NZ729207B2 - Neutralization of inhibitory pathways in lymphocytes - Google Patents

Neutralization of inhibitory pathways in lymphocytes Download PDF

Info

Publication number
NZ729207B2
NZ729207B2 NZ729207A NZ72920715A NZ729207B2 NZ 729207 B2 NZ729207 B2 NZ 729207B2 NZ 729207 A NZ729207 A NZ 729207A NZ 72920715 A NZ72920715 A NZ 72920715A NZ 729207 B2 NZ729207 B2 NZ 729207B2
Authority
NZ
New Zealand
Prior art keywords
antibody
nkg2a
cells
nkgza
tumor
Prior art date
Application number
NZ729207A
Other versions
NZ729207A (en
Inventor
Pascale Andre
Mathieu Blery
Carine Paturel
Nicolai Wagtmann
Original Assignee
Innate Pharma
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Innate Pharma filed Critical Innate Pharma
Priority claimed from PCT/EP2015/071069 external-priority patent/WO2016041945A1/en
Publication of NZ729207A publication Critical patent/NZ729207A/en
Publication of NZ729207B2 publication Critical patent/NZ729207B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Abstract

The present invention relates to use of a non-depleting anti-NKG2A antibody Z720 in the manufacture of a medicament for treating an HLA-E expressing cancer, wherein the medicament is to be administered with an anti-PD-L1 antibody, and wherein the anti-NKG2A antibody comprises HCDR1 of SYWMN, HCDR2 of RIDPYDSETHY, HCDR3 of GGYDFDVGTLYWFFDV, LCDR1 of RASENIYSYLA, LCDR2 of NAKTLAE, and LCDR3 of QHHYGTPRT. f RIDPYDSETHY, HCDR3 of GGYDFDVGTLYWFFDV, LCDR1 of RASENIYSYLA, LCDR2 of NAKTLAE, and LCDR3 of QHHYGTPRT.

Description

(12) Granted patent caon (19) NZ (11) 729207 (13) B2 (47) Publicaon date: 2021.12.24 (54) NEUTRALIZATION OF INHIBITORY PATHWAYS IN LYMPHOCYTES (51) Internaonal Patent Classificaon(s): A61K 39/395 C07K 16/28 A61P 35/00 A61K 39/00 (22) Filing date: (73) Owner(s): 2015.09.15 INNATE PHARMA (23) Complete specificaon filing date: (74) Contact: 9.15 DAVIES COLLISON CAVE PTY LTD (30) Internaonal Priority Data: (72) Inventor(s): US 62/050,948 2014.09.16 ANDRE, Pascale US 62/083,929 2014.11.25 BLERY, Mathieu US 62/093,141 2014.12.17 PATUREL, Carine WAGTMANN, NicolaÏ (86) Internaonal Applicaon No.: (87) aonal Publicaon number: WO/2016/041945 (57) ct: The t invenon s to use of a non-depleng an-NKG2A anbody Z720 in the manufacture of a medicament for treang an HLA-E expressing cancer, wherein the medicament is to be administered with an an-PD-L1 anbody, and wherein the an-NKG2A anbody comprises HCDR1 of SYWMN, HCDR2 of RIDPYDSETHY, HCDR3 of GGYDFDVGTLYWFFDV, LCDR1 of RASENIYSYLA, LCDR2 of NAKTLAE, and LCDR3 of QHHYGTPRT.
NZ 729207 B2 WO 41945 2015/071069 LIZATION OF INHIBITORY PATHWAYS IN LYMPHOCYTES CROSS-REFERENCE TO RELATED APPLICATIONS This application claims the benefit of US. Provisional Application Nos. 62/050,948, filed 16 September 2014; 62/083,929 filed 25 November 2014; and 62/093,141 filed 17 De— cember 2014; all of which are incorporated herein by reference in their entirety; ing any drawings.
REFERENCE TO SEQUENCE LISTING The present application is being filed along with a Sequence g in onic format. The Sequence Listing is provided as a file entitled “NKGZA—PD1_ST25”, created 15 September 2015, which is 38 KB in size. The information in the electronic format of the Se- quence Listing is incorporated herein by reference in its ty.
FIELD OF THE INVENTION This invention relates to the combined use of NKG2A-neutraliing agents and PD-1 neutralizing agents for the treatment of cancer.
BACKGROUND OF THE INVENTION NK cell activity is regulated by a complex mechanism that involves both activating and inhibitory signals. Several distinct NK-specific receptors have been identified that play an important role in the NK cell mediated ition and killing of HLA Class | deficient target cells. Natural xicity ors (NCR) refers to a class of activating receptor proteins, and the genes expressing them, that are specifically expressed in NK cells. Examples of NCRs include NKp30, NKp44, and NKp46 (see, 9.9., Lanier (2001) Nat Immunol 2:23-27, Pende et al. (1999) J Exp Med. 190:1505—1516, Cantoni et al. (1999) J Exp Med. 189:787— 796, Sivori et al (1997) J. Exp. Med. 186:1129-1136, Pessino et al. (1998) J Exp Med. 188(5):953-60; Mandelboim et al. (2001) Nature 409:1055-1060, the entire disclosures of which are herein orated by reference). These receptors are members of the Ig super- family, and their cross—linking, induced by specific mAbs, leads to a strong NK cell activation resulting in increased intracellular Ca” levels, triggering of cytotoxicity, and lymphokine re- lease, and an activation of NK cytotoxicity against many types of target cells. 2015/071069 CD94/NKGZA is an inhibitory receptor found on subsets of cytes.
CD94/NKG2A restricts cytokine release and cytotoxic ses of certain cytes to— wards cells expressing the CD94/NKG2A—Iigand HLA-E (see, e.g., W099/28748). HLA—E has also been found to be secreted in soluble form by certain tumor cells (Derre et al., J Immunol 2006;177:3100—7) and activated elial cells (Coupel et al., Blood 09:2806—14).
Antibodies that inhibit CD94/NKGZA signalling may increase the ne release and cyto- lytic activity of lymphocytes towards HLA-E positive target cells, such as responses of CD94/NKGZA—positive NK cells towards HLA—E sing tumor cells or virally infected cells. ore, therapeutic antibodies that inhibit CD94/NKGZA but that do not provoke the killing of CD94/NKGZA—expressing cells (i.e. non-depleting antibodies), may induce control of tumor—growth in cancer patients.
PD—1 is an inhibitory member of the CD28 family of receptors that also includes CD28, CTLA—4, ICOS and BTLA. PD-1 is expressed on activated B cells, T cells, and mye- loid cells Okazaki et al. (2002) Curr. Opin. Immunol. 14: 391779-82; Bennett et al. (2003) J Immunol 170:711—8). Two ligands for PD—l have been identified, PD— L1 and PD—L2, that have been shown to downregulate T cell activation upon binding to PD-1 (Freeman et al. (2000) J Exp Med 192:1027-34; Latchman et al. (2001) Nat Immunol 2:261-8; Carter et al. (2002) Eur J Immunol —43). PD—L1 is abundant in a variety of human cancers (Dong et al. (2002) Nat. Med. 8:787-9). The interaction between PD-1 and PD-L1 results in a decrease in tumor infiltrating lymphocytes, a decrease in T-cell receptor mediated proliferation, and immune evasion by the cancerous cells. Immune ssion can be reversed by inhibiting the local interaction of PD-l with PD-L1, and the effect is additive when the interaction of PD- 1 with PD-L2 is blocked as well.
PD-1 blockade has resulted in impressive anti-tumor ses in numerous clinical trials. However, not all patients respond to treatment with anti-tumor responses, and further- more some patients have cancers that relapse after treatment. Consequently, there is a need in the art for improved benefit to ts treated with inhibitors of the PD-1 axis.
SUMMARY OF THE INVENTION The present invention provides improved methods of enhancing an umor im- mune response through the combined neutralization of inhibitory receptors NKG2A and PD- 1, e.g. via the use of antibodies. While CD8T cells and NK cells (in the periphery) do not ex- press both NKGZA and PD-1, it has been found that tumor infiltrating lymphocytes that medi- ate elimination of tumor cells are capable of expressing both the inhibitory receptor PD-1 and the inhibitory receptor NKG2A. Additionally, ent with anti-PD1 can cause upregulation 2015/071069 of NKGZA ors on tumor infiltrating lymphocytes, such that NKGZA may be restricting the efficacy of agents that block the PD1 axis. Since these receptors can both restrict the cy— totoxic activities of tumor infiltrating lymphocytes, neutralization of the inhibitory activity of both these two receptors by antibodies enables PD1+ lymphocytes to effectively eliminate cancer cells. In one embodiment, the NKG2A+PD1+ lymphocytes are cytotoxic cytes, ally CD8+ T cells or NK cells.
Inhibition or neutralization the inhibitory activity of PD-1 can advantageously involve use of a polypeptide (e.g. an antibody, a polypeptide fused to an F0 domain, an immunoad- hesin, etc.) that prevents PD-L1-induced PD-1 signalling, e.g. by blocking the interaction with its natural ligand PD-L1 (and optionally further blocking the interaction between PD-1 and PD-L2. In one aspect the polypeptide is an antibody that binds PD-1 (an anti-PD—1 antibody); such dy may block the interaction between PD—1 and PD—L1 and/or between PD—1 and PD-L2. In another aspect the polypeptide is an antibody that binds PD-L1 (an anti-PD-L1 an- tibody) and blocks the interaction between PD-1 and PD-L1.
Accordingly, in one embodiment, provided is a method for treating or preventing a cancer in an individual, the method comprising administering to an individual: (a) a therapeu- tically active amount of a compound that inhibits a human NKGZA polypeptide, and (b) a therapeutically active amount of a compound that inhibits a human PD—1 polypeptide. In one embodiment, the cancer is a solid tumor. In one embodiment, the compound that inhibits a human NKGZA polypeptide is an antibody that neutralizes the inhibitory activity of NKGZA. In one embodiment, the compound that inhibits a human PD—1 polypeptide is an anti-PD-1 or anti-PDL-1 antibody that lizes the inhibitory activity of PD-1. The individual can be specified to be a human.
In one embodiment, ed is method of activating or iating the activity of a CD8+ tumor-infiltrating T cell in an individual, the method comprising administering to an in- dividual: (a) a therapeutically active amount of a compound that inhibits a human NKGZA polypeptide, and (b) a therapeutically active amount of a compound that inhibits a human PD—1 polypeptide. In one embodiment, ed is method of activating or potentiating the activity of a tumor-infiltrating NK cell in an individual, the method comprising stering to an individual: (a) a therapeutically active amount of a compound that inhibits a human NKG2A polypeptide, and (b) a therapeutically active amount of a compound that inhibits a human PD-1 polypeptide.
In one , provided is a treatment comprising administering a combination of an antibody that neutralizes the inhibitory activity of NKGZA, and dy that neutralizes the inhibitory activity of PD-1.
In one aspect provided is a composition comprising an antibody that inhibits a hu- man NKGZA polypeptide and an antibody that inhibits a human PD—1 polypeptide. In one as— pect, the composition is for use in the treatment or prevention of a cancer, optionally a solid tumor, optionally a haematological malignancy. in one embodiment, the anti—NKGZA dy is administered in an amount that re— sults in the neutralization of the inhibitory activity of human CD94/NKG2A in the human pa- tient (in vivo), e.g., an amount that results in the lization of the inhibitory ty of hu- man CD94/NKGZA on CD8 T cells and NK cells in the human patient. In one embodiment, the amount that results in the neutralization of the inhibitory ty of human CD94/NKG2A in the human patient is at least “IO-fold (e.g., 10-20 fold, 10-50 fold, 10-100 fold, 20-50 fold, -100 fold, 30-100 fold, 50-100 fold), ally at least 50-, 60-, 80- or 100-fold, the mini- mum concentration required to substantially saturate NKGZA receptors on the surface of NKG2A+ cells (e.g., in a binding assay where antibody is titrated on PBMC). In one embodi- ment, the anti-NKGZA antibody competes with HLA-E for binding to human NKGZA.
In one embodiment, the anti—NKG2A dy is stered for at least one ad— ministration cycle, the administration cycle comprising at least a first and second (and op- tionally a 3rd, 4th, 5th, 6th, 7th and/or 8th or further) administration of the anti-NKG2A antibody, wherein the anti—NKGZA antibody is administered in an amount effective to achieve a contin— uous (minimum) blood concentration of KGZA antibody of at least 10 ug/ml (or, option- ally at least 20, 30, 40 or 50 ug/mL) between the first and second (and optionally the further) administrations. Achieving or maintaining a specified continuous blood concentration means that the blood concentration does not drop substantially below the specified blood concentra- tion for the duration of the specified time period (e.g. between two administrations of anti- body, number of weeks), i.e. although the blood concentration can vary during the specified time period, the specified blood concentration represents a minimum or ”trough” concentra- tion. in one embodiment, the anti-NKGZA antibody is administered in an amount ive to achieve a peak blood concentration of about or at least about 50, 60, 70 or 80 ug/ml, op— tionally at least about 100 ug/ml, upon administration (e.g. within 1 or 2 days of administra- tion). in one embodiment, the anti—NKGZA antibody is administered in an amount effective to achieve a uous (minimum) blood concentration of anti-NKG2A dy of about or at least about 10, 20, 30, 40, 50, 60, 70 or 80 ug/ml, optionally at least about 100 ug/ml, for at least one week, or at least two weeks, following administration of the antibody.
In one embodiment, the anti-NKG2A antibody is administered in an amount effective WO 41945 to e a continuous (minimum) blood concentration of KGZA antibody of about or at least about 50, 60, 70 or 80 pg/ml, optionally at least about 100 pg/ml, between two suc— cessive administrations. In one embodiment, the first and second administrations are sepa- rated in time by about two weeks, optionally about one week.
The KGZA antibody can optionally be administered in an amount effective and according to a frequency that achieves a continuous (minimum) blood concentration as spec- ified for the entire duration of an administration cycle. in one embodiment, the anti-NKGZA antibody is administered in combination with antibody that neutralizes a human PD-1 polypeptide, for the treatment of a solid tumor in an individual, wherein the administration cycle comprising least two administrations of the anti- NKG2A antibody, n the anti-NKG2A antibody in administered in an amount effective to achieve a continuous um) concentration in an extravascular tissue (e.g. in the tumor environment) of at least 4 pg/mL, optionally at least 10 pg/mL between two successive ad- ministrations. Optionally, the anti-NKGZA antibody is administered in an amount effective to achieve a continuous (minimum) concentration in an extravascular tissue (e.g. in the tumor environment) of at least 4 pg/mL, optionally at least 10 ug/mL, for the entire duration of the administration cycle. In one embodiment, the KGZA antibody is administered in an amount effective to achieve a continuous (minimum) blood concentration of anti—NKG2A an— tibody of at least 40 pg/mL, ally at least 100 pg/mL, between two successive admin- istrations, or for the duration of the administration cycle. in one embodiment, the antibody that neutralizes a human PD-1 polypeptide is ad— ministered in an amount that s in the neutralization of the inhibitory activity of human PD-1 in the human patient (in vivo), e.g. an amount that results in the neutralization of the inhibitory activity of human PD-1 on CD8 T cells and NK cells in the human patient. In one aspect, the combination is stered (or is for stration) according to a particular clinical dosage regimen, notably at a particular dose amount and according to a specific dos- ing schedule.
In one aspect, an antibody that neutralizes NKGZA is a non—depleting antibody, e.g. an antibody that does not kill, ate, lyse or induce such killing, elimination or lysis, so as to negatively affect the number of NKG2A—expressing cells present in a sample or in a sub- ject. In one aspect an antibody that neutralizes PD—1 is a non—depleting antibody. A non— depleting antibody can, for example, lack an F0 domain or have an F0 domain with minimal or no binding to one or more ch receptors (e.g. CD16). Example include dies with nt regions from human lgG4 isotype antibodies, antibodies of any isotype (e.g. lgG1, lgGZ, IgG3) with constant s modified to reduce or abolish binding to one or more ch receptors (e.g. CD16).
In one embodiment the cancer is an advanced and/or refractory solid tumor. In one miting embodiment, the cancer (e.g., the ed refractory solid tumor) is selected from the group ting of non-small cell lung cancer (NSCLC), kidney cancer, pancreatic or esophagus adenocarcinoma, breast cancer, renal cell carcinoma (RCC), melanoma, colo— rectal , and ovarian cancer.
The compound that inhibits a NKGZA polypeptide (anti-NKGZA agent) is a com- pound that increases the y of an NKGZA—expressing NK and/or T cells to cause the death of the HLA—E-expressing cell. Optionally, the compound that inhibits a NKG2A poly- peptide is a polypeptide, ally an antibody (e.g. monoclonal antibody), that binds a NKG2A polypeptide.
In one embodiment, the anti—NKG2A agent reduces the inhibitory activity of NKG2A by blocking binding of its ligand, HLA—E, i.e., the KGZA agent interferes with the bind- ing of NKGZA by HLA—E. Antibody having the heavy chain of any of SEQ ID NOS: 4-8 and the light chain of SEQ ID NO: 9 is an example of such an antibody. In one ment, the anti-NKG2A agent reduces the inhibitory activity of NKGZA without ng binding of its lig- and, HLA—E, i.e., the anti-NKGZA agent is a non-competitive antagonist and does not inter- fere with the binding of NKG2A by HLA—E. The antibody having the heavy and light chain var— iable regions of SEQ ID N08: 10 and 11 respectively is an example of such an antibody.
In one embodiment, the anti-NKG2A agent is antibody which binds with a signifi- cantly higher affinity to NKGZA than to one or more activating NKGZ receptors. For example, in one embodiment, the agent is antibody which binds with a significantly higher affinity to NKGZA than to NKGZC. In an additional or alternative embodiment, the agent is antibody which binds with a significantly higher affinity to NKGZA than to NKGZE. In an additional or alternative embodiment, the agent is dy which binds with a significantly higher affinity to NKGZA than to NKG2H.
In one embodiment, the anti-NKGZA agent competes with the antibody having the heavy and light chains of SEQ ID NOS: 4—8 and 9 respectively, or the antibody having the heavy and light chain variable regions of SEQ ID NOS: 10 and 11 respectively, in g to CD94/NKG2A. The agent can be, e.g., a human or humanized anti—NKG2A antibody.
In one embodiment, the anti—NKGZA antibody is a humanized antibody having the heavy chain CDRs of any of the heavy chains of any of SEQ ID NOS: 4-8 and the light chain CDRs of the light chain of SEQ ID NO: 9 respectively. In one ment, the anti-NKG2A antibody is a humanized antibody having the heavy chain variable region of any of the heavy chains of any of SEQ ID NOS: 4-8 and the light chain variable region of the light chain of 2015/071069 SEQ ID NO: 9 respectively. Exemplary complementarity-determining region (CDR) residues or sequences and/or sites for amino acid substitutions in framework region (FR) of such hu— manized antibodies having ed properties such as, e.g., lower immunogenicity, im- proved antigen-binding or other functional properties, and/or improved physicochemical properties such as, e.g., better stability, are provided.
In certain optional s, patients can be identified for treatment with an anti- NKG2A agent and PD1-neutralizing agent by assessing the presence in a tumor sample (e.g. tumor tissue and/or tumor adjacent tissue) of ligands for NKGZA, optionally further a ligand of PD-1. In one embodiment of any of the therapeutic uses or cancer treatment or prevention methods herein, the treatment or prevention of a cancer in an individual comprises: a) determining the HLA—E polypeptide status of ant cells within the individual having a cancer, and b) upon a determination that HLA—E polypeptides are prominently expressed by (e.g. on the surface of) malignant cells (e.g. tumor cells), administering to the individual a com- pound that neutralizes the inhibitory activity of a human NKGZA polypeptide and an agent that inhibits a human PD-1 polypeptide.
In one embodiment of any of the therapeutic uses or cancer treatment or tion methods herein, the treatment or prevention of a cancer in an individual comprises: a) determining the HLA—E polypeptide status and PD-L1 ptide status of malig- nant cells (e.g. tumor cells) within the dual having a cancer, and b) upon a determination that HLA—E and PD-L1 polypeptides are prominently ex- pressed on the surface of malignant cells, administering to the individual a compound that neutralizes the inhibitory activity of a human NKGZA polypeptide and an agent that ts a human PD-1 polypeptide.
In one embodiment, a determination that a biological sample (e.g., a sample com- prising tumor cells, tumor tissue and/or tumor adjacent tissue) prominently expresses HLA—E nucleic acid or polypeptide indicates that the dual has a cancer that can be treated with an agent that inhibits NKGZA in combination with an agent that inhibits a human PD—1 poly— ln one ment of any of the s, ining the HLA—E polypeptide sta- tus or determining the level of sion in step (a) comprises determining the level of ex— pression of a HLA—E nucleic acid or polypeptide of malignant cells in a biological sample and comparing the level to a reference level (e.g. a value, weak or strong cell surface staining, etc.). The reference level may, for example, correspond to a healthy individual, to an individ- ual deriving no/low clinical benefit from treatment with an anti-NKGZA antibody (optionally in combination with an agent that ts a human PD-1 polypeptide), or to an individual deriv- ing substantial clinical benefit from treatment with an anti—NKGZA antibody (optionally in ation with an agent that inhibits a human PD-1 polypeptide). A determination that a biological sample expresses HLA—E nucleic acid or polypeptide at a level that is increased (e.g. a high value, strong surface ng, a level that corresponds to that of an individual deriving substantial clinical benefit from treatment with an anti-NKGZA antibody, a level that is higher than that corresponding to an individual ng no/low clinical t from treat- ment with an anti-NKGZA antibody, etc.) tes that the individual has a cancer that can be treated with an anti-NKGZA antibody in combination with an agent that inhibits a human PD-1 polypeptide, e.g. according to the treatment methods described herein. in one embodiment provided is a method for identifying NKG2A—inhibited PD expressing lymphocytes, the method comprising: a) determining the NKGZA and PD-1 polypeptide status of NK and/or CD8 T lym- phocytes in a biological sample, and b) wherein a determination that NKG2A and PD—1 polypeptides are expressed on the surface of a significant tion of the lymphocytes, indicates that the lymphocytes are NKG2A—inhibited PDexpressing lymphocytes. Optionally the lymphocytes are tumor infil- trating lymphocytes. Optionally the biological sample is a sample that comprises tumor tissue and/or tumor nt tissue. in one embodiment ed is a method for identifying an individual having a can- cer for whom treatment with an anti-NKGZA agent is suitable, the method comprising: a) determining the NKGZA and PD-1 polypeptide status of tumor infiltrating lymphocytes from the individual, and b) n a determination that NKGZA and PD-1 polypeptides are expressed on the surface of a significant proportion of tumor infiltrating lymphocytes from the individual, optionally TlLs of a pre-defined subset (e.g. CD8 T cells, NK , indicates that treatment with a compound that neutralizes the inhibitory activity of a human NKGZA polypeptide and an agent that inhibits a human PD—1 polypeptide is suitable for the individual.
In one embodiment provided is a method for treatment or prevention of a cancer in an individual comprises: a) determining the NKGZA and PD—1 polypeptide status of tumor infiltrating lympho— cytes from the individual, and b) upon a determination that NKG2A and PD—1 polypeptides are expressed on the surface of a icant proportion of tumor infiltrating lymphocytes, optionally TlLs of a pre- defined subset (e.g. CD8 T cells, NK cells), from the individual, administering to the individual a eutic regimen that comprises a compound that neutralizes the inhibitory activity of a human NKG2A polypeptide and an agent that inhibits a human PD—1 polypeptide.
In one embodiment, the tumor infiltrating lymphocytes are CD8 T cells. In one em- bodiment, the tumor infiltrating lymphocytes are NK cells. In one embodiment, at least 10, 15, , 25% of CD8 T cells are NKG2A+PD+. In one embodiment, at least 10%, 15%, 20% or % of CD8 T cells are NKG2A+PD-1+. In one ment, at least 20%, 25%, 30% or 35% of NK cells are PD-1+.
In other embodiments, pharmaceutical compositions and kits are provided, as well as methods for using them. In one embodiment, provided is a pharmaceutical composition comprising a compound that neutralizes the inhibitory activity of a human NKG2A polypep- tide and an agent that inhibits a human PD-1 polypeptide. In one embodiment, provided is a kit sing a compound that neutralizes the inhibitory activity of a human NKG2A poly— peptide and an agent that inhibits a human PD—1 polypeptide.
These aspects are more fully described in, and additional aspects, features, and advantages will be apparent from, the ption of the invention provided herein.
BRIEF DESCRIPTION OF THE DRAWINGS Figures 1A and 1B shows PD—L1+ Qa—1+ RMA—S Qa—1 Qdm 82m and A20 tumor cells are infiltrated by NK cells expressing NKG2A and CD8 T cells expressing NKG2A and/or PD-1.RMA-S Qa-1 Qdm B2m (top row) and A20 (bottom row) tumor bearing mice were sacrificed when tumor volumes were around 500 mm3. Tumor cells (Figure 1A) and tu- mor infiltrating lymphocytes—TIL- e 18) were analyzed by flow cytometry respectively for the sion of Qa-1 and PDL-1 for tumor cells and NKG2A/C/E and PD-1 for TIL.
MF|:Median of scence intensity.
Figure 2 shows distribution of NKG2A and PD-1 on NK and T cell subsets in mice.
Lymphocytes were taken from spleen, from tumor ng lymph nodes, and from within sol- id tumor masses. PD-1 expression was infrequent or absent among all cell subsets from spleen and lymph nodes, however among tumor infiltrating lymphocytes (TIL), all cells sub— sets had relatively high percentages of cells expressing PD—1. NKG2A on the other hand was found on NK cells but not on T cell s in spleen and lymph nodes, yet in the tumor was found on a significant tage of the TlLs, with a mean of more than 30% of NK cells and more than 19% of CD8 T cells double positive for NKG2A and PD-1.
Figures 3A and 3B show NKG2A and PD-1 expression in tumor bearing mice. RMA Rae1 (top row), MC38 (medium row) and RMA (bottom row) tumor bearing mice were sacri- ficed when their tumors reached respectively the volumes of 500, 2000 and 800 mm3. NK cells (Figure 3A) and CD8 T cells (Figure 38) were analyzed by flow cytometry in spleen, tu- mor draining lymph node (LN) and tumor for C/E and PD—1 expression.
Figure 4 shows treatment of mice with anti-PD-1 mAb increases the frequency of NKG2A sing TCD8 cells in M038 tumors. M038 tumor bearing mice were either treat- ed with 200 ug of rat lgGZa isotype control (IC) or anti—mouse PD—1 antibodies on days 11, 14 and 17 after cells engraftment. Mice were sacrificed on day 31 and CD8 T cells were characterized by flow cytometry in spleen, tumor draining lymph node (LN) and tumor.
Figure 5 shows median tumor volume over time in mice d with isotype control, anti-mouse NKG2A mAb (200 ug, iv), anti-mouse PD-L1 mAb (200 ug, ip) or anti- mNKG2A/mPDL-1 combination on days 11, 14 and 18. While anti-NKG2A yielded only a modest umor effect ed to isotype control in this model and anti-PD-L1 yielded a substantial anti—tumor effect but with tumor volume increasing toward day 28, the combined treatment with anti-NKG2A and anti-PD-L1 completely abolished tumor growth, with no sig- nificant growth in tumor volume observed at day 28.
DETAILED DESCRIPTION Definitions As used in the specification, "3" or "an" may mean one or more. As used in the claim(s), when used in ction with the word "comprising", the words "a" or "an" may mean one or more than one. As used herein "another" may mean at least a second or more.
Where "comprising" is used, this can optionally be replaced by "consisting ial- ly of" or by "consisting of".
NKG2A (OMIM 161555, the entire disclosure of which is herein incorporated by ref- erence) is a member of the NKGZ group of transcripts (Houchins, et al. (1991) J. Exp. Med. 173:1017—1020). NKG2A is encoded by 7 exons spanning 25 kb, showing some ential splicing. Together with CD94, NKGZA forms the heterodimeric inhibitory receptor CD94/NKG2A, found on the surface of subsets of NK cells, or/B T cells, v/6 T cells, and NKT cells. Similar to inhibitory KIR receptors, it possesses an ITIM in its cytoplasmic . As used herein, “NKG2A” refers to any variant, derivative, or isoform of the NKG2A gene or en- coded protein. Human NKGZA comprises 233 amino acids in 3 s, with a cytoplasmic domain comprising residues 1-70, a transmembrane region comprising residues 71-93, and an extracellular region comprising residues 94—233, of the ing sequence: MDNQGVIYSDLNLPPNPKRQQRKPKGNKSSILATEQEITYAELNLQKASQDFQGND- KTYHCKDLPSAPEKLIVGILGIICLILMASVVTIVVIPSTLIQRHNNSSLNTRTQKARHCGHCP EEWITYSNSCYYIGKERRTWEESLLACTSKNSSLLSIDNEEEMKFLSIISPSSWIGVFRNSS HHPWVTMNGLAFKHEIKDSDNAELNCAVLQVNRLKSAQCGSSIIYHCKHKL (SEQ ID NO:1).
NKGZC (OMIM 602891, the entire disclosure of which is herein incorporated by ref- erence) and NKG2E (OMIM 602892, the entire disclosure of which is herein incorporated by reference) are two other members of the NKGZ group of transcripts (Gilenke, et al. (1998) Immunogenetics 482163—173). The CD94/NKGZC and CD94/NKGZE receptors are activating receptors found on the surface of subsets of cytes such as NK cells and T-cells.
HLA—E (OMIM 143010, the entire disclosure of which is herein incorporated by ref- erence) is a nonclassical MHC molecule that is expressed on the cell surface and regulated by the binding of peptides, e.g such as fragments derived from the signal sequence of other MHC class I molecules. Soluble versions of HLA-E have also been identified. In on to its T—cell receptor binding properties, HLA—E binds s of natural killer (NK) cells, natural killer T-cells (NKT) and T cells (or/B and v/6), by binding specifically to KGZA, KGZB, and KGZC (see, e.g., Braud et al. (1998) Nature 391:795—799, the entire disclosure of which is herein incorporated by reference). e expression of HLA—E protects target cells from lysis by CD94/NKG2A+ NK, T, or NKT cell clones. As used herein, “HLA—E” refers to any variant, derivative, or isoform of the HLA-E gene or encoded protein.
In the context of the t invention, “NKGZA” or “CD94/NKGZA positive lympho— cyte” refers to cells of the lymphoid lineage (e.g. NK-, NKT- and T-cells) expressing CD94/NKG2A on the cell-surface, which can be detected by e.g. flow-cytometry using anti- bodies that specifically recognize a combined epitope on CD94 and NKGZA or and epitope on NKGZA alone. “NKGZA ve lymphocyte” also includes al cell lines of lymphoid origin (e.g. NKL, NK-92).
In the t of the present invention, “reduces the inhibitory activity of NKGZA”, “neutralizes NKGZA” or “neutralizes the inhibitory activity of NKGZA” refers to a process in which CD94/NKGZA is inhibited in its capacity to negatively affect intracellular processes leading to lymphocyte ses such as cytokine release and xic responses. This can be measured for example in a NK— or T—cell based xicity assay, in which the capacity of a therapeutic compound to stimulate killing of HLA—E positive cells by CD94/NKG2A positive lymphocytes is measured. In one embodiment, an antibody preparation causes at least a % augmentation in the cytotoxicity of a CD94/NKGZA—restricted cyte, optionally at least a 40% or 50% augmentation in lymphocyte cytotoxicity, optionally at least a 70% aug- mentation in NK cytotoxicity”, and referring to the cytotoxicity assays described. If an anti- NKGZA antibody reduces or blocks CD94/NKGZA interactions with HLA—E, it may increase the cytotoxicity of CD94/N KG2A—restricted lymphocytes. This can be evaluated, for example, in a standard 4-hour in vitro cytotoxicity assay using, e.g., NK cells that express CD94/NKG2A, and target cells that express HLA—E. Such NK cells do not efficiently kill tar— gets that s HLA—E because CD94/NKGZA recognizes HLA—E, leading to initiation and propagation of inhibitory signaling that prevents lymphocyte-mediated cytolysis. Such an in vitro cytotoxicity assay can be carried out by standard methods that are well known in the art, as described for e in Coligan et al., eds., Current Protocols in logy, Greene hing Assoc. and Wiley Interscience, N.Y., (1992, 1993). Chromium release and/or oth- er parameters to assess the ability of the antibody to stimulate lymphocytes to kill target cells such as P815, K562 cells, or riate tumor cells are also disclosed in Sivori et al., J.
Exp. Med. 1997;186:1129-1136; Vitale et al., J. Exp. Med. 1998; 187:2065-2072; Pessino et al. J. Exp. Med. 1998;188:953-960; Neri et al. Clin. Diag. Lab. Immun. 2001;8:1131-1135; Pende et al. J. Exp. Med. 1999;190:1505—1516, the entire disclosures of each of which are herein orated by reference. The target cells are labeled with 51Cr prior to addition of NK cells, and then the killing is estimated as proportional to the release of 51Cr from the cells to the medium, as a result of killing. The addition of an antibody that prevents CD94/NKG2A from binding to HLA—E results in prevention of the tion and ation of inhibitory sig- naling via CD94/NKG2A. Therefore, addition of such agents results in increases in lympho- cyte—mediated killing of the target cells. This step thereby identifies agents that prevent CD94/NKG2A—induced negative ing by, e.g., blocking ligand binding. In a particular 51Cr-release cytotoxicity assay, CD94/NKG2A—expressing NK effector-cells can kill HLA—E- negative LCL 721.221 target cells, but less well HLA—E-expressing LCL 721 .221-Cw3 control cells. In contrast, YTS effector-cells that lack CD94/NKG2A kill both cell—lines efficiently.
Thus, NK or cells kill less efficiently HLA—E+ LCL 721.221-CW3 cells due to HLA—E- d inhibitory signaling via CD94/NKG2A. When NK cells are pre-incubated with block- ing anti-CD94/NKG2A antibodies ing to the present invention in such a 51Cr—release cytotoxicity assay, HLA—E-expressing LCL 721.221-Cw3 cells are more efficiently killed, in an antibody-concentration-dependent fashion. The inhibitory activity (i.e. cytotoxicity enhancing potential) of an anti—NKG2A antibody can also be assessed in any of a number of other ways, e.g., by its effect on intracellular free calcium as described, e.g., in Sivori et al., J. Exp.
Med. 1997;186:1129-1136, the disclosure of which is herein orated by reference. . Ac- tivation of NK cell cytotoxicity can be assessed for example by measuring an increase in cy— tokine production (e.g. IFN-v tion) or xicity markers (e.g. CD107 or CD137 mobi- lization). In an exemplary protocol, IFN-y production from PBMC is assessed by cell surface and intracytoplasmic staining and analysis by flow cytometry after 4 days in culture. Briefly, Brefeldin A (Sigma Aldrich) is added at a final concentration of 5 ug/ml for the last 4 hours of culture. The cells are then incubated with anti-CD3 and anti-CD56 mAb prior to permeabiliza- tion (lntraPrepTM; Beckman Coulter) and staining with PE—anti—lFN—y or PE—lgG1 (Pharmingen). GM-CSF and IFN-y production from polyclonal activated NK cells are meas- ured in supernatants using ELISA (GM-CSF: DuoSet Elisa, R&D Systems, Minneapolis, MN, IFN—y: OptElA set, Pharmingen).
As used herein, the terms “PD-1” refers to the protein mmed Death 1 (PD-1) (also ed to as “Programmed Cell Death 1”), an inhibitory member of the CD28 family of receptors, that also es CD28, CTLA—4, ICOS and BTLA. The te human PD-1 sequence can be found under GenBank Accession No. , shown as follows: MQII VVWAVLQLGWRPGWF."LIBS PDRIPWNPPTFFPALLVVT3G )— NATFTCSFSNTSTS:TVNWYRMSRSWQTDKLAAFPI1..)RSQ .RGQDCR:TRVTQL PNGRD— FHVISVVRARRNDSGTYLCGAII SLARKAQLKL'SLRAL'LRVTL'RRAJL‘VRTAHP- SPSPRPAGQFQTLVVGVVGGLLGSLVLLVWVLAV. CS RAARGT.GARRTGQPLKEIDPSAV— PVFSVDYGI3LDFQWREKTPEPPV.RCVPTQTTYAT IIVFRSGVIGTSS .RARRG— SADGPRSAQPLRPE'DG {CSWPL (SEQ ID NO: 2).
“PD-1” also es any variant, derivative, or isoform of the PD-1 gene or encoded protein.
PD-1 is expressed on activated B cells, T cells, and myeloid cells Okazaki et al. (2002) Curr.
Opin. Immunol. 14: 391779-82; t et al. (2003) J Immunol 170:711-8). The initial mem— bers of the family, CD28 and ICOS, were discovered by onal effects on augmenting T cell proliferation ing the addition of monoclonal antibodies (Hutloff et al. (1999) Nature 397:263-266; Hansen et al. (1980) lmmunogenics 10:247-260). Two ligands for PD-1 have been identified, PD— L1 and PD—L2, that have been shown to downregulate T cell activation upon binding to PD-1 (Freeman et al. (2000) J Exp Med 192:1027-34; Latchman et al. (2001) Nat Immunol 2:261-8; Carter et al. (2002) EurJ Immunol 32:634-43). Both PD-L1 and PD-L2 are B7 homologs that bind to PD—1, but do not bind to other CD28 family members.
The complete human PD-L1 sequence can be found under UniProtKB/Swiss-Prot, identifier Q9NZQ7-1, shown as follows: MRIFAVFIFVI TYWHLLNAFT VTVPRDLYVV TIEC KFPVEKQ'.D'.
AAUIVYWLML 3K IIQhVHG L.DL<VQ{SS YRQRARLL<D AALQ IT DVKLQDAG VY'RCMISYGG ADYKRITVKV NAPYN<I QR ILWDPVTSE‘.
HLHTCQALGY RKAEVIWTSS DHQVLSG<TT TTNSKRLL<L :NVTSTLRI TTTNLIEYCT hRRHDPLLNH TALLVIPLLP LAHPP FRTH HVILGAIHLC LGVALTFIFR ILR<GRM__DVK (CGIQDT SK KQSDTiLLLT (SEQID NO: 3).
PD-L1 is abundant in a variety of human cancers (Dong et al. (2002) Nat. Med. 8:787-9). The interaction between PD-1 and PD-L1 results in a decrease in tumor infiltrating lymphocytes, a decrease in T-cell receptor ed proliferation, and immune evasion by the cancerous cells (Dong et al. (2003) J. Mol. Med. 81:281- 7; Blank et al. (2005) Cancer Immunol. Immunother. 54:307-314; Konishi et al. (2004) Clin. Cancer Res. 10:5094-100).
Immune suppression can be ed by ting the local interaction of PD—1 with PD—L1, and the effect is additive when the interaction of PD—1 with PD-L2 is blocked as well.
In the context of the present invention, “reduces the inhibitory activity of human PD- 1”, “neutralizes PD—1” or “neutralizes the inhibitory activity of human PD—1” refers to a pro— cess in which PD-1 is inhibited in its signal transduction capacity resulting from the interac- tion of PD-1 with one or more of its binding rs, such as PD-L1 or PD-L2. An agent that neutralizes the tory activity of PD-1 ses, blocks, ts, abrogates or interferes with signal transduction resulting from the interaction of PD-1 with one or more of its binding partners, such as PD-L1, PD-L2. Such an agent can thereby reduce the negative co- stimulatory signal mediated by or through cell e proteins expressed on T lymphocytes, so as to enhance T—cell effector functions such as proliferation, cytokine tion and/or cytotoxicity.
Whenever within this whole specification "treatment of " or the like is men- tioned with reference to anti—NKG2A and anti—PD—1 or anti—PD—L1 binding agent (e.g. anti— body), are comprised: (a) method of treatment of cancer, said method comprising the step of administering (for at least one treatment) an NKG2A and anti-PD-1 or anti-PD-L1 binding agent, (e.g., together or each separately in a pharmaceutically acceptable carrier material) to an individual, a mammal, ally a human, in need of such treatment, in a dose that al- lows for the treatment of , (a therapeutically effective amount), optionally in a dose (amount) as specified herein; (b) the use of an anti-NKGZA and anti-PD-1 or anti-PD-L1 binding agent for the treatment of , or an anti-NKGZA binding agent, for use in said treatment (especially in a human); (c) the use of an KGZA and anti-PD-1 or anti-PD-L1 binding agent for the manufacture of a pharmaceutical preparation for the treatment of can- cer, a method of using an anti-NKGZA and anti—PD-1 or anti-PD—L1 binding agent for the manufacture of a pharmaceutical preparation for the treatment of cancer, comprising admix- ing an anti-NKGZA and anti-PD-1 or anti-PD-L1 binding agent with a pharmaceutically ac- ceptable carrier, or a pharmaceutical preparation comprising an effective dose of an anti— NKG2A and anti-PD-1 or anti-PD-L1 binding agent that is appropriate for the treatment of cancer; or (d) any ation of a), b), and c), in accordance with the subject matter allow- able for patenting in a country where this application is filed.
The term "biopsy" as used herein is defined as removal of a tissue for the purpose of examination, such as to establish diagnosis. Examples of types of biopsies include by ap- plication of suction, such as through a needle attached to a e; by instrumental removal of a nt of tissue; by removal with appropriate instruments h an endoscope; by 2015/071069 surgical excision, such as of the whole lesion; and the like.
The term ody,” as used herein, refers to polyclonal and onal antibodies.
Depending on the type of constant domain in the heavy chains, antibodies are assigned to one of five major classes: lgA, lgD, lgE, lgG, and lgM. l of these are further divided into subclasses or isotypes, such as lgG1, lgG2, lgG3, lgG4, and the like. An exemplary im— munoglobulin (antibody) structural unit comprises a er. Each er is composed of two cal pairs of polypeptide chains, each pair having one “light” (about 25 kDa) and one ” chain (about 50-70 kDa). The N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids that is primarily responsible for antigen recognition.
The terms variable light chain (VL) and variable heavy chain (VH) refer to these light and heavy chains respectively. The heavy-chain constant domains that correspond to the differ— ent classes of immunoglobulins are termed “alpha,” “delta,” “epsilon,” “gamma” and “mu,” re— spectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known. lgG are the exemplary classes of antibodies employed herein because they are the most common dies in the physiological situation and be— cause they are most easily made in a tory setting. Optionally the antibody is a mono- clonal antibody. Particular examples of dies are humanized, chimeric, human, or oth- erwise—human—suitable antibodies. “Antibodies” also includes any fragment or derivative of any of the herein described antibodies.
The term “specifically binds to” means that an antibody can bind preferably in a competitive binding assay to the binding partner, e.g. NKGZA, PD-1, PD—Ll, as assessed using either recombinant forms of the proteins, es therein, or native proteins present on the surface of isolated target cells. Competitive g assays and other methods for de- termining specific binding are well known in the art. For example binding can be detected via radiolabels, physical methods such as mass spectrometry, or direct or indirect fluorescent labels detected using, e.g., uorometric analysis (e.g. FACScan). Binding above the amount seen with a control, non-specific agent indicates that the agent binds to the target.
An agent that ically binds NKGZA may bind NKG2A alone or NKG2A as a dimer with CD94.
When an antibody is said to “compete with” a particular monoclonal antibody, it means that the antibody competes with the monoclonal antibody in a binding assay using either recombinant molecules (e.g., NKGZA, PD-1, PD-L1) or surface expressed mole- cules(e.g., NKG2A, PD-1, PD-L1). For example, if a test antibody reduces the binding of an antibody having a heavy chain of any of SEQ ID NO: 4-8 and a light chain of SEQ ID NO: 9 to a NKGZA polypeptide or NKG2A—expressing cell in a binding assay, the antibody is said to 2015/071069 “compete” respectively with such dy.
The term ity”, as used herein, means the strength of the binding of an antibody to an epitope. The affinity of an antibody is given by the iation constant Kd, defined as [Ab] x [Ag] / [Ab-Ag], where [Ab-Ag] is the molar concentration of the antibody-antigen com- plex, [Ab] is the molar concentration of the unbound antibody and [Ag] is the molar concen— tration of the unbound antigen. The affinity constant Ka is defined by 1/Kd. Methods for de- termining the affinity of mAbs can be found in Harlow, et al., Antibodies: A Laboratory Manu- al, Cold Spring Harbor tory Press, Cold Spring Harbor, N.Y., 1988), Coligan et al., eds., Current Protocols in Immunology, Greene Publishing Assoc. and Wiley lnterscience, N.Y., (1992, 1993), and Muller, Meth. Enzymol. 92:589-601 , which references are en- tirely incorporated herein by reference. One rd method well known in the art for de- termining the affinity of mAbs is the use of surface plasmon resonance (SPR) screening (such as by analysis with a BlAcoreT'V' SPR analytical device).
Within the context herein a “determinant” designates a site of interaction or binding on a polypeptide.
The term “epitope” refers to an antigenic determinant, and is the area or region on an n to which an antibody binds. A protein epitope may comprise amino acid residues directly involved in the g as well as amino acid residues which are effectively blocked by the specific antigen binding antibody or peptide, i.e., amino acid residues within the "foot- print" of the antibody. It is the simplest form or smallest structural area on a complex antigen le that can combine with e.g., an antibody or a receptor. Epitopes can be linear or conformationallstructural. The term r epitope” is defined as an epitope composed of amino acid residues that are contiguous on the linear sequence of amino acids (primary structure). The term “conformational or structural e” is defined as an epitope composed of amino acid residues that are not all contiguous and thus represent separated parts of the linear ce of amino acids that are brought into proximity to one r by folding of the le (secondary, tertiary and/or quaternary structures). A conformational epitope is dependent on the 3—dimensional structure. The term ‘conformational’ is therefore often used interchangeably with ‘structural’.
The term "agent" is used herein to denote a chemical compound, a mixture of chem- ical compounds, a biological macromolecule, or an extract made from biological materials.
The term "therapeutic agent" refers to an agent that has biological activity.
For the purposes herein, a “humanized” or “human” antibody refers to an antibody in which the constant and variable framework region of one or more human immunoglobulins is fused with the binding , e.g. the CDR, of an animal immunoglobulin. Such antibodies are designed to maintain the binding specificity of the non—human antibody from which the binding regions are derived, but to avoid an immune reaction against the non—human anti— body. Such antibodies can be obtained from enic mice or other animals that have been “engineered” to produce specific human dies in response to antigenic challenge (see, e.g., Green et al. (1994) Nature Genet 7:13; Lonberg et al. (1994) Nature 368:856; Taylor et al. (1994) Int Immun 6:579, the entire teachings of which are herein incorporated by refer- ence). A fully human antibody also can be constructed by genetic or chromosomal transfec— tion methods, as well as phage display technology, all of which are known in the art (see, e.g., McCafferty et al. (1990) Nature 348:552—553). Human antibodies may also be generat- ed by in vitro activated B cells (see, e.g., US. Pat. Nos. 5,567,610 and 5,229,275, which are incorporated in their entirety by reference).
A “chimeric antibody” is an antibody molecule in which (a) the constant region, or a portion thereof, is d, replaced or exchanged so that the antigen binding site (variable region) is linked to a nt region of a different or altered class, effector function and/or species, or an entirely different molecule which confers new properties to the chimeric anti— body, e.g., an , toxin, hormone, growth factor, drug, etc.; or (b) the variable region, or a portion thereof, is altered, replaced or exchanged with a variable region having a different or altered antigen specificity.
The terms "Fc domain," "Fc portion," and "Fc region" refer to a C-terminal fragment of an antibody heavy chain, e.g., from about amino acid (aa) 230 to about aa 450 of human y (gamma) heavy chain or its counterpart sequence in other types of antibody heavy chains (e.g., or, 6, e and u for human dies), or a naturally occurring allotype thereof. Unless ise specified, the commonly accepted Kabat amino acid numbering for immunoglobu- lins is used hout this disclosure (see Kabat et al. (1991 ) Sequences of Protein of Im- munological st, 5th ed., United States Public Health Service, National Institute of Health, Bethesda, MD).
The terms “isolated”, “purified” or "biologically pure” refer to material that is n- tially or essentially free from ents which normally accompany it as found in its native state. Purity and homogeneity are lly determined using analytical chemistry ques such as polyacrylamide gel ophoresis or high performance liquid chromatography. A protein that is the predominant species present in a preparation is substantially purified.
The terms “polypeptide,” “peptide” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues. The terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally ing amino acid polymer.
The term “recombinant” when used with reference, e.g., to a cell, or nucleic acid, n, or , indicates that the cell, nucleic acid, protein or vector, has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified. Thus, for example, recom— binant cells express genes that are not found within the native (nonrecombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all.
Within the context herein, the term antibody that ” a polypeptide or epitope designates an antibody that binds said determinant with icity and/or ty.
The term “identity” or “identical”, when used in a relationship between the sequenc- es of two or more polypeptides, refers to the degree of sequence relatedness between poly— peptides, as determined by the number of matches between strings of two or more amino acid residues. "Identity" measures the percent of identical matches between the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer program (i.e., "algorithms"). Identity of related polypeptides can be readily calculated by known methods. Such methods include, but are not limited to, those described in Computational lar Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome ts, Smith, D. W., ed., ic Press, New York, 1993; Computer Analysis of Sequence Data, Part 1, Griffin, A. M., and Griffin, H.
G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; Sequence is Primer, ov, M. and Devereux, J., eds., M. Stockton Press, New York, 1991; and Carillo et al., SIAM J. Applied Math. 48, 1073 (1988).
Methods for determining identity are designed to give the largest match between the ces tested. Methods of determining identity are described in ly available com- puter programs. Computer program methods for determining identity between two sequenc- es include the GCG program package, including GAP (Devereux et al., Nucl. Acid. Res. 12, 387 (1984); Genetics Computer Group, University of Wisconsin, Madison, Wis.), , BLASTN, and FASTA (Altschul et al., J. Mol. Biol. 215, 403-410 (1990)). The BLASTX pro- gram is publicly available from the National Center for Biotechnology Information (NCBI) and other sources (BLAST , Altschul et al. NCB/NLM/NIH Bethesda, Md. 20894; Altschul et al., supra). The nown Smith Waterman algorithm may also be used to determine identity.
NKG2A-neutra/izing therapeutic agents The anti—NKGZA agent binds an extra—cellular portion of human CD94/NKGZA re— ceptor and reduces the inhibitory activity of human CD94/NKG2A receptor expressed on the surface of a KG2A positive lymphocyte. In one ment the agent competes with HLA—E in binding to KGZA, i.e. the agent blocks the interaction between CD94/NKG2A and its ligand HLA—E. In another embodiment the agent does not compete with HLA—E in binding to CD94/NKG2A; i.e. the agent is capable of binding CD94/NKG2A simul- taneously with HLA—E. The antibody may bind a combined epitope on CD94 and NKGZA or and epitope on NKG2A alone.
In one aspect the anti-NKGZA agent is an antibody selected from a fully human an- tibody, a humanized dy, and a chimeric antibody. In one aspect, the agent comprises a constant domain d from a human lgG1, lgG2, lgG3 or lgG4 antibody.ln one aspect, the agent is a fragment of an antibody selected from lgA, an lgD, an lgG, an lgE and an lgM an- tibody. In one , the agent is an antibody fragment ed from a Fab fragment, a Fab' fragment, a Fab'—SH fragment, a F(ab)2 fragment, a F(ab')2 fragment, an Fv fragment, a Heavy chain lg (a llama or camel lg), a VHH nt, a single domain FV, and a single-chain dy fragment.ln one aspect, the agent is a synthetic or semisynthetic antibody-derived le selected from a scFV, a dsFV, a minibody, a diabody, a triabody, a kappa body, an lgNAR; and a multispecific antibody.
Optionally, the anti-NKGZA antibodies do not demonstrate substantial specific bind- ing to Fey receptors, e.g. CD16. Such antibodies may comprise nt regions of various heavy chains that are known not to bind Fc receptors. One such example is a human lgG4 nt region. In one embodiment, the lgG4 antibody comprises a modification to prevent the ion of half antibodies (fab arm exchange) in vivo, e.g., the antibody comprises an lgG4 heavy chain comprising a serine to proline mutation in residue 241, corresponding to position 228 according to the EU-index (Kabat et a/., “Sequences of proteins of immunologi- cal interest”, 5th ed., NIH, Bethesda, ML, 1991). Such modified lgG4 antibodies will remain intact in vivo and maintain a bivalent (high affinity) binding to NKGZA, as opposed to native lgG4 that will undergo fab arm exchange in vivo such that they bind to NKG2A in lent manner which can alter binding affinity. Alternatively, antibody fragments that do not com- prise nt regions, such as Fab or F(ab’)2 fragments, can be used to avoid Fc receptor binding. Fc receptor binding can be assessed according to methods known in the art, including for example testing binding of an antibody to F0 receptor protein in a BIACORE assay.
Also, any human antibody type (e.g. lgG1, lgGZ, |gG3 or lgG4) can be used in which the Fc portion is modified to minimize or eliminate binding to Fc receptors (see, e.g., W003101485, the disclosure of which is herein incorporated by reference). Assays such as, e.g., cell based assays, to assess Fc receptor binding are well known in the art, and are described in, e.g., WOO3101485.
The present invention thus concerns antibodies or other agents binding to NKGZA.
In one aspect, the antibody binds to NKGZA with a KD at least 100—fold lower than to human NKGZC and/or NKGZE.
In one aspect of the invention, the agent s CD94/NKGZA—mediated inhibition of a CD94/NKGZA—expressing lymphocyte by interfering with KGZA signalling by, e.g., interfering with the binding of HLA—E by NKGZA, preventing or inducing conformational changes in the CD94/NKG2A receptor, and/or affecting dimerization and/or clustering of the CD94/NKGZA receptor.
In one aspect of the invention, the agent binds to an extracellular portion of NKG2A with a KD at least 100 fold lower than to NKG2C. In a further preferred aspect, the agent binds to an extracellular portion of NKG2A with a KD at least 150, 200, 300, 400, or 10,000 fold lower than to NKG2C. In another aspect of the invention, the agent binds to an extracel— lular portion of NKGZA with a KD at least 100 fold lower than to NKG2C, NKGZE and/or NKG2H molecules. In a further preferred aspect, the agent binds to an extracellular portion of NKG2A with a KD at least 150, 200, 300, 400, or 10,000 fold lower than to NKG2C, NKGZC and/or NKGZH molecules. This can be measured, for ce, in BiaCore experi- ments, in which the capacity of agents to bind the extracellular portion of immobilized CD94/NKGZA (e.g. purified from CD94/NKGZ expressing cells, or produced in a bio-system) is measured and ed to the g of agents to similarly produced CD94/NKG2C and/or other CD94/NKG2 variants in the same assay. Alternatively, the binding of agents to cells that either naturally express, or over-express (e.g. after transient or stable transfection), CD94/NKG2A can be measured and compared to binding of cells sing CD94/NKG2C and/or other CD94/NKG2 variants. Anti-NKG2A antibodies may optionally bind NKGZB, which is an NKGZA splice variant forming an inhibitory receptor together with CD94. In one embodiment, affinity can be ed using the methods sed in U.S. Patent No 8,206,709, for e by ing binding to covalently immobilized CD94-Fc fu- sion protein by Biacore as shown in Example 8 of U.S. Patent No 8,206,709, the disclosure of which is incorporate herein by reference.
The anti-NKGZA antibody can be a humanized dy, for example comprising a VH human acceptor framework from a human acceptor sequence selected from, e.g., VH1_18, VH5_a, VH5_51, VH1_f, and VH1_46, and a JH6 J—segment, or other human ne VH framework sequences known in the art. The VL region human acceptor se- quence may be, e.g., VK|_02/JK4.
In one embodiment, the antibody is a humanized dy based on dy 2270.
Different zed 2270VH chains are shown in SEQ ID NOS: 4-8 (variable region domain amino acid underlined). HumZZ70VH6 (SEQ ID NO: 4) is based on VH5_51; HumZZ70VH1 (SEQ ID NO: 5) is based on VH1_18; 0VH5 (SEQ ID NO: 6) is based on VH5_a; hum2270VH7 (SEQ ID NO: 7) is based on VH1_f; and humZZ70VH8 (SEQ ID NO: 8) is based on VH1_46; all with a JH6 J-segment. Each of these antibodies retains high affinity binding to NKGZA, with low likelihood of a host immune response against the antibody as the 6 C-terminal amino acid residues of the Kabat CDR-HZ of each of the humanized constructs are identical to the human acceptor framework. Using the alignment program VectorNTl, the following sequence identities between humZZ70VH1 and 0VH5, —6, —7, and —8 were obtained: 78,2% (VH1 vs. VH5), 79,0% (VH1 vs. VH6), 88,7% (VH1 vs. VH7), and 96,0% (VH1 vs. VH8).
In one aspect, the agent comprises (i) a heavy chain le region of any of SEQ ID NOS: 4-8, or an amino acid sequence at least 50%, 60%, 70%, 80%, 90%, 95%, 98% or 99% identical thereto, and (ii) a light chain variable region of SEQ ID NO: 9, or an amino acid sequence at least 50%, 60%, 70%, 80%, 90%, 95%, 98% or 99% identical thereto. In one aspect, the agent comprises (i) a heavy chain comprising the amino acid sequence of any of SEQ ID NOS: 4-8, or an amino acid sequence at least 50%, 60%, 70%, 80%, 90%, 95%, 98% or 99% identical thereto, and (ii) a light chain comprising the amino acid sequence of SEQ ID NO: 9, or an amino acid sequence at least 50%, 60%, 70%, 80%, 90%, 95%, 98% or 99% identical thereto. The dy having the heavy chain of any of SEQ ID NOS: 4-8 and a light chain of SEQ ID NO: 9 neutralizes the inhibitory activity of NKGZA, but does not sub- stantially bind the activating receptors NKGZC, NKGE or NKG2H. This antibody furthermore competes with HLA—E for binding to NKGZA on the surface of a cell. In one aspect, the agent comprises HCDR1, HCDR2 and/or HCDR3 sequences d from the heavy chain having the amino acid sequence of any of SEQ ID NO: 4—8. In one aspect of the invention, the agent comprises LCDR1, LCDR2 and/or LCDR3 sequences derived from the light chain having the amino acid sequence of SEQ ID NO: 9.
Heavy Chains VH6: EVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWMNWVRQMPGKGLEWMGRIDPYD- SETHYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARGGYDFDVGTLY— WFFDVWGQGTTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGV HTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKP- SNTKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMIS— RTPEVTCWVDVSQEDPEVQFNWWDGVEVHNAK- QFNSTYRWSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEK— TISKAKGQPREPQWTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPEN- NYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK (SEQ ID NO: 4) VH1: QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYWMNWVRQAPGQGLEWMGRIDPYDSETHYA— QKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCARGGYDFDVGTLYWFFDVWGQGTTVTVS SASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSG LYS— LSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFPPKPKD TLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDW LNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAV EWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLS LSLGK (SEQ ID NO: 5) VH5: EVQLVQSGAEVKKPGESLRISCKGSGYSFTSYWMNWVRQMPGKGLEWMGRIDPYD- SETHYSPSFQGHVTISADKSISTAYLQWSSLKASDTAMYYCARGGYDFDVGTLY- WFFDVWGQGTTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGV HTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKP- SNTKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMIS- CWVDVSQEDPEVQFNWWDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDW LNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAV EWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLS LSLGK (SEQ ID NO: 6) VH7: EVQLVQSGAEVKKPGATVKISCKVSGYTFTSYWMNWVQQAPGKGLEWMGRIDPYDSETHY RVTITADTSTDTAYMELSSLRSEDTAVYYCATGGYDFDVGTLY— WFFDVWGQGTTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGV HTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKP- SNTKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMIS- RTPEVTCWVDVSQEDPEVQFNWWDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDW LNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAV EWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLS LSLGK (SEQ ID NO: 7) VH8: QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYWMNWVRQAPGQGLEWMGRIDPYDSETHY AQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARGGYDFDVGTLYWFFDVWGQGTTVTVS SASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHT- FPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPE- FLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWWDGVEVHNAK- TKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEK— TISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPEN- NYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK (SEQ ID NO: Light chain SPSSLSASVGDRVTITCRASENIYSYLAWYQQKPGKAPKLLIYNAKTLAEGVPSRFSGS GSGTDFTLTISSLQPEDFATYYCQHHYGTPRTFGGGTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVC LLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQG LSSPVTKSFNRGEC (SEQ ID NO: 9) In one aspect, the anti-NKGZA antibody is an antibody comprising a CDR—H1 corre- sponding to residues 31-35 of SEQ ID NOS: 4-8, a CDR-H2 corresponding to residues 50-60 (optionally 50-66 when including amino acids of human origin) of SEQ ID NOS: 4-8, and a CDR-H3 ponding to residues 99-114 (95-102 according to Kabat) of SEQ ID NOS: 4-8.
In one embodiment, the CDR-H2 corresponding to es 50-66 of SEQ ID NOS: 4-8. Op- tionally, a CDR may comprise one, two, three, four, or more amino acid substitutions.
In one aspect, the KGZA antibody is an antibody comprising a CDR-L1 corre- sponding to residues 24—34 of SEQ ID NO: 9, a CDR—L2 corresponding to residues 50—56 of SEQ ID NO: 9, and an CDR-L3 corresponding to residues 89-97 of SEQ ID NO: 9. Optional- ly, a CDR may comprise one, two, three, four, or more amino acid tutions.
In one aspect, the anti—NKGZA antibody is an antibody comprising a CDR—H1 corre— sponding to residues 31—35 of SEQ ID NOS: 4-8, a CDR-H2 corresponding to residues 50-60 (optionally 50-66) of SEQ ID NOS: 4-8, and a CDR-H3 corresponding to residues 99-114 (95—102 according to Kabat) of SEQ ID NOS: 4—8, a CDR—L1 corresponding to residues 24— 34 of SEQ ID NO: 9, a CDR-L2 ponding to residues 50-56 of SEQ ID NO: 9, and an CDR-L3 corresponding to residues 89-97 of SEQ ID NO: 9.
In one aspect, the agent comprises HCDR1, HCDR2 and/or HCDR3 sequences de- rived from the VH having the amino acid sequence of SEQ ID NO: 10. In one aspect of the invention, the agent comprises , LCDR2 and/or LCDR3 sequences derived from the VL having the amino acid sequence of SEQ ID NO: 11. In one aspect, the agent comprises HCDR1, HCDR2 and/or HCDR3 sequences derived from the VH having the amino acid se— quence of SEQ ID NO: 10, and LCDR1, LCDR2 and/or LCDR3 sequences derived from the VL having the amino acid sequence of SEQ ID NO: 11. The antibody having the heavy chain of SEQ ID NO: 10 and a light chain of SEQ ID NO: 11 neutralizes the inhibitory activity of NKGZA, and also binds the activating receptors NKGZC, NKGZE or NKGZH. The antibody does not competes with HLA—E for binding to NKGZA on the surface of a cell (i.e. it is a non- competitive antagonist of NKGZA).
EVQLVESGGGLVKPGGSLKLSCAASGFTFSSYAMSWVRQSPEKRLEWVAEISSGGSYTYY PDTVTGRFTISRDNAKNTLYLEISSLRSEDTAMYYCTRHGDYPRFFDVWGAGTTVTVSS (SEQ ID NO: 10) QIVLTQSPALMSASPGEKVTMTCSASSSVSYIYWYQQKPRSSPKPWIYLTSNLASGVPAR FSGSGSGTSYSLTISSMEAEDAATYYCQQWSGNPYTFGGGTKLEIKR (SEQ ID NO:11) In one aspect, the agent comprises amino acid residues 31-35, 50-60, 62, 64, 66, and 99-108 of the le-heavy (VH) domain (SEQ ID NO: 10) and amino acid residues 24- 33, 49—55, and 88-96 of the variable-light (VL) domain (SEQ ID NO: 11), ally with one, two, three, four, or more amino acid substitutions.
In one aspect, the agent is a fully human antibody which has been raised against the CD94/NKGZA epitope to which any of the aforementioned antibodies bind.
It will be appreciated that, while the aforementioned antibodies can be used, other antibodies can recognize and be raised against any part of the NKGZA ptide so long as the dy causes the neutralization of the tory activity of NKGZA. For example, any fragment of NKG2A, preferably but not exclusively human NKG2A, or any ation of NKG2A fragments, can be used as immunogens to raise antibodies, and the antibodies can recognize epitopes at any location within the NKG2A polypeptide, so long as they can do so on NKGZA expressing NK cells as described herein. Optionally, the epitope is the epitope specifically recognized by antibody having the heavy chain of SEQ ID NOS: 4—8 and the light chain of SEQ ID NO: 9.
In one aspect, the agent es with humZZ7O antibody disclosed in US. Patent No 8,206,709 (the disclosure of which is incorporated herein by nce) in binding to the extra—cellular portion of human CD94/NKG2A receptor. Competitive binding can be meas- ured, for instance, in BiaCore ments, in which the capacity of agents is measured, for binding the extracellular portion of immobilized CD94/NKG2A or (e.g. purified from CD94/NKG2 expressing cells, or produced in a bio-system) saturated with humZZ70. Alter- natively, the binding of agents to cells is ed that either naturally express, or over- express (e.g. after transient or stable transfection), CD94/NKG2A receptor, and which have been cubated with saturating doses of Z270. In one embodiment, competitive binding can be measured using the methods disclosed in U.S. Patent No 8,206,709, for example by assessing binding to Ba/F3—CD94—NKG2A cells by flow cytometry as shown in Example 15 of U.S. Patent No 8,206,709, the disclosure of which is incorporate herein by reference.
PD-1 lizing therapeutic agents There are tly at least six agents blocking the D-L1 pathway that are ed or in clinical evaluation. One agent is EMS-936558 (Nivolumab/ONO-4538, Bristol- Myers Squibb; formerly MDX—1106). Nivolumab, (Trade name Opdivo®) is an FDA-approved fully human lgG4 anti—PD—L1 mAb that inhibits the binding of the PD—L1 ligand to both PD—1 and CD80 and is described as antibody 5C4 in , the disclosure of which is incorporated herein by reference. For melanoma patients, the most significant OR was ob- served at a dose of 3 mg/kg, while for other cancer types it was at 10 mg/kg. Nivolumab is generally dosed at 10 mg/kg every 3 weeks until cancer progression.
MK-3475 (human lgG4 anti—PD1 mAb from Merck), also referred to as iambroli- zumab or lizumab (Trade name Keytruda®) has been approved by the FDA for the treatment of melanoma and is being tested in other cancers. Pembrolizumab was tested at 2 mg/kg or 10 mg/kg every 2 or 3 weeks until disease progression. DNA constructs encoding the variable regions of the heavy and light chains of the humanized antibodies | have been deposited with the American Type Culture Collection Patent Depository (10801 Univer- sity Blvd., Manassas, VA). The plasmid containing the DNA ng the heavy chain of l 1 was deposited on June 9, 2008 and identified as 081469_SPD—H and the plasmid containing the DNA encoding the light chain of h409Al 1 was deposited on June 9, 2008 and identified as 0801470_SPD-L-l 1. 5, also known as Merck 37450r SCH-900475, is also described in WO2009/114335.
MPDL3280A/RG7446 (anti—PD—L1 from Roche/Genentech) is a human anti—PD—L1 mAb that contains an engineered Fc domain designed to optimize cy and safety by min- imizing FC’yR binding and consequential antibody-dependent cellular cytotoxicity (ADCC).
Doses of S1, 10, 15, and 25 mg/kg MPDL3280A were administered every 3 weeks for up to 1 year. In phase 3 trial, MPDL3280A is administered at 1200 mg by intravenous infusion every three weeks in NSCLC.
AMP-224 (Amplimmune and GSK) is an immunoadhesin comprising a PD-L2 extra- cellular domain fused to an F0 domain. Other es of agents that neutralize PD-1 may include an dy that binds PD-L2 (an anti-PD-L2 antibody) and blocks the interaction be- tween PD—1 and PD—L2.
Pidlizumab (CT-011; CureTech) (humanized IgG1 anti-PD1 mAb from Cu- reTech/Teva), Pidlizumab (CT-011; CureTech) (see e.g., W02009/101611) Thirty patients with rituximab—sensitive relapsed FL were d with 3 mg/kg intravenous CT—011 every 4 weeks for 4 infusions in combination with rituximab dosed at 375 mg/m2 weekly for 4 weeks, starting 2 weeks after the first infusion of CT-011.
Further known PD-1 antibodies and other PD-1 inhibitors include AMP-224 (a B7- DC/IgGI fusion n licensed to GSK), AMP- 514 described in , antibody MEDI-4736 (an anti-PD-L1 developed by AstraZeneca/Medimmune) described in WO2011/066389 and U82013/034559, antibody YW243.55.S7O (an D-L1) described in W02010/077634, MDX—1105, also known as BMS—936559, is an anti—PD—L1 antibody devel— oped by Bristol-Myers Squibb described in WO2007/005874, and antibodies and inhibitors described in W02006/121168, /014708, W02009/114335 and WO2013/019906, the sures of which are hereby incorporated by reference. Further examples of D1 an— tibodies are disclosed in W02015/085847 (Shanghai Hengrui Pharmaceutical Co. Ltd.), for example antibodies having light chain variable domain CDR1, 2 and 3 of SEQ ID NO: 6, SEQ ID NO: 7 and/or SEQ ID NO: 8, respectively, and antibody heavy chain variable domain CDR1, 2 and 3 of SEQ ID NO: 3, SEQ ID NO: 4 or SEQ ID NO: 5, respectively, n the SEQ ID NO references are the numbering according to WO2015/085847, the disclosure of which is orated herein by reference. Antibodies that compete with any of these anti- bodies for binding to PD-1 or PD-L1 also can be used.
An exemplary D-1 antibody is pembrolizumab (see, e.g., the disclosure of which is incorporated herein by reference). The anti-PD-1 antibody may be the antibody h409AI 1 in , comprising heavy chain variable regions d by the DNA deposited at the ATCC as 081469_SPD-H and light chain variable regions d by the DNA deposited at the ATCC asO801470_SPD-L-l 1. In other embodiments, the anti- body comprises the heavy and light chain CDRs or variable regions of pembrolizumab. Ac— cordingly, in one embodiment, the antibody comprises the CDR1, CDR2, and CDR3 s of the VH of pembrolizumab encoded by the DNA deposited at the ATCC as 081469_SPD-H, and the CDR1, CDR2 and CDR3 domains of the VL of pembrolizumab encoded by the DNA deposited at the ATCC as 0801470_SPD-L-I 1.
In some embodiments, the PD-1 neutralizing agent is an anti-PD-L1 mAb that inhib- its the binding of PD-L1 to PD-1. In some embodiments, the PD—1 neutralizing agent is an anti-PD1 mAb that inhibits the g of PD-1 to PD-L1. In some embodiments, the PD-1 neutralizing agent is an immunoadhesin (e.g., an immunoadhesin comprising an extracellular or PD—1 binding portion of PD—L1 or PD—L2 fused to a constant region (e.g., an Fc region of an immunoglobulin sequence).
Another exemplary anti-PD-1 antibody is nivolumab comprising heavy and light chains having the respective sequences shown in SEQ ID N08: 12 and 13 or a respective amino acid sequence at least 50%, 60%, 70%, 80%, 90%, 95%, 98% or 99% identical there- to, or n binding nts and variants thereof. In other embodiments, the antibody comprises the heavy and light chain CDRs or variable regions of nivolumab. Accordingly, in one embodiment, the antibody comprises the CDR1, CDR2, and CDR3 domains of the heavy chain of nivolumab having the sequence set forth in SEQ ID NO: 12, and the CDR1, CDR2 and CDR3 domains of the light chain of nivolumab having the sequences set forth in SEQ ID NO: 13.
QVQLVESGGGWQPGRSLRLDCKASGITFSNSGMHWVRQAPGKGLEWVAVrWY DGSKRYYADSVKGRFTISRDNSKNTLFLQMNSLRAEDTAVYYCATNDDYWGQGTLVT VSSASTKGPSVFPLAPCSRSTSESTAALGCLV DYFPEPVTVSWNSGALTSGVHTFPAVL QSSGLYSLSSVVTVPSSSLGT TYTCNVDHKPSNTKVD RVES YG PAPEFLGG PSVFLFPPKPKDTLMlSRTPEVTCWVDVSQEDPEVQFNWYYDGVEVHNA TKPREEQF NSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKA GQPREPQVYTLPPSQ EEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPEKNYKTTPPVLDSDGSFFLYSRLTVDK SRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK (SEQ ID NO: 12).
EIVLTQSPATLSLSPGERATLSCRASQSVSSYLAWYQQ PGQAPRLLIYDASNRATGIPARFSGSGSG TDFTLTISSLEPEDFAVYYCQQSSNWPRTFGQGTKVEI RTVAAPSVFI FPPSDEQL SGTASVVCLLN NFYPREA VQWKVDNALQSGNSQESVTEQDS DSTYSL SSTLLSKADYEKHKVYACEVTHQGLSS RGEC (SEQ ID NO: 13).
An exemplary anti-PD-L1 antibody comprises heavy and light chain variable regions having the respective sequences shown in SEQ ID N03: 14 and 15, or an amino acid se— quence at least 50%, 60%, 70%, 80%, 90%, 95%, 98% or 99% cal o respectively, or antigen binding fragments and variants thereof. In other embodiments, the antibody com- prises the heavy and light chain CDRs or variable regions of 80A. Accordingly, in one ment, the dy comprises the CDR1, CDR2, and CDR3 domains of the heavy chain having the sequence set forth in SEQ ID NO: 14, and the CDR1, CDR2 and CDR3 domains of the light chain having the sequences set forth in SEQ ID NO: 15.
EVQLVESGGGLVQPGGSLRLSCAASGFTFSDSWIHWVRQAPGKGLEWVAWIS PYGGSTY- GRFTISADTSKNTAYLQ NSLRAEDTAVYYCARRHWPGGFDYWG QGTLVTVSS (SEQ ID NO: 14) DIQMTQSPSSLSASVGDRVTITCRASQDVSTAVAWYQQ PGKAPKLLIY SASF LYSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQYLYHPATFGQGTKVEIKR (SEQ ID NO: 15) The anti-PD-1 or D-L1 antibody can be selected from a fully human antibody, a humanized antibody, and a chimeric antibody. In one aspect of the invention, the agent comprises a constant domain derived from a human lgG1, lgG2, |gG3 or lgG4 antibody. In one aspect of the invention, the agent is a fragment of an antibody selected from IgA, an IgD, an lgG, an IgE and an lgM dy. In one aspect of the invention, the agent is an antibody fragment selected from a Fab fragment, a Fab' fragment, a Fab'-SH nt, a F(ab)2 frag- ment, a F(ab')2 fragment, an Fv fragment, a Heavy chain lg (a llama or camel lg), a VHH fragment, a single domain FV, and a —chain antibody fragment. In one aspect of the invention, the agent is a synthetic or nthetic antibody-derived molecule selected from a scFV, a dsFV, a minibody, a diabody, a triabody, a kappa body, an IgNAR; and a multi- specific antibody.
The anti-PD-‘I or anti—PD-L1 antibody can lack ntial specific binding to F0y re- ceptors, e.g. CD16. Such antibodies may comprise constant regions of various heavy chains that are known not to bind Fc receptors. One such example is an |gG4 constant region. lgG4 Alternatively, antibody fragments that do not comprise nt s, such as Fab or F(ab’)2 fragments, can be used to avoid Fc receptor binding. Fc receptor binding can be as- sessed according to methods known in the art, including for example testing binding of an antibody to Fc receptor protein in a BIACORE assay. Also, any human antibody type (e.g.
IgG1, IgGZ, IgG3 or IgG4) can be used in which the Fc portion is modified to minimize or eliminate g to ch receptors.The anti-PD-1 or anti-PDL1 antibody, the antibody will therefore typically have reduced or minimal effector on. In one aspect, the minimal ef— fector function results from production in prokaryotic cells. In one aspect the l effector function results from an "effector-less Fc mutation" or aglycosylation. In still a further embod- iment, the effector—less Fc on is an N297A or D265A/N297A substitution in the con— stant region.
Formulations An anti-NKG2A or anti-PD-1 or anti-PD-L1 agent such as an antibody can be incor- porated in a pharmaceutical formulation comprising in a concentration from 1 mg/ml to 500 mg/ml, wherein said ation has a pH from 2.0 to 10.0. The formulation may further se a buffer system, preservative(s), ty agent(s), chelating agent(s), stabilizers and surfactants. In one embodiment, the pharmaceutical formulation is an aqueous formula- tion, i.e., formulation comprising water. Such formulation is typically a solution or a - sion. In a further embodiment, the ceutical formulation is an aqueous solution. The term “aqueous formulation” is defined as a formulation comprising at least 50 %w/w water.
Likewise, the term “aqueous solution” is defined as a solution comprising at least 50 %w/w water, and the term “aqueous sion” is defined as a suspension comprising at least 50 %w/w water.
In another embodiment, the pharmaceutical formulation is a -dried formula- tion, whereto the physician or the t adds solvents and/or diluents prior to use.
In another embodiment, the pharmaceutical formulation is a dried formulation (e.g. freeze-dried or spray-dried) ready for use without any prior ution.
In a further aspect, the pharmaceutical formulation comprises an aqueous solution of such an antibody, and a buffer, wherein the antibody is present in a concentration from 1 mg/ml or above, and wherein said formulation has a pH from about 2.0 to about 10.0.
In a r embodiment, the pH of the formulation is in the range selected from the list consisting of from about 2.0 to about 10.0, about 3.0 to about 9.0, about 4.0 to about 8.5, about 5.0 to about 8.0, and about 5.5 to about 7.5.
In a further ment, the buffer is selected from the group consisting of sodium acetate, sodium carbonate, citrate, glycylglycine, histidine, glycine, lysine, ne, sodium dihydrogen phosphate, disodium hydrogen phosphate, sodium phosphate, and tris(hydroxymethyl)—aminomethan, bicine, tricine, malic acid, succinate, maleic acid, fumaric acid, tartaric acid, aspartic acid or mixtures thereof. Each one of these specific buffers consti- tutes an alternative embodiment of the ion.
In a further embodiment, the formulation further comprises a pharmaceutically ac- ceptable vative. In a further embodiment, the formulation further comprises an isotonic agent. In a r embodiment, the formulation also comprises a chelating agent. In a fur- ther embodiment of the invention the ation further comprises a stabilizer. In a further embodiment, the formulation further comprises a surfactant.For convenience nce is made to Remington: The Science and Practice ofPharmacy, 19‘h edition, 1995.
It is possible that other ingredients may be present in the peptide pharmaceutical formulation of the present invention. Such additional ingredients may include wetting agents, fiers, antioxidants, bulking agents, tonicity modifiers, chelating agents, metal ions, ole— s vehicles, proteins (e.g., human serum albumin, gelatine or ns) and a zwitterion (e.g., an amino acid such as betaine, taurine, arginine, glycine, lysine and histidine). Such onal ingredients, of course, should not adversely affect the overall stability of the phar- maceutical formulation of the present invention. stration of pharmaceutical compositions according to the invention may be through several routes of administration, for example, enous. Suitable antibody formu— lations can also be determined by examining experiences with other already developed ther- apeutic onal antibodies. l onal antibodies have been shown to be effi- cient in clinical situations, such as Rituxan (Rituximab), Herceptin (Trastuzumab) Xolair (Omalizumab), Bexxar (Tositumomab), Campath (Alemtuzumab), Zevalin, Oncolym and simi- lar formulations may be used with the antibodies of this invention. For example, a monoclo- nal antibody can be supplied at a concentration of 10 mg/mL in either 100 mg (10 mL) or 500 mg (50 mL) single-use vials, formulated for IV administration in 9.0 mg/mL sodium chloride, 7.35 mg/mL sodium citrate ate, 0.7 mg/mL polysorbate 80, and Sterile Water for Injec- tion. The pH is adjusted to 6.5. In another embodiment, the antibody is supplied in a formu- lation comprising about 20 mM Na—Citrate, about 150 mM NaCl, at pH of about 6.0.
Also ed are kits which include a ceutical composition containing an an- ti-NKGZA antibody, and an anti—PD-1 or anti-PD-L1 antibody, and a pharmaceutically— acceptable carrier, in a therapeutically effective amount adapted for use in the preceding methods. The kits ally also can include instructions, e.g., comprising administration schedules, to allow a practitioner (e.g., a physician, nurse, or patient) to administer the com- position contained therein to administer the composition to a patient having cancer (e.g., a solid tumor). The kit also can include a e.
Optionally, the kits include multiple packages of the single-dose pharmaceutical compositions each ning an effective amount of the anti- NKGZA, anti-PD-1 or PD-L1 dy for a single administration in ance with the methods provided above. Instru— ments or devices necessary for administering the pharmaceutical composition(s) also may be included in the kits. For instance, a kit may provide one or more pre-filled syringes con- taining an amount of the anti-NKGZA, anti-PD-1 or anti-PD-L1 antibody. in one embodiment, the present invention provides a kit for treating a cancer in a human patient, the kit comprising: (a) a dose of an KGZA antibody comprising the CDR1, CDR2 and CDR3 do— mains of a heavy chain having the sequence set forth in any of SEQ ID NOS: 4-8, and the CDR1, CDR2 and CDR3 domains of a light chain having the sequence set forth in SEQ ID NO: 9; (b) a dose of an anti-PD-1 antibody or an anti-PD-L1 antibody; and (c) optionally, instructions for using the KGZA antibody and anti-PD-1 antibody in any of the methods described herein.
Diagnostics, prognostics, and treatment of malignancies Described are methods useful in the diagnosis, prognosis, monitoring, treatment and prevention of a cancer in an individual. While the treatment regimens and methods described herein are particularly useful for the ent of solid tumors, the treatment regimens and methods described herein can also be used for a y of hematological cancers, as well as infectious disease, and inflammation and autoimmune disorders. The methods and composi- tions of the present invention are utilized for example the treatment of a variety of cancers and other proliferative diseases including, but not limited to: carcinoma, ing that of the bladder, , colon, kidney, liver, lung, ovary, prostate, as, stomach, cervix, d and skin; hematopoietic tumors of lymphoid lineage, including leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, acute lymphoblastic ia, B-cell lymphoma, T- cell lymphoma, Hodgkins lymphoma, non—Hodgkins lymphoma, hairy cell ma and Burketts lymphoma, and multiple myeloma; hematopoietic tumors of myeloid lineage, includ- ing acute and chronic myelogenous leukemias, promyelocytic leukemia, and ysplastic syndrome; tumors of hymal origin, ing fibrosarcoma and rhabdomyoscarcoma; other tumors, including melanoma, ma, terato—carcinoma, neuroblastoma and glioma; tumors of the l and peripheral nervous system, including astrocytoma, neuroblastoma, glioma, and schwannomas; tumors of mesenchymal origin, including fibrosarcoma, rhabdo— myoscaroma, and osteosarcoma; and other tumors, including melanoma, xeroderma pig- mentosum, keratoacanthoma, seminoma, and d follicular cancer. ation therapies for the treatment of cancer provided herein involve admin- istration of a neutralizing anti-NKG2A antibody and a eutralizing agent, e.g. neutraliz— ing anti-PD-1 or anti-PD-L1 antibody, to treat subjects afflicted with cancer (e.g., advanced refractory solid tumors). In one embodiment, the invention provides an anti-NKGZA antibody and an anti-PD-1 antibody in ation to treat subjects having a solid tumor (e.g., a solid tumor, an advanced tory solid tumor) or subjects having a logical tumor. In a particular embodiment, the KG2A antibody comprises a heavy chain of any of SEQ ID NOS: 4—8 and a light chain of SEQ ID NO: 9. In one embodiment, the antibody that neutraliz— es the inhibitory activity of PD-1 is selected from the group consisting of pembrolizumab, nivolumab, AMP-514, MEDl-4736, CT-011 and MPDL3280A.
As used herein, adjunctive or combined administration (co—administration) includes simultaneous administration of the compounds in the same or different dosage form, or sepa- rate administration of the compounds (e.g., sequential administration). Thus, the anti-NKGZA and anti-PD—1 or anti—PD-L1 antibodies can be simultaneously administered in a single for- mulation. Alternatively, the anti-NKGZA and anti-PD-1 or anti-PD—L1 antibodies can be for— WO 41945 mulated for separate administration and are administered concurrently or sequentially.
In one embodiment, the cancer treated with the s disclosed herein is an HLA—E-expressing cancer. In one embodiment, the cancer is selected from the group con- sisting of lung cancer (e.g. non-small cell lung cancer (NSCLC)), renal cell carcinoma (RCC), melanoma, colorectal cancer, and ovarian cancer.
A patient having a cancer can be treated with the anti-NKG2A agents with or t a prior detection step to assess expression of HLA—E on the surface of tumor cells. Advanta- geously, the treatment methods can comprises a step of detecting a HLA—E nucleic acid or polypeptide in a biological sample of a tumor (e.g. on a tumor cell) from an individual. Exam- ple of biological samples include any suitable biological fluid (for example serum, lymph, blood), cell sample, or tissue . For example, a tissue sample may be a sample of tu- mor tissue or tumor—adjacent tissue. Optionally, HLA—E polypeptide is detected on the sur— face of a malignant cell. A determination that a biological sample expresses HLA—E (e.g. prominently expresses; expresses HLA—E at a high level, high intensity of staining with an anti—HLA—E antibody, compared to a reference) indicates that the individual has a cancer that may have a strong benefit from treatment with an agent that inhibits NKGZA. In one embod- iment, the method comprises ining the level of expression of a HLA-E nucleic acid or polypeptide in a biological sample and comparing the level to a reference level (e.g. a value, weak cell surface staining, etc.) corresponding to a healthy individual. A determination that a ical sample expresses an HLA—E c acid or polypeptide at a level that is increased compared to the reference level indicates that the individual has a cancer that can be treated with an agent that inhibits NKGZA.
In one ment, a determination that a biological sample (e.g a sample compris- ing tumor cells, tumor tissue and/or tumor adjacent ) prominently expresses HLA—E nu- cleic acid or polypeptide tes that the individual has a cancer that can be treated with an agent that inhibits NKG2A. “Prominently expressed”, when referring to a HLA—E polypeptide, means that the HLA—E polypeptide is expressed in a substantial number of tumor cells taken from a given individual. While the definition of the term “prominently expressed” is not bound by a precise percentage value, in some examples a receptor said to be nently ex- pressed” will be present on at least 30%, 40%, 50°%, 60%, 70%, 80%, or more of the tumor cells taken from a patient (in a sample). ining whether an individual has cancer cells that express an HLA—E polypep— tide can for example comprise obtaining a biological sample (e.g. by performing a biopsy) from the dual that comprises cancer cells, bringing said cells into contact with an anti- body that binds an HLA—E polypeptide, and ing whether the cells express HLA—E on their surface. Optionally, determining r an individual has cancer cells that express HLA—E comprises conducting an immunohistochemistry assay. Optionally determining whether an individual has cancer cells that express HLA—E comprises conducting a flow cy- tometry assay.
In the ent methods, the anti—NKG2A antibody and the anti—PD—1 or anti—PD—L1 antibodies can be administered separately, together or sequentially, or in a cocktail. In some embodiments, the antigen-binding compound is administered prior to the administration of the anti-PD—1 or anti-PD-L1 antibodies. For example, the anti—NKGZA antibody can be ad— ministered approximately 0 to 30 days prior to the administration of the anti-PD-1 or D- L1 antibodies. In some embodiments, an anti-NKG2A antibody is administered from about 30 minutes to about 2 weeks, from about 30 minutes to about 1 week, from about 1 hour to about 2 hours, from about 2 hours to about 4 hours, from about 4 hours to about 6 hours, from about 6 hours to about 8 hours, from about 8 hours to 1 day, or from about 1 to 5 days prior to the administration of the anti-PD-1 or anti-PD-L1 antibodies. In some embodiments, an anti—NKGZA antibody is administered concurrently with the administration of the D—1 or anti-PD-L1 antibodies. In some embodiments, an anti-NKGZA dy is stered after the administration of the anti-PD-1 or anti—PD-L1 antibodies. For example, an anti- NKGZA antibody can be administered approximately 0 to 30 days after the administration of the anti-PD-1 or D—L1 antibodies. In some embodiments, an anti-NKG2A antibody is administered from about 30 minutes to about 2 weeks, from about 30 minutes to about 1 week, from about 1 hour to about 2 hours, from about 2 hours to about 4 hours, from about 4 hours to about 6 hours, from about 6 hours to about 8 hours, from about 8 hours to 1 day, or from about 1 to 5 days after the administration of the anti-PD-1 or anti-PD-L1 antibodies.
Suitable treatment protocols for treating a human having cancer include, for exam- ple, administering to the patient an effective amount of each of an antibody that inhibits NKG2A and an dy that neutralizes the inhibitory activity of human PD-1, n the method comprises at least one administration cycle in which at least one dose of the anti- NKGZA antibody is administered at a dose of 1—10 mg/kg body weight and at least one dose of the D-1 or anti-PD-L1 antibody is administered at a dose of 1-20 mg/kg body weight.
In one embodiment, the administration cycle is n 2 weeks and 8 weeks.
In one embodiment, the method comprises at least one administration cycle, where— in the cycle is a period of eight weeks or less, n for each of the at least one cycles, two, three or four doses of the anti-NKG2A antibody are administered at a dose of 1-10 mg/kg body weight and two, three or four doses of the anti—PD-1 or anti-PD-L1 antibody are administered at a dose of 1-20 mg/kg body weight.
The anti-NKGZA antibody can advantageously be administered in an amount that achieves a concentration in circulation that is at least 10, 20, or 30 times higher than the concentration required for substantially full (e.g., 90%, 95%) receptor saturation (e.g., as as- sessed by titrating anti-NKGZA antibody on NKG2A—expressing cells, for e in PBMC), or optionally in an amount that achieves a tration in a extravascular tissue (e.g. the tumor tissue or environment) that is at least 10, 20, or 30 times higher than the concentration required for substantially full or saturation (e.g., as assessed by titrating anti-NKG2A antibody on NKGZA—expressing cells, for example in PBMC).
NKGZA+ NK cell response can be assessed using a suitable assay of cytotoxic ac- tivity of NKG2A—expressing NK cells toward HLA—E expressing target cells. Examples include assays based on markers of NK cell activation, for example CD107 or CD137 sion.
The EC50 for NKG2A+ NK cell response (e.g., as assessed in a CD107 mobilization assay) of blocking anti-NKGZA antibody humZZ70 used in the Examples herein (e.g. having the heavy chain of any of SEQ ID NOS: 4-8 and a light chain of SEQ ID NO: 9) is about 4 ug/ml, and the EC100 is about 10 ug/ml. Thus an amount of anti—NKGZA antibody is administered so at to maintain a continuous (minimum) blood concentration of at least 4 ug/ml. Advantageously an amount of anti-NKGZA antibody can be administered so at to achieve and/or maintain a continuous (minimum) blood concentration of at least 10 ug/ml. For example, the blood con— tion to be achieved and/or maintained can be between 10-12 ug/ml, 10-15 pg/ml, 10- 20 ug/ml, 10-30 ug/ml, 10-40 pg/ml, 10-50 ug/ml, 10-70 ug/ml, 10—100 ug/ml, 10-150 ug/ml or 10-200 pg/ml. When tissues outside of the vasculature are targeted (e.g. in the treatment of solid tumors), an amount of anti-NKGZA antibody is stered so at to achieve and/or maintain a tissue tration of at least 10 ug/ml; for example, administering an amount of KGZA antibody to achieve a blood concentration of at least 100 ug/ml is expected to e a tissue tration of at least 10 ug/ml. For e, the blood concentration to be achieved and/or maintained in order to achieve/maintain 10 ug/ml in a tissue can be be- tween 100-110 ug/ml, 100-120 ug/ml, 100-130 ug/ml, 100-140 ug/ml, 100-150 ug/ml, 100- 200 ug/ml, 100—250 ug/ml or 100—300 pg/ml. in some embodiments, an amount of anti-NKG2A antibody is administered so as to obtain a concentration in blood (e.g., blood serum) that corresponds to at least the EC50 for NKG2A+ lymphocyte cell response (e.g., the NKG2A+ NK cell response), optionally at about or at least about, the EC100. "EC50" (or “EC100”) with t to NKG2A+ cell response (e.g.
NK cell response), refers to the efficient concentration of anti-NKG2A antibody which pro- duces 50% (or 100% when referring to the E0100) of its maximum response or effect with re- spect to such NKGZA+ cells response (e.g. NK cell response). In some ments, partic- ularly for the treatment of solid , the concentration achieved is designed to lead to a concentration in tissues (outside of the vasculature, e.g. in the tumor nment) that cor— responds to at least the E050 for NKGZA+ NK cell response, optionally at about, or at least about, the E0100 for NKG2A+ NK cell response.
Suitable ent protocols for an anti—NKG2A antibody such as humZ270 used in the Examples herein having an E0100 for NKG2A+ NK cell response of about 10 ug/ml com- prise at least one administration cycle in which at least one dose of the anti-NKG2A antibody is administered at a dose of 2-10 mg/kg, optionally 4-10 mg/kg, ally 6-10 mg/kg, op- tionally 2-6 mg/kg, optionally 2-8 mg/kg, or ally 2-4 mg/kg body weight. Optionally, at least 2, 3, 4, 5, 6, 7 or 8 doses of the anti-NKGZA antibody are administered. In one embod- iment, the administration cycle is between 2 weeks and 8 weeks. In one embodiment, the administration cycle is 8 weeks. In one embodiment, the administration cycle is 8 weeks and comprises administering one dose of the anti-NKGZA antibody every two weeks (Le. a total of four doses).
In one aspect of any of the embodiments herein, the anti—NKG2A antibody is admin— istered once about every two weeks.
Suitable treatment protocols for use with an anti-NKG2A antibody, particularly for the treatment of a hematopoietic tumor, include for example, stering to the patient an anti-NKG2A antibody two times per month in an amount effective to maintain a continuous blood concentration of anti-NKG2A antibody of at least 10 ug/ml between at least two suc- cessive administrations of the anti-NKGZA antibody is between 2-10 mg/kg, optionally 2—6 mg/kg, optionally 2-8 mg/kg, optionally 2-4 mg/kg, optionally 2-6 mg/kg, optionally 2-4 mg/kg, optionally about 4 mg/kg body weight. These doses can optionally be administered so as to e for continued blood concentration of anti-NKGZA antibody of at least 10 pg/ml throughout the treatment cycle. Achieving blood concentration of anti-NKGZA antibody of 10 ug/ml corresponds to the E0100 for an antibody such as humanized Z270. le treatment protocols for use with an anti-NKGZA antibody, ularly for the treatment of a solid tumor where anti—NKG2A antibody E050 tration is sought in extravascular tissue (e.g., in the tumor or tumor environment), include for e, adminis- tering to the patient an anti-NKG2A antibody two times per month in an amount effective to maintain a continuous blood concentration of anti—NKGZA antibody of at least 40 ug/ml be— tween at least two successive administrations of the KGZA antibody is between 2-10 mg/kg, optionally 2-6 mg/kg, ally 2-4 mg/kg, optionally about 4 mg/kg body weight.
These doses can optionally be administered so as to provide for continued blood concentra- tion of KGZA antibody of at least 40 ug/ml throughout the treatment cycle. Achieving blood concentration of anti-NKGZA antibody of 40 ug/ml is expected to provide a tissue (e.g., ascular tissue, tumor environment) concentration of about 4 ug/ml, in turn correspond— ing to the E050 for an antibody such as humanized 2270.
Suitable treatment protocols for use with an anti-NKG2A antibody, particularly for the treatment of a solid tumor where anti—NKGZA antibody E050 concentration is sought in extravascular tissue (e.g., in the tumor or tumor environment), include for example, adminis- tering to the t an effective amount of an anti-NKGZA antibody, wherein the antibody is administered 2 times per month and the amount effective to maintain a continuous blood concentration of KGZA antibody of at least 100 ug/ml between at least two successive administrations of the KGZA dy is between 4-10 mg/kg, optionally 4—6 mg/kg, op- ly 4-8 mg/kg, optionally about 4 mg/kg, optionally about 6 mg/kg, optionally about 8 mg/kg, or optionally about 10 mg/kg. These doses can optionally be administered so as to provide for continued blood concentration of anti-NKGZA antibody of at least 100 ug/ml throughout the treatment cycle. Achieving blood concentration of anti-NKGZA antibody of 100 ug/ml is expected to provide a tissue (e.g., extravascular, tumor environment) concentra— tion of about 10 ug/ml, in turn corresponding to the E0100 for an dy such as humanized 2270.
Further suitable ent protocols for use with an anti—NKGZA antibody include regimens that employ a loading period with a higher dose, followed by a maintenance period.
For example, a g period may comprise administering to the patient an effective amount of an anti-NKGZA antibody, wherein the antibody is administered one or more times in an amount effective to maintain a continuous blood concentration of anti-NKGZA antibody of at least 100 ug/ml until the first administration of anti-NKGZA antibody in the maintenance reg- imen. For example, when administered once, a loading dose of 10 mg/kg of anti-NKGZA an- tibody can be administered, wherein the first administration of anti-NKGZA antibody within the maintenance regimen occurs about two weeks (or less) after the loading dose. The nance regimen can then employ a lower dose and/or lower frequency of administra- tion in order to maintain a continuous blood concentration of anti—NKGZA antibody of at least 100 ug/ml between successive administrations within the maintenance regimen. For exam- ple, a nance regimen can comprise administering anti-NKGZA dy every two weeks at a dose of n 2—10 mg/kg, optionally 4—10 mg/kg, optionally 2—4 mg/kg, option— ally 4-6 mg/kg, optionally 4-8 mg/kg, ally about 4 mg/kg, optionally about 6 mg/kg, optionally about 8 mg/kg.
In certain embodiments, a dose (e.g. each dose) of the anti-NKGZA antibody is ad- ministered at 4, 6, 8 or 10 mg/kg. In certain embodiments, a dose (e.g. each dose) of the an- ti-PD—1 antibody is administered at 1-20 mg/kg, optionally at ‘IO mg/kg. In certain embodi- ments, a dose (e.g. each dose) of the anti—PD—L1 dy is administered at 10, 15, 20 or 25 mg/kg, optionally at 1200 mg total dose. In certain embodiments, the combined therapy per- mits the anti-PD-1 or PD—L1 dy to be administered at a lower dose; in one embodi- ment, each dose of the D—1 antibody is administered at 2 or 3 mg/kg.
In one embodiment, the anti-NKGZA antibody and anti-PD-1 or anti-PD-L1 antibody are administered at the following doses: (a) 1-10 mg/kg anti-NKGZA antibody and (i) 1-10 mg/kg of anti-PD-1 antibody or (ii) 1-20 mg/kg of anti-PD-L1 antibody; (b) 4, 6, 8 or 10 mg/kg anti-NKGZA antibody and 10 mg/kg of anti-PD-1 or anti-PD- L1 antibody; (0) 4, 6, 8 or 10 mg/kg anti—NKGZA dy and 3 mg/kg of anti—PD—1 antibody; or (d) 4, 6, 8 or 10 mg/kg anti-NKGZA antibody and 2 mg/kg of anti-PD-1 antibody.
In one aspect of any of the embodiments herein, the anti-NKGZA antibody is admin- istered once about every two weeks. In one aspect of any of the ments herein, the anti-PD-‘I or anti-PD—L1 antibody is administered once about every three weeks. In one as- pect of any of the embodiments herein, the anti-PD-1 or anti-PD-L1 antibody is administered once about every two weeks. In one aspect of any of the embodiments herein, the anti—PD—1 or anti-PD-L1 antibody is administered once about every four weeks.
In one embodiment the anti-PD-1 or anti-PD-L1 antibody and the anti-NKGZA anti- body are administered by i.v. In one embodiment the anti-PD-1 or anti-PD-L1 antibody and the anti-NKGZA dy are administered on the same day, optionally further once about every two weeks, optionally further by i.v.
In other aspects, s are provided for identifying NKGZA+PD1+ NK cells and/or T cells. ing the co-expression of NKGZA and PD-1 on NK cells and/or T cells can be used in diagnostic or prognostic methods. For example, a biological sample can be obtained from an individual (e.g. from cancer or cancer-adjacent tissue obtained from a can- cer patient) and analyzed for the presence of PD1+ NK and/or T cells. The expres— sion of both NKG2A and PD-1 on such cells can, for example, be used to identify individuals having tumor rating NK and/or T cells which are inhibited by both NKGZA and PD1 poly- peptides. The method can, for example, be useful as a prognostic for se to treatment with an agent that neutralizes NKGZA, as a prognostic for response to treatment with an agent that neutralizes PD1, or as a prognostic for se for combined treatment with an agent that neutralizes NKGZA and an agent that neutralizes PD1.
In one embodiment, provided is a method for assessing r an individual is suitable for treatment with an agent that inhibits NKG2A and an agent that neutralizes the tory activity of human PD—1, the method comprising detecting a lymphocyte tion (e.g. CD8+ T cells) that express both an NKG2A nucleic acid or polypeptide and a PD-1 nu- cleic acid or polypeptide in a biological sample from an individual. A determination that the individual has a cyte population that express both an NKGZA nucleic acid or polypep— tide and a PD-1 nucleic acid or ptide tes that the patient has a cancer that can be treated with an agent that inhibits NKGZA in combination with an agent that neutralizes the inhibitory activity of human PD-1.
In other s, methods are provided for identifying PD1+ NK cells and/or T cells. The finding that tumor infiltrating or lymphocytes can express both inhibi- tory receptors NKG2A and PD-1 gives rise to improved treatment methods as well as meth- ods to detect such double—restricted/inhibited effector cells that can be useful in diagnostics and prognostics.
For example, a biological sample can be obtained from an individual (e.g. from can- cer or cancer—adjacent tissue obtained from a cancer patient) and analyzed for the presence of NKG2A+PD1+ NK and/or T cells. The expression of both NKG2A and PD-1 on such cells can, for example, be used to identify individuals having tumor infiltrating NK and/or T cells which are inhibited by both NKGZA and PD1 polypeptides. The method can, for example, be useful as a prognostic for se to treatment with an agent that neutralizes NKG2A, as a prognostic for response to treatment with an agent that neutralizes PD1, or as a prognostic for response for combined treatment with an agent that lizes NKGZA and an agent that neutralizes PD1.
Detecting NKG2A— and PD-1 restricted NK and/or CD8 T cells within biological sam- ples can more generally have ages for use in the study, evaluation, diagnosis, prog- nosis and/or prediction of pathologies where characterization of NK and/or CD8 T cells is of interest. For example, favorable or unfavorable cancer prognosis can be made by assessing whether tumor or tumor adjacent tissues are characterized by infiltrating NK and/or CD8 T cells that express both NKG2A and PD—1.
For example, cancer in patients can be characterized or assessed using anti- NKGZA and anti—PD1 antibodies to assess whether tumor-infiltrating NK and/or CD8 T cells are NKG2A+PD1+, including whether such NK and/or CD8 T cells are present at the tumor periphery (in cancer adjacent tissue). The s can be useful to determine whether a pa- tient has a pathology characterized by NK and/or CD8 T cells which could be amenable to modulation by therapeutic agents that directly act on such NK and/or CD8 T cells (e.g. by binding to NKG2A and/or PD—1, or their respective ligands HLA—E or PD-L1) or that indirectly WO 41945 act on such NK and/or CD8 T cells (e.g., by producing cytokines or other signalling mole- cules that can modulate the activity of the NK and/or CD8 T cells). Optionally, in any embod— iment, the patient has been treated with an agent that neutralizes PD-1. The methods de- scribed herein can optionally further comprise administering to an dual such a therapeu- tic agent if it determined that the individual has a pathology which could be amenable to modulation by therapeutic agents that act on the tumor infiltrating NK and/or CD8 T cells.
In one aspect the inventors provides an in vitro method for ing a NKG2A+ PD- 1+ lymphocyte, optionally an NK or CD8+ T cell, the method comprising providing a biologi- cal sample sing tumor infiltrating lymphocytes and determining whether the lympho- cytes express NKGZA and PD-1.
In one aspect the inventors provides an in vitro method of detecting NKG2A and PD- 1—expressing (i.e. double positive) CD8 T cells or NK cells within a sample from a human in— dividual, said method comprising providing a sample from an individual, contacting cells in said sample using a monoclonal antibody that specifically binds to a human NKGZA polypep- tide and a monoclonal antibody that specifically binds to a human PD—1 polypeptide in the samples, and detecting binding of the antibodies to cells. In one embodiment the cells are CD8 T cells and/or NK cells.
In one aspect the inventors provides an in vitro method of detecting tissue infiltrating human CD8 T cells and/or NK cells that are inhibited by both NKG2A and PD-1 within a sample from a human individual, said method sing providing a sample from an indi- vidual, and ing tissue infiltrating CD8 T cells and/or NK cells in said sample using a monoclonal dy that specifically binds to a human NKGZA polypeptide and a o- nal antibody that specifically binds to a human PD-1 polypeptide in the s, wherein a detection of NKGZA and PD-1 polypeptide indicates the presence of NKGZA— and PD inhbited tissue infiltrating CD8 T cells and/or NK cells. Optionally, in any embodiment, the patient has been treated with an agent that neutralizes PD-1. In one embodiment, the sample comprises tumor cells, tumor tissue or tumor adjacent tissue. In one embodiment, the CD8 T cells and/or NK cells are identified using immunohistochemistry methods. In one embodi— ment, the sample is a paraffin-embedded sample; optionally the paraffin-embedded sample has been fixed, embedded in in, sectioned, deparaffinized, and transferred to a slide before being brought into t with the monoclonal antibody. In one embodiment, the CD8 T cells and/or NK cells are identified using flow try methods.
Examples WO 41945 e 1 —RMA-S and A20 tumor cells are infiltrated by NK cells expressing NKGZA and CD8 T cells expressing NKGZA and/or PD-1 Lymphocytes generally are not found to co-express NKGZA and PD—1. To i- gate the expression of these receptors on tumor-infiltrating lymphocytes, distribution of NKG2A and PD—1 were studied on NK and T cell subsets in tumor from mice. Lymphocytes were taken from spleen, from tumor draining lymph nodes, as well as from within solid tu- mors. 057/BL6 mice were engrafted ($0) with PDL-1+ Qa-1+ RMA-S cells (Qa-1, Qdm, 82m) or with A20 tumor cells. RMA—S Qa-1 Qdm BZm (top row) and A20 (bottom row) tumor bearing mice were sacrificed when tumor volumes were around 500 mm3.
Results are shown in Figures 1A and 18. Tumor cells (Figure 1 A) and tumor infil- trating lymphocytes—TIL— (Figure 1 B) were analyzed by flow cytometry respectively for the expression of Qa-1 and PDL-1 for tumor cells and NKG2A and PD-1 for TIL. One enta- tive mouse out of 3 is shown. MFleedian of fluorescence intensity.
More than half of the rating NK cells from both tumor types expressed NKG2A, suggesting that tumor-infiltrating NK cells are inhibited by NKGZA. The NKG2A+ NK cells generally did not express significant s of PD-1. However, CD8 T cells that were posi- tive for both NKG2A and PD—1 were found, suggesting that the CD8 T cells may be restricted by both inhibitory receptors NKGZA and PD-1.
Example 2 —NK and T cell subsets from mice g rma-rae1 tumors are capable of NKGZA and PD-1 co-expression To further investigate the expression of receptors NKG2A and PD-1, bution of NKGZA and PD—1 were studied on NK and T cell subsets in mice. Lymphocytes were taken from spleen, from tumor draining lymph nodes, as well as from within solid tumors. 057/BL6 mice were engrafted (80) with RMA— Rae clone 6 (2 million cells). These tumor cells express CD94/NKGZA ligand, Qa—1. Mice were sacrificed at day 12 with a mean tumor volume: 723 mm3, SD: 161 mms, n=4. Following cell suspension preparation from spleen, LN and tumor, cells were stained as follows: CD3e PerCP Cy5.5, NKP46 Alexa 647, NKGZA/C/E FITC, PD1 PE, CD8 Pacific Blue. s are shown in Figure 2 and Tables 1-3.
In the NK cell subset, cells in both the draining lymph nodes and spleen were about half NKGZA—positive and half NKGZA—negative, however in neither case was there significant expression of PD1. NK cells from lymph nodes were NKG2A+ PD-1' (49.2%) and NKG2A‘ PD-1' (49.5%), and less than 1% (mean) of NK cells were NKGZA+ PD-1+. NK cells from spleen were NKG2A+ PD-1' (44.1%) and NKGZA' PD-1' ) and a mean of 0.1% (mean) of NK cells were NKG2A+ PD—1+.
In the T cell subset most cells were NKG2A-negative (only 1.1% in lymph nodes and 4.7% in spleen are ), and a small fraction of cells were PD-1+ (3.5% in lymph nodes and 10% in spleen were PD-1+ ), without significant double positive NKG2A PD—1 cells. Only 0.1% (mean) of T cells in lymph nodes were NKG2A+ PD-1+ and only 0.4% (mean) of T cells in spleen were NKG2A+ PD-1+. 95.1% of T cells from lymph nodes were double negative and 85.6% of T cells from spleen were double negative.
In the CD8 T cell subset, most cells were again NKGZA—negative (only 1.6% in lymph nodes and 3.9% in spleen are NKG2A+), and a small on of cells were PD-1+ (1.1% in lymph nodes and 2.5% in spleen were PD-1+ NKGZA'), without significant double positive NKG2A PD—1 cells. Only 0.2% (mean) of T cells in lymph nodes were NKG2A+ PD—1+ and only 0.3% (mean) of T cells in spleen were NKG2A+ PD-1+. 97.3% of T cells from lymph nodes were double negative and 93.6% of T cells from spleen were double negative.
However, among tumor infiltrating lymphocytes (TIL), all cells subsets had cells ex— pressing PD-1. NK cells, which were not usly found in significant percentages to ex- press PD-1, were observed in tumor to be PDpositive, including within the NKG2A+ sub- set, with 31.8% (mean) of NK cells that were NKG2A+PD—1+. While almost no CD8 T cells outside the tumor had NKG2A expression, CD8 T cells expressing PD-1 were frequent in the tumor (the tumor infiltrating CD8 T cell subset had a mean of 26.3% NKG2A+ positive cells).
Moreover, within this NKGZA—positive CD8 T cell subset, most of the cells were NKG2A+ PD- 1+ (19.4% (mean). Yet, among the CD8‘ T cell subset, there was little difference in NKG2A expression observed between TlLs and spleen or lymph node cells, as only 5.1% of T cells in the tumor expressed NKG2A, and only 3.6% of T cells were double positive NKGZA PD-1.
Table 1: Spleen % among NK %NK %NK %NK NKG2A— NKG2A+ NKG2A+ % NK NKG2A- PD1+ PD1+ PD1- PD1- %NK NKG2A+ Mean 0.1 0.1 44.1 55.7 44.2 SD 0.1 0.1 2.2 2.2 2.2 % among T lymphocytes %T NKG2A- % T NKG2A+ % T NKG2A+ % T NKG2A- PD1+ PD1+ PD1- PD1- % T NKGZA+ Mice1 Spleen 8.08 0.38 4.52 4.54 Mice2 Spleen 9.64 0.32 4.22 6.25 Mice4 Spleen 12.3 0.41 3.21 3.37 Mean 10.0 0.4 4.0 4.7 SD 2.13 0.05 0.69 1.45 % among CD8+ T cytes %TCD8+ %TCD8+ %TCD8+ NKG2A- NKG2A+ NKG2A+ %TCD8+ % T CD8+ PD1+ PD1+ PD1- NKG2A— PD 1 - NKG2A+ Mice1 Spleen 1.8 MiceZ Spleen 2.08 Mice4 Spleen 3.67 Mean 2.5 SD 1.01 Table 2: Tumor Draining Lymph Nodes % among NK %NK %NK %NK NKG2A- NKG2A+ NKG2A+ % NK NKG2A- PD1+ PD1+ PD1- PD1- %NK NKG2A+ Mice1 LN 0.68 0.85 45.10 Mice3 LN 0.20 0.40 54.70 Mice4 LN 0.61 1.21 47.90 Mean 0.50 0.82 49.23 SD 0.26 0.41 4.94 % among T lymphocytes %T NKG2A- % T NKG2A+ °/o T NKG2A+ % T NKG2A- PD1+ PD1+ PD1- PD1- % T NKG2A+ Mice1 LN 2.8 0.1 1.8 95.3 0.5 Mice3 LN 6.6 0.4 2.4 90.6 0.7 2015/071069 Mice4 LN 2.1 0.0 0.6 97.2 0.6 Mean 3.5 0.1 1.3 95.1 1.1 SD 2.1 0.2 0.9 3.1 1.1 % among CD8+ T lymphocytes %TCD8+ %TCD8+ %TCD8+ NKG2A— NKG2A+ NKG2A+ %TCD8+ % T CD8+ PD1+ PD1+ PD1- NKG2A- PD1- NKG2A+ Mice1 LN 1.0 Mice3 LN 2.1 Mice4 LN 0.6 Mean 1.1 SD 0.7 Table 3: Tumor lnfiltrating Lymphocytes % among NK %NK %NK %NK NKG2A— NKG2A+ NKG2A+ % NK NKG2A- PD1+ PD1+ PD1- PD1- %NK NKG2A+ Mice1 TIL 18.8 25.6 26.6 28.9 Mice2 TIL 22.8 31 13.5 32.7 Mice3 TIL 26 38.2 13.8 22 52 Mice4 TIL 24.4 32.5 22.5 20.7 55 Mean 23 31.8 19.1 26.1 50.9 SD 3.1 5.2 6.5 5.7 4.5 % among T lymphocytes %T NKG2A— % T NKG2A+ % T NKG2A+ % T NKG2A- PD1+ PD1+ PD1- PD1- % T NKG2A+ Mice1 T|L 51.7 4.85 7.8 35.6 2.89 Mice2 TIL 86.5 1.47 1.42 10.7 8.22 Mice3 T|L 75 4.93 3.29 16.8 9.3 Mice4 TIL 66 3.1 6.2 24.7 0 Mean 69.8 3.6 4.7 22.0 5.1 SD 14.7 1. 6 2.9 10.8 4.4 % among CD8+ T lymphocytes %TCD8+ %TCD8+ %TCD8+ %TCD8+ ”/0 T CD8+ NKG2A— NKG2A+ NKG2A+ NKG2A— PD1- NKG2A+ 2015/071069 PD1+ PD1+ PD1- Mice1 TIL 49.5 16 10.8 23.7 26.8 Mice2 TIL 52.2 21.7 4.35 21.7 26.05 Mice3 TIL 44.3 28.6 5.71 21.4 34.31 Mice4 TIL 52.8 11.3 6. 6 29.2 17.9 Mean 49.7 19 4. 6 .9 24 0. 26 3.
SD 3.9 7.5 2. 8 3.6 6.7 Example 3 —NKG2A and PD-1 expression in tumor bearing mice To further investigate NKGZA and PD—1 expression in tumor—bearing mice, C57/BL6 mice were engrafted (so) with different tumor cells, either RMA- Rae1, MC38 or RMA lines.
To evaluate the influence of tumor volume, mice were sacrificed when their tumors reached respectively the volumes of 500, 2000 and 800 mm3.
Results are shown in Figures 3A and 3B, with RMA Rae1 (top row), MC38 (medium row) and RMA (bottom row). NK cells e 3A) and CD8 T cells (Figure 3B) were analyzed by flow cytometry in spleen, tumor draining lymph node (LN) and tumor for NKGZA and PD-1 expression. One entative mouse out of 2 to 4 is shown.
In the NK cell subset, cells in the tumor, lymph nodes and spleen were about half NKG2A-positive and half negative. Neither NK cells (regardless of their NKG2A expression ) from the draining lymph nodes nor the spleen showed any significant expression of PD1. However, the tumor infiltrating NK cells from RMA-Rae1 and RMA expressed signifi- cant levels of both NKG2A and PD1. Tumor rating NK cells from tumor line MC38 that were sacrificed with particularly large volume (2000 mm3) expressed NKGZA (50%) but did not significantly express PD1 (3%).
Unlike NK cells which express NKG2A in about half the population, the CD8 T cells from spleen and lymph nodes generally expressed neither NKG2A nor PD1. However, in tu— mors, a large proportion of CD8 T cells expressed both NKGZA and PD1 (28% in RMA- Rae1, 43% of MC38 and 40% of RMA were double positive). The results again suggest that tumor infiltrating CD8 T cells as well as NK cells may be capable of being cted by both tory receptor NKG2A and PD1, furthermore across different types of tumor cells.
Example 4 —Treatment with anti-PD-1 mAb increases the frequency of NKG2A express- ing CD8 T cells in tumors To evaluate the effect of ent with anti-PD1 antibody on CD8 T cells, M038 tumor bearing mice were either treated with 200 pg of rat lgG2a isotype control (IC) or neu— tralizing anti-mouse PD-1 monoclonal dies on days 11, 14 and 17 after cells engraft- ment. Mice (n=3/group) were sacrificed on day 31 and CD8 T cells were characterized by flow cytometry in , tumor draining lymph node (LN) and tumor. Means +/— SD (n=3) of the percentages of CD8 NKG2A+ among CD8 T cells are represented. P<0.005 (***), P<0.0005 (****), statistical analysis performed with Two way ANOVA followed by Tukey’s le ison test.
Results are shown in Figure 4. Similarly to that observed in other experiments, CD8 T cells from spleen of lymph nodes did not significantly express NKGZA. stration of anti-PD1 antibody did not cause any change in the level of NKG2A expression in the spleen or lymph node T cells. However, in the tumor infiltrating CD8 T cell population, administra— tion of anti-PD1 antibody caused a more than 50% increase in NKGZA—expressing CD8 T cells. The results suggest that upon treatment with anti-PD1 antibody, NKGZA receptor may have an increased contribution to the inhibition of the CD8 T cell response toward tumors in vivo. Neutralization of NKG2A may therefore be useful to reverse the inhibition of the NKG2A restricted T cells in individuals treated with an agent that neutralizes the PD-1 axis such as an anti—PD1 or PDL1 antibody.
Example 5 — Combinatorial anti-NKGZA/anti-PD1 blockade inhibits tumor growth To evaluate the effect of combination treatment with neutralizing anti-PD1 dy and neutralizing anti-NKGZA antibody, C57BL/6 mice were engrafted (so) with RMA—S Qa-1 Qdm B2m tumor cells and treated with neutralizing anti-PD1 agent (a neutralizing anti-PD-L1 antibody) and neutralizing anti—NKGZA antibody.
Briefly, 6 mice were randomized on day 11 when RMA—S Qa—1 Qdm B2m tumor volume were about 85 mm3 (n=8 mice/group) and treated with isotype control, anti- mouse NKGZA mAb (200 pg, iv), anti—mouse PD—L1 mAb (200 ug, ip) or anti— mNKG2A/mPDL-1 combination on days 11, 14 and 18. Tumor volume was measured twice a week with a caliper. Animals were euthanized when tumor became large e >2000 mm3), ulcerated or necrotic. Data represent median tumor volume per experiment.
The evolution of median tumor volume over time is shown in Figure 5. While anti- NKGZA yielded only a modest umor effect compared to isotype control in this model and D-L1 yielded a substantial anti-tumor effect but with tumor volume increasing to- ward day 28, the combined treatment with anti-NKGZA and D-L1 completely abolished tumor growth, with no significant growth in tumor volume observed at day 28.
All nces, including publications, patent ations, and patents, cited herein are hereby orated by reference in their entirety and to the same extent as if each refer- ence were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein (to the maximum extent permitted by law), regardless of any separately provided incorporation of particular documents made elsewhere herein.
The use of the terms “a” and “an” and “the” and similar referents in the context of describing the invention are to be construed to cover both the singular and the plural, unless othenNise indicated herein or y dicted by context.
Unless otherwise stated, all exact values provided herein are representative of cor- responding approximate values (e.g., all exact exemplary values provided with t to a particular factor or measurement can be considered to also provide a corresponding approx— imate measurement, modified by "about," where appropriate). Where "about" is used in con- nection with a number, this can be ied as including values corresponding to +/-10% of the ied number.
The description herein of any aspect or embodiment of the ion using terms such as “comprising 71 H “-Including,” , having, 11 or “containing” with reference to an element or elements is intended to provide support for a similar aspect or embodiment of the invention that “consists of”, “consists essentially of”, or antially comprises” that particular element or elements, unless otherwise stated or clearly contradicted by context (e.g., a composition bed herein as comprising a particular element should be understood as also describing a composition consisting of that element, unless otherwise stated or clearly contradicted by context).
The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise d. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the inven- tion.

Claims (11)

THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS:
1. Use of an anti-natural killer cell lectin-like receptor C1 (NKG2A) antibody sing the CDR1, CDR2 and CDR3 domains of a heavy chain having the ce set forth in amino acid residues 31-35, 50-66 and 99-114 of any one of SEQ ID NOS: 4-8, and the CDR1, CDR2 and CDR3 domains of a light chain having the sequence set forth in amino acid residues 24-34, 50-56 and 89-97 of SEQ ID NO: 9 in the manufacture of a medicament for ng a major histocompatability complex IE (HLA-E)-expressing cancer, wherein the medicament is to be administered with an anti-programmed cell death 1 ligand 1 (PD-L1) antibody, and wherein the anti-NKG2A antibody is a non-depleting antibody.
2. The use of claim 1, wherein the medicament is to be administered in at least two doses in an amount effective to achieve a continuous blood concentration of anti-NKG2A antibody of at least 10 μg/ml for at least one week following administration thereof.
3. The use of claim 1 or claim 2, wherein the medicament is to be administered in at least one administration cycle, wherein for each cycle the medicament is to be stered in two, three or four doses and the D-L1 antibody is to be administered in two, three or four doses.
4. The use of claim 3, wherein the cycle is a period of eight weeks.
5. The use any one of claims 1 to 4, wherein the medicament and the D-L1 are for separate administration and are to be administered sequentially.
6. The use of any one of claims 1 to 4, wherein the medicament and the D-L1 are to be administered concurrently.
7. The use of any one of claims 1 to 6, wherein the cancer is a solid tumor.
8. The use of any one of claims 1 to 6, n the cancer is hematological tumor.
9. The use of claim 7, wherein the cancer is selected from the group consisting of lung cancer, renal cell carcinoma, melanoma, colorectal cancer, and ovarian cancer.
10. The use of any one of claims 1 to 9 , n the anti-NKG2A antibody is an lgG4 antibody or wherein the antibody lacks an Fc domain.
11. The use of any one of claims 1 to 10, wherein the anti-PD-L1 antibody is selected from the group consisting of nivolumab and atezolizumab.
NZ729207A 2014-09-16 2015-09-15 Neutralization of inhibitory pathways in lymphocytes NZ729207B2 (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US201462050948P 2014-09-16 2014-09-16
US62/050,948 2014-09-16
US201462083929P 2014-11-25 2014-11-25
US62/083,929 2014-11-25
US201462093141P 2014-12-17 2014-12-17
US62/093,141 2014-12-17
PCT/EP2015/071069 WO2016041945A1 (en) 2014-09-16 2015-09-15 Neutralization of inhibitory pathways in lymphocytes

Publications (2)

Publication Number Publication Date
NZ729207A NZ729207A (en) 2021-08-27
NZ729207B2 true NZ729207B2 (en) 2021-11-30

Family

ID=

Similar Documents

Publication Publication Date Title
US11572410B2 (en) Neutralization of inhibitory pathways in lymphocytes
US20230303691A1 (en) Neutralization of inhibitory pathways in lymphocytes
AU2015316993B2 (en) Treatment regimens using anti-NKG2A antibodies
AU2019270277A1 (en) Treatment of cancer
NZ729207B2 (en) Neutralization of inhibitory pathways in lymphocytes