US20160084839A1 - Immunohistochemical assay for detecting expression of programmed death ligand 1 (pd-l1) in tumor tissue - Google Patents

Immunohistochemical assay for detecting expression of programmed death ligand 1 (pd-l1) in tumor tissue Download PDF

Info

Publication number
US20160084839A1
US20160084839A1 US14/782,067 US201414782067A US2016084839A1 US 20160084839 A1 US20160084839 A1 US 20160084839A1 US 201414782067 A US201414782067 A US 201414782067A US 2016084839 A1 US2016084839 A1 US 2016084839A1
Authority
US
United States
Prior art keywords
mps
tumor
mhs
staining
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/782,067
Other languages
English (en)
Inventor
Marisa Dolled-Filhart
Kenneth Emancipator, JR.
Frank Lynch
Robert H. Pierce
Dianna Wu
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Merck Sharp and Dohme LLC
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=51659153&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20160084839(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Individual filed Critical Individual
Priority to US14/782,067 priority Critical patent/US20160084839A1/en
Publication of US20160084839A1 publication Critical patent/US20160084839A1/en
Assigned to QUALTEK MOLECULAR LABORATORIES reassignment QUALTEK MOLECULAR LABORATORIES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LYNCH, FRANK
Assigned to MERCK SHARP & DOHME CORP. reassignment MERCK SHARP & DOHME CORP. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PIERCE, ROBERT H., DOLLED-FILHART, MARISA, EMANCIPATOR, Kenneth, QUALTEK MOLECULAR LABORATORIES, WU, Dianna
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57492Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds localized on the membrane of tumor or cancer cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57415Specifically defined cancers of breast
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57423Specifically defined cancers of lung
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/5743Specifically defined cancers of skin, e.g. melanoma
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70596Molecules with a "CD"-designation not provided for elsewhere in G01N2333/705
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the present invention relates to the quantitation of PD-L1 expression in animal tissue samples by immunohistochemical (IHC) assay.
  • the invention also relates to the identification of PD-L1 expression levels that correlate with response of cancer patients to therapy with antagonists of Programmed Death 1 (PD-1).
  • IHC immunohistochemical
  • PD-L1 is a cell surface glycoprotein that is one of two known ligands for Programmed Death 1 (PD-1), which is recognized as an important player in immune regulation and the maintenance of peripheral tolerance. Expression of PD-L1 has been observed on the surface of a variety of immune cells, including naive lymphocytes and activated B and T cells, monocytes and dendritic cells (Id.). Furthermore, PD-L1 mRNA is expressed by non-lymphoid tissues including vascular endothelial cells, epithelial cells, muscle cells, and in tonsil and placental tissue. See, e.g., Keir, M. E. et al., Annu Rev Immunol. 26:677-704 (2008); Sharp A. H. et al., Nature Immunol. 8:239-245 (2007); Okazaki T and Honjo T, Internat. immunol. 19:813-824 (2007).
  • PD-L1 expression has also been observed in a variety of human cancers, and interaction of tumor-cell expressed PD-L1 with PD-1 can induce inhibition or apoptosis of tumor-specific T cells.
  • mAbs monoclonal antibodies
  • mAbs that block binding of PD-L1 to PD-1 have been shown to have anti-tumor activity against a variety of tumor types, with early human clinical data suggesting that patients whose tumors express PD-L1 are more likely to respond to anti-PD-1 therapy.
  • One approach employed a simple binary end-point of positive or negative for PD-L1 expression, with a positive result defined in terms of the percentage of tumor cells that exhibited histologic evidence of cell-surface membrane staining.
  • the minimum threshold of membrane-stained tumor cells selected as the cut-off point at which a tumor tissue section was counted as positive for PD-L1 expression was either 1% of total tumor cells (Gadiot et al.) or 5% (Toplian et al., supra,) of total tumor cells.
  • PD-L1 expression in the tumor tissue section was quantified in the tumor cells as well as in infiltrating immune cells, which predominantly comprise lymphocytes (Taube et al., supra; Thompson).
  • the percentage of tumor cells and infiltrating immune cells that exhibited membrane staining were separately quantified as ⁇ 5%, 5 to 9%, and then in 10% increments up to 100%.
  • PD-L1 expression was counted as negative if the score was ⁇ 5% score and positive if the score was ⁇ 5%.
  • PD-L1 expression in the immune infiltrate was reported as a semi-quantitative measurement called the adjusted inflammation score (AIS), which is determined by multiplying the percent of membrane staining cells by the intensity of the infiltrate, which was graded as none (0), mild (score of 1, rare lymphocytes), moderate (score of 2, focal infiltration of tumor by lymphohistiocytic aggregates), or severe (score of 3, diffuse infiltration).
  • AIS adjusted inflammation score
  • Diagnostic pathologists have devised other objective criteria for assigning a semi-quantitative score to the observed expression level of a target protein detected in IHC assays, including a three—to four—point scale, an HSCORE (McCarty, K. S. Jr, et al., Cancer Res. 46(suppl 8):4244s-4248s (1986) and the criteria of Allred et al. (Allred D C, Harvey J M, Berardo M, Clark G M. Mod Pathol. 11:155-168 (1998).
  • a 4-point HSCORE (McCarty et al) was calculated based on (a) intensity of staining ranging from 0 (no staining), 1+ (weak staining), 2+ (distinct staining), 3+ (strong staining) and 4+ (stained cells have minimal light transmission through them, meaning extremely strong/saturated signal) and multiplying by the percent of cells staining at each intensity (0 to 100%). Allred scoring (Allred et al) is based on calculating a Total Score (TS, range 0 to 8) by adding together a proportion score (PS) and an intensity score (IS).
  • TS Total Score
  • PS proportion score
  • IS intensity score
  • the inventors have surprisingly discovered that quantifying PD-L1 expression using IHC analysis, beyond a simple binary positive/negative characterization, can provide valuable predictive information to identify patients who are more likely to respond to treatment with anti-PD-1 therapy. Furthermore, the inventors have identified a unique scoring process that is intended to facilitate standardizing the quantitation of PD-L1 expression across multiple stained tissue sections and pathology laboratories. A PD-L1 expression score produced by performing this scoring process may be useful in selecting patients to receive anti-PD-1 therapy.
  • the present invention provides a novel process for scoring the expression of a PD-L1 protein in a tumor sample removed from an animal.
  • the process comprises obtaining a tissue section from the tumor that has been stained with an antibody that binds to the PD-L1 protein (anti-PD-L1 Ab), examining each tumor nest in the tissue section for staining, and assigning to the tissue section one or both of a modified H score (MHS) and a modified proportion score (MPS). If both of the MHS and MPS are assigned, the assignments may be performed in either order or simultaneously.
  • MHS modified H score
  • MPS modified proportion score
  • the estimated percentages, the sum of which is 100%, are then input into the formula of 1 ⁇ (percent of weak staining cells)+2 ⁇ (percent of moderate staining cells)+3 ⁇ (percent of strong staining cells), and the result is assigned to the tissue section as the MHS.
  • the MPS is assigned by estimating, across all of the viable tumor cells and stained mononuclear inflammatory cells in all of the examined tumor nests, the percentage of cells that have at least partial membrane staining of any intensity, and the resulting percentage is assigned to the tissue section as the MPS.
  • estimating the percentage comprises selecting the percentage from the group consisting of: 0, 1, 2, 5, 10, 15, 20, 30, 40, 50, 60, 70, 80, 85, 90, 95 and 100 that the observer determines best represents the proportion of the examined cells are positive for membrane staining.
  • the scoring process of the invention further comprises examining the stroma surrounding each tumor nest for the presence or absence of a band of anti-membrane-stained cells within the stroma. If the band is detected in the stroma surrounding at least one tumor nest, then the tissue section is assigned a stroma score (SS) of positive. A stroma score (SS) of negative is assigned to the tissue section if the band is not detected.
  • SS stroma score
  • the scoring process of the invention is used to score PD-L1 expression in a tissue section of a tumor sample obtained from a patient diagnosed with non-small cell lung carcinoma (NSCLC), head and neck squamous carcinoma, or transitional cell carcinoma of the bladder (TCC), and in this embodiment, the process further comprises designating the tumor sample as positive or negative for PD-L1 expression.
  • the tumor sample is designated as positive if the tissue section has any one or more for the following score assignments: (i) the SS is positive; (ii) the MHS is greater than zero, and (iii) the MPS is greater than zero.
  • the tumor sample is designated as negative if the tissue section has any one or more of the following score assignments: (iv) the SS is negative and the MHS is zero; (v) the SS is negative and the MPS is zero; and (vi) the SS is negative, the MHS is zero and the MPS is zero.
  • the scoring process of the invention is used to score PD-L1 expression in a tissue section of a tumor sample obtained from a patient diagnosed with melanoma or breast cancer, and the process further comprises examining each tumor nest for the presence or absence of a lattice pattern of membrane-stained dendriform cells within the tumor nest. If the lattice pattern is detected in at least one of the tumor nests, the tissue section is assigned a dendriform pattern score (DS) of positive. A DS of negative is assigned to the tissue section if the lattice pattern is not detected in any of the tumor nests.
  • DS dendriform pattern score
  • the stroma, DS, and one or both of the MHS and MPS scores assigned to a tissue section from melanoma or breast cancer are used to designate the tumor sample as positive or negative for PD-L1 expression.
  • the tumor sample is designated as positive if the tissue section has any one or more for the following score assignments: (i) the stroma score is positive; (ii) the DS is positive; (iii) the MHS is greater than zero, and (iv) the MPS is greater than zero.
  • the tumor sample is designated as negative if the tissue section has any one or more of the following score assignments: (v) the stroma score is negative, the DS is negative and the MHS is zero; (vi) the stroma score is negative, the DS is negative and the MPS is zero; and (vii) the stroma score is negative, the DS is negative, the MHS is zero and the MPS is zero.
  • the invention provides a method of designating a tumor sample as PD-L1 positive or PD-L1 negative.
  • the method comprises (a) obtaining a tissue section from the tumor sample that has been stained with an antibody that binds to the PD-L1 protein (anti-PD-L1 Ab), (b) examining each tumor nest in the tissue section for staining, (c) assigning to the tissue section a modified proportion score (MPS), (d) examining the stroma surrounding each tumor nest for the presence or absence of a band of anti-membrane-stained cells within the stroma, (e) assigning to the tissue section a stroma score (SS) of positive or negative, wherein a positive SS is assigned if the band is detected and a negative stroma score (SS) is assigned to the tissue section if the band is not detected, and designating the tumor sample as PD-L1 positive if the assigned MPS is greater than 0 or if the assigned SS is positive, and designating the tumor sample as PD
  • the tumor sample is selected from the group consisting of gastric cancer, bladder cancer, melanoma, NSCLC, head and neck cancer, colon or rectal adenocarcinoma, Hodgkins lymphoma, non-Hodgkins lymphoma, multiple myeloma, Myelodysplastic syndrome (MDS), anal canal squamous cell carcinoma, pancreas adenocarcinoma, esophageal squamous cell carcinoma or adenocarcinoma, biliary tract adenocarcinoma (gallbladder and biliary tree but excluding ampulla of vater cancers), carcinoid tumors, neuroendocrine carcinomas, well or moderately differentiated pancreatic neuroendocrine tumor, estrogen receptor (ER) positive HER2 negative breast cancer, HER2 positive breast cancer, triple negative breast cancer, ovarian epithelial, fallopian tube or primary peritoneal carcinoma, endometrial carcinoma, cervical a,
  • the present invention provides a method for predicting if a human patient diagnosed with non-small cell lung carcinoma (NSCLC) is likely to respond to treatment with a PD-1 antagonist.
  • the process comprises obtaining one or both of an MHS and an MPS for a tumor sample removed from the patient, wherein the MHS and MPS are obtained by performing the scoring process described above, and predicting the patient as likely to respond to the treatment if the obtained MHS and/or MPS is at or above a specified threshold or predicting the patient as not likely to respond to the treatment if each of the obtained MHS and MPS is below the specified threshold.
  • the MHS and/or the MPS determined in accordance with the scoring process described above are used in a method of treating a human patient diagnosed with NSCLC.
  • the method comprises obtaining a diagnostic report that provides one or both of a modified H score (MHS) and a modified proportion score (MPS) for PD-L1 expression for a NSCLC tumor sample removed from the patient.
  • MHS modified H score
  • MPS modified proportion score
  • the patient is treated with a chemotherapeutic regimen comprising a PD-1 antagonist if one or both of the MHS and the MPS is at or above a specified threshold. However, if each of the MHS and MPS are below the specified threshold, the patient is treated with a chemotherapeutic regimen that does not include a PD-1 antagonist.
  • the estimated percentages used in assigning one of both of the MHS and the MPS is preferably confined to a discrete estimate selected from the group consisting of 0, 1, 2, 5, 10, 15, 20, 30, 40, 50, 60, 70, 80, 85, 90, 95 and 100.
  • the tissue section is preferably a formalin-fixed, paraffin-embedded tissue section.
  • the anti-PD-L1 Ab used for staining the tissue section is preferably the monoclonal antibody 22C3.
  • the PD-1 antagonist is preferably an anti-PD-1 antibody which comprises a heavy chain and a light chain, and wherein the heavy chain comprises the amino acid sequence shown in FIG. 9 (SEQ ID NO:32) and the light chain comprises the amino acid sequence shown in FIG. 10 (SEQ ID NO:33).
  • FIG. 1 shows images of an FFPE tissue section of NSCLC stained by IHC with the antibody 22C3, with the box indicating a particular field viewed at 1 ⁇ , 4 ⁇ and 20 ⁇ magnification, and the arrows indicating PD-L1 expressing tumor cells and PD-L1 positive stroma in this field at 20 ⁇ magnification.
  • FIG. 2 shows images of an FFPE tissue section of NSCLC stained by IHC with the antibody 22C3, with the box indicating a particular field viewed at 1 ⁇ , 4 ⁇ and 20 ⁇ magnification, the arrows indicating PD-L1 expressing tumor cells and PD-L1 negative stroma in this field at 20 ⁇ magnification, with PD-L1 staining intensity scored as 1+ for weak staining, 2+ for moderate staining and 3+ for strong staining.
  • FIG. 3 shows images of IHC staining by the antibody 22C3 in FFPE tissue sections of squamous cell carcinoma of the head and neck at 20 ⁇ magnification, with FIG. 3A showing a field that has no PD-L1 staining in the stroma or tumor cells, FIG. 3B showing a field that has no PD-L1 staining in the tumor, but PD-L1 staining in the stroma, FIG. 3C showing a field that has moderate PD-L1 staining in the tumor with no staining of the stroma, and FIG. 3D showing strong staining of tumor with no staining of the stroma.
  • FIG. 4 shows images of IHC staining by the antibody 22C3 in FFPE tissue sections of melanoma at 20 ⁇ magnification, with FIG. 4A showing a field that has staining of melanin, but no PD-L1 membrane staining, FIGS. 4B and 4C showing fields that have PD-L1 membrane staining in the tumor, FIG. 4D showing a field that has PD-L1 staining in the stroma and the tumor, and FIG. 4E showing a dendriform pattern of PD-L1 staining.
  • FIG. 5 shows images of IHC staining by the antibody 22C3 in FFPE tissue sections of breast cancer at 20 ⁇ magnification, with FIG. 5A showing a field that has a stained dendriform pattern, FIG. 5B showing a field that has stromal PD-L1 staining within the smaller intercalating regions; and FIG. 5C showing PD-L1 staining in the tumor.
  • FIG. 6 shows images of IHC staining by the antibody 22C3 in FFPE tissue sections of transitional cell carcinoma of the bladder at 40 ⁇ magnification, with FIG. 6A showing a field that has no PD-L1 staining, and FIGS. 6B and 6C showing a field that has PD-L1 staining in the tumor.
  • FIG. 7 shows the nucleotide sequences for antibody variable light and heavy chain cDNA (SEQ ID NO:7 and SEQ ID NO:15) prepared from total RNA isolated from hybridoma MEB037.20C3 and the predicted amino acid sequences encoded thereby (bold font) (SEQ ID NO:5 and SEQ ID NO:13), with brackets indicating nucleotide and amino acid sequences for the leader peptide and underlining indicating the CDR sequences.
  • FIG. 8 shows the nucleotide sequences for antibody variable light and heavy chain cDNA (SEQ ID NO:22 and SEQ ID NO:30) prepared from total RNA isolated from hybridoma MEB037.22C3 and the predicted amino acid sequences encoded thereby (bold font) (SEQ ID NO:20 and SEQ ID NO:28, with brackets indicating nucleotide and amino acid sequences for the leader peptide and underlining indicating the CDR sequences.
  • FIGS. 9A and 9B show the amino acid sequence (SEQ ID NO:32) for the heavy chain of a preferred anti-PD-1 antibody useful in the treatment methods of the present invention, which corresponds to amino acid residues 20 to 466 of SEQ ID NO:31 set forth in U.S. Pat. No. 8,354,509.
  • FIG. 10 shows the amino acid sequence (SEQ ID NO:33) for the light chain of a preferred anti-PD-1 antibody useful in the treatment methods of the present invention, which corresponds to amino acid residues 20 to 237 of SEQ ID NO:36 set forth in U.S. Pat. No. 8,354,509.
  • FR Antibody framework region the immunoglobulin variable regions excluding the CDR regions.
  • V region The segment of IgG chains which is variable in sequence between different antibodies. It extends to Kabat residue 109 in the light chain and 113 in the heavy chain.
  • administering refers to contact of an exogenous pharmaceutical, therapeutic, diagnostic agent, or composition to the animal, human, subject, cell, tissue, organ, or biological fluid.
  • Treatment of a cell encompasses contact of a reagent to the cell, as well as contact of a reagent to a fluid, where the fluid is in contact with the cell.
  • administering and “treatment” also means in vitro and ex vivo treatments, e.g., of a cell, by a reagent, diagnostic, binding compound, or by another cell.
  • subject includes any organism, preferably an animal, more preferably a mammal (e.g., rat, mouse, dog, cat, rabbit) and most preferably a human.
  • antibody refers to any form of antibody that exhibits the desired biological or binding activity. Thus, it is used in the broadest sense and specifically covers, but is not limited to, monoclonal antibodies (including full length monoclonal antibodies), polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), humanized, fully human antibodies, chimeric antibodies and camelized single domain antibodies.
  • parent antibodies are antibodies obtained by exposure of an immune system to an antigen prior to modification of the antibodies for an intended use, such as humanization of an antibody for use as a human therapeutic antibody.
  • the basic antibody structural unit comprises a tetramer.
  • Each tetramer includes two identical pairs of polypeptide chains, each pair having one “light” (about 25 kDa) and one “heavy” chain (about 50-70 kDa).
  • the amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the carboxy-terminal portion of the heavy chain may define a constant region primarily responsible for effector function.
  • human light chains are classified as kappa and lambda light chains.
  • human heavy chains are typically classified as mu, delta, gamma, alpha, or epsilon, and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively.
  • the variable and constant regions are joined by a “J” region of about 12 or more amino acids, with the heavy chain also including a “D” region of about 10 more amino acids. See generally, Fundamental Immunology Ch. 7 (Paul, W., ed., 2nd ed. Raven Press, N.Y. (1989).
  • variable regions of each light/heavy chain pair form the antibody binding site.
  • an intact antibody has two binding sites.
  • the two binding sites are, in general, the same.
  • variable domains of both the heavy and light chains comprise three hypervariable regions, also called complementarity determining regions (CDRs), located within relatively conserved framework regions (FR).
  • CDRs complementarity determining regions
  • FR framework regions
  • the CDRs are usually aligned by the framework regions, enabling binding to a specific epitope.
  • both light and heavy chains variable domains comprise FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4.
  • the assignment of amino acids to each domain is, generally, in accordance with the definitions of Sequences of Proteins of Immunological Interest , Kabat, et al.; National Institutes of Health, Bethesda, Md.; 5 th ed.; NIH Publ. No.
  • hypervariable region refers to the amino acid residues of an antibody that are responsible for antigen-binding.
  • the hypervariable region comprises amino acid residues from a “complementarity determining region” or “CDR” (i.e. CDRL1, CDRL2 and CDRL3 in the light chain variable domain and CDRH1, CDRH2 and CDRH3 in the heavy chain variable domain).
  • CDR complementarity determining region
  • CDR complementarity determining region
  • antibody fragment or “antigen binding fragment” refers to antigen binding fragments of antibodies, i.e. antibody fragments that retain the ability to bind specifically to the antigen bound by the full-length antibody, e.g. fragments that retain one or more CDR regions.
  • antibody binding fragments include, but are not limited to, Fab, Fab′, F(ab′) 2 , and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules, e.g., sc-Fv; nanobodies and multispecific antibodies formed from antibody fragments.
  • antibody 20C3 is (i) the antibody produced by hybridoma subclone MEB037.20C3.116 or (ii) a monoclonal antibody comprising the light chain and heavy chain mature variable regions set forth in Table 1 below.
  • antibody 22C3 is (i) the antibody produced by hybridoma subclone MEB037.22C3.138 or (ii) a monoclonal antibody comprising the light chain and heavy chain mature variable regions set forth in Table 2 below.
  • cancer refers to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • examples of cancer include but are not limited to, carcinoma, lymphoma, leukemia, blastoma, and sarcoma.
  • cancers include squamous cell carcinoma, myeloma, small-cell lung cancer, non-small cell lung cancer, glioma, hodgkin's lymphoma, non-hodgkin's lymphoma, acute myeloid leukemia (AML), multiple myeloma, gastrointestinal (tract) cancer, renal cancer, ovarian cancer, liver cancer, lymphoblastic leukemia, lymphocytic leukemia, colorectal cancer, endometrial cancer, kidney cancer, prostate cancer, thyroid cancer, melanoma, chondrosarcoma, neuroblastoma, pancreatic cancer, glioblastoma multiforme, cervical cancer, brain cancer, stomach cancer, bladder cancer, hepatoma, breast cancer, colon carcinoma, and head and neck cancer.
  • tissue samples such as ovarian, renal, colorectal, pancreatic, breast, liver, glioblastoma, non-small cell lung cancer, gastric, esophageal cancers, melanoma, head and neck cancer, and transitional cell cancer of the bladder.
  • a “chemotherapeutic agent” is a chemical compound or biological molecule useful in the treatment of cancer.
  • Classes of chemotherapeutic agents include, but are not limited to: alkylating agents, antimetabolites, kinase inhibitors, spindle poison plant alkaloids, cytoxic/antitumor antibiotics, topoisomerase inhibitors, photosensitizers, and antibodies and fusion proteins that block ligand/receptor signaling in any biological pathway that supports tumor maintenance and/or growth.
  • Chemotherapeutic agents useful in the treatment methods of the present invention include cytostatic agents, cytotoxic agents and immunotherapeutic agents.
  • Constantly modified variants or “conservative substitution” refers to substitutions of amino acids in a protein with other amino acids having similar characteristics (e.g. charge, side-chain size, hydrophobicity/hydrophilicity, backbone conformation and rigidity, etc.), such that the changes can frequently be made without altering the biological activity of the protein.
  • Those of skill in this art recognize that, in general, single amino acid substitutions in non-essential regions of a polypeptide do not substantially alter biological activity (see, e.g., Watson et al. (1987) Molecular Biology of the Gene , The Benjamin/Cummings Pub. Co., p. 224 (4th Ed.)).
  • substitutions of structurally or functionally similar amino acids are less likely to disrupt biological activity. Exemplary conservative substitutions are set forth in Table 3.
  • “Function-conservative variants,” as used herein, refers to antibodies or fragments in which one or more amino acid residues have been changed without altering a desired property, such an antigen affinity and/or specificity. Such variants include, but are not limited to, replacement of an amino acid with one having similar properties, such as the conservative amino acid substitutions of Table 3.
  • Dendriform cell in the context of a tumor tissue section stained in an IHC assay with an anti-PD-L1 Ab means a cell having numerous, branched surface projections resembling a tree.
  • a dendriform cell may include, but is not limited to, any type of dendritic cell that forms part of the mammalian immune system.
  • “Homology” refers to sequence similarity between two polypeptide sequences when they are optimally aligned. When a position in both of the two compared sequences is occupied by the same amino acid monomer subunit, e.g., if a position in a light chain CDR of two different Abs is occupied by alanine, then the two Abs are homologous at that position. The percent of homology is the number of homologous positions shared by the two sequences divided by the total number of positions compared ⁇ 100. For example, if 8 of 10 of the positions in two sequences are matched or homologous when the sequences are optimally aligned then the two sequences are 80% homologous.
  • the comparison is made when two sequences are aligned to give maximum percent homology.
  • the comparison can be performed by a BLAST algorithm wherein the parameters of the algorithm are selected to give the largest match between the respective sequences over the entire length of the respective reference sequences.
  • BLAST ALGORITHMS Altschul, S. F., et al., (1990) J. Mol. Biol. 215:403-410; Gish, W., et al., (1993) Nature Genet. 3:266-272; Madden, T. L., et al., (1996) Meth. Enzymol. 266:131-141; Altschul, S. F., et al., (1997) Nucleic Acids Res. 25:3389-3402; Zhang, J., et al., (1997) Genome Res. 7:649-656; Wootton, J. C., et al., (1993) Comput. Chem.
  • isolated antibody refers to the purification status and in such context means the molecule is substantially free of other biological molecules such as nucleic acids, proteins, lipids, carbohydrates, or other material such as cellular debris and growth media. Generally, the term “isolated” is not intended to refer to a complete absence of such material or to an absence of water, buffers, or salts, unless they are present in amounts that substantially interfere with experimental or therapeutic use of the binding compound as described herein.
  • the term “monoclonal antibody”, as used herein, refers to a population of substantially homogeneous antibodies, i.e., the antibody molecules comprising the population are identical in amino acid sequence except for possible naturally occurring mutations that may be present in minor amounts.
  • conventional (polyclonal) antibody preparations typically include a multitude of different antibodies having different amino acid sequences in their variable domains, particularly their CDRs, which are often specific for different epitopes.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al. (1975) Nature 256: 495, or may be made by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567).
  • the “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al. (1991) Nature 352: 624-628 and Marks et al. (1991) J. Mol. Biol. 222: 581-597, for example. See also Presta (2005) J. Allergy Clin. Immunol. 116:731.
  • Patient or “subject” refers to any single subject for which therapy is desired or that is participating in a clinical trial, epidemiological study or used as a control, including humans and mammalian veterinary patients such as cattle, horses, dogs, and cats.
  • PD-1 antagonist means any chemical compound or biological molecule that blocks binding of PD-L1 to PD-1, and includes antibodies and fusion proteins that bind to PD-1 or PD-L1.
  • Examples of PD-1 antagonists include any anti-PD-1 antibody described in U.S. Pat. No. 8,008,449, U.S. Pat. No. 7,521,051 and U.S. Pat. No. 8,354,509, any anti-PD-L1 antibody described in U.S. Pat. No.
  • BMS-936558 (nivolumab), MPDL3280A, CT-011, ONO-4538, BMS-936559, MEDI4736, AMP-224, and an anti-PD-1 antibody comprising the heavy and light chain amino acid sequences shown in FIGS. 9 and 10 herein.
  • Primary anti-PD-L1 antibody refers to an antibody that binds specifically to PD-L1 in a tissue section, and is generally the first antibody used in an IHC assay of PD-L1 expression in a tumor sample. In one embodiment, the primary antibody is the only antibody used in the IHC assay.
  • Secondary antibody refers to an antibody that binds specifically to a primary anti-PD-L1 antibody, thereby forming a bridge between the primary antibody and a subsequent detection reagent, if any.
  • the secondary antibody is generally the second antibody used in an IHC assay of PD-L1 expression in a tumor sample.
  • terapéuticaally effective amount refers to an amount of a chemotherapeutic effective to “treat” a cancer in a subject or mammal by achieving at least one positive therapeutic effect, such as for example, reduced number of cancer cells, reduced tumor size, reduced rate of cancer cell infiltration into peripheral organs, and reduced rate of tumor metastasis or tumor growth.
  • Positive therapeutic effects in cancer can be measured in a number of ways (See, W. A. Weber, J. Nucl. Med. 50:1S-10S (2009)).
  • a T/C ⁇ 42% is the minimum level of anti-tumor activity.
  • the treatment achieved by a therapeutically effective amount is progression free survival (PFS), disease free survival (DFS) or overall survival (OS).
  • PFS also referred to as “Time to Tumor Progression” indicates the length of time during and after treatment that the cancer does not grow, and includes the amount of time patients have experienced a complete response or a partial response, as well as the amount of time patients have experienced stable disease.
  • DFS refers to the length of time during and after treatment that the patient remains free of disease.
  • OS refers to a prolongation in life expectancy as compared to naive or untreated individuals or patients.
  • tissue Section refers to a single part or piece of a tissue sample, e.g., a thin slice of tissue cut from a sample of a normal tissue or of a tumor. It is understood that multiple sections of a single tissue sample may be prepared and analyzed in accordance with the present invention.
  • Treat” or “treating” means to administer a therapeutic agent, such as a composition containing any of the antibodies or antigen binding fragments of the present invention, internally or externally to a subject or patient having one or more disease symptoms, or being suspected of having a disease, for which the agent has therapeutic activity.
  • a therapeutic agent such as a composition containing any of the antibodies or antigen binding fragments of the present invention
  • the agent is administered in an amount effective to alleviate one or more disease symptoms in the treated subject or population, whether by inducing the regression of or inhibiting the progression of such symptom(s) by any clinically measurable degree.
  • the amount of a therapeutic agent that is effective to alleviate any particular disease symptom may vary according to factors such as the disease state, age, and weight of the patient, and the ability of the drug to elicit a desired response in the subject. Whether a disease symptom has been alleviated can be assessed by any clinical measurement typically used by physicians or other skilled healthcare providers to assess the severity or progression status of that symptom.
  • an embodiment of the present invention may not be effective in alleviating the target disease symptom(s) in every subject, it should alleviate the target disease symptom(s) in a statistically significant number of subjects as determined by any statistical test known in the art such as the Student's t-test, the chi 2 -test, the U-test according to Mann and Whitney, the Kruskal-Wallis test (H-test), Jonckheere-Terpstra-test and the Wilcoxon-test.
  • any statistical test known in the art such as the Student's t-test, the chi 2 -test, the U-test according to Mann and Whitney, the Kruskal-Wallis test (H-test), Jonckheere-Terpstra-test and the Wilcoxon-test.
  • Treatment refers to therapeutic treatment, as well as research and diagnostic applications. “Treatment” as it applies to a human, veterinary, or research subject, or cell, tissue, or organ, encompasses contact of the antibodies or antigen binding fragments of the present invention to a human or animal subject, a cell, tissue, physiological compartment, or physiological fluid.
  • Tumor as it applies to a subject diagnosed with, or suspected of having, a cancer, means a malignant or potentially malignant neoplasm or tissue mass of any size, and includes primary tumors and secondary neoplasms.
  • the present invention provides processes for scoring PD-L1 expression in tumor tissue sections that have been stained with an anti-PD-L1 antibody in an IHC assay.
  • the results of these scoring processes may be used to select patients for treatment with a PD-1 antagonist, e.g., as enrollment criteria in a clinical trial, to predict response of a subject to a PD-1 antagonist, and in methods of treating a patient for cancer.
  • a tumor tissue sample used to prepare stained tissue sections for scoring PD-L1 expression can be collected from a subject before and/or after exposure of the subject to one or more therapeutic agents, e.g. a PD-1 antagonist or other chemotherapeutic agent. Accordingly, tumor samples may be collected from a subject over a period of time.
  • the tumor sample can be obtained by a variety of procedures including, but not limited to, surgical excision, aspiration or biopsy.
  • the tissue sample may be sectioned and examined for PD-L1 as a fresh specimen. In other embodiments, the tissue sample is frozen for further sectioning. In other embodiments, the tissue sample is preserved by fixing and embedding in paraffin or the like.
  • the tissue sample may be fixed by conventional methodology, with the length of fixation depending on the size of the tissue sample and the fixative used. Neutral buffered formalin, glutaraldehyde, Bouin's or paraformaldehyde are nonlimiting examples of fixatives.
  • the tissue sample is fixed with formalin.
  • the fixed tissue sample is also embedded in paraffin to prepare a formalin-fixed and paraffin-embedded (FFPE) tissue sample.
  • FFPE formalin-fixed and paraffin-embedded
  • paraffin include, but are not limited to, Paraplast, Broloid and Tissuemay.
  • the tissue sample is fixed and dehydrated through an ascending series of alcohols, infiltrated and embedded with paraffin or other sectioning media so that the tissue sample may be sectioned.
  • the tumor tissue sample is first sectioned and then the individual sections are fixed.
  • the scoring process of the invention is performed on FFPE tissue sections of about 3-4 millimeters, and preferably 4 micrometers, which are mounted and dried on a microscope slide.
  • An IHC assay typically begins with antigen retrieval, which may vary in terms of reagents and methods.
  • the antigen retrieval process may involve pressure cooking, protease treatment, microwaving, or heating histologic sections in baths of appropriate buffers, with the standard goal of unmasking antigens hidden by formalin crosslinks or other fixation. See, e.g., Leong et al. Appl. Immnunohistochem. 4(3):201 (1996).
  • IHC Two general methods of IHC may be used; direct and indirect assays.
  • a direct IHC assay binding of antibody to the target antigen is determined directly.
  • This direct assay uses a labeled reagent, such as a fluorescent tag or an enzyme-labeled primary antibody, which can be visualized without further antibody interaction.
  • a labeled reagent such as a fluorescent tag or an enzyme-labeled primary antibody, which can be visualized without further antibody interaction.
  • unconjugated primary antibody binds to the antigen and then a labeled secondary antibody binds to the primary antibody.
  • a chromagenic or fluorogenic substrate is added to provide visualization of the antigen. Signal amplification occurs because several secondary antibodies may react with different epitopes on the primary antibody.
  • the primary and/or secondary antibody used for immunohistochemistry typically will be labeled with a detectable moiety.
  • Numerous labels are available which can be generally grouped into the following categories:
  • Radioisotopes such as 35 S, 14 C, 125 I, 3 H, and 131 I.
  • the antibody can be labeled with the radioisotope using the techniques described in Current Protocols in Immunology , Volumes 1 and 2, Coligen et al., Ed. Wiley-Interscience, New York, N.Y., Pubs. (1991) for example and radioactivity can be measured using scintillation counting.
  • radionuclides include 99 Tc, 90 Y, 111 In, 32 P, 11 C, 15 O, 13 N, 18 F, 51 Cr, 57 To, 226 Ra, 60 Co, 59 Fe, 57 Se, 152 Eu, 67 CU, 217 Ci, 211 At, 212 Pb, 47 Sc, 109 Pd, 234 Th, and 40 K, 157 Gd, 55 Mn 52 Tr, and 56 Fe.
  • Colloidal gold particles Colloidal gold particles.
  • Fluorescent or chemiluminescent labels including, but not limited to, rare earth chelates (europium chelates), fluorescein and its derivatives, rhodamine and its derivatives, isothiocyanate, phycoerythrin, phycocyanin, allophycocyanin, o-phthaladehyde, fluorescamine, dansyl, umbelliferone, luciferin, luminal label, isoluminal label, an aromatic acridinium ester label, an imidazole label, an acridimium salt label, an oxalate ester label, an aequorin label, 2,3-dihydrophthalazinediones, Texas Red, dansyl, Lissamine, umbelliferone, phycocrytherin, phycocyanin, or commercially available fluorophores such SPECTRUM ORANGE® and SPECTRUM GREEN® and/or derivatives of any one or more of the
  • the enzyme generally catalyzes a chemical alteration of the chromogenic substrate that can be measured using various techniques. For example, the enzyme may catalyze a color change in a substrate, which can be measured spectrophotometrically. Alternatively, the enzyme may alter the fluorescence or chemiluminescence of the substrate. Techniques for quantifying a change in fluorescence are described above.
  • the chemiluminescent substrate becomes electronically excited by a chemical reaction and may then emit light which can be measured (using a chemiluminometer, for example) or donates energy to a fluorescent acceptor.
  • enzymatic labels include luciferases (e.g. firefly luciferase and bacterial luciferase; U.S. Pat. No. 4,737,456), luciferin, 2,3-dihydrophthalazinediones, malate dehydrogenase, urease, peroxidase such as horseradish peroxidase (HRPO), alkaline phosphatase, ⁇ -galactosidase, glucoamylase, lysozyme, saccharide oxidases (e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase), heterocyclic oxidases (such as uricase and xanthine oxidase), lactoperoxidase, microperoxidase, and the like.
  • luciferases e.g. firefly luciferase and bacterial luciferase
  • HRP Horseradish peroxidase
  • DAB 3,3′ diamino benzidine
  • AEC 3-amino-9-ethylcarbazole
  • CN 4-chloro-1-napthol
  • p-Phenylenediamine dihydrochloride/pyrocatecol which generates a blue-black product
  • OPD orthophenylene diamine
  • TMB 3,3′,5,5′-tetramethyl benzidine hydrochloride
  • the label is indirectly conjugated with the antibody.
  • the antibody can be conjugated with biotin and any of the four broad categories of labels mentioned above can be conjugated with avidin, or vice versa. Biotin binds selectively to avidin and thus, the label can be conjugated with the antibody in this indirect manner.
  • the antibody is conjugated with a small hapten and one of the different types of labels mentioned above is conjugated with an anti-hapten antibody. Thus, indirect conjugation of the label with the antibody can be achieved.
  • the tissue section is exposed to an anti-PD-L1 antibody as the primary antibody for a sufficient period of time and under suitable conditions to allow the primary antibody to bind to the PD-L1 protein in the tissue section.
  • suitable conditions for achieving this can be determined by routine experimentation, with one example of suitable conditions described in Example 1 below.
  • the slide is then washed to remove unbound and excess amounts of the primary antibody.
  • the primary antibody is linked to a detectable label, such as paramagnetic ions, radioactive isotopes, fluorochromes, and NMR-detectable substances, and the slide is evaluated for PD-L1 staining using the appropriate imaging apparatus.
  • a detectable label such as paramagnetic ions, radioactive isotopes, fluorochromes, and NMR-detectable substances
  • immune complexes between PD-L1 and the primary antibody may be detected using a second binding agent that is linked to a detectable label.
  • the second binding agent is preferably a secondary antibody, which is applied to the slide at a concentration and for a period of time sufficient to allow the formation of secondary immune complexes. The slide is then typically washed to remove any non-specifically bound secondary antibody, and the label in the secondary immune complexes is detected.
  • the secondary antibody may be labeled using avidin, strepavidin or biotin, which is independently labeled with a detectable moiety, such as a fluorescent dye (stain), a luminescent dye or a non-fluorescent dye.
  • a detectable moiety such as a fluorescent dye (stain), a luminescent dye or a non-fluorescent dye.
  • any enzyme that can be conjugated to or can bind indirectly to the secondary antibody e.g., via conjugated avidin, strepavidin, biotin
  • the enzyme employed can be, for example, alkaline phosphatase (AP), horseradish peroxidase (HRP), beta-gal actosidase and/or glucose oxidase.
  • the enzyme can also be directed at catalyzing a luminescence reaction of a substrate, such as, but not limited to, luciferase and aequorin, having a substantially non-soluble reaction product capable of luminescencing or of directing a second reaction of a second substrate, such as but not limited to, luciferine and ATP or coelenterazine and Ca.sup.++, having a luminescencing product.
  • a detection reagent is applied that includes a chromagen or a fluorescently tagged molecule to visualize the localization of the immune complexes.
  • the IHC assay may be performed using an automated pathology system, which may include automated staining (conventional stains, histochemical techniques, immunostainers); automated in situ hybridization systems; automatic slide preparation (coverslip, slide drying) and integrated slide and cassette labeling, as described in Roja et al., Review of imaging solutions for integrated, quantitative immunohistochemistry in the Pathology daily practice, Folia Histochemica et Cytobiologica, Vol. 47, No. 3, 349-354, 2009.
  • automated pathology system may include automated staining (conventional stains, histochemical techniques, immunostainers); automated in situ hybridization systems; automatic slide preparation (coverslip, slide drying) and integrated slide and cassette labeling, as described in Roja et al., Review of imaging solutions for integrated, quantitative immunohistochemistry in the Pathology daily practice, Folia Histochemica et Cytobiologica, Vol. 47, No. 3, 349-354, 2009.
  • a preferred IHC assay employs the commercially available Dako EnVisionTM FLEX detection system, which is intended for use together with a Dako Autostainer instrument (Dako, an Agilent Technologies Company, Glostrup, Denmark). These reagents can be used off the shelf for other autostainers or for manually-performed staining (not performed with an autostainer)
  • the primary antibody is an anti-PD-L1 antibody, or antigen binding fragment thereof, which binds to the mature form of PD-L1 (lacking the presecretory leader sequence, also referred to as leader peptide) that is expressed on the surface of certain mammalian cells.
  • PD-L1 and “mature PD-L1” are used interchangeably herein, and shall be understood to mean the same molecule unless otherwise indicated or readily apparent from the context.
  • an anti-human PD-L1 antibody or an anti-hPD-L1 antibody refers to an antibody that specifically binds to mature human PD-L1.
  • a mature human PD-L1 molecule consists of amino acids 19-290 of the following sequence:
  • An antibody that “specifically binds to human PD-L1,” or an antibody that “specifically binds to a polypeptide comprising the amino acid sequence of human PD-L1,” is an antibody that exhibits preferential binding to human PD-L1 as compared to other antigens, but this specificity does not require absolute binding specificity.
  • An anti-hPD-L1 antibody is considered “specific” for human PD-L1 if its binding is determinative of the presence of human PD-L1 in a sample, e.g. without producing undesired results such as false positives in an IHC diagnostic assay.
  • Antibodies, or binding fragments thereof, useful as a primary antibody in the processes and methods of the present invention will bind to human PD-L1 with an affinity that is at least two fold greater, preferably at least ten times greater, more preferably at least 20-times greater, and most preferably at least 100-times greater than the affinity with any non-PD-L1 protein.
  • an antibody is said to bind specifically to a polypeptide comprising a given sequence, e.g. mature human PD-L1, if it binds to polypeptides comprising that sequence but does not bind to proteins lacking that sequence.
  • Tissue sections of tumor samples from human subjects may be scored for PD-L1 expression using any anti-hPD-L1 Ab that produces essentially the same staining results on an FFPE or frozen tissue section of a tumor sample from a human as produced by the 22C3 Ab.
  • the scoring process of the present invention may be used to quantify PD-L1 expression detected by the 5H-1 antibody developed at Mayo Foundation for Medical Education and Research (Toplian et al., supra).
  • an anti-PD-L1 Ab or antigen binding fragment useful in scoring expression of human PD-L1 by IHC assay will exhibit the same degree of specificity for human PD-L1 as the 22C3 antibody and retain at least 80%, 85%, 90%, 95% or 100% of its human PD-L1 binding affinity when that affinity is expressed on a molar basis. It is also intended that an anti-PD-L1 antibody or antigen binding fragment useful in the invention can include conservative or non conservative amino acid substitutions from the 22C3 Ab or 20C3 Ab (referred to as “conservative variants” or “function conserved variants” of the antibody) that do not substantially alter its binding specificity or affinity.
  • Anti-PD-L1 antibodies may be purified from a hybridoma culture by the following process.
  • the hybridoma culture is clarified by depth filtration using 1.2 micrometer glass fiber and 0.2 micrometer cellulose acetate filter.
  • An equal volume of 2 ⁇ ProSepA Buffer (100 mM Boric Acid, 5M NaCl, pH 8.5) is added to the clarified harvest and the diluted harvest is loaded onto a 170 mL bed volume Protein-A column.
  • the column is washed with 5 column volumes (CV) of 1 ⁇ ProSepA Buffer (50 mM Boric Acid, 2.5M NaCl, pH 8.5), then washed with 2CV of 1 ⁇ PBS, and the anti-hPD-L1 antibody eluted with 5CV of Elution Buffer (0.1M Glycine, pH 3.0).
  • the elution fractions containing IgG are combined and the pH neutralized by adding 1/10th volume of 1.0M Tris, pH buffer.
  • the neutralized antibody composition is then sterile filtered using a 10 kDa disposable TFF cassette.
  • the antibody may be formulated for storage by diafiltration against 10 liter of formulation buffer (20 mM sodium acetate, 9% sucrose, pH 5.0) and using 20 volume changes.
  • antibody 22C3 at a concentration of about 5.0 mg/ml can be prepared with a purity of at least 98% by SDS-PAGE, SEC HPLC and C8 RP-HPLC measurements, and with endotoxin levels of less than 0.1 EU/ml and less than 0.02 EU/mg.
  • the anti-PD-L1 antibodies described herein may also be produced recombinantly (e.g., in an E. coli /T7 expression system as discussed above).
  • nucleic acids encoding the antibody molecules e.g., V H or V L
  • V H or V L may be inserted into a pET-based plasmid and expressed in the E. coli /T7 system.
  • Methods by which to produce recombinant antibodies which are known in the art.
  • One example of a method for recombinant production of antibodies is disclosed in U.S. Pat. No. 4,816,567. Transformation can be by any known method for introducing polynucleotides into a host cell.
  • Methods for introduction of heterologous polynucleotides into mammalian cells include dextran-mediated transfection, calcium phosphate precipitation, polybrene-mediated transfection, protoplast fusion, electroporation, encapsulation of the polynucleotide(s) in liposomes, biolistic injection and direct microinjection of the DNA into nuclei.
  • nucleic acid molecules may be introduced into mammalian cells by viral vectors. Methods of transforming cells are well known in the art. See, for example, U.S. Pat. Nos. 4,399,216; 4,912,040; 4,740,461 and 4,959,455.
  • Anti-PD-L1 antibodies can also be synthesized by any of the methods set forth in U.S. Pat. No. 6,331,415.
  • Mammalian cell lines available as hosts for expression of the antibodies or fragments described herein are well known in the art and include many immortalized cell lines available from the American Type Culture Collection (ATCC). These include, inter alia, Chinese hamster ovary (CHO) cells, NSO, SP2 cells, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells (e.g., Hep G2), A549 cells, 3T3 cells, HEK-293 cells and a number of other cell lines.
  • Mammalian host cells include human, mouse, rat, dog, monkey, pig, goat, bovine, horse and hamster cells. Cell lines of particular preference are selected through determining which cell lines have high expression levels.
  • insect cell lines such as Sf9 cells, amphibian cells, bacterial cells, plant cells and fungal cells.
  • the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or, more preferably, secretion of the antibody into the culture medium in which the host cells are grown.
  • Antibodies can be recovered from the culture medium using standard protein purification methods. Further, expression of antibodies of the invention (or other moieties therefrom) from production cell lines can be enhanced using a number of known techniques. For example, the glutamine synthetase gene expression system (the GS system) is a common approach for enhancing expression under certain conditions. The GS system is discussed in whole or part in connection with European Patent Nos. 0 216 846, 0 256 055, and 0 323 997 and European Patent Application No. 89303964.4.
  • a polyclonal antibody is an antibody which was produced among or in the presence of one or more other, non-identical antibodies.
  • polyclonal antibodies are produced from collections of different B-lymphocytes, e.g. the B-lymphocyte of an animal treated with an immunogen of interest, which produces a population of different antibodies that are all directed to the immunogen.
  • polyclonal antibodies are obtained directly from an immunized animal, e.g. spleen, serum or ascites fluid.
  • the processes of the present invention further may use as the primary antibody an antigen-binding fragment of an anti-PD-L1 antibody.
  • the antibody fragments include F(ab) 2 fragments, which may be produced by enzymatic cleavage of an IgG by, for example, pepsin.
  • Fab fragments may be produced by, for example, reduction of F(ab) 2 with dithiothreitol or mercaptoethylamine.
  • a Fab fragment is a V L -C L chain appended to a V H -C H1 chain by a disulfide bridge.
  • a F(ab) 2 fragment is two Fab fragments which, in turn, are appended by two disulfide bridges.
  • the Fab portion of an F(ab) 2 molecule includes a portion of the F c region between which disulfide bridges are located.
  • An F V fragment is a V L or V H region.
  • Immunoglobulins may be assigned to different classes depending on the amino acid sequences of the constant domain of their heavy chains. There are at least five major classes of immunoglobulins: IgA, IgD, IgE, IgG and IgM, and several of these may be further divided into subclasses (isotypes), e.g. IgG-1, IgG-2, IgG-3 and IgG-4; IgA-1 and IgA-2.
  • the invention may employ antibodies and antigen binding fragments of any of these classes or subclasses of antibodies.
  • the primary antibody comprises a heavy chain constant region, e.g. a human constant region, such as ⁇ 1, ⁇ 2, ⁇ 3, or ⁇ 4 human heavy chain constant region or a variant thereof.
  • the primary antibody comprises a light chain constant region, e.g. a human light chain constant region, such as lambda or kappa human light chain region or variant thereof.
  • the human heavy chain constant region can be ⁇ 1 and the human light chain constant region can be kappa.
  • the Fc region of the antibody is ⁇ 4 with a Ser228Pro mutation (Schuurman, J et. al., Mol. Immunol. 38: 1-8, 2001).
  • the slide is analyzed for PD-L1 staining, either by a human, e.g., a pathologist, or a computer programmed to distinguish between specific and non-specific staining results.
  • the analysis may be performed directly by viewing the slide through a microscope at low, medium (10-20 ⁇ ) and high power (40-60 ⁇ ), or by viewing high resolution images of the slide taken at low, medium and high power.
  • Low and medium power is typically used to detect stained tumor nests, as well as stroma staining and dendriform pattern staining.
  • Medium and high power is typically used to examine individual tumor nests to estimate the number and intensity of viable tumor and mononuclear inflammatory cells that exhibit at least partial membrane staining.
  • stained tumor cells that are outside of tumor nests are included in the percentages used for calculating the MHS and/or the MPS.
  • Each of the steps of obtaining a tissue sample, preparing one or more tissue sections therefrom for IHC assay, performing the IHC staining, and scoring the results may be performed by separate individuals/entities at the same or separate locations. For example, a surgeon may obtain by biopsy a tissue sample from a cancer patient's tumor and then send the tissue sample to a pathology lab, which may fix the tissue sample and then prepare one or more slides, each with a single tissue section, for IHC assay. The slide(s) may be analyzed by IHC soon after preparation, or stored for future assay. The lab that prepared a tissue section for IHC assay may conduct the assay or send the slide(s) to a different lab to conduct the assay.
  • a pathologist or trained professional who scores the stained slide(s) for PD-L1 staining may work for the diagnostic lab, or may be an independent contractor.
  • a single diagnostic lab obtains the tissue sample from the subject's physician or surgeon and then performs all of the steps involved in preparing tissue sections, staining the slide(s) and scoring PD-L1 expression in the stained tissue section(s) or sending the stained slide(s) to a trained professional for PD-L1 scoring.
  • the individuals involved with preparing and analyzing the tissue section by IHC assay do not know the identity of the subject whose sample is being tested; i.e., the sample received by the laboratory is made anonymous in some manner before being sent to the laboratory.
  • the sample may be merely identified by a number or some other code (a “sample ID”) and the results of the IHC assay is reported to the party ordering the test using the sample ID.
  • sample ID a number or some other code
  • the link between the identity of a subject and the subject's tissue sample is known only to the individual or to the individual's physician.
  • the diagnostic laboratory after the test results have been obtained, the diagnostic laboratory generates a test report, which may include any one or more of the following results: the tissue sample was positive or negative for PD-L1 expression, the MPS, the MHS, the tissue sample was positive or negative for stromal staining, and the tissue sample was positive or negative for dendriform pattern.
  • the test report may also include guidance on how to interpret the results for predicting if a subject is likely to respond to a PD-1 antagonist.
  • the patient's tumor is from a NSLC and if the MPS or MHS is at or above a prespecified threshold, the test report may indicate that the patient has a PD-L1 expression score that is correlated with response or better response to treatment with a PD-1 antagonist, while if the MPS or MHS is below the threshold, then the test report indicates that the patient has a PD-L1 expression score that is correlated with no response or poor response to treatment with a PD-1 antagonist.
  • the prespecified threshold for PD-L1 expression in NSLC tissue samples is an MHS of 170, 175, 180, 185, 190, 195, 200, 205, 210, or any number between 170 and 210, and is preferably between 185 and 195. In other embodiments, the prespecified threshold for PD-L1 expression in NSLC tissue samples is an MPS of 80, 85, 90, 95, 100 or any number between 80 and 100, and is preferably between 85 and 95.
  • the test report is a written document prepared by the diagnostic laboratory and sent to the patient or the patient's physician as a hard copy or via electronic mail.
  • the test report is generated by a computer program and displayed on a video monitor in the physician's office.
  • the test report may also comprise an oral transmission of the test results directly to the patient or the patient's physician or an authorized employee in the physician's office.
  • the test report may comprise a record of the test results that the physician makes in the patient's file.
  • a physician may use PD-L1 expression score(s) as a guide in deciding how to treat a patient who has been diagnosed with a type of cancer that is susceptible to treatment with a PD-1 antagonist or other chemotherapeutic agent(s).
  • the physician would order a diagnostic test to determine PD-L1 expression in a tumor tissue sample removed from the patient prior to initiation of treatment with the PD-1 antagonist or the other chemotherapeutic agent(s), but it is envisioned that the physician could order the first or subsequent tests at any time after the individual is administered the first dose of the PD-1 antagonist or other chemotherapeutic agent(s).
  • a physician may be considering whether to treat the patient with a pharmaceutical product that is indicated for patients whose tumor tests positive for PD-L1 expression. In some embodiments, if the patient is diagnosed with a type of cancer that is susceptible to treatment with a PD-1 antagonist and at least one chemotherapeutic agent that is not a PD-1 antagonist, the physician may select which chemotherapeutic agent to use based on the PD-L1 expression score for the patients tumor obtained from the diagnostic laboratory.
  • the physician selects a chemotherapeutic regimen that includes at least the PD-1 antagonist (optionally in combination with one or more additional chemotherapeutic agents), and if the reported PD-L1 expression score is below a threshold score that is correlated with no response or poor response to treatment with a PD-1 antagonist, the physician selects a regimen that does not include any PD-1 antagonist.
  • the physician may also take into account other relevant circumstances, such as the type of cancer to be treated, the age, weight, gender, genetic background and race of the patient, including inputting a combination of these factors and the test results into a model that helps guide the physician in choosing a therapy and/or treatment regimen with that therapy.
  • the reagents useful to perform immunohistochemistry on an FFPE tumor tissue section are provided in a kit along with instructions for performing the IHC assay and scoring PD-L1 expression on the tissue section in accordance with a scoring process described herein.
  • the kit comprises a primary antibody, and may further comprise a secondary antibody.
  • the kit will include substrates and cofactors required by the enzyme (e.g., a substrate precursor which provides the detectable chromophore or fluorophore).
  • substrates and cofactors required by the enzyme e.g., a substrate precursor which provides the detectable chromophore or fluorophore.
  • other additives may be included such as stabilizers, buffers (e.g., a block buffer or lysis buffer) and the like.
  • the relative amounts of the various reagents may be varied widely to provide for concentrations in solution of the reagents which substantially optimize the sensitivity of the assay.
  • the reagents may be provided as dry powders, usually lyophilized, including excipients which on dissolution will provide a reagent solution having the appropriate concentration.
  • a process for scoring the expression of a programmed death ligand 1 (PD-L1) protein in a tumor sample removed from an animal comprising:
  • assigning the MHS comprises (i) estimating, across all of the viable tumor cells and stained mononuclear inflammatory cells in all of the examined tumor nests, four separate percentages for cells that have no staining, weak staining (+1), moderate staining (+2) and strong staining (+3), wherein a cell must have at least partial membrane staining to be included in the weak, moderate or strong staining percentages, and wherein the sum of all four percentages equals 100; and (ii) inputting the estimated percentages into the formula of 1 ⁇ (percent of weak staining cells)+2 ⁇ (percent of moderate staining cells)+3 ⁇ (percent of strong staining cells), and assigning the result of the formula to the tissue section as the MHS;
  • assigning the MPS comprises estimating, across all of the viable tumor cells and mononuclear inflammatory cells in all of the examined tumor nests, the percentage of cells that have at least partial membrane staining of any intensity, and assigning the resulting percentage to the tissue section as the MPS;
  • the assignments may be made in either order or simultaneously.
  • the tumor sample is from a cancer selected from the group consisting of non-small cell lung carcinoma (NSCLC), head and neck squamous carcinoma, and transitional bladder carcinoma, and the process further comprises designating the tumor sample as positive or negative for PD-L1 expression, wherein
  • NSCLC non-small cell lung carcinoma
  • PD-L1 transitional bladder carcinoma
  • the tumor sample is designated as positive for PD-L1 expression if either of the MHS or the MPS is greater than zero, and
  • the tumor sample is designated as negative for PD-L1 expression if the MHS is zero or the MPS is zero.
  • the tumor sample is from a cancer selected from the group consisting of non-small cell lung carcinoma (NSCLC), head and neck squamous carcinoma, and transitional bladder carcinoma, and the process further comprises designating the tumor sample as positive or negative for PD-L1 expression,
  • NSCLC non-small cell lung carcinoma
  • PD-L1 transitional bladder carcinoma
  • tumor sample is designated as positive for PD-L1 expression if the tissue section has any one or more of the following score assignments:
  • tumor sample is designated as negative for PD-L1 expression if the tissue section has any one or more of the following score assignments:
  • DS dendriform pattern score
  • tumor sample is designated as positive for PD-L1 expression if the tissue section has any one or more of the following score assignments:
  • tumor sample is designated as negative for PD-L1 expression if the tissue section has any one or more of the following score assignments:
  • MPS modified P score
  • the anti-PD-L1 Ab is selected from the group consisting of, the monoclonal antibody 20C3, the monoclonal antibody 22C3 or the monoclonal antibody 5H-1. 19. The method of any of the above embodiments, wherein the anti-PD-L1 Ab is the monoclonal antibody 22C3. 20. The process or method of any of the above embodiments, wherein the tumor sample is from a human patient who is being evaluated for treatment with a PD-1 antagonist. 21. The process or method of embodiment 20, wherein the PD-1 antagonist is an anti-PD-1 monoclonal antibody that comprises a heavy chain and a light chain, and wherein the heavy chain comprises SEQ ID NO:32 and the light chain comprises SEQ ID NO:33.
  • NSCLC non small cell lung carcinoma
  • the specified threshold is selected from the group consisting of: the MHS is greater than zero; the MPS is greater than zero; and the MHS and the MPS is greater than zero.
  • MHS and MPS were determined by a scoring process comprising:
  • This Example describes the IHC assay used for the stained tumor tissue sections shown in FIGS. 1-6 .
  • Step 2 Dewax/Pre-Antigen Retrieval/Unmasking:
  • a Four (4) changes of room temperature (25° C.) absolute xylene for 5 min each with no agitation; b. Two (2) changes of room temperature (25° C.) absolute alcohol for 2 min each with no agitation; c. Two (2) changes of room temperature (25° C.) 70% alcohol for 2 min each with no agitation; d. Two (2) changes of room temperature (25° C.) 30% alcohol for 2 min each with no agitation; e. Two (2) changes of room temperature (25° C.) distilled water for rinsing using a minimum of 16 dips in-out; and f. Slides are immersed in room temperature (25° C.) distilled water and then transferred to antigen/epitope retrieval/unmasking platform.
  • a Commercial steamer pre-heated to 97° C.
  • b Heat induced antigen/epitope retrieval/unmasking using 1 ⁇ DAKO 51699 low pH Target Retrieval Solution, 97° C. for 20 min.
  • Ten (10) tissue sections are face-paired with clean blank slides in TechMate reagent trays containing exactly 10 mL antigen/epitope retrieval/unmasking solution to capillary draw reagent up and over the tissues. Tissue sections on slides are bathed in antigen/epitope retrieval/unmasking solution in presence of steam heat.
  • c
  • Post-antigen/epitope retrieval/unmasking cool for 5 min, slide pairs firmly inserted into a TechMate slide holder and drained of DAKO S1699 low pH Target Retrieval Solution with an absorbent wick pad. d. Wash two (2) times manually using capillary action (drain-draw) with FLEX+ wash buffer (part of DAKO K8012 EnVision FLEX+ kit [no more than 1 wk diluted from 20 ⁇ concentrate]).
  • EnVision FLEX+ reagents are stored at 2-8° C., dispense or dilute as necessary, with all procedures below automated at room temperature (25° C.). Reagent changes (washes, incubations) take place by capillary action (drain-draw) using absorbent wick pads (drain) and TechMate reagent trays (draw). a. Wash three (3) times with FLEX+ wash buffer (part of DAKO K8012 [no more than 1 wk diluted from 20 ⁇ concentrate]) b. DAKO 53020 Proteinase K diluted 1/160 in FLEX+ wash buffer, 10 mins c. Wash three (3) times with FLEX+ wash buffer d.
  • Immunohistochemical applications may be manually operated or automated. Automated systems can perform all the staining steps as described above. Laboratory equipment necessary to perform the assay include a heat source (steamer) and automated staining platform (such as TechMate).
  • FIGS. 1 to 6 illustrate how FFPE tissue sections stained with an anti-PD-L1 antibody in the IHC assay described in Example 1 would be scored for PD-L1 expression using the scoring process of the present invention.
  • the Brief Description of the Figures lists the various scores that were assigned to individual fields of each tissue section for illustrative purposes only. However, a scoring process of the invention is to be performed by examining the entire tissue section on the slide, and in practice, the pathologist would score a slide for PD-L1 expression by viewing the tissue section on the slide at low, medium and high magnification.
  • the scoring process described above was used to score PD-L1 expression in tissue sections of NSCLC tumor samples removed from patients prior to treatment with an anti-PD-1 antibody, which comprised the heavy and light chains set forth in SEQ ID NOs: 33 and 32, respectively.
  • an anti-PD-1 antibody which comprised the heavy and light chains set forth in SEQ ID NOs: 33 and 32, respectively.
  • the results indicated that higher MHS and MPS values predict tumor response, irrespective of stroma score, and also that the MPS was as predictive as the MHS in predicting tumor response in this patient group.
  • the following table summarizes the results of these correlations.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Urology & Nephrology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Oncology (AREA)
  • Medicinal Chemistry (AREA)
  • Hospice & Palliative Care (AREA)
  • Biotechnology (AREA)
  • Food Science & Technology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
US14/782,067 2013-04-02 2014-03-31 Immunohistochemical assay for detecting expression of programmed death ligand 1 (pd-l1) in tumor tissue Abandoned US20160084839A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/782,067 US20160084839A1 (en) 2013-04-02 2014-03-31 Immunohistochemical assay for detecting expression of programmed death ligand 1 (pd-l1) in tumor tissue

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201361807581P 2013-04-02 2013-04-02
PCT/US2014/032305 WO2014165422A1 (en) 2013-04-02 2014-03-31 Immunohistochemical assay for detecting expression of programmed death ligand 1 (pd-l1) in tumor tissue
US14/782,067 US20160084839A1 (en) 2013-04-02 2014-03-31 Immunohistochemical assay for detecting expression of programmed death ligand 1 (pd-l1) in tumor tissue

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/032305 A-371-Of-International WO2014165422A1 (en) 2013-04-02 2014-03-31 Immunohistochemical assay for detecting expression of programmed death ligand 1 (pd-l1) in tumor tissue

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/712,246 Continuation US11193937B2 (en) 2013-04-02 2017-09-22 Immunohistochemical assay for detecting expression of programmed death ligand 1 (PD-L1) in tumor tissue

Publications (1)

Publication Number Publication Date
US20160084839A1 true US20160084839A1 (en) 2016-03-24

Family

ID=51659153

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/782,067 Abandoned US20160084839A1 (en) 2013-04-02 2014-03-31 Immunohistochemical assay for detecting expression of programmed death ligand 1 (pd-l1) in tumor tissue
US15/712,246 Active 2034-06-23 US11193937B2 (en) 2013-04-02 2017-09-22 Immunohistochemical assay for detecting expression of programmed death ligand 1 (PD-L1) in tumor tissue

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/712,246 Active 2034-06-23 US11193937B2 (en) 2013-04-02 2017-09-22 Immunohistochemical assay for detecting expression of programmed death ligand 1 (PD-L1) in tumor tissue

Country Status (3)

Country Link
US (2) US20160084839A1 (de)
EP (1) EP2981821B2 (de)
WO (1) WO2014165422A1 (de)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109311989A (zh) * 2016-04-14 2019-02-05 创新微技术公司 在癌症疗法的治疗决定中的pd-l1表达的应用方法
CN109963872A (zh) * 2016-09-20 2019-07-02 默克专利股份有限公司 诊断抗pd-l1抗体及其用途
WO2019241730A2 (en) 2018-06-15 2019-12-19 Flagship Pioneering Innovations V, Inc. Increasing immune activity through modulation of postcellular signaling factors
US10628658B2 (en) * 2014-11-10 2020-04-21 Ventana Medical Systems, Inc. Classifying nuclei in histology images
US10695426B2 (en) * 2014-08-25 2020-06-30 Pfizer Inc. Combination of a PD-1 antagonist and an ALK inhibitor for treating cancer
WO2020227159A2 (en) 2019-05-03 2020-11-12 Flagship Pioneering Innovations V, Inc. Methods of modulating immune activity
CN112462072A (zh) * 2020-09-24 2021-03-09 浙江大学 一种tmub1蛋白在制备肿瘤免疫抑制分子检测剂中的应用
WO2021127217A1 (en) 2019-12-17 2021-06-24 Flagship Pioneering Innovations V, Inc. Combination anti-cancer therapies with inducers of iron-dependent cellular disassembly
WO2022006179A1 (en) 2020-06-29 2022-01-06 Flagship Pioneering Innovations V, Inc. Viruses engineered to promote thanotransmission and their use in treating cancer
CN113933505A (zh) * 2021-12-15 2022-01-14 北京市肿瘤防治研究所 一组多维分析预测胃癌免疫治疗疗效的tiic指标及其应用
WO2022212784A1 (en) 2021-03-31 2022-10-06 Flagship Pioneering Innovations V, Inc. Thanotransmission polypeptides and their use in treating cancer
WO2023278641A1 (en) 2021-06-29 2023-01-05 Flagship Pioneering Innovations V, Inc. Immune cells engineered to promote thanotransmission and uses thereof
US11585817B2 (en) 2016-04-01 2023-02-21 Agilent Technologies, Inc. Scoring methods for anti-PD therapy eligibility and compositions for performing same
WO2024077191A1 (en) 2022-10-05 2024-04-11 Flagship Pioneering Innovations V, Inc. Nucleic acid molecules encoding trif and additionalpolypeptides and their use in treating cancer

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL2170959T3 (pl) 2007-06-18 2014-03-31 Merck Sharp & Dohme Przeciwciała przeciwko ludzkiemu receptorowi programowanej śmierci PD-1
KR20210060670A (ko) 2008-12-09 2021-05-26 제넨테크, 인크. 항-pd-l1 항체 및 t 세포 기능을 향상시키기 위한 그의 용도
AR093984A1 (es) 2012-12-21 2015-07-01 Merck Sharp & Dohme Anticuerpos que se unen a ligando 1 de muerte programada (pd-l1) humano
WO2014151006A2 (en) 2013-03-15 2014-09-25 Genentech, Inc. Biomarkers and methods of treating pd-1 and pd-l1 related conditions
WO2015048312A1 (en) 2013-09-26 2015-04-02 Costim Pharmaceuticals Inc. Methods for treating hematologic cancers
JOP20200094A1 (ar) 2014-01-24 2017-06-16 Dana Farber Cancer Inst Inc جزيئات جسم مضاد لـ pd-1 واستخداماتها
JOP20200096A1 (ar) 2014-01-31 2017-06-16 Children’S Medical Center Corp جزيئات جسم مضاد لـ tim-3 واستخداماتها
EP3149042B1 (de) 2014-05-29 2019-08-28 Spring Bioscience Corporation Pd-l1-antikörper und verwendungen davon
MX2017000419A (es) 2014-07-11 2017-08-16 Genentech Inc Anticuerpos anti-pd-l1 y sus usos de diagnóstico.
US9535074B2 (en) * 2014-09-08 2017-01-03 Merck Sharp & Dohme Corp. Immunoassay for soluble PD-L1
RU2718914C2 (ru) 2014-09-13 2020-04-15 Новартис Аг Сочетанные способы лечения с использованием ингибиторов alk
CR20170143A (es) 2014-10-14 2017-06-19 Dana Farber Cancer Inst Inc Moléculas de anticuerpo que se unen a pd-l1 y usos de las mismas
WO2016070001A1 (en) * 2014-10-31 2016-05-06 Jounce Therapeutics, Inc. Methods of treating conditions with antibodies that bind b7-h4
ES2791950T3 (es) 2015-02-03 2020-11-06 Ventana Med Syst Inc Ensayo histoquímico para evaluar la expresión del ligando de muerte programada 1 (PD-L1)
US10478494B2 (en) * 2015-04-03 2019-11-19 Astex Therapeutics Ltd FGFR/PD-1 combination therapy for the treatment of cancer
EP3283882B1 (de) 2015-04-17 2020-12-16 Merck Sharp & Dohme Corp. Blutbasierte biomarker der tumorempfindlichkeit gegen pd-1-antagonisten
WO2016172624A1 (en) * 2015-04-22 2016-10-27 Agenus Inc. Methods for treating cancer
EP3892284B1 (de) 2015-05-29 2024-05-22 Merck Sharp & Dohme LLC Kombination aus einem pd-1-antagonist und cpg-c-typ-oligonukleotid zur behandlung von krebs
US11090358B2 (en) 2015-09-09 2021-08-17 Fred Hutchinson Cancer Research Center Cartilage-homing peptides
CN105717298A (zh) * 2016-01-29 2016-06-29 浙江数问生物技术有限公司 Pd-l1免疫组化试剂盒
AU2017273726B2 (en) 2016-06-01 2023-10-19 Epizyme, Inc. Use of EZH2 inhibitors for treating cancer
EP3464369A1 (de) * 2016-06-03 2019-04-10 Bristol-Myers Squibb Company Anti-pd-1-antikörper zur verwendung in einem verfahren zur behandlung eines tumors
CN110944628A (zh) * 2017-06-02 2020-03-31 Epizyme股份有限公司 使用ezh2抑制剂治疗癌症
AU2018313744C1 (en) 2017-08-08 2023-07-27 Chemocentryx, Inc. Macrocyclic immunomodulators
WO2019178573A1 (en) * 2018-03-16 2019-09-19 Blaze Bioscience, Inc. Truncated cartilage-homing peptides and peptide complexes and methods of use thereof
WO2020081463A1 (en) * 2018-10-18 2020-04-23 Verily Life Sciences Llc Systems and methods for using image processing to generate inferences of biomarker for immunotherapy
AU2019369222A1 (en) 2018-10-31 2021-05-27 Merck Sharp & Dohme Llc Anti-human PD-1 antibody crystals and methods of use thereof
WO2023010057A1 (en) * 2021-07-28 2023-02-02 Atreca, Inc. Atrc-101 target expression assay
WO2023091909A1 (en) 2021-11-16 2023-05-25 Sotio Biotech Inc. Treatment of myxoid/round cell liposarcoma patients
WO2023114346A2 (en) * 2021-12-16 2023-06-22 Merck Sharp & Dohme Llc Biomarkers for predicting eligibility for an anti-ilt4 and anti-pd-1 combination therapy
CN115165511B (zh) * 2022-09-09 2023-01-06 迈杰转化医学研究(苏州)有限公司 Pd-l1免疫组化检测质控品和参考品

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100266617A1 (en) * 2007-06-18 2010-10-21 N.V. Organon Antibodies to human programmed death receptor pd-1
WO2014100079A1 (en) * 2012-12-21 2014-06-26 Merck Sharp & Dohme Corp. Antibodies that bind to human programmed death ligand 1 (pd-l1)

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4318980A (en) 1978-04-10 1982-03-09 Miles Laboratories, Inc. Heterogenous specific binding assay employing a cycling reactant as label
US4399216A (en) 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4740461A (en) 1983-12-27 1988-04-26 Genetics Institute, Inc. Vectors and methods for transformation of eucaryotic cells
WO1986005807A1 (en) 1985-04-01 1986-10-09 Celltech Limited Transformed myeloma cell-line and a process for the expression of a gene coding for a eukaryotic polypeptide employing same
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
GB8601597D0 (en) 1986-01-23 1986-02-26 Wilson R H Nucleotide sequences
US4959455A (en) 1986-07-14 1990-09-25 Genetics Institute, Inc. Primate hematopoietic growth factors IL-3 and pharmaceutical compositions
US4912040A (en) 1986-11-14 1990-03-27 Genetics Institute, Inc. Eucaryotic expression system
GB8717430D0 (en) 1987-07-23 1987-08-26 Celltech Ltd Recombinant dna product
EP1576014B1 (de) 2002-12-23 2011-06-29 Wyeth LLC Antikörper gegen pd-1 und ihre verwendung
KR101339628B1 (ko) 2005-05-09 2013-12-09 메다렉스, 인코포레이티드 예정 사멸 인자 1(pd-1)에 대한 인간 모노클로날 항체, 및 항-pd-1 항체를 단독 사용하거나 기타 면역 요법제와 병용한 암 치료 방법
KR101888321B1 (ko) 2005-07-01 2018-08-13 이. 알. 스퀴부 앤드 선즈, 엘.엘.씨. 예정 사멸 리간드 1 (피디-엘1)에 대한 인간 모노클로날 항체
WO2007082154A2 (en) 2006-01-05 2007-07-19 Mayo Foundation For Medical Education And Research B7-h1 and b7-h4 in cancer
EP1835285A1 (de) * 2006-03-14 2007-09-19 Institut Gustave Roussy ERCC1-Expression bei der Prognose der Reaktion auf eine Krebs-Chemotherapie
MX351975B (es) * 2006-12-27 2017-11-06 Emory Univ Star Composiciones y metodos para el tratamiento de infecciones y tumores.
WO2010132958A1 (en) * 2009-05-22 2010-11-25 Garvan Institute Of Medical Research Methods for predicting responsiveness to treatment
US8969304B2 (en) * 2010-04-09 2015-03-03 The Institute For Cancer Research Compositions and methods for the prevention of cancer in high risk patients
CN104470949A (zh) 2012-05-15 2015-03-25 百时美施贵宝公司 通过破坏pd-1/pd-l1信号传输的免疫治疗

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100266617A1 (en) * 2007-06-18 2010-10-21 N.V. Organon Antibodies to human programmed death receptor pd-1
WO2014100079A1 (en) * 2012-12-21 2014-06-26 Merck Sharp & Dohme Corp. Antibodies that bind to human programmed death ligand 1 (pd-l1)

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Bremnes et al J Thora Oncol 6:209-17, 2011, abstract only. *
Mu et al (Med Oncol 28:682-688, 2011 *
Sun et al, Tissue Antigens 69:19-27, 2007 *

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10695426B2 (en) * 2014-08-25 2020-06-30 Pfizer Inc. Combination of a PD-1 antagonist and an ALK inhibitor for treating cancer
US10628658B2 (en) * 2014-11-10 2020-04-21 Ventana Medical Systems, Inc. Classifying nuclei in histology images
US11585817B2 (en) 2016-04-01 2023-02-21 Agilent Technologies, Inc. Scoring methods for anti-PD therapy eligibility and compositions for performing same
CN109311989A (zh) * 2016-04-14 2019-02-05 创新微技术公司 在癌症疗法的治疗决定中的pd-l1表达的应用方法
CN109963872A (zh) * 2016-09-20 2019-07-02 默克专利股份有限公司 诊断抗pd-l1抗体及其用途
WO2019241730A2 (en) 2018-06-15 2019-12-19 Flagship Pioneering Innovations V, Inc. Increasing immune activity through modulation of postcellular signaling factors
WO2020227159A2 (en) 2019-05-03 2020-11-12 Flagship Pioneering Innovations V, Inc. Methods of modulating immune activity
WO2021127217A1 (en) 2019-12-17 2021-06-24 Flagship Pioneering Innovations V, Inc. Combination anti-cancer therapies with inducers of iron-dependent cellular disassembly
WO2022006179A1 (en) 2020-06-29 2022-01-06 Flagship Pioneering Innovations V, Inc. Viruses engineered to promote thanotransmission and their use in treating cancer
CN112462072A (zh) * 2020-09-24 2021-03-09 浙江大学 一种tmub1蛋白在制备肿瘤免疫抑制分子检测剂中的应用
WO2022212784A1 (en) 2021-03-31 2022-10-06 Flagship Pioneering Innovations V, Inc. Thanotransmission polypeptides and their use in treating cancer
WO2023278641A1 (en) 2021-06-29 2023-01-05 Flagship Pioneering Innovations V, Inc. Immune cells engineered to promote thanotransmission and uses thereof
CN113933505A (zh) * 2021-12-15 2022-01-14 北京市肿瘤防治研究所 一组多维分析预测胃癌免疫治疗疗效的tiic指标及其应用
WO2024077191A1 (en) 2022-10-05 2024-04-11 Flagship Pioneering Innovations V, Inc. Nucleic acid molecules encoding trif and additionalpolypeptides and their use in treating cancer

Also Published As

Publication number Publication date
EP2981821B1 (de) 2018-12-19
US11193937B2 (en) 2021-12-07
EP2981821B2 (de) 2021-11-03
EP2981821A1 (de) 2016-02-10
US20180080938A1 (en) 2018-03-22
WO2014165422A1 (en) 2014-10-09
EP2981821A4 (de) 2016-10-26

Similar Documents

Publication Publication Date Title
US11193937B2 (en) Immunohistochemical assay for detecting expression of programmed death ligand 1 (PD-L1) in tumor tissue
US11932701B2 (en) Method for increasing the efficacy of cancer therapy by administering an anti-FOLR1 immunoconjugate
US10273308B2 (en) Methods of producing antibodies specific for p95
EP3254110B1 (de) Histochemischer test zur beurteilung der expression des liganden 1 für programmierten zelltod (pd-l1)
US9535074B2 (en) Immunoassay for soluble PD-L1
US20210373027A1 (en) Compositions and methods for assessing the risk of cancer occurrence
JP2020534540A (ja) Napi2b標的化療法に対する応答を予測するための組成物および方法
KR20110091476A (ko) 항―tmap/ckap2 항체를 포함하는 암의 예후 진단용 조성물
JP2024095796A (ja) 葉酸受容体1の検出用の抗体及びアッセイ
CN117836627A (zh) 从脱石蜡细胞开始的单细胞elisa用于检测感兴趣分子的用途
KR102680356B1 (ko) 엽산 수용체 1의 검출을 위한 항체 및 분석
WO2020198699A9 (en) Companion diagnostic assay for globo-h related cancer therapy

Legal Events

Date Code Title Description
AS Assignment

Owner name: MERCK SHARP & DOHME CORP., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DOLLED-FILHART, MARISA;EMANCIPATOR, KENNETH;PIERCE, ROBERT H.;AND OTHERS;SIGNING DATES FROM 20140326 TO 20140403;REEL/FRAME:038275/0597

Owner name: QUALTEK MOLECULAR LABORATORIES, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LYNCH, FRANK;REEL/FRAME:038275/0445

Effective date: 20140326

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION