Criteria in Solid Tumors version 1.1 (RECIST v 1.1), RECIST immuno- related or modified Cheson / Lugano Classification for lymphomas; response time; response duration; progression-free survival; incidence of anti-drug antibodies; pharmacokinetic profile of single and multiple dose of the PL07-2001-C5H9v2 activable antibody alone and of the PL07-2001-C5H9v2 activable antibody in combination with ipilimumab, or vemurafenib; and / or overall survival.
[00298] [00298] The additional goals / objectives of this study may include any one or more of the following, or any combination thereof: potential predictive markers of activity of the activable antibody PL07-2001-C5H9v2; protease activity and degree of antibody cleavage activable by PL07-2001-C5H9v2 in the tumor and in the peripheral blood; and / or immunomodulatory activity of the activable antibody PL07- 2001-C5H9v2 in biopsies under treatment.
[00299] [00299] The following evaluations are exemplary and are not intended to be limiting. They are performed, in some modalities, at each study visit: adverse events, physical examination, vital signs, hematology, serum chemistry, B symptoms (lymphoma patients), performance status of the Eastern Cooperative Oncology Group (ECOG ) and concomitant drugs. Images for assessing the tumor response are taken every 8 weeks for the first 12 months and then every 12 weeks thereafter. Blood samples for pharmacokinetic, pharmacodynamic and biomarker analyzes are obtained at pre-specified times. After the last dose of study medication, patients are assessed every 3 months for disease progression and overall survival until withdrawal from the study or death. In some modalities, biopsies are collected. In some modalities, samples of archival tissue or fresh biopsy are provided at the baseline. In some modalities, Part A2 patients are subjected to at least one tumor biopsy under treatment. In some modalities, Part B2 patients undergo at least one tumor biopsy under treatment. In some modalities, these patients have measurable diseases.
[00300] [00300] Several translational strategies / methods are used to investigate, for example, the presence of activating antibody-activating protease activity, activation of activable antibodies, for example, activation of protease-dependent activable antibodies, presence of target (PDL1), target involvement, inhibition of PDL1 or other inhibition of the PD-1 pathway, pattern of immune response in the tumor and other biological effects. Such strategies / methods may include any one or more of the following, or any combination thereof: (a) activation of antibody activable in, for example, biopsies or blood samples, for example, plasma, using, for example , (i) a WES assay, which comprises capillary electrophoresis with immunodetection; see, for example, the WES ProteinSimple’s Simple Western brochure, and / or (ii) an assay that detects protease activation of activable antibodies, such as one of the assays described in WO / 2014/107599, ibid .; (b) evaluation of the pharmacodynamic biomarker by, for example: (i) NANOSTRING gene expression panel from, for example, a biopsy, (ii) IHC, for example, from a biopsy, to detect the infiltration of immune cells and / or (iii) evaluation of the LUMINEX cytokine panel, for example, plasma; and / or (c) evaluation of the expression of PD-L1 from, for example, a biopsy, by, for example, IHC. In some embodiments, the immunoPET image will be used.
[00301] [00301] An example of a WES assay that compares the amount of activable antibody PL07-2001-C5H9v2 cleaved and intact in preclinical tumor and plasma samples is shown in Figure 2)
[00302] [00302] In the dose increase segment described above the test, pharmacokinetics (PK) in patients who received a single dose of monotherapy with PL07-2001-C5H9v2 was evaluated. PK samples were collected intensively after the first dose of PL07-2001- C5H9v2, with sparse collection later. The analyzed ones quantified in plasma samples were intact (non-cleaved) activable antibody (ie, the prodrug form) and the total sum of the intact and cleaved PL07-2001-C5H9v2 (representing the sum of intact and activated species). Preliminary single dose PK data were collected for patients enrolled in the dose increase segment of the studies described above, receiving a single dose of 0.03-30.0 mg / kg of PL07-2001-C5H9v2 as a single agent.
[00303] [00303] The concentrations of PL07-2001-C5H9v2 both intact and total (most activated intact) were determined in plasma samples using a validated tandem mass chromatography method (HPLC MS / MS) with a lower limit of quantification for each analyzed of 0.657 nM. Magnetic beads coated with protein A were used to enrich the immunoglobulin (including PL07-2001-C5H9v2 intact and activated) in K2EDTA plasma samples. Bound proteins were digested with trypsin and two peptide fragments exclusive to PL07-2001-C5H9v2 were monitored: a heavy chain peptide present in both intact and activated forms of PL07-2001- C5H9v2 (for quantification of total PL07-2001-C5H9v2) and a peptide that is present in the intact (activable form) of PL07-2001- C5H9v2, however, not in the activated PL07-2001-C5H9v2 (for quantification of intact PL07-2001-C5H9v2). After the steps of immunocapture and digestion, the final extract was analyzed by HPLC with detection by MS / MS using positive ion electrospray. The results are shown in Figures 8A and 8B, which show the mean plasma concentration of PL07-2001-C5H9v2 (nM) intact (Figure 8A) and total PL07-2001-C5H9v2 (ie, intact and activated (nM), Figure 8B), respectively, versus the time (day) after administration of up to 30 mg / kg of q2w in cohorts A and A2 Cycle 1 Dose 1.
[00304] [00304] Preliminary single dose PK data suggests that PL07-2001-C5H9v2 circulates predominantly as the prodrug species, intact. There does not seem to be a monotonic tendency for clearance estimates and volume of distribution at dose levels of 0.1 to 30 mg / kg. A mechanistic PK model suggests that target-mediated drug disposition (TMDD) may not be an important contributor to the clearance of intact and protected PL07-2001-C5H9v2 across the range of doses assessed.
[00305] [00305] Regarding the assessment of PL07-2001-C5H9v2 as a monotherapy in a dose-increasing cohort in patients with intensively pretreated advanced solid tumors, eligible patients include those who are naive to the PD-1 inhibitor, PD-L1 and CTLA-4 with immunotherapy (IMT) unavailable as a standard of care for your disease. PL07-2001-C5H9v2 was administered every 14 days in cohorts of doses ranging from 0.03 to 30 mg / kg IV. Vin and two patients with a median age of 65 years (range 32-81) were enrolled with a median of three previous anticancer treatments (range 1-13).
[00306] [00306] The following preliminary results were observed: 1 dose-limiting toxicity (DLT) was observed (Grade 3 febrile neutropenia; 3 mg / kg); the maximum tolerated dose (BAT) has not been reached. Grade 3-4 treatment-related events were observed in 2 patients, respectively: febrile neutropenia / thromboctonpenia (3 mg / kg) and elevated AST / ALT (30 mg / kg). At all dose levels, the best response based on the alteration of baseline target lesions in 17 patients evaluated included 2 PR (thymoma and TNBC negative for PD-L1), 11 SD and 4 SD. 7/17 (41%) of the patients evaluated had a decrease in the target lesions since the baseline, according to RECIST v1.1. At dose levels ≥ 3 mg / kg, 5/8 subjects (63%) had decreased target lesions from baseline. Thus, preliminary data suggest that PL07-2001-C5H9v2 in heavily pre-treated patients with solid tumors naive to IMT, where the checkpoint block is not available, as the SOC for their disease shows a profile manageable security system with signs of anti-tumor activity.
[00307] [00307] After approximately four months, a new data cut was made after obtaining the above results. At the date of the last data cut, Part A had enrolled 22 patients, including 2 patients still undergoing treatment. Twenty patients interrupted treatment for the following reasons: radiological or clinical progression of the disease (n = 16), voluntary withdrawal (n = 2) or adverse event (n = 2). Subjects had any of several different types of cancer, including, for example, uterine carcinoma, esophageal carcinoma, pancreatic carcinoma, castration-resistant prostate carcinoma, rectal carcinoma, thymoma or thymic cancers, and triple negative breast cancer . The baseline characteristics of patients treated with PL07-2001-C5H9v2 are provided in Table 3
[00308] [00308] The average durations (range) of the treatment are given in Table 4.
[00309] [00309] Preliminary single-dose, single-agent, PL07-2001-C5H9v2 pharmacokinetic data suggests that PL07-2001-C5H9v2: (a) circulates predominantly as the intact prodrug species (96% intact at 30 mg / kg); and (b) it is probably only minimally influenced by the target-mediated drug disposition at low doses. In comparison, the PD-L1 inhibitor atezolizumab appears to exhibit non-linear PK below the dose level of 1 mg / kg. See, R.S. Herbst, et al., "Predictive correlates of response to the anti-PD-L1 anti-body MPDL3280A in cancer patients," Nature (2014 November 27) 515 (7528): 563-567. Tumor response rates among evaluated patients (n = 20) are provided in Table 5. Table 5. Best Tumor Response in Patients Evaluated by RE- CIST1 v1.1, n (%) PL07-2001- 0.03 dose 0.1 0.3 1.0 3.0 10.0 30.0 All Patients Av- C5H9v2, mg / kg n = 2 n = 2 n = 2 n = 3 n = 5 n = 3 n = 3 = 20 Best global response, n (%) Partial response 0 0 0 0 1 (20.0) 2 (66.7) 0 3 (15.0) Stable disease 1 (50.0) 1 (50.0) 1 (50.0) 1 (33.3) 2 (40.0) c 0 2 (66.7) 8 (40.0) Progressive disease 1 (50.0) 1 (50.0) 1 (50.0 ) 2 (66.7) 1 (20.0) 1 (33.3) 0 7 (35.0) Not assessable 0 0 0 0 1 (20.0) 0 1 (33.3) 2 (10.0 ) 1 RECIST: Response Assessment Criteria in Solid Tumors. a Patients assessed are those with an adequate assessment of the disease at baseline and ≥ 1 assessment of the tumor after the baseline. bIncludes 2 patients with unconfirmed partial response. c Includes 1 patient with incomplete response / non-progressive disease who had no measurable disease at baseline.
[00310] [00310] The increase to 30 mg / kg has been completed and the maximum tolerated dose (BAT) has not been reached. Target lesions decreased from baseline in 8 of 19 patients (42%) with measurable disease at baseline, as shown in Figure 9A. Target lesions decreased from baseline by dose levels ≥ 3 mg / kg in 6 out of 10 patients (60%). The percentage change in tumor burden over time is shown in Figure 9B. Sample Case Studies
[00311] [00311] Patient A has thymic cancer with high expression of baseline PD-L1 and received treatment with PL07-2001-C5H9v2 at a dose of 3 mg / kg. The patient responded to treatment after 2 weeks and had a 48% reduction in medi- tinal mass. The patient stopped treatment due to neutropenia.
[00312] [00312] Patient B has triple negative breast cancer with low stable mutational tumor load in microsatellites (4 mutations / megabase) and PD-L1 negative and received treatment with PL07- 2001-C5H9v2 at a dose of 10 mg / kg. Follow-up staging revealed a confirmed partial response. The results are shown in Table 6. Table 6. Tracking Node C2D56 C4D56 Aug 14, 2017 Dec 5, 2017 Mar 27, 2018 Right axillary 30 mm 12 mm 9 mm Precarinal lymph 17 mm 9 mm 6 mm Subcutaneous 25 mm 14 mm 19 mm
[00313] [00313] The analysis of biomarkers from patient biopsy pairs of patient C (esophageal cancer; PL07-2001-C5H9v2, 30 mg / kg) demonstrated a 3-fold increase in the infiltration of CD8 + T cells after 4 weeks of treatment. Conclusions
[00314] [00314] BAT has not been determined with doses up to 30 mg / kg. PL07-2001-C5H9v2 is activated in vivo and exerts biological activity, as evidenced by: (a) 3 objective responses in 20 patients evaluated
[00315] [00315] The main objectives of Part B1 of the study are to assess safety and tolerability and to determine the maximum tolerated dose (BAT) and dose limiting toxicity (DLT) of PL07-2001-C5H9v2 when administered in combination programming with ipilimumab. The secondary objectives are to obtain preliminary evidence of anticancer activity in patients treated with PL07- 001-C5H9v2 combined with ipilimumab using response rate (Response Evaluation Criteria in Solid Tumors (RECIST) v
[00316] [00316] Patients with advanced solid tumors received
[00317] [00317] Planned doses: PL07-2001-C5H9v2 0.3-30 mg / kg intravenously (IV) every 21 days + ipilimumab 3 mg / kg or 10 mg / kg IV every 21 days for 4 cycles, in a row monotherapy with PL07- 2001-C5H9v2 every 14 days. In preliminary results, Part B1 registered 9 patients. The median age was 44 years (range, 28-70); 6 patients (67%) were male. The average number of previous anticancer treatments was 4 (range, 2-18). At the time of data cut, 6 patients remained on treatment. The average number of doses of PL07-2001-C5H9v2 (0.3 and 1 mg / kg) and ipilimumab (3 mg / kg) was 2 (range, 2-10) and 2 (range, 2-4), respectively. 1 DLT was observed (grade 3 dyspnoea, 0.3 mg / kg PL07-2001-C5H9v2 + 3 mg / kg ipilimumab). BAT was not achieved and the dose increase continues. Grade 1-2 treatment-related adverse events (TRAEs) occurred in 6 patients (67%). Four grade 3 TRAEs were experienced by 2 patients (22%) and included colitis, pneumonitis and increases in AST and ALT (0.3 mg / kg of PL07-2001-C5H9v2) + 3 mg / kg of ipilimumab). On the data cut-off date, 1 out of 4 patients
[00318] [00318] A new data cut was made after the preliminary results above were obtained. In this last cut of data, N = 16 subjects received the following doses of PL07-2001-C5H9v2 + ipilimumab, 3.0 mg / kg. 0.3, n = 6, 1.0, n = 3, 3.0, n = 3, 10, n = 4. The baseline characteristics are shown in Table 7. Table 7. Baseline characteristics All Patients N = 16 Mean age, years (range) 60 (28-70) Sex, n (%) Male 8 (50.0) Female 8 (50.0) Race, n (%) White 13 (81, 3) Asian 1 (6.3) Not reported / unknown / other 2 (12.5) ECOG performance status, n (%) 0 6 (37.5) 1 10 (62.5) No. of previous treatment cancer, median (range) 3 (1-12) Types of cancer, an (%) Pancreatic carcinoma 2 (12.5) Other 14 (87.5) a One patient had anal squamous cell carcinoma, breast carcinoma (ER + ), cervical carcinoma, colon carcinoma, gastric cancer, glioblastoma, osteosarcoma, salivary gland carcinoma, cancer of unknown primary origin (CUP), small cell lung cancer, neuroendocrine cell cancer small cells, testicular carcinoma, triple negative breast cancer and squamous cell carcinoma of the head and neck.
[00319] [00319] At the time of the analysis, 4 patients (25.0%) were still receiving treatment. 12 patients discontinued treatment because of disease progression (n = 8), symptomatic deterioration (n = 3) or death n = 1).
[00320] [00320] The average durations (range) of the treatment are given in Table 8. Table 8 Duration of PL07-2001-C5H9v2. PL07-2001-C5H9v2 dose (mg / kg) 0.3 1.0 3.0 10.0 All Patients + Ipilimumab 3.0 mg / kg n = 6 n = 3 n = 3 n = 4 N = 16 Treatment, mean (range), 3.0 (1-10) 4.6 (3-6) 3.4 (1-4) 1.8 (1-3) 3.1 (1-10) months Response to Tumor
[00321] [00321] The best responses to the tumor are shown in Table 9. Table 9. Best Response to Tumor in Patients Evaluated by RECIST v1.1, n (%) Dose of PL07-2001-C5H9v2 0.3 + 3.0 1.0 + 3.0 3.0 + 3.0 10.0 + 3.0 All Patients (mg / kg) + Ipilimumab (mg / kg) n = 5 n = 3 n = 2 n = 2 Evaluated N = 12 Objective response taxab 1 (20.0) 1 (33.3) 1 (50.0) 0 3 (25.0) Complete response 1 (20.0) 0 0 0 1 (8.3) Partial response 0 1 (33.3) 1 (50.0) 0 2 (16.7) Stable disease 0 1 (33.3) 0 0 1 (8.3) Progressive disease 4 (80.0) 1 (33.3 ) 1 (50.0) 2 (100.0) 8 (66.7) aPatients evaluated are those with an adequate assessment of the disease at baseline and ≥ 1 assessment of the tumor after the baseline. bIncludes patients with an unconfirmed response.
[00322] [00322] Among the evaluated patients (n = 12), the best response to the tumor was: (a) Complete response (n = 1): anal squamous cell carcinoma (0.3 mg / kg of PL07-2001- C5H9v2, 3 mg / kg of ipilimumab); negative for PD-L1, MSS, low TMB, HPV-pos; and (b) Partial response (n = 2): unknown testicular and primary cancer (probably small intestine). Target lesions decreased from baseline in 3 out of 10 (30%) patients with measurable disease at baseline, as shown in Figure 10A. The percentage change in tumor load over time is shown in Figure 10B. Sample Case Studies
[00323] [00323] Patient A has anal squamous cell carcinoma with mutational intermediary tumor load (9 megabase mutations), stable to microsatellites, positive for HPV and uncovered PD-L1 status.
[00324] [00324] Patient B has carcinoma of the small intestine and negative PD-L1 status. The patient was treated with PL07-2001-C5H9v2, 3 mg / kg + ipilimumab, 3 mg / kg, and had a partial response not confirmed at follow-up. Conclusions
[00325] [00325] Early safety observations in this dose-increasing study of the combination of the activating anti-PD-L1 antibody, PL07-2001-C5H9v2 and ipilimumab, 3 mg / kg, report an AE rate related to treatment below the reported level for other PD-1 pathway inhibitors in combination with ipilimumab. No new safety signs were observed with the combination of the active anti-PD-L1 antibody, PL07-2001-C5H9v2 + ipilimumab, 3 mg / kg. The preliminary results of efficacy show 1 complete response and 2 partial responses (3/12, 25%). EXAMPLE 2. Generation of Antibodies that Bind Activated and Activated Anti-PDL1 Antibodies
[00326] [00326] The studies provided here are designed to generate and evaluate antibodies that bind to activable anti-PDL1 antibodies of the description.
[00327] [00327] The studies presented here used the active anti-PDL1 antibody referred to here as PL07-2001-C5H9v2, which comprises the heavy chain sequence of SEQ ID NO: 432 and the light chain sequence of SEQ ID NO : 428, as shown below.
[00328] [00328] The mice were immunized by GenScript Biotech Corporation with the CQQDNGYPSTFGGGT peptide antigen (SEQ ID NO: 1203), comprising the VL CDR3 of the activable anti-PDL1 antibody PL07-2001-C5H9v2, which was conjugated to the vein protein - Hemocyanine Lapa in Keyhole (KLH) circle using the procedure shown below in Table 3. Six mice aged three months (3 Balb / c 3 C56) were immunized according to the protocol listed below. At the time of each injection, the antigen aliquot was thawed and combined with Freund's Complete Adjuvant (CFA) for the first injection or with Freund's incomplete Adjuvant (IFA) for subsequent injections. Table 10. Immunization schedule Procedure Schedule Dosage and routine Preimmune bleeding T = -4 days Primary immunization T = 0 days 50 μg / animal, sc Reinforcement 1 T = 14 days 25 μg / animal, sc Bleeding Test 1 T = 21 days Reinforcement 2 T = 28 days 25 μg / animal, sc Bleeding Test 2 T = 35 days Final Reinforcement T = 50 ± 7 days 25 μg / animal, iv Cell Fusion 4 days after the final reinforcement
[00329] [00329] Serum titers against free peptide, as well as antigen
[00330] [00330] Mouse monoclonal antibodies were generated as follows: Lymphocytes from two mice were used for hybridoma fusion and seeded in forty 96-well plates (400 million lymphocytes per mouse). The plates were kept in tissue culture incubators under standard conditions. EXAMPLE 3. Screening of Hybridoma Clones and Characterization of Antibodies
[00331] [00331] This Example describes the screening and characterization of hybridoma clones and resulting antibodies generated against the activable anti-PDL1 antibody PL07-2001-C5H9v2.
[00332] [00332] The hybridoma supernatant of parental clones was screened by GenScript against a short peptide containing the VL CDR3 of the PL07-2001-C5H9v2 activable antibody by indirect ELISA. Briefly, the GenScript high-binding plates were coated with peptide-BSA at a concentration of 1 µg / mL, 100 µL / well. The supernatant was used without dilution. Antiserum at a 1: 1000 dilution was used as a positive control. Goat Anti-Mouse IgG Peroxidase-AffiniPure, Fractional Specific FC (minimal cross-reactivity with human, bovine or cava serum albumin, also known as min X Hu, Bov, Hrs Sr Prot) was used co- secondary hand. Twenty clones with positive signals were also screened against the anti-PDL1 antibody C5H9v2, the parent antibody of the activable antibody PL07-2001-C5H9v2 and 5 µg / ml of human IgG.
[00333] [00333] This example describes the ability of the antibodies in the description to bind to the activable anti-PDL1 antibody PL07-2001- C5H9v2.
[00334] [00334] To test the specificity of 17G1 binding to the activating anti-PDL1 antibody PL07-2001-C5H9v2, 160 ng / ml of activating anti-PDL1 antibody activated by a PL07-2001-C5H9v2 arm were reinforced in human plasma (dilution from 1 to 100 in PBS) or lung tumor lysate. Briefly, tumor homogenates were prepared in Thermo Scientific Pierce ™ IP Lysis Buffer (Catalog # 87788) with the Thermo Scientific Halt ™ Protease Inhibitor Single Use Cocktail Kit (catalog # 78430) using Barocycler (Pres- sure Biosciences). The 17G1 antibody was also tested against the same plasma and tumor that were not boosted with the active anti-PDL1 antibody activated by a PL07-2001-C5H9v2 arm. The test samples were then analyzed using the method based on Wes capillary electrophoresis immunoassay, in which the separation was performed by electrophoresis based on SDS (Protein Simple), also known as Wes system. Figures 4A-4D demonstrate high specificity of binding of the 17G1 antibody to the activatable anti-PDL1 antibody PL07- 2001-C5H9v2 reinforced in human plasma (Figure 4C) and samples of lung tumor lysate (Figure 4D). Figures 4A and 4B demonstrate antecedent binding of the 17G1 antibody in samples of human plasma and lung tumor lysate, respectively, in the absence of activable anti-PDL1 antibody PL07-2001-C5H9v2. EXAMPLE 5. Quantification of Activated and Intact Activated Anti-PDL1 Antibodies in Biological Samples
[00335] [00335] This example describes the ability of the 17G1 antibody to detect the activated and intact anti-PDL1 activable antibody PL07- 2001-C5H9v2 in plasma tumor samples and carcinoma xenograft
[00336] [00336] The activable anti-PDL1 antibody PL07-2001-C5H9v2 is designed to be cleaved (ie, activated) by various serine proteases and matrix metalloproteinases (MMPs) that are generally associated with human tumors (LeBeau et al., Imaging a functional tumorigenic bio-marker in the transformed epithelium. Proc Natl Acad Sci 2013; 110: 93–98; Overall & Kleifeld, 2006, Validating Matrix Metalloproteinases as Drug Targets and Anti-Targets for Cancer Therapy. Nature Review Cancer, 6, 227-239), and with low activity in the blood or normal tissues. To evaluate and measure the activation of the activable antibody in tumor and plasma samples, the samples were analyzed by the Wes system that allows the detection of the active and activated anti-PDL1 antibody activated PL07-2001-C5H9v2, as described here. Using this system, it has been shown that the activable antibodies remain practically intact (that is, inactivated) in the circulation, however, they are activated in the tumors of the mouse xenograft.
[00337] [00337] In general, the following protocol was used: a mouse xenograft tumor model was developed by SC implantation of 3x106 MDA-MB-231-luc2-4D3LN cells in 30 μl of serum-free medium containing matrigel (1 : 1) for 7-8 weeks old female nude mice. Body weights and tumor measurements were measured and recorded twice a week during the study period. After the tumors reached a volume of 200-500 mm3, the mice were randomized into 3 groups of equivalent average tumor volume and dosed with the activable anti-PDL1 antibody PL07-2001-C5H9v2. Four days after treatment, the tumor and plasma (heparin) were collected and stored at -80ºC before analysis. Tumor homogenates (ie, lysates) were prepared in Thermo Scientific PierceTM IP Lysis Buffer (Catalog #
[00338] [00338] The samples were analyzed using a protocol similar to that described by ProteinSimple in the Simple Western Size Assay Development Guide (http://www.proteinsimple.com/documents/042- 889_Rev1_Size_Assay_Development_Guide.pdf), as long as this method whole allows the separation of intact and activated species. In some modalities, the variation of one or more of the following using the methods can be used to facilitate the separation of intact and activated species: variation, for example, increase or decrease, stacking time, variation, for example, increase or decrease, sampling time, and / or variation, for example, increase or decrease, separation time.
[00339] [00339] In general, one part (for example, 1μL) of 5X Fluorescent Master Mix (ProteinSimple) was combined with the 4-part lysate (for example, 4 μL) to be tested in a microcentrifuge tube. A range of 1 ng to 5 µg of activable anti-PDL1 antibody PL07-2001- C5H9v2 was used for antibody screening and characterization. For biological samples comprising tumor tissue, 0.8 mg / ml protein lysate in IP lysis buffer with protease inhibitor HALT / EDTA was used. Plasma samples were diluted 1 in 100 in PBS. The primary antibodies were used in a concentration of 1.7 ng / ml (diluted in Antibody Diluent 2 (Protein- Simple Cat # 042-203). The secondary mouse antibody (ProteinSimple) was used neat. plates with samples prepared according to the Simple Western Size Assay Development Guide were centrifuged for 5 minutes at 2500 rpm (~ 1000xg) at room temperature before analysis in the Wes system (ProteinSimple).
[00340] [00340] Figures 5A and 5B compare the specific detection of activable anti-PDL1 antibody intact and activated PL07-2001-C5H9v2 by the anti-idiotypic antibody 17G1 of the description and commercial anti-human IgG A110UK (goat anti-human IgG adsorbed from a canine monkey) from American Qualex. The 17G1 antibody of the description was able to detect the activable anti-PDL1 antibody PL07-2001-C5H9v2 in the plasma of mice treated with only 0.1 mg / kg of activable anti-PDL1 antibody PL07-2001-C5H9v2 (Figure 5B) in comparison with the commercial human IgG antibody only being able to minimally detect the activable anti-PDL1 antibody PL07-2001-C5H9v2 in the plasma of mice treated with 10 mg / kg of activable anti-PDL1 antibody PL07-2001-C5H9v2 (Figure 5A).
[00341] [00341] Figures 6A and 6B show the preferential activation of the activable anti-PDL1 antibody PL07-2001-C5H9v2 in tumor versus plasma samples. In this study, the MDA-MD-231 xenograft mice were treated with 1 mg / kg of activable anti-PDL1 antibody PL07-2001-C5H9v2. The tumor and plasma samples were collected on day 4 (96 hours). The tumor homogenate and plasma samples were analyzed in the Wes system using the 17G1 antibody for detection. Plasma samples exhibited intact anti-PDL1 antibody PL07-2001-C5H9v2 intact (Figure 6B) while the tumor microenvironment activated at least a portion of the active anti-PDL1 antibody PL07-2001-C5H9v2 (Figure 6A) . EXAMPLE 6. Quantification of Activated and Intact Anti-PDL1 Activated Antibodies in Biological Samples
[00342] [00342] This example demonstrates that the Wes system can be applied to different types of xenograft tumors and different dosage concentrations.
[00343] [00343] Briefly, a mouse xenograft tumor model was developed by SC implantation of 5x106 SAS cells in 100 μL of serum-free medium for 7-8 week old female nude mice. Body weights and tumor measurements were measured and recorded twice a week during the study period. After the tumors reached a volume of 450-550 mm3, the mice were randomized into 3 groups of average tumor equivalent volume and dosed with 0.1 mg / kg of activable anti-PDL1 antibody PL07-2001-C5H9v2. Four days after treatment, the tumor and plasma samples (heparin) were collected and stored at -80ºC before analysis. Tumor homogenates (ie, lysates) were prepared in Thermo Scientific PierceTM IP Lysis Buffer (Catalog # 87788) with the Thermo Scientific Halt ™ Protease Inhibitor Single Use Cocktail Kit (Catalog # 78430) using Bacyclercler (Pressure Biosciences ). Approximately 0.8 mg / mL of protein level in IP lysis buffer with protease inhibitor HALT / EDTA and plasma samples diluted 1 in 250 in PBS were analyzed by the Wes system using the 17G1 antibody for detection. Figures 7A and 7B indicate preferential activation of activable antibody therapies in tumor versus plasma samples. Other Modalities
[00344] [00344] Although the invention has been described in conjunction with its detailed description, the above description is intended to illustrate and not to limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages and modifications are within the scope below.
权利要求:
Claims (132)
[1]
1. Activable anti-PDL1 antibody, characterized by the fact that it comprises: a. an antibody (AB) that specifically binds to human PDL1, where AB comprises: i. a heavy chain variable region comprising a complementarity determining region 1 (CDRH1) comprising the amino acid sequence of SEQ ID NO: 212, a complementarity determining region 2 (CDRH2) comprising the amino acid sequence of SEQ ID NO: 246 and a complementarity determining region 3 (CDRH3) comprising the amino acid sequence or SEQ ID NO: 235; and ii. a light chain variable region comprising a light chain complementarity determining region 1 (CDRL1) comprising the amino acid sequence of SEQ ID NO: 209, a light chain complementarity determining region 2 (CDRL2) comprising the SEQ ID amino acid sequence ID NO: 215, a light chain complementarity determining region 3 (CDRL3) comprising the amino acid sequence of SEQ ID NO: 228; B. a cleavable portion (CM) linked to AB, where CM is a polypeptide that functions as a substrate for a protease; and c. a masking portion (MM) attached to the CM, for use in treatment, alleviating a symptom of, or delaying the progression of cancer in an individual, and in which the activable antibody is administered intravenously at a dose of about 0.3 mg / kg to 30 mg / kg
[2]
2. Activable anti-PDL1 antibody, characterized by the fact that it comprises:
The. an antibody (AB) that specifically binds to human PDL1, where AB comprises: i. a heavy chain variable region comprising a complementarity determining region 1 (CDRH1) comprising the amino acid sequence of SEQ ID NO: 212, a complementarity determining region 2 (CDRH2) comprising the amino acid sequence of SEQ ID NO: 246 and a complementarity determining region 3 (CDRH3) comprising the amino acid sequence or SEQ ID NO: 235; and ii. a light chain variable region comprising a light chain complementarity determining region 1 (CDRL1) comprising the amino acid sequence of SEQ ID NO: 209, a light chain complementarity determining region 2 (CDRL2) comprising the SEQ amino acid sequence ID ID NO: 215, a light chain complementarity determining region 3 (CDRL3) comprising the amino acid sequence of SEQ ID NO: 228; B. a cleavable portion (CM) linked to AB, where CM is a polypeptide that functions as a substrate for a protease; and c. a masking portion (MM) attached to the CM, for use in treatment, alleviating a symptom of, or delaying the progression of cancer in an individual, and in which the activable antibody is administered intravenously at a fixed dose of about from 24 to 2400 mg.
[3]
3. Activable anti-PDL1 antibody according to claim 1 or 2, characterized by the fact that MM inhibits the binding of AB to human PDL1 when the activable antibody is in an uncleaved state.
[4]
4. Activable anti-PDL1 antibody according to any of claims 1 to 3, characterized by the fact that MM comprises the amino acid sequence of SEQ ID NO: 63.
[5]
An anti-PDL-1 antibody activable according to any one of claims 1 to 4, characterized by the fact that the CM comprises the amino acid sequence of SEQ ID NO: 377.
[6]
An anti-PDL1 antibody activable according to any one of claims 1 to 5, characterized by the fact that AB comprises a heavy chain variable region (VH) comprising the amino acid sequence of SEQ ID NO: 46 and a light chain variable (VL) comprising the amino acid sequence of SEQ ID NO: 58 or SEQ ID NO: 137.
[7]
7. Activable anti-PDL1 antibody according to claim 1 or 2, characterized in that the activable antibody comprises a light chain comprising the amino acid sequence of SEQ ID NO: 1008 and a heavy chain comprising the sequence - amino acid class of SEQ ID NO: 432.
[8]
8. Activable anti-PDL1 antibody according to claim 1 or 2, characterized in that the activable antibody comprises a light chain comprising the amino acid sequence of SEQ ID NO: 428 and a heavy chain comprising the sequence - amino acid class of SEQ ID NO: 432.
[9]
9. Activable anti-PDL-1 antibody according to any one of claims 1 and 3 to 8, characterized in that the dose is approximately between 3 mg / kg to 10 mg / kg.
[10]
10. Activated anti-PDL-1 antibody according to any of claims 1 and 3 to 8, characterized in that the dose is approximately between 3 mg / kg to 15 mg / kg.
[11]
11. Activated anti-PDL-1 antibody according to any one of claims 1 and 3 to 8, characterized by the fact that the dose is 0.3 mg / kg.
[12]
12. Activated anti-PDL-1 antibody according to any one of claims 1 and 3 to 8, characterized in that the dose is 1 mg / kg.
[13]
13. Activated anti-PDL-1 antibody according to any one of claims 1 and 3 to 8, characterized by the fact that the dose is 3 mg / kg.
[14]
14. Activated anti-PDL-1 antibody according to any one of claims 1 and 3 to 8, characterized in that the dose is 6 mg / kg.
[15]
15. Activated anti-PDL-1 antibody according to any one of claims 1 and 3 to 8, characterized in that the dose is 10 mg / kg.
[16]
16. Activable anti-PDL-1 antibody according to any one of claims 1 and 3 to 8, characterized by the fact that the dose is 15 mg / kg.
[17]
17. Activated anti-PDL-1 antibody according to any one of claims 1 and 3 to 8, characterized in that the dose is 30 mg / kg.
[18]
18. Anti-PDL1 antibody activable according to claims 2 to 8, characterized by the fact that the fixed dose is 240 mg.
[19]
19. Anti-PDL1 antibody activable according to claims 2 to 8, characterized by the fact that the fixed dose is 480 mg.
[20]
20. Anti-PDL1 antibody activable according to claims 2 to 8, characterized by the fact that the fixed dose is 800 mg.
[21]
21. Anti-PDL1 antibody activable according to claims 2 to 8, characterized by the fact that the fixed dose is 1200 mg.
[22]
22. Anti-PDL1 antibody activable according to claims 2 to 8, characterized by the fact that the fixed dose is 2400 mg.
[23]
23. Activable anti-PDL-1 antibody according to any of the preceding claims, characterized by the fact that the activable antibody is administered on a dose schedule every 7-30 days.
[24]
24. Activable anti-PDL-1 antibody according to claim 23, characterized by the fact that the activable antibody is administered on a dose schedule every 14 days.
[25]
25. Activable anti-PDL-1 antibody according to claim 23, characterized by the fact that the activable antibody is administered on a dose schedule every 21 days.
[26]
26. Activable anti-PDL-1 antibody according to any of the preceding claims, characterized by the fact that the activable antibody is administered as a monotherapy.
[27]
27. Anti-PDL-1 activable antibody according to any of the preceding claims, characterized by the fact that the activable antibody is administered as a component of a combination therapy.
[28]
28. Activable anti-PDL-1 antibody according to claim 27, characterized in that the combination therapy comprises administering a dose of an anti-CTLA-4 antibody or a B-RAF inhibitor.
[29]
29. Anti-PDL1 antibody activable according to claim 28, characterized by the fact that the anti-CTLA-4 antibody is ipilimumab.
[30]
30. Anti-PDL1 antibody activable according to claim 27 or 28, characterized in that the anti-CTLA-4 antibody is administered intravenously.
[31]
31. Anti-PDL1 antibody activable according to any of claims 28 to 30, characterized in that the anti-CTLA-4 antibody is administered in a dose of 3 mg / kg, 6 mg / kg or 10 mg / kg.
[32]
32. Activated anti-PDL1 antibody according to any of claims 28 to 30, characterized in that the anti-CTLA-4 antibody is administered in a fixed dose of 240 mg, 480 mg or 800 mg.
[33]
33. Anti-PDL1 antibody activable according to claim 28, characterized by the fact that the B-RAF inhibitor is virafenib.
[34]
34. Anti-PDL1 antibody activable according to claim 28 or 33, characterized by the fact that the B-RAF inhibitor is administered orally.
[35]
35. Anti-PDL1 antibody activable according to claim 28, 33 or 34, characterized by the fact that the B-RAF inhibitor is administered in a dose of 960 mg.
[36]
36. Anti-PDL1 antibody activable according to claim 28, 33 or 34, characterized by the fact that the B-RAF inhibitor is administered in a dose of 875 mg.
[37]
37. Activable anti-PDL1 antibody according to claim 28, 33 to 36, characterized by the fact that the administration step comprises administering the activable antibody and the B-RAF inhibitor over the same period of time.
[38]
38. Activated anti-PDL-1 antibody according to any of claims 28, 33 to 37, characterized in that the dose of the B-RAF inhibitor is administered twice a day.
[39]
39. Anti-PDL-1 activable antibody according to any of claims 28, 33 to 38, characterized in that at least 4 doses of each of the activable antibody and the B-RAF inhibitor are administered.
[40]
40. Anti-PDL-1 activable antibody according to any of claims 28 to 32, characterized in that the administration steps comprise the administration of multiple doses of the activable antibody and the anti-CTLA-4 antibody during a first period of time, followed by administration of multiple doses of the activable antibody as a monotherapy for a second period of time.
[41]
41. Activable anti-PDL1 antibody according to any one of claims 28 to 32, characterized in that the dose of the activable antibody and a dose of the anti-CTLA-4 antibody are administered concomitantly as a combination therapy. - gives 21 days for 4 doses, followed by administration of a dose of the activable antibody as a monotherapy every 14 days.
[42]
42. Anti-PDL-1 activable antibody according to any of claims 28 to 32, characterized in that the administration steps comprise the administration of multiple doses of the activatable antibody as a monotherapy during a first period of time, followed by the concomitant administration of multiple doses of the activatable antibody and the anti-CTLA-4 antibody as a combination therapy over a second period of time.
[43]
43. Anti-PDL-1 activable antibody according to any one of claims 28 to 32, characterized in that the administration step comprises (i) administering multiple doses of the activatable antibody as a monotherapy during a first period of time , (ii) subsequently administering multiple doses of the activable antibody and anti-CTLA-4 antibody as a combination therapy over a second period of time, and (iii) subsequently administering multiple doses of the activable antibody as a monotherapy for a third period of time.
[44]
44. Activable anti-PDL-1 antibody according to any one of claims 28 to 32, characterized in that the dose of the activable antibody is administered as a monotherapy every 14 days for 4 doses, followed by the administration of one dose of the activable antibody and a dose of anti-CTLA-4 antibody is administered as a combination therapy every 21 days, for 4 doses, followed by administration of a dose of an activable antibody as a monotherapy every 14 days.
[45]
45. Activated anti-PDL-1 antibody according to any of the preceding claims, characterized by the fact that cancer is an advanced, unresectable solid tumor or lymphoma.
[46]
46. Activable anti-PDL1 antibody according to claim 45, characterized by the fact that the advanced, non-resectable solid tumor is a type of tumor responsive to PDL1.
[47]
47. Anti-PDL-1 antibody activable according to any of the preceding claims, characterized by the fact that the cancer is a carcinoma.
[48]
48. Anti-PDL1 antibody activable according to claim 47, characterized by the fact that the carcinoma is squamous cell carcinoma.
[49]
49. Activated anti-PDL-1 antibody according to any of the preceding claims, characterized by the fact that the cancer is anal squamous cell carcinoma, basal cell carcinoma, bladder cancer, bone cancer, intestinal carcinoma, breast cancer , carcinoid, castration-resistant prostate cancer (CRPC), cervical carcinoma, colorectal cancer (CRC), cutaneous squamous cell carcinoma, colon cancer, endometrial cancer, esophageal cancer, gastric carcinoma, gastroesophageal junction cancer, glioblastoma / mixed glioma, glioma, head and neck cancer, hepatocellular carcinoma, hematological malignancy, liver cancer, lung cancer, melanoma, Merkel cell carcinoma, multiple myeloma, nasopharyngeal cancer, osteosarcoma, ovarian cancer , pancreatic cancer, peritoneal carcinoma, undifferentiated pleomorphic sarcoma, prostate cancer, rectal carcinoma, kidney cancer, sarcoma, salivary gland carcinoma, squamous cell carcinoma
sas, stomach cancer, testicular cancer, thymic carcinoma, thymic epithelial tumor, thymoma, thyroid cancer, urogenital cancer, urothelial cancer, uterine carcinoma or uterine sarcoma.
[50]
50. Anti-PDL-1 antibody activable according to any of the preceding claims, characterized by the fact that the cancer is a High Tumor Mutational Charge Cancer (hTMB).
[51]
51. Activated anti-PDL1 antibody according to claim 49, characterized by the fact that breast cancer is triple negative breast cancer or estrogen receptor positive breast cancer.
[52]
52. Anti-PDL1 antibody activable according to claim 49, characterized by the fact that the hematological malignancy is a lymphoma or leukemia.
[53]
53. Activated anti-PDL1 antibody according to claim 52, characterized in that the lymphoma is a B cell lymphoma, a T cell lymphoma, a Hodgkin lymphoma or an EBV lymphoma, a lymphoma primary mediastinal of B cells.
[54]
54. Anti-PDL1 antibody activable according to claim 49, characterized by the fact that cancer is melanoma.
[55]
55. Anti-PDL1 antibody activable according to claim 49, characterized by the fact that intestinal carcinoma is small intestine carcinoma or small intestine adenocarcinoma.
[56]
56. Anti-PDL1 antibody activable according to claim 49, characterized by the fact that colon cancer is colon adenocarcinoma.
[57]
57. Anti-PDL1 antibody activable according to claim 49, characterized by the fact that lung cancer is a non-small cell lung cancer (NSCLC) or small cell lung cancer.
[58]
58. Anti-PDL1 antibody activable according to claim 57, characterized by the fact that the NSCLC is non-squamous NSCLC or scaly NSCLC.
[59]
59. Anti-PDL1 antibody activable according to claim 49, characterized by the fact that prostate cancer is small cell neuroendocrine prostate cancer.
[60]
60. Anti-PDL1 antibody activable according to claim 49, characterized by the fact that renal cancer is renal cell carcinoma or renal sarcoma.
[61]
61. Activated anti-PDL1 antibody according to claim 49, characterized by the fact that the cancer is non-differentiated pleomorphic sarcoma, small bowel adenocarcinoma, Merkel cell carcinoma, thymic carcinoma, squamous cell carcinoma anal infections, cutaneous squamous cell carcinoma or triple negative cancer.
[62]
62. Anti-PDL-1 antibody activable according to any of the preceding claims, characterized by the fact that the individual exhibits one or more of the following characteristics: a. Naive to PD-1 / PDL1 inhibitor, b. Naive to the CTLA-4 inhibitor, c. Positive for BRAFV600E mutation, d. Naive to the BRAF inhibitor, e.g. Positive for PDL1, f. Unknown for PDL1 and g. was previously treated with a PD1 / PDL1 inhibitor.
[63]
63. Anti-PDL-1 antibody activable according to any of the preceding claims, characterized by the fact that the individual has no additional standard of care available.
[64]
64. Anti-PDL-1 antibody activable according to any of the preceding claims, characterized by the fact that PD1 / PDL1 inhibitor therapy is not approved for the individual's cancer.
[65]
65. Anti-PDL-1 antibody activable according to any of the preceding claims, characterized by the fact that the subject has previously been treated with a PD-1 / PDL1 inhibitor; in which treatment with the PD-1 / PDL1 inhibitor was interrupted for reasons other than toxicity; and where the individual is naive to the CTLA-4 inhibitor.
[66]
66. Anti-PDL-1 antibody activable according to any of the preceding claims, characterized by the fact that the individual is naive to immunotherapy.
[67]
67. Method of treating, relieving a symptom of, or delaying the progression of cancer in an individual, characterized by the fact that it comprises intravenous administration at a dose of approximately 0.3 mg / kg to 30 mg / kg an anti-PDL1 antibody activable to the subject, wherein the activable antibody comprises: a. an antibody (AB) that specifically binds to human PDL1, wherein AB comprises: i. a heavy chain variable region comprising a complementarity determining region 1 (CDRH1) comprising the amino acid sequence of SEQ ID NO: 212, a complementarity determining region 2 (CDRH2) comprising the amino acid sequence of SEQ ID NO: 246 , and a complementarity determining region 3 (CDRH3) comprising the amino acid sequence or SEQ ID NO: 235; and ii. a light chain variable region comprising a light chain complementarity determining region 1 (CDRL1) comprising the amino acid sequence of SEQ ID NO: 209, a light chain complementarity determining region 2 (CDRL2) comprising the amino acid sequence of SEQ ID
NO: 215, a light chain complementarity determining region 3 (CDRL3) comprising the amino acid sequence of SEQ ID NO: 228; B. a cleavable portion (CM) linked to AB, where CM is a polypeptide that functions as a substrate for a protease; and c. a masking portion (MM) attached to the CM.
[68]
68. Method of treating, alleviating a symptom of, or delaying the progression of cancer in an individual, characterized by the fact that comprising administering intravenously in a fixed dose of approximately between 24 and 2400 mg of an activable anti-PDL-1 antibody to the individual, in which the activable antibody comprises: a. an antibody (AB) that specifically binds to human PDL1, wherein AB comprises: i. a heavy chain variable region comprising a complementarity determining region 1 (CDRH1) comprising the amino acid sequence of SEQ ID NO: 212, a complementarity determining region 2 (CDRH2) comprising the amino acid sequence of SEQ ID NO: 246 , and a complementarity determining region 3 (CDRH3) comprising the amino acid sequence or SEQ ID NO: 235; and ii. a light chain variable region comprising a light chain complementarity determining region 1 (CDRL1) comprising the amino acid sequence of SEQ ID NO: 209, a light chain complementarity determining region 2 (CDRL2) comprising the amino acid sequence of SEQ ID NO: 215, a light chain complementarity determining region 3 (CDRL3) comprising the amino acid sequence of SEQ ID NO: 228; B. a cleavable portion (CM) linked to AB, where CM is a polypeptide that functions as a substrate for a protease; and c. a masking portion (MM) attached to the AB.
[69]
69. The method of claim 67 or 68, characterized by the fact that MM inhibits the binding of AB to human PDL1 when the activable antibody is in an uncleaved state.
[70]
70. Method according to any of claims 67 to 69, characterized by the fact that MM comprises the amino acid sequence of SEQ ID NO: 63.
[71]
71. Method according to any of claims 67 to 70, characterized by the fact that the CM comprises the amino acid sequence of SEQ ID NO: 377.
[72]
72. Method according to any of claims 67 to 71, characterized in that the AB comprises a heavy chain variable region (VH) comprising the amino acid sequence of SEQ ID NO: 46 and a light chain variable (VL) comprising the amino acid sequence of SEQ ID NO: 58 or SEQ ID NO: 137.
[73]
73. The method of claim 67 or 68, characterized in that the activable antibody comprises a light chain comprising the amino acid sequence of SEQ ID NO: 1008 and a heavy chain comprising the amino acid sequence of SEQ ID NO. : 432.
[74]
74. The method of claim 67 or 68, characterized in that the activable antibody comprises a light chain comprising the amino acid sequence of SEQ ID NO: 428 and a heavy chain comprising the amino acid sequence of SEQ ID NO : 432.
[75]
75. Method according to any of claims 67 and 69 to 74, characterized by the fact that the dose is approximately
between 3 mg / kg to 10 mg / kg.
[76]
76. Method according to any of claims 67 and 69 to 74, characterized by the fact that the dose is approximately between 3 mg / kg to 15 mg / kg.
[77]
77. Method according to any one of claims 67 and 69 to 74, characterized in that the dose is 0.3 mg / kg.
[78]
78. Method according to any one of claims 67 and 69 to 74, characterized in that the dose is 1 mg / kg.
[79]
79. Method according to any of claims 67 and 69 to 74, characterized in that the dose is 3 mg / kg.
[80]
80. Method according to any of claims 67 and 69 to 74, characterized in that the dose is 6 mg / kg.
[81]
81. Method according to any of claims 67 and 69 to 74, characterized in that the dose is 10 mg / kg.
[82]
82. Method according to any of claims 67 and 69 to 74, characterized in that the dose is 15 mg / kg.
[83]
83. Method according to any of claims 67 and 69 to 74, characterized in that the dose is 30 mg / kg.
[84]
84. Method according to claims 68 to 74, characterized by the fact that the fixed dose is 240 mg
[85]
85. Method according to claims 68 to 74, characterized in that the fixed dose is 480 mg.
[86]
86. Method according to claims 68 to 74, characterized in that the fixed dose is 800 mg.
[87]
87. Method according to claims 68 to 74, characterized by the fact that the fixed dose is 1200 mg.
[88]
88. Method according to claims 68 to 74, characterized by the fact that the fixed dose is 2400 mg.
[89]
89. Method according to any one of the preceding claims, characterized by the fact that the activable antibody is administered on a dose schedule every 7-30 days.
[90]
90. Method according to claim 89, characterized by the fact that the activable antibody is administered in a dose schedule every 14 days.
[91]
91. Method according to claim 89, characterized by the fact that the activable antibody is administered in a dose schedule every 21 days.
[92]
92. Method according to any one of the preceding claims, characterized in that the activable antibody is administered as a monotherapy.
[93]
93. Method according to any one of the preceding claims, characterized in that the activable antibody is administered as a component of a combination therapy.
[94]
94. The method of claim 93, characterized in that the combination therapy comprises administering a dose of an anti-CTLA-4 antibody or a B-RAF inhibitor
[95]
95. The method of claim 94, characterized by the fact that the anti-CTLA-4 antibody is ipilimumab.
[96]
96. Method according to claim 93 or 94, characterized in that the anti-CTLA-4 antibody is administered intravenously.
[97]
97. Method according to any one of claims 94 to 96, characterized in that the anti-CTLA-4 antibody is administered in a dose of 3 mg / kg, 6 mg / kg or 10 mg / kg.
[98]
98. Method according to any of claims 94 to 96, characterized in that the anti-CTLA-4 antibody is administered in a fixed dose of 240 mg, 480 mg or 800 mg.
[99]
99. Method according to claim 94, characterized by the fact that the B-RAF inhibitor is vemurafenib.
[100]
100. The method of claim 94 or 99, comprising
characterized by the fact that the B-RAF inhibitor is administered orally.
[101]
101. Method according to claim 94, 99 or 100, characterized in that the B-RAF inhibitor is administered in a dose of 960 mg.
[102]
102. Method according to claim 94, 99 or 100, characterized in that the B-RAF inhibitor is administered in a dose of 875 mg.
[103]
103. Method according to claims 94, 99 to 102, characterized in that the administration step comprises administering the activable antibody and the B-RAF inhibitor over the same period of time.
[104]
104. Method according to any of claims 94, 99 to 102, characterized in that the dose of the B-RAF inhibitor is administered twice a day.
[105]
105. Method according to any of claims 94, 99 to 102, characterized by the fact that at least 4 doses of each of the activable antibody and the B-RAF inhibitor are administered.
[106]
106. Method according to any of claims 94 to 98, characterized in that the administration steps comprise the administration of multiple doses of the activable antibody and the anti-CTLA-4 antibody during a first period of time , followed by administration of multiple doses of the active antibody as a monotherapy for a second period of time.
[107]
107. Method according to any of claims 94 to 98, characterized in that the dose of the active antibody and a dose of the anti-CTLA-4 antibody are administered concomitantly as a combination therapy to each 21 days for 4 doses, followed by administration of a dose of the activable antibody as a monotherapy every 14 days.
[108]
108. Method according to any of claims 94 to 98, characterized by the fact that the administration steps comprise the administration of multiple doses of the activable antibody as a monotherapy during a first period of time, followed by the concomitant administration of multiple doses of the activatable antibody and anti-CTLA-4 antibody as a combination therapy over a second period of time.
[109]
109. Method according to any one of claims 94 to 98, characterized by the fact that the administration step comprises (i) administering multiple doses of the activable antibody as a monotherapy during a first period of time, (ii) subsequently quently administer multiple doses of the activatable antibody and anti-CTLA-4 antibody as a combination therapy over a second period of time, and (iii) subsequently administer multiple doses of the activable antibody as a monotherapy over a third period of time.
[110]
110. Method according to any one of claims 94 to 98, characterized in that the dose of the activable antibody is administered as a monotherapy every 14 days for 4 doses, followed by the administration of a dose of the activable antibody and a dose of anti-CTLA-4 antibody is administered as a combination therapy every 21 days, for 4 doses, followed by the administration of a dose of an activable antibody as a monotherapy every 14 days.
[111]
111. Method according to any one of the preceding claims, characterized by the fact that cancer is an advanced, unresectable solid tumor or lymphoma.
[112]
112. Method according to claim 111, characterized by the fact that the advanced, unresectable solid tumor is a type of tumor responsive to PDL1.
[113]
113. Method according to any one of the preceding claims, characterized by the fact that cancer is a carcinoma.
[114]
114. Method according to claim 113, characterized by the fact that the carcinoma is squamous cell carcinoma.
[115]
115. Method according to any one of the preceding claims, characterized by the fact that the cancer is anal squamous cell carcinoma, basal cell carcinoma, bladder cancer, bone cancer, intestinal carcinoma, breast cancer, carcinoid, cancer castration-resistant prostate cancer (CRPC), cervical carcinoma, colorectal cancer (CRC), colon cancer, cutaneous squamous cell carcinoma, endometrial cancer, esophageal cancer, gastric carcinoma, gastroesophageal junction cancer, glioblastoma / mixed glioma , glioma, head and neck cancer, hepatocellular carcinoma, hematological malignancy, liver cancer, lung cancer, melanoma, Merkel cell carcinoma, multiple myeloma, nasopharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer , peritoneal carcinoma, undifferentiated pleomorphic sarcoma, prostate cancer, rectal carcinoma, kidney cancer, sarcoma, salivary gland carcinoma, squamous cell carcinoma, stoma cancer go, testicular cancer, thymic carcinoma, thymic epithelial tumor, thymoma, thyroid cancer, urogenital cancer, urothelial cancer, uterine carcinoma or uterine sarcoma.
[116]
116. Method according to any one of the preceding claims, characterized by the fact that the cancer is a High Tumor Mutational Charge Cancer (hTMB).
[117]
117. Method according to claim 115, characterized by the fact that breast cancer is triple negative breast cancer or estrogen receptor positive breast cancer.
[118]
118. Method according to claim 115, characterized by the fact that the hematological malignancy is lymphoma or leukemia.
[119]
119. Method according to claim 118, characterized in that the lymphoma is a B-cell lymphoma, a T-cell lymphoma, a Hodgkin's lymphoma or an EBV lymphoma, a primary mediastinal lymphoma of cells B.
[120]
120. Method according to claim 115, characterized by the fact that cancer is melanoma
[121]
121. Method according to claim 115, characterized by the fact that the carcinoma of the intestine is carcinoma of the small intestine or adenocarcinoma of the small intestine.
[122]
122. Method according to claim 115, characterized by the fact that colon cancer is colon adenocarcinoma.
[123]
123. Method according to claim 115, characterized by the fact that lung cancer is non-small cell lung cancer (NSCLC) or small cell lung cancer.
[124]
124. Method according to claim 123, characterized by the fact that the NSCLC is non-scaly NSCLC or scaly NSCLC.
[125]
125. Method according to claim 115, characterized by the fact that prostate cancer is small cell neuroendocrine prostate cancer.
[126]
126. Method according to claim 115, characterized by the fact that renal cancer is renal cell carcinoma or renal sarcoma.
[127]
127. Method according to claim 115, characterized by the fact that the cancer is non-differentiated pleomorphic sarcoma, adenocarcinoma of the small intestine, cell carcinoma of
Merkel, thymic carcinoma, anal squamous cell carcinoma, cutaneous squamous cell carcinoma or triple negative breast cancer.
[128]
128. Method according to any one of the preceding claims, characterized by the fact that the individual exhibits one or more of the following characteristics: a. Naive to PD-1 / PDL1 inhibitor, b. Naive to the CTLA-4 inhibitor, c. Positive for BRAFV600E mutation, d. Naive to the BRAF inhibitor, e.g. Positive for PDL1, f. Unknown for PDL1 and g. was previously treated with a PD1 / PDL1 inhibitor.
[129]
129. Method according to any one of the previous claims, characterized by the fact that the individual has no additional standard of care available.
[130]
130. Method according to any one of the preceding claims, characterized by the fact that therapy with PD1 / PDL1 inhibitor is not approved for the individual's cancer.
[131]
131. Method according to any one of the preceding claims, characterized in that the individual has been previously treated with a PD-1 / PDL1 inhibitor; where treatment with the PD-1 / PDL1 inhibitor was stopped for reasons other than toxicity, and where the individual is naive to the CTLA-4 inhibitor.
[132]
132. Method according to any one of the preceding claims, characterized by the fact that the individual is naive to immunotherapy.
Part A: Monotherapy
Petition 870190124893, of 11/28/2019, p. 160/330 with AA
Part B1: AA + ipilimumab (3 mg / kg IV) Concomitant programming 1/17
Part B2: AA + ipilimumab (3 mg / kg IV) Phased programming
Part C: AA + vemurafenib (960 mg / kg PO)
Part D: AA monotherapy Dose expansion
Part A: AA monotherapy
Petition 870190124893, of 11/28/2019, p. 161/330 Part A2: Monotherapy with AA Biomarkers and efficacy in PD L1 + tumors
Part B1: 2/17
AA + ipilimumab (3 mg / kg IV) Concomitant programming
Part B2: AA + ipilimumab (3 mg / kg IV) Phased programming
Part C: AA + vemurafenib (960 mg / kg PO)
Part D: AA monotherapy Dose expansion
Petition 870190124893, of 11/28/2019, p. 162/330 Intact Activable Antibody (Blue) Intact Activable Antibody (Blue) 3/17
Activated Antibody Cleaved (Green)
Chemiluminescence Chemiluminescence Molecular Weight, kDa Molecular Weight, kDa
White Intact Cleaved
Chemiluminescence
% Cleaved in Human Plasma at 1: 100
Human Plasma Lung Tumor Lysate
Chemiluminescence Chemiluminescence
AA Tx Intact
AA Tx Intact AA Tx Cleaved
AA Tx Clived
Chemiluminescence Chemiluminescence
Detection of commercial anti-human IgG
Petition 870190124893, of 11/28/2019, p. 167/330 Mouse plasma treated with 10 mg / kg of PL07-2001-C5H9V2
Anti-Human IgG absorbed by Intact Cleaved Cino 8/17
Chemiluminescence
Anti-idiotypic Ab detection
Petition 870190124893, of 11/28/2019, p. 168/330 Mouse plasma treated with 0.1 mg / kg of PL07-2001-C5H9V2
Intact Cleaved Anti-id
AA Tx Intact 9/17
Chemiluminescence AA Tx Cleaved
AA Tx Intact AA Tx Intact
AA Tx Clived
Chemiluminescence Chemiluminescence
AA Tx Intact
AA Tx Intact
AA Tx Clived AA Tx Cleaved
Chemiluminescence Chemiluminescence
Time (day)
Concentration (nM)
Time (day)
Concentration (nM)
Esophageal Thymoma / Thymic Cancer Uterine Pancreatic Colon
Petition 870190124893, of 11/28/2019, p. 173/330 Peritoneal Rectal Salivary Gland Prostate CR 14/17
Change in Baseline Target Injuries (%) Patient Treatment
CR, resistant to castration; ER + BC, estrogen receptor positive breast cancer; HNSCC, squamous cell carcinoma of the head and neck; PD, progressive disease; PR, partial response; RECIST, Response Evaluation Criteria in Solid Tumors; DS, stable disease; TNBC, triple negative breast cancer.
Esophageal Uterine Salivary Gland Prostate CR Pancreatic Thymoma / Peritoneal thymic cancer
Petition 870190124893, of 11/28/2019, p. 174/330 Rectal Colon 15/17
Treatment
Percentage of Change in Baseline Tumor Load (%) Weeks Since Beginning of Treatment
Researcher's Response to Time Point Response
CR, resistant to castration; ER + BC, estrogen receptor positive breast cancer; HNSCC, squamous cell carcinoma of the head and neck; PD, progressive disease; PR, partial response; RECIST, Response Evaluation Criteria in Solid Tumors; DS, stable disease; TNBC, triple negative breast cancer.
Petition 870190124893, of 11/28/2019, p. 175/330 Colon osteosarcoma 16/17
Small Cell Prostate Cancer
Testicular Small Intestine
Change in Baseline Target Injuries (%) SCC Anal
Patient Treatment AA + 3 mg / kg of Ipilimumab AA + 3 mg / kg of Ipilimumab AA + 3 mg / kg of Ipilimumab AA + 3 mg / kg of Ipilimumab
CR, complete response; ER + BC, estrogen receptor positive breast cancer; HNSCC, squamous cell carcinoma of the head and neck; PD, progressive disease; PR, partial response; RECIST, Response Evaluation Criteria in Solid Tumors; SCC, squamous cell carcinoma; SCLC, small cell lung cancer; DS, stable disease; TNBC, triple negative breast cancer.
Colon osteosarcoma
Petition 870190124893, of 11/28/2019, p. 176/330 Small Cell Prostate Cancer
Small Intestine Testicular SCC Anal 17/17
Treatment AA + 3 mg / kg of Ipilimumab AA + 3 mg / kg of Ipilimumab AA + 3 mg / kg of Ipilimumab AA + 3 mg / kg of Ipilimumab
Percentage of Change in Baseline Tumor Load (%) Weeks Since Beginning of Treatment
Researcher's Response to Time Point Response:
CR, complete response; ER + BC, estrogen receptor positive breast cancer; HNSCC, squamous cell carcinoma of the head and neck; PD, progressive disease; PR, partial response; RECIST, Response Evaluation Criteria in Solid Tumors; SCC, squamous cell carcinoma; SCLC, small cell lung cancer; DS, stable disease; TNBC, triple negative breast cancer.
类似技术:
公开号 | 公开日 | 专利标题
US11168144B2|2021-11-09|Activatable anti-PDL1 antibodies, and methods of use thereof
JP6960485B2|2021-11-05|Monoclonal antibody against fibroblast growth factor receptor 2
BR112019014694A2|2020-04-07|anti-cd47 antibodies and their uses
EP1660010B1|2013-07-24|Compositions and methods for restoring sensitivity to treatment with her2 antagonists
EP3180356B1|2019-11-27|Human anti-fgfr4 antibody
CN107708666A|2018-02-16|The conjoint therapy for the treatment of cancer
AU2017204686B2|2020-04-30|Compositions and methods for detecting and treating esophageal cancer
WO2019024911A1|2019-02-07|B7h3 antibody-drug conjugate and medical use thereof
KR20180105642A|2018-09-28|Compositions and methods for the detection and treatment of gastric cancer
CN111212662A|2020-05-29|Combination therapy for the treatment of cancer
TW201902514A|2019-01-16|Use of PD-1 antibody in combination with VEGF ligand or VEGF receptor inhibitor for the preparation of a medicament for treating tumor
US20190002582A1|2019-01-03|Compositions and methods for detecting and treating ovarian cancer
CN110366562A|2019-10-22|Use the method for anti-PD-L1 antibody and anti-androgen therapy cancer
US20180057595A1|2018-03-01|Anti-MET Antibodies and Methods of Use Thereof
AU2017347822A1|2019-05-09|Use of beta-catenin as a biomarker for treating cancers using anti-Dkk-1 antibody
WO2020228604A1|2020-11-19|Anti-trop-2 antibody, antigen binding fragment thereof and medical use thereof
TW201716439A|2017-05-16|HER3 antibodies
KR20200072507A|2020-06-22|Combination products for cancer treatment
JP2021530428A|2021-11-11|Treatment of head and neck cancer
CA3064588A1|2018-12-06|Activatable anti-pdl1 antibodies, and methods of use thereof
JP2021107384A|2021-07-29|Treatment with site specific her2 antibody-drug conjugates
WO2020221198A1|2020-11-05|Bispecific antibody with double her2 sites for tumor immunotherapy
US20200255506A1|2020-08-13|Treatment of ck8 positive cancers in relation with k-ras gene status
BR112019019505A2|2020-05-26|ANTI-DR5 ANTIBODY AND THE USE OF THE SAME
JP2019505516A|2019-02-28|Method of treating acne using an interleukin-17 | antagonist
同族专利:
公开号 | 公开日
IL270921D0|2020-01-30|
WO2018222949A1|2018-12-06|
US11168144B2|2021-11-09|
CN110914302A|2020-03-24|
US20190016814A1|2019-01-17|
JP2020522486A|2020-07-30|
AU2018278327A1|2019-12-12|
EP3630838A1|2020-04-08|
KR20200016899A|2020-02-17|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题

US3773919A|1969-10-23|1973-11-20|Du Pont|Polylactide-drug mixtures|
US4485045A|1981-07-06|1984-11-27|Research Corporation|Synthetic phosphatidyl cholines useful in forming liposomes|
US4522811A|1982-07-08|1985-06-11|Syntex Inc.|Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides|
US4544545A|1983-06-20|1985-10-01|Trustees University Of Massachusetts|Liposomes containing modified cholesterol for organ targeting|
DE3788058T2|1986-08-28|1994-04-21|Teijin Ltd|CELL TOXIC ANTIBODY COMPLEX AND METHOD FOR THE PRODUCTION THEREOF.|
US5013556A|1989-10-20|1991-05-07|Liposome Technology, Inc.|Liposomes with enhanced circulation time|
US5151510A|1990-04-20|1992-09-29|Applied Biosystems, Inc.|Method of synethesizing sulfurized oligonucleotide analogs|
DE69329503T2|1992-11-13|2001-05-03|Idec Pharma Corp|Therapeutic use of chimeric and labeled antibodies directed against a differentiation antigen, the expression of which is restricted to human B lymphocyte, for the treatment of B cell lymphoma|
PL354286A1|1999-08-23|2003-12-29|Dana-Farber Cancer Institutedana-Farber Cancer Institute|Pd-1, a receptor for b7-4, and uses therefor|
AT353365T|1999-08-23|2007-02-15|Dana Farber Cancer Inst Inc|NEW B7-4 MOLECULES AND THEIR USES|
CA2392477A1|1999-11-30|2001-06-07|Mayo Foundation For Medical Education And Research|B7-h1, a novel immunoregulatory molecule|
US6803192B1|1999-11-30|2004-10-12|Mayo Foundation For Medical Education And Research|B7-H1, a novel immunoregulatory molecule|
WO2001091798A2|2000-06-01|2001-12-06|Universite Catholique De Louvain|Tumor activated prodrug compounds|
AU7309601A|2000-06-28|2002-01-08|Genetics Inst|Pd-l2 molecules: novel pd-1 ligands and uses therefor|
AT513563T|2000-10-09|2011-07-15|Isis Innovation|THERAPEUTIC AND TOLERANT INDUCING ANTIBODIES|
US7465790B2|2000-10-09|2008-12-16|Isis Innovation, Inc.|Therapeutic antibodies|
WO2002079499A1|2001-04-02|2002-10-10|Wyeth|Pd-1, a receptor for b7-4, and uses therefor|
WO2002086083A2|2001-04-20|2002-10-31|Mayo Foundation For Medical Education And Research|Methods of enhancing cell responsiveness|
JP4488740B2|2001-11-13|2010-06-23|ダナ−ファーバーキャンサーインスティテュート,インコーポレイテッド|Agents that modulate immune cell activation and methods of use thereof|
EP2206517A1|2002-07-03|2010-07-14|Ono Pharmaceutical CO., LTD.|Immunopotentiating compositions comprising anti-PD-L1 antibodies|
US20040109855A1|2002-07-23|2004-06-10|Herman Waldmann|Therapeutic antibodies with reduced side effect|
US8809504B2|2002-09-03|2014-08-19|Vit Lauermann|Inhibitor which is deactivatable by a reagent produced by a target cell|
US7432351B1|2002-10-04|2008-10-07|Mayo Foundation For Medical Education And Research|B7-H1 variants|
US7449300B2|2002-11-21|2008-11-11|Mayo Foundation For Medical Education And Research|Detection of antibodies specific for B7-H1 in subjects with diseases or pathological conditions mediated by activated T cells|
PL1810026T3|2004-10-06|2018-08-31|Mayo Foundation For Medical Education And Research|B7-h1 and pd-1 in treatment of renal cell carcinoma|
EP3530736A3|2005-05-09|2019-11-06|ONO Pharmaceutical Co., Ltd.|Human monoclonal antibodies to programmed death 1 and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics|
MX351401B|2005-06-08|2017-10-13|Dana Farber Cancer Inst Inc|Methods and compositions for the treatment of persistent infections and cancer by inhibiting the programmed cell death 1 pathway.|
CA2612241C|2005-07-01|2018-11-06|Medarex, Inc.|Human monoclonal antibodies to programmed death ligand 1 |
EP1919931A4|2005-08-31|2010-01-20|Univ California|Cellular libraries of peptide sequences and methods of using the same|
WO2007105027A1|2006-03-10|2007-09-20|Diatos|Anticancer drugs conjugated to antibody via an enzyme cleavable linker|
MX2009006786A|2006-12-27|2009-10-19|Univ Emory|Compositions and methods for the treatment of infections and tumors.|
JP5481199B2|2006-12-27|2014-04-23|ザジョンズホプキンスユニバーシティー|Compositions and methods for treating inflammation and autoimmune diseases|
WO2008156712A1|2007-06-18|2008-12-24|N. V. Organon|Antibodies to human programmed death receptor pd-1|
US20090304719A1|2007-08-22|2009-12-10|Patrick Daugherty|Activatable binding polypeptides and methods of identification and use thereof|
US8062852B2|2007-10-01|2011-11-22|The Children's Hospital And Regional Medical Center|Detection and treatment of autoimmune disorders|
US8552154B2|2008-09-26|2013-10-08|Emory University|Anti-PD-L1 antibodies and uses therefor|
KR101050829B1|2008-10-02|2011-07-20|서울대학교산학협력단|Anticancer agents comprising an anti-PD-1 antibody or an anti-PD-L1 antibody|
JP5520961B2|2008-11-28|2014-06-11|エモリーユニバーシティ|Methods for treating infections and tumors|
US20100189727A1|2008-12-08|2010-07-29|Tegopharm Corporation|Masking Ligands For Reversible Inhibition Of Multivalent Compounds|
PE20141722A1|2008-12-09|2014-12-02|Genentech Inc|ANTI-PD-L1 ANTIBODIES AND ITS USE TO IMPROVE T-CELL FUNCTION|
RU2017136814A3|2009-01-12|2021-10-21|
US8741295B2|2009-02-09|2014-06-03|Universite De La Mediterranee|PD-1 antibodies and PD-L1 antibodies and uses thereof|
RS56469B1|2009-11-24|2018-01-31|Medimmune Ltd|Targeted binding agents against b7-h1|
PL2542590T5|2010-03-05|2020-08-10|The Johns Hopkins University|Compositions and methods for targeted immunomodulatory antibodies and fusion proteins|
EP2555778A4|2010-04-06|2014-05-21|Alnylam Pharmaceuticals Inc|Compositions and methods for inhibiting expression of cd274/pd-l1 gene|
US20110280877A1|2010-05-11|2011-11-17|Koji Tamada|Inhibition of B7-H1/CD80 interaction and uses thereof|
US8962804B2|2010-10-08|2015-02-24|City Of Hope|Meditopes and meditope-binding antibodies and uses thereof|
TWI743461B|2011-03-28|2021-10-21|法商賽諾菲公司|Dual variable region antibody-like binding proteins having cross-over binding region orientation|
AU2012245477C1|2011-04-20|2017-06-15|Medimmune, Llc|Antibodies and other molecules that bind B7-H1 and PD-1|
TWI654994B|2011-08-01|2019-04-01|建南德克公司|Methods of treating cancer using pd-1 axis binding antagonists and mek inhibitors|
WO2013043569A1|2011-09-20|2013-03-28|Vical Incorporated|Synergistic anti-tumor efficacy using alloantigen combination immunotherapy|
US8956619B2|2011-10-25|2015-02-17|University Of Maryland, Baltimore County|Soluble CD80 as a therapeutic to reverse immune supression in cancer patients|
KR101981873B1|2011-11-28|2019-05-23|메르크 파텐트 게엠베하|Anti-pd-l1 antibodies and uses thereof|
GB201203442D0|2012-02-28|2012-04-11|Univ Birmingham|Immunotherapeutic molecules and uses|
RU2713121C2|2012-04-27|2020-02-03|Сайтомкс Терапьютикс, Инк.|Activated antibodies which bind to epidermal growth factor receptor, and methods for use thereof|
JP6448533B2|2012-05-15|2019-01-09|ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company|Cancer immunotherapy by disrupting PD-1 / PD-L1 signaling|
KR102163408B1|2012-05-31|2020-10-08|소렌토 쎄라퓨틱스, 인코포레이티드|Antigen binding proteins that bind pd-l1|
WO2013192546A1|2012-06-22|2013-12-27|Cytomx Therapeutics, Inc.|Activatable antibodies having non-binding steric moieties and mehtods of using the same|
WO2014022758A1|2012-08-03|2014-02-06|Dana-Farber Cancer Institute, Inc.|Single agent anti-pd-l1 and pd-l2 dual binding antibodies and methods of use|
EP2882844B1|2012-08-10|2018-10-03|Cytomx Therapeutics Inc.|Protease-resistant systems for polypeptide display and methods of making and using thereof|
CN107892719B|2012-10-04|2022-01-14|达纳-法伯癌症研究所公司|Human monoclonal anti-PD-L1 antibodies and methods of use|
CA2889182A1|2012-10-26|2014-05-01|The University Of Chicago|Synergistic combination of immunologic inhibitors for the treatment of cancer|
WO2014074852A1|2012-11-09|2014-05-15|President And Fellows Of Harvard College|Compositions and methods for modulating an immune response|
KR102291355B1|2012-11-30|2021-08-19|에프. 호프만-라 로슈 아게|Identification of patients in need of pd-l1 inhibitor cotherapy|
EP2934576A1|2012-12-19|2015-10-28|The Johns Hopkins University|Anti-human b7-h4 antibodies and their uses|
AR093984A1|2012-12-21|2015-07-01|Merck Sharp & Dohme|ANTIBODIES THAT JOIN LEGEND 1 OF SCHEDULED DEATH HUMAN|
CN105008918A|2013-01-04|2015-10-28|西托姆克斯治疗公司|Compositions and methods for detecting protease activity in biological systems|
US20150344577A1|2013-01-11|2015-12-03|Dingfu Biotarget Co., Ltd|Agents for treating tumors, use and method thereof|
EP2948475A2|2013-01-23|2015-12-02|AbbVie Inc.|Methods and compositions for modulating an immune response|
EP2953645A4|2013-02-07|2016-12-28|Immunomedics Inc|Pro-drug form of the highly potent 2-pyrrolinodoxorubicin conjugated to antibodies for targeted therapy of cancer|
WO2014165082A2|2013-03-13|2014-10-09|Medimmune, Llc|Antibodies and methods of detection|
US20150071910A1|2013-03-15|2015-03-12|Genentech, Inc.|Biomarkers and methods of treating pd-1 and pd-l1 related conditions|
AU2014235453A1|2013-03-15|2015-10-08|Genentech, Inc.|Biomarkers and methods of treating PD-1 and PD-L1 related conditions|
WO2014194302A2|2013-05-31|2014-12-04|Sorrento Therapeutics, Inc.|Antigen binding proteins that bind pd-1|
RU2015154275A3|2013-06-03|2018-05-11|
WO2014197369A1|2013-06-06|2014-12-11|Dana-Farber Cancer Institute, Inc.|Compositions and methods for identification, assessment prevention, and treatment of cancer using pd-l1 isoforms|
CN104250302B|2013-06-26|2017-11-14|上海君实生物医药科技股份有限公司|The anti-antibody of PD 1 and its application|
FR3008408B1|2013-07-11|2018-03-09|Mc Saf|NOVEL ANTIBODY-MEDICAMENT CONJUGATES AND THEIR USE IN THERAPY|
RU2020123264A|2013-07-25|2020-10-14|Сайтомкс Терапьютикс, Инк.|MULTI-SPECIFIC ANTIBODIES, MULTI-SPECIFIC ACTIVATED ANTIBODIES AND METHODS OF THEIR APPLICATION|
WO2015035112A1|2013-09-05|2015-03-12|The Johns Hopkins University|Cancer therapy via a combination of epigenetic modulation and immune modulation|
WO2015038538A1|2013-09-10|2015-03-19|Medimmune, Llc|Compositions and methods for treating sepsis|
EP3656398A1|2013-09-11|2020-05-27|MedImmune Limited|Anti-b7-h1 antibodies for treating tumors|
AR097584A1|2013-09-12|2016-03-23|F Hoffmann-La Roche Ag|ANTIBODY COMBINATION THERAPY AGAINST HUMAN CSF-1R AND ANTIBODIES AGAINST HUMAN PD-L1|
WO2015035606A1|2013-09-13|2015-03-19|Beigene, Ltd.|Anti-pd1 antibodies and their use as therapeutics and diagnostics|
US10570204B2|2013-09-26|2020-02-25|The Medical College Of Wisconsin, Inc.|Methods for treating hematologic cancers|
UA122478C2|2013-09-27|2020-11-25|Дженентек, Інк.|Anti-pdl1 antibody formulations|
EP3052131B1|2013-10-01|2018-12-05|Mayo Foundation for Medical Education and Research|Methods for treating cancer in patients with elevated levels of bim|
EP3060581A4|2013-10-25|2017-06-07|Dana-Farber Cancer Institute, Inc.|Anti-pd-l1 monoclonal antibodies and fragments thereof|
CA2929407A1|2013-11-05|2015-05-14|Cognate Bioservices, Inc.|Combinations of checkpoint inhibitors and therapeutics to treat cancer|
US20150165021A1|2013-11-05|2015-06-18|Nkt Therapeutics Inc.|Combination therapy|
KR102362803B1|2013-11-25|2022-02-14|페임웨이브 리미티드|Compositions comprising anti-ceacam1 and anti-pd antibodies for cancer therapy|
WO2015081158A1|2013-11-26|2015-06-04|Bristol-Myers Squibb Company|Method of treating hiv by disrupting pd-1/pd-l1 signaling|
EP3079772B1|2013-12-10|2020-02-05|Merck Sharp & Dohme Corp.|Immunohistochemical proximity assay for pd-1 positive cells and pd-ligand positive cells in tumor tissue|
EP3079699A4|2013-12-11|2017-07-19|Glaxosmithkline LLC|Treating cancer with a combination of a pd-1 antagonist and a vegfr inhibitor|
WO2016071701A1|2014-11-07|2016-05-12|Kymab Limited|Treatment of disease using ligand binding to targets of interest|
HUE047699T2|2013-12-17|2020-05-28|Hoffmann La Roche|Methods of treating cancers using pd-1 axis binding antagonists and taxanes|
SG11201604875PA|2013-12-17|2016-07-28|Genentech Inc|Methods of treating cancer using pd-1 axis binding antagonists and an anti-cd20 antibody|
WO2015092393A2|2013-12-17|2015-06-25|Kymab Limited|Human targets|
EP3083671B1|2013-12-20|2020-09-30|Fred Hutchinson Cancer Research Center|Tagged chimeric effector molecules and receptors thereof|
JP6706578B2|2013-12-30|2020-06-10|エピムアブ バイオセラピューティクス インコーポレイテッド|Tandem Fab immunoglobulins and uses thereof|
WO2015103602A1|2014-01-06|2015-07-09|The Trustees Of The University Of Pennsylvania|Pd1 and pdl1 antibodies and vaccine combinations and use of same for immunotherapy|
WO2015104406A2|2014-01-13|2015-07-16|Pieris Ag|Multi-specific polypeptide useful for localized tumor immunomodulation|
WO2015108907A2|2014-01-14|2015-07-23|Dana-Farber Cancer Institute, Inc.|Compositions and methods for identification, assessment, prevention, and treatment of melanoma using pd-l1 isoforms|
US10407502B2|2014-01-15|2019-09-10|Kadmon Corporation, Llc|Immunomodulatory agents|
EP3096782A4|2014-01-21|2017-07-26|Medlmmune, LLC|Compositions and methods for modulating and redirecting immune responses|
TWI680138B|2014-01-23|2019-12-21|美商再生元醫藥公司|Human antibodies to pd-l1|
RU2752424C2|2014-02-10|2021-07-28|Мерк Патент Гмбх|Targeted tgf beta inhibition|
US10519237B2|2014-03-12|2019-12-31|Yeda Research And Development Co. Ltd|Reducing systemic regulatory T cell levels or activity for treatment of disease and injury of the CNS|
US9987258B2|2014-04-06|2018-06-05|H. Lee Moffitt Cancer Center And Research Institute, Inc.|Histone deacetylase as a modulator of PDL1 expression and activity|
TW201623333A|2014-05-13|2016-07-01|Chugai Pharmaceutical Co Ltd|T cell-redirected antigen-binding molecule for cells having immunosuppression function|
WO2016030455A1|2014-08-28|2016-03-03|Medimmune Limited|Anti-b7-h1 and anti-ctla-4 antibodies for treating non-small lung cancer|
SG10201810108PA|2014-05-13|2018-12-28|Medimmune Ltd|Anti-b7-h1 and anti-ctla-4 antibodies for treating non-small cell lung cancer|
WO2015179654A1|2014-05-22|2015-11-26|Mayo Foundation For Medical Education And Research|Distinguishing antagonistic and agonistic anti b7-h1 antibodies|
KR20150135148A|2014-05-23|2015-12-02|주식회사 제넥신|PD-L1 Fused protein and use thereof|
JP6666905B2|2014-05-29|2020-03-18|スプリング バイオサイエンス コーポレーション|PD-L1 antibody and use thereof|
JP2017517525A|2014-05-29|2017-06-29|メディミューン リミテッド|PDL-1 and PD-1 antagonists for the treatment of HPV negative cancer|
CA2947687C|2014-05-30|2021-07-06|Ventana Medical Systems, Inc.|Multiplex assay for improved scoring of tumor tissues stained for pd-l1|
WO2015195163A1|2014-06-20|2015-12-23|R-Pharm Overseas, Inc.|Pd-l1 antagonist fully human antibody|
US10449227B2|2014-06-27|2019-10-22|H. Lee Moffitt Cancer Center And Research Institute, Inc.|Conjugates for immunotherapy|
EP3160505A4|2014-07-03|2018-01-24|BeiGene, Ltd.|Anti-pd-l1 antibodies and their use as therapeutics and diagnostics|
CN105233291A|2014-07-09|2016-01-13|博笛生物科技有限公司|Combined therapy composition and combined therapy method for treating cancers|
JP6279733B2|2014-07-09|2018-02-14|日本全薬工業株式会社|Anti-canine PD-1 antibody or anti-canine PD-L1 antibody|
TWI727427B|2014-07-09|2021-05-11|英屬開曼群島商博笛生物科技有限公司|Anti-pd-1 combinations for treating tumors|
EP3309174A1|2014-07-11|2018-04-18|Genentech, Inc.|Anti-pd-l1 antibodies and diagnostic uses thereof|
US8992927B1|2014-07-15|2015-03-31|Kymab Limited|Targeting human NAV1.7 variants for treatment of pain|
US9139648B1|2014-07-15|2015-09-22|Kymab Limited|Precision medicine by targeting human NAV1.9 variants for treatment of pain|
US9067998B1|2014-07-15|2015-06-30|Kymab Limited|Targeting PD-1 variants for treatment of cancer|
US9045545B1|2014-07-15|2015-06-02|Kymab Limited|Precision medicine by targeting PD-L1 variants for treatment of cancer|
US9914769B2|2014-07-15|2018-03-13|Kymab Limited|Precision medicine for cholesterol treatment|
US10765710B2|2014-07-16|2020-09-08|Institut Gustave-Roussy|Combination of oncolytic virus with immune checkpoint modulators|
EP3193921A4|2014-07-18|2018-04-25|Advaxis, Inc.|Combination of a pd-1 antagonist and a listeria-based vaccine for treating prostate cancer|
US10435470B2|2014-08-05|2019-10-08|Cb Therapeutics, Inc.|Anti-PD-L1 antibodies|
JP6245622B2|2014-08-07|2017-12-13|学校法人兵庫医科大学|Cancer therapeutic agent using IL-18 and molecular target antibody in combination|
WO2016128912A1|2015-02-12|2016-08-18|Acerta Pharma B.V.|Therapeutic combinations of a btk inhibitor, a pi3k inhibitor, a jak-2 inhibitor, a pd-1 inhibitor, and/or a pd-l1 inhibitor|
MX2017001976A|2014-08-14|2017-08-02|F Hoffmann-La Roche Ag|Combination therapy of antibodies activating human cd40 and antibodies against human pd-l1.|
RU2710354C2|2014-08-29|2019-12-25|Ф.Хоффманн-Ля Рош Аг|Combined therapy based on tumour-targeted immunocytokines containing variant il-2, and antibodies to human pd-l1|
CN112546238A|2014-09-01|2021-03-26|博笛生物科技有限公司|anti-PD-L1 conjugates for the treatment of tumors|
US20160067336A1|2014-09-08|2016-03-10|Celgene Corporation|Methods for treating a disease or disorder using oral formulations of cytidine analogs in combination with an anti-pd1 or anti-pdl1 monoclonal antibody|
US10550194B2|2014-09-30|2020-02-04|Intervet Inc.|PD-L1 antibodies binding canine PD-L1|
CR20170143A|2014-10-14|2017-06-19|Dana-Farber Cancer Inst Inc|ANTIBODY MOLECULES THAT JOIN PD-L1 AND USES OF THE SAME|
WO2016059602A2|2014-10-16|2016-04-21|Glaxo Group Limited|Methods of treating cancer and related compositions|
JP2017533207A|2014-10-23|2017-11-09|ファイヴ プライム セラピューティクス インク|SLAMF1 antagonist and use thereof|
JP6687612B2|2014-10-24|2020-04-22|アストラゼネカ アクチボラグ|combination|
BR112017008914A2|2014-10-29|2018-01-16|Squibb Bristol Myers Co|method for treating cancer, makeup and use of makeup|
PT3212233T|2014-10-31|2020-07-16|Oncomed Pharm Inc|Combination therapy for treatment of disease|
WO2016075174A1|2014-11-11|2016-05-19|Medimmune Limited|Therapeutic combinations for treating neoplasia|
WO2016079049A1|2014-11-17|2016-05-26|Medimmune Limited|Therapeutic combinations and methods for treating neoplasia|
CN105777906B|2014-12-19|2019-04-23|苏州丁孚靶点生物技术有限公司|Anti- PD-L1 human antibody and its application|
US10849862B2|2014-12-26|2020-12-01|Theravalues Corporation|Formulation of curcumin and anti-PD-1 antibody|
CA2972757A1|2014-12-31|2016-07-07|Arthur M. Krieg|Combination tumor immunotherapy|
EP3286213B1|2015-04-20|2021-08-04|Etubics Corporation|Methods and compositions for combination immunotherapy|
GB201500319D0|2015-01-09|2015-02-25|Agency Science Tech & Res|Anti-PD-L1 antibodies|
AU2016206707A1|2015-01-14|2017-08-10|Compass Therapeutics Llc|Multispecific immunomodulatory antigen-binding constructs|
RU2696378C2|2015-01-16|2019-08-01|Академиа Синика|Molecular structures with targeting and effector elements|
EP3250229A4|2015-01-29|2018-08-01|The Trustees of the University of Pennsylvania|Checkpoint inhibitor and vaccine combinations and use of same for immunotherapy|
DK3254110T3|2015-02-03|2020-05-18|Ventana Med Syst Inc|Histochemical test to assess programmed death ligand 1 expression |
MA41460A|2015-02-03|2017-12-12|Oncomed Pharm Inc|TNFRSF LIAISON AGENTS AND THEIR USES|
RU2714233C2|2015-02-26|2020-02-13|Мерк Патент Гмбх|Pd-1/pd-l1 inhibitors for treating cancer|
CA2977532A1|2015-03-06|2016-09-15|Dana-Farber Cancer Institute, Inc.|Pd-l2 biomarkers predictive of pd-1 pathway inhibitor responses in esophagogastric cancers|
MA42971A|2015-03-13|2018-08-15|Cytomx Therapeutics Inc|ANTI-PDL1 ANTIBODIES, ACTIVABLE ANTI-PLD1 ANTIBODIES, AND METHODS OF USE THEREOF|
EP3067062A1|2015-03-13|2016-09-14|Ipsen Pharma S.A.S.|Combination of tasquinimod or a pharmaceutically acceptable salt thereof and a pd1 and/or pdl1 inhibitor, for use as a medicament|
EP3070102A1|2015-03-18|2016-09-21|F. Hoffmann-La Roche AG|Combination therapy of antibodies humancd40 activating antibodies and anti human pld-1 antibodies|
WO2016154412A2|2015-03-26|2016-09-29|The Trustees Of The University Of Pennsylvania|Combination of a pd-1 antagonist and a listeria based vaccine for treating pancreatic cancer|
US20180085398A1|2015-03-30|2018-03-29|Dana-Farber Cancer Institute, Inc.|Compositions and methods of treating cancer|
US11026921B2|2015-03-30|2021-06-08|Dana-Farber Cancer Institute, Inc.|Compositions and methods of treating cancer|
US20180078650A1|2015-03-30|2018-03-22|Dana-Farber Cancer Institute, Inc.|Compositions and methods of treating acute myeloid leukemia|
EP3277290A1|2015-03-30|2018-02-07|Dana-Farber Cancer Institute, Inc.|Compositions and methods of treating renal cell cancer|
WO2016160792A1|2015-03-30|2016-10-06|Stcube & Co., Inc.|Antibodies specific to glycosylated pd-l1 and methods of use thereof|
US20180071340A1|2015-03-30|2018-03-15|Dana-Farber Cancer Institute, Inc.|Compositions and methods of treating multiple myeloma|
WO2016156501A1|2015-04-01|2016-10-06|Medimmune Limited|Combined anti-pld-1 and anti-ctla-4 antibodies for treating non-small lung cancer|
JP2018515450A|2015-04-23|2018-06-14|エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト|Combination therapy of antibodies that bind to angiopoietin 2 with antibodies that bind to programmed death ligand 1|
EP3290517B1|2015-04-30|2020-01-01|Kyoto University|Method for predicting therapeutic effect of pd-1/pd-l1 inhibitor using abnormality in pd-l1 as index|
JP2018520996A|2015-05-12|2018-08-02|ジェネンテック, インコーポレイテッド|Methods of treatment and diagnosis for cancer|
TW201705955A|2015-05-14|2017-02-16|輝瑞大藥廠|Combination therapies comprising a pyrrolidine-2,5-dione IDO1 inhibitor and an anti-PD1/anti-PD-L1 antibody|
US20160347848A1|2015-05-28|2016-12-01|Medimmune Limited|Therapeutic combinations and methods for treating neoplasia|
US11078278B2|2015-05-29|2021-08-03|Bristol-Myers Squibb Company|Treatment of renal cell carcinoma|
EP3763827A1|2015-05-29|2021-01-13|F. Hoffmann-La Roche AG|Pd-l1 promoter methylation in cancer|
CN106243225B|2015-06-11|2021-01-19|智翔(上海)医药科技有限公司|Novel anti-PD-L1 antibodies|
US10696745B2|2015-06-11|2020-06-30|Wuxi Biologics Co. Ltd.|Anti-PD-L1 antibodies|
US10869924B2|2015-06-16|2020-12-22|Merck Patent Gmbh|PD-L1 antagonist combination treatments|
BR112017028519A2|2015-06-29|2018-08-28|Verastem Inc|therapeutic compositions, combinations and methods of use|
CN107849131A|2015-07-06|2018-03-27|瑞泽恩制药公司|Polyspecific antigen binding molecules and application thereof|
KR20180027576A|2015-07-14|2018-03-14|브리스톨-마이어스 스큅 컴퍼니|How to treat cancer using immune checkpoint inhibitors|
AU2016297583A1|2015-07-22|2018-02-01|Hznp Limited|Combination of immunomodulatory agent with PD-1-or PD-L1 checkpoint inhibitors in the treatment of cancer|
CN106397592A|2015-07-31|2017-02-15|苏州康宁杰瑞生物科技有限公司|Single-domain antibody directed at programmed death ligand and derived protein thereof|
WO2017020291A1|2015-08-06|2017-02-09|Wuxi Biologics Co. Ltd.|Novel anti-pd-l1 antibodies|
EP3331919A1|2015-08-07|2018-06-13|GlaxoSmithKline Intellectual Property Development Limited|Combination therapy comprising anti ctla-4 antibodies|
AR105654A1|2015-08-24|2017-10-25|Lilly Co Eli|ANTIBODIES PD-L1 |
WO2017059387A1|2015-09-30|2017-04-06|Igm Biosciences, Inc.|Binding molecules with modified j-chain|
US20180296561A1|2015-10-07|2018-10-18|The University Of North Carolina At Chapel Hill|The Methods For Treatment Of Tumors|
CN108697788A|2015-10-15|2018-10-23|杜克大学|Combined therapy|
WO2017072273A1|2015-10-30|2017-05-04|Affibody Ab|New polypeptide|
MY187827A|2015-10-30|2021-10-26|Affibody Ab|New polypeptide having affinity to pd-l1|
MX2018005720A|2015-11-17|2018-11-09|Suzhou Suncadia Biopharmaceuticals Co Ltd|Pd-l1 antibody, antigen fragment binding thereof and pharmaceutical use thereof.|
WO2017087547A1|2015-11-17|2017-05-26|Oncomed Pharmaceuticals, Inc.|Pd-l1-binding agents and uses thereof|
EP3377107B1|2015-11-19|2020-08-12|H. Hoffnabb-La Roche Ag|Methods of treating cancer using b-raf inhibitors and immune checkpoint inhibitors|
HUE050811T2|2015-12-07|2021-01-28|Merck Patent Gmbh|Aqueous pharmaceutical formulation comprising anti-pd-1 antibody avelumab|
CA3007613A1|2015-12-07|2017-06-15|Kyoto University|Combination therapy based on pd-1 signal inhibitors|
CN105461808B|2015-12-24|2019-03-19|长春金赛药业股份有限公司|Monoclonal antibody and its application|
US20190022129A1|2016-01-08|2019-01-24|Biothera, Inc.|Beta-glucan immunotherapies affecting the immune microenvironment|
WO2017123673A2|2016-01-11|2017-07-20|Inhibrx Lp|Multivalent and multispecific ox40-binding fusion proteins|
EA201891732A1|2016-02-02|2019-02-28|Кадмон Корпорейшн, Ллк|BISPECIFIC BINDING PROTEINS FOR PD-L1 AND KDR|
WO2017136820A2|2016-02-06|2017-08-10|Epimab Biotherapeutics, Inc.|Fabs-in-tandem immunoglobulin and uses thereof|
CA2987118C|2016-03-04|2020-03-24|Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd.|A pdl-1 antibody, pharmaceutical composition thereof and use thereof|
WO2017161154A2|2016-03-16|2017-09-21|H. Lee Moffitt Cancer Center & Research Institute, Inc.|Pd1 and pdl-1 expression during progression from myelodysplastic syndrome to acute myelogenous leukemia|
WO2017161976A1|2016-03-23|2017-09-28|Mabspace Biosciences Co., Ltd|Novel anti-pd-l1 antibodies|
WO2017172518A1|2016-03-29|2017-10-05|Stcube, Inc.|Dual function antibodies specific to glycosylated pd-l1 and methods of use thereof|
TW201801724A|2016-04-07|2018-01-16|卡默森屈有限公司|Reducing tumor burden by administering CCR1 antagonists in combination with PD-1 inhibitors or PD-L1 inhibitors|
JP2019513725A|2016-04-07|2019-05-30|キャンサー・リサーチ・テクノロジー・リミテッドCancer Research Technology Limited|Anti-CD25 FC.GAMMA. Receptor Bispecific Antibodies for Depletion of Tumor-Specific Cells|
WO2017177179A1|2016-04-08|2017-10-12|Gilead Sciences, Inc.|Compositions and methods for treating cancer, inflammatory diseases and autoimmune diseases|
KR20190003958A|2016-04-15|2019-01-10|제넨테크, 인크.|Treatment and monitoring of cancer|
US10421811B2|2016-04-25|2019-09-24|Medimmune, Llc|Compositions comprising coformulation of anti-PD-L1 and anti-CTLA-4 antibodies|
CA3023225A1|2016-05-04|2017-11-09|H. Lee Moffitt Cancer Center And Research Institute, Inc.|A delta-opioid receptor targeted agent for molecular imaging and immunotherapy of cancer|
CN109789225A|2016-05-05|2019-05-21|宾夕法尼亚大学理事会|Target the DNA monoclonal antibody of checkpoint molecule|
AR108377A1|2016-05-06|2018-08-15|Medimmune Llc|BISPECIFIC UNION PROTEINS AND ITS USES|
JP6979971B2|2016-05-09|2021-12-15|アイジーエム バイオサイエンシズ インコーポレイテッド|Anti-PD-L1 antibody|
TW201808286A|2016-05-11|2018-03-16|滬亞生物國際有限公司|Combination therapies of HDAC inhibitors and PD-L1 inhibitors|
EP3458481A4|2016-05-18|2020-07-29|Mayo Foundation for Medical Education and Research|Targeting pd-l1 on tumor cells|
SG11201809669RA|2016-05-20|2018-11-29|Biohaven Pharm Holding Co Ltd|Use of glutamate modulating agents with immunotherapies to treat cancer|
CA3025391A1|2016-05-26|2017-11-30|Merck Patent Gmbh|Pd-1 / pd-l1 inhibitors for cancer treatment|
WO2017205801A1|2016-05-27|2017-11-30|Takeda Pharmaceutical Company Limited|Combination of immunotherapy agents and spleen tyrosine kinase inhibitors|
CN107459578B|2016-05-31|2021-11-26|泰州迈博太科药业有限公司|Difunctional fusion protein targeting CD47 and PD-L1|
WO2017210335A1|2016-06-01|2017-12-07|Bristol-Myers Squibb Company|Imaging methods using 18f-radiolabeled biologics|
US20190184042A1|2016-06-01|2019-06-20|Bristol-Myers Squibb Company|Pet imaging with pd-l1 binding polypeptides|
US20190106494A1|2016-06-13|2019-04-11|Askgene Pharma Inc.|PD-L1 Specific Monoclonal Antibodies for Disease Treatment and Diagnosis|
WO2017218435A1|2016-06-13|2017-12-21|Askgene Pharma Inc.|PD-L1 Specific Monoclonal Antibodies for Disease Treatment and Diagnosis|
KR101996019B1|2016-06-13|2019-07-03|아이-맵|Anti-PD-L1 antibodies and their use|
WO2018115859A1|2016-12-20|2018-06-28|Kymab Limited|Multispecific antibody with combination therapy for immuno-oncology|
EP3471754A1|2016-06-20|2019-04-24|Kymab Limited|Anti-pd-l1 antibodies|
US9567399B1|2016-06-20|2017-02-14|Kymab Limited|Antibodies and immunocytokines|
SG10202100100UA|2016-06-29|2021-02-25|Checkpoint Therapeutics Inc|Pd-l1-specific antibodies and methods of using the same|
WO2018009894A1|2016-07-07|2018-01-11|Iovance Biotherapeutics, Inc.|Programmed death 1 ligand 1 binding proteins and methods of use thereof|
CN106243223B|2016-07-28|2019-03-05|北京百特美博生物科技有限公司|Anti-human PDL1 antibody and application thereof|
KR102048477B1|2016-08-05|2019-11-26|주식회사 와이바이오로직스|Antibodies Against Programmed death-ligand 1 and Uses Thereof|
CN106977602B|2016-08-23|2018-09-25|中山康方生物医药有限公司|A kind of anti-PD1 monoclonal antibodies, its medical composition and its use|
EP3515456A4|2016-09-20|2020-06-17|Medimmune, LLC|Compositions and methods for characterizing solid tumors responsiveness to anti-pd-l1 antibody monotherapy|
CA3037230A1|2016-09-20|2018-03-29|Merck Patent Gmbh|Diagnostic anti-pd-l1 antibody and use thereof|
BR112019006504A2|2016-10-06|2019-06-25|Merck Patent Gmbh|Avelumab Dosage Regimen For Cancer Treatment|
CN106478819B|2016-10-27|2018-12-07|常州费洛斯药业科技有限公司|A kind of monoclonal antibody or antibody fragment for PD-L1|
KR20190088986A|2016-10-30|2019-07-29|상하이 헨리우스 바이오테크, 인크.|Anti-PD-L1 antibodies and variants|
US20190256603A1|2016-11-11|2019-08-22|Medimmune, Llc|Anti-pd-l1 and anti-ctla-4 antibodies for treating non-small cell lung cancer|
MX2019006448A|2016-12-01|2020-02-05|Regeneron Pharma|Radiolabeled anti-pd-l1 antibodies for immuno-pet imaging.|
AU2017368923A1|2016-12-01|2019-06-13|Glaxosmithkline Intellectual Property Development Limited|Combination therapy|
CN110023338A|2016-12-08|2019-07-16|伊莱利利公司|For the anti-TIM-3 antibody with anti-PD-L1 antibody combination|
TW201828993A|2016-12-12|2018-08-16|日商第一三共股份有限公司|Combination of antibody-drug conjugate and immune checkpoint inhibitor|
KR20190095921A|2016-12-12|2019-08-16|제넨테크, 인크.|How to Treat Cancer Using Anti-PD-L1 Antibody and Antiandrogen|
CN106978400A|2016-12-13|2017-07-25|无锡傲锐东源生物科技有限公司|Anti- PD L1 protein monoclonal antibodies and application thereof|
JP2020502231A|2016-12-22|2020-01-23|エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト|Treatment of tumor with anti-CSF-1R antibody in combination with anti-PD-L1 antibody after unsuccessful anti-PD-L1 / PD1 treatment|
JP2020514277A|2016-12-23|2020-05-21|アールイーエムディー バイオセラピューティクス,インコーポレイテッドREMD Biotherapeutics,Inc|Immunization with an antibody that binds to programmed death ligand-1 |
CN109071656B|2017-01-05|2021-05-18|璟尚生物制药公司|Checkpoint modulator antagonists|
CN110312736A|2017-01-23|2019-10-08|苏州康宁杰瑞生物科技有限公司|PD-L1 combination polypeptide or compound|
CN106699891B|2017-01-25|2019-04-09|北京天广实生物技术股份有限公司|A kind of anti-PD-L1 antibody, its medical composition and its use|
US20200165340A1|2017-02-16|2020-05-28|Ying Zhang|Anti-programmed death-ligand 1 antibodies and therapeutic uses thereof|
WO2018152415A1|2017-02-16|2018-08-23|Medimmune, Llc|Anti-pd-l1 antibody treatment of bladder cancer|
CN108456251A|2017-02-21|2018-08-28|上海君实生物医药科技股份有限公司|Anti- PD-L1 antibody and its application|
JP2020511133A|2017-03-09|2020-04-16|ゲンマブ エー/エス|Antibody against PD-L1|
WO2018170021A1|2017-03-16|2018-09-20|Alpine Immune Sciences, Inc.|Pd-l1 variant immunomodulatory proteins and uses thereof|
US20200095323A1|2017-03-20|2020-03-26|The General Hospital Corporation|MITIGATING Fc-Fc RECEPTOR INTERACTIONS IN CANCER IMMUNOTHERAPY|
WO2018178123A1|2017-03-29|2018-10-04|Glycotope Gmbh|BISPECIFIC MUC-1 x PD-L1 ANTIBODIES|
AU2018241916A1|2017-03-29|2019-10-17|Glycotope Gmbh|PD-L1 and TA-MUC1 antibodies|
SG10202110707UA|2017-03-30|2021-11-29|Merck Patent Gmbh|Combination of an anti-pd-l1 antibody and a dna-pk inhibitor for the treatment of cancer|
RU2665790C1|2017-04-17|2018-09-04|Закрытое Акционерное Общество "Биокад"|Monoclonal pd-l1 antibody|
CN107973854B|2017-12-11|2021-05-04|苏州银河生物医药有限公司|PDL1 monoclonal antibody and application thereof|
CN108250296B|2018-01-17|2020-07-07|长春金赛药业有限责任公司|Fully human anti-human PD-L1 monoclonal antibody and application thereof|
CN108276492B|2018-01-31|2021-12-14|中国药科大学|anti-PD-L1 monoclonal antibody and application thereof|EP3099319A2|2014-01-31|2016-12-07|Cytomx Therapeutics Inc.|Matriptase and u-plasminogen activator substrates and other cleavable moieties and methods of use thereof|
MA41374A|2015-01-20|2017-11-28|Cytomx Therapeutics Inc|MATRIX METALLOPROTEASE CLIVABLE AND SERINE PROTEASE CLIVABLE SUBSTRATES AND METHODS OF USE THEREOF|
MA42971A|2015-03-13|2018-08-15|Cytomx Therapeutics Inc|ANTI-PDL1 ANTIBODIES, ACTIVABLE ANTI-PLD1 ANTIBODIES, AND METHODS OF USE THEREOF|
CN106397592A|2015-07-31|2017-02-15|苏州康宁杰瑞生物科技有限公司|Single-domain antibody directed at programmed death ligandand derived protein thereof|
US9567399B1|2016-06-20|2017-02-14|Kymab Limited|Antibodies and immunocytokines|
SG11202011349PA|2018-05-14|2020-12-30|Werewolf Therapeutics Inc|Activatable interleukin-2 polypeptides and methods of use thereof|
JP2021523741A|2018-05-14|2021-09-09|ウェアウルフ セラピューティクス, インコーポレイテッド|Activateable interleukin 12 polypeptide and how to use it|
CN113677372A|2019-02-26|2021-11-19|西托姆克斯治疗公司|Combination therapy of activatable immune checkpoint inhibitors and conjugated activatable antibodies|
WO2020215020A1|2019-04-18|2020-10-22|Qlsf Biotherapeutics Inc.|Humanized anti-pd-l1 antibodies|
CN114173818A|2019-06-13|2022-03-11|西托姆克斯治疗公司|Use of activatable anti-PDL 1 antibody and anti-CTLA-4 antibody in a neoadjuvant combination therapy for the treatment of cancer|
CN114173815A|2019-06-13|2022-03-11|西托姆克斯治疗公司|Use of activatable anti-PDL 1 antibody and anti-CTLA-4 antibody in a combination therapy for treating cancer|
法律状态:
2021-11-03| B350| Update of information on the portal [chapter 15.35 patent gazette]|
优先权:
申请号 | 申请日 | 专利标题
US201762513937P| true| 2017-06-01|2017-06-01|
US62/513,937|2017-06-01|
US201762534950P| true| 2017-07-20|2017-07-20|
US62/534,950|2017-07-20|
US201762555598P| true| 2017-09-07|2017-09-07|
US62/555,598|2017-09-07|
US201862657567P| true| 2018-04-13|2018-04-13|
US62/657,567|2018-04-13|
PCT/US2018/035508|WO2018222949A1|2017-06-01|2018-05-31|Activatable anti-pdl1 antibodies, and methods of use thereof|
[返回顶部]