WO2019211370A1 - Méthodes et compositions pharmaceutiques pour le traitement du cancer - Google Patents

Méthodes et compositions pharmaceutiques pour le traitement du cancer Download PDF

Info

Publication number
WO2019211370A1
WO2019211370A1 PCT/EP2019/061228 EP2019061228W WO2019211370A1 WO 2019211370 A1 WO2019211370 A1 WO 2019211370A1 EP 2019061228 W EP2019061228 W EP 2019061228W WO 2019211370 A1 WO2019211370 A1 WO 2019211370A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
btn3
btla
antibody
cancer
Prior art date
Application number
PCT/EP2019/061228
Other languages
English (en)
Inventor
Daniel Olive
Christine Pasero
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Centre National De La Recherche Scientifique (Cnrs)
Institut Jean Paoli & Irene Calmettes
Université D'aix Marseille
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 INSERM (Institut National de la Santé et de la Recherche Médicale), Centre National De La Recherche Scientifique (Cnrs), Institut Jean Paoli & Irene Calmettes, Université D'aix Marseille filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2019211370A1 publication Critical patent/WO2019211370A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to methods and pharmaceutical compositions for treating cancer in a subject in need thereof.
  • White blood cells are cells of the immune system involved in defending the body against pathogens. Among these cells, lymphocytes, monocytes, and dendritic cells can be cited. Monocytes may migrate from the bloodstream to other tissues and differentiate into tissue resident macrophages or dendritic cells. Dendritic cells play a role as antigen presenting cells (APC) that activate lymphocytes. Among lymphocytes, T cells can be divided into gd T cells and ab T cells.
  • APC antigen presenting cells
  • Vy9V02 cells represent a major peripheral blood T-cell subset in humans with broad reactivity against microbial agents and tumors. Unlike abT cells, gdT cells do not require the help of conventional major histocompatibility complex (MHC) class I and class II molecules for recognizing the antigens. Strikingly, gdT cells have emerged as the most favorable prognostic immune population among many cancer types. However, the mechanisms of gd T- cell immune responses to tumors remain poorly understood.
  • MHC major histocompatibility complex
  • Vy9V02 cells reserve a significant intracellular pool of PD- 1 located in early endosomes, which is mobilized at the cell membrane after bisphosphonate activation.
  • PD-l is a major regulator of Vy9Vd2 T-cell homeostasis, similarly to BTLA, another member of the Ig superfamily, for which the inventors previously demonstrated a key role in the regulation of TCR-independent and TCR-dependent Vy9V02 T- cell proliferation.
  • PD-l and BTLA differ in their expression pattern in Vy9V02 T cells and interestingly, combined blockade with monoclonal antibodies synergizes to improve Vy9V02 T cell expansion compared to single receptor blockade.
  • the inventors show that gdT cells infiltrate prostate tumors and overexpress PD-l, in addition to BTLA and other co-inhibitory molecules.
  • the upregulation of PD-l intracellular pool appears as a possible mechanism of immune escape by tumor cells, that upregulate PD-L1 and HVEM, by interfering with gdT cell antitumor activity.
  • the inventors show that PD-l and BTLA blockade improves Vy9V02 T cell proliferation and antitumor functions toward tumor cells.
  • the present invention highlight a new role for PD-l and the potent combination with BTLA in the control of Ug9U ⁇ 2 cells.
  • the present invention relates to methods and pharmaceutical compositions for treating cancer in a subject in need thereof.
  • the present invention also relates to a compound selected from the group consisting of PD1 antagonist or PD1 expression inhibitor in combination with a compound selected from the group consisting of BTLA antagonist or BTLA expression inhibitor and/or compound selected from the group consisting of BTN3 agonist or BTN3 expression activator for use in the treatment of cancer in a subject in need thereof
  • the present invention highlights a new role for PD-l and the potent combination with BTLA and BTN3 in the control of Vy9V52 cells.
  • the present invention relates to a compound selected from the group consisting of PD-l antagonist or PD-l expression inhibitor in combination with a compound selected from the group consisting of BTLA antagonist or BTLA expression inhibitor and/or compound selected from the group consisting of BTN3 agonist or BTN3 expression activator for use in the treatment of cancer in a subject in need thereof.
  • the combination of the invention is particularly suitable for increasing the proliferation of Vy9V52 T cells.
  • the combination of the invention is particularly suitable for Vy9V02 T-cell activation.
  • the term“combination” refers to either a fixed dose combination in one unit dosage form, non- fixed dose combination, or a kit of parts for the combined administration where a compound of the present invention, typically a PD-l antagonist, and one or more combination partners (e.g. another compound selected from BTN3 agonist and/or BTLA antagonist as explained below) may be administered independently at the same time or separately within time intervals, especially where these time intervals allow the combination partners show a cooperative, e.g.
  • fixed dose combination means that the active ingredients are both administered to a patient simultaneously in the form of a single entity or dosage.
  • non-fixed dose combination means that the active ingredients, e.g. a compound of the present invention such PD- 1 antagonist and one or more combination partners, e.g. BTN3 agonist and/or BTLA antagonist, are both administered to a patient as separate entities either simultaneously or sequentially, with no specific time limits, wherein such administration provides therapeutically effective levels of the two compounds in the body of the patient.
  • active ingredients e.g. a compound of the present invention such PD- 1 antagonist and one or more combination partners, e.g. BTN3 agonist and/or BTLA antagonist
  • joint therapeutically effective means that the therapeutic agents may be given separately (in a chronologically staggered manner, especially a sequence-specific manner) in such time intervals to show a (preferably synergistic) interaction (i.e. joint therapeutic effect).
  • the term“medicament” means a pharmaceutical composition, or a combination of several pharmaceutical compositions, which contains one or more active ingredients in the presence of one or more excipients.
  • the term“subject” denotes a mammal.
  • a subject according to the invention refers to any subject (preferably human) afflicted or at risk to be afflicted with cancer.
  • the term“subject” refers to a subject afflicted or at risk to be afflicted with pancreatic cancer.
  • the term“subject” refers to a subject afflicted or at risk to be afflicted with prostate cancer.
  • treatment refers to both prophylactic or preventive treatment as well as curative or disease modifying treatment, including treatment of subjects at risk of contracting the disease or suspected to have contracted the disease as well as subjects who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse.
  • the treatment may be administered to a subject having a medical disorder or who ultimately may acquire the disorder, in order to prevent, cure, delay the onset of, reduce the severity of, or ameliorate one or more symptoms of a disorder or recurring disorder, or in order to prolong the survival of a subject beyond that expected in the absence of such treatment.
  • therapeutic regimen is meant the pattern of treatment of an illness, e.g., the pattern of dosing used during therapy.
  • a therapeutic regimen may include an induction regimen and a maintenance regimen.
  • the phrase “induction regimen” or “induction period” refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the initial treatment of a disease.
  • the general goal of an induction regimen is to provide a high level of drug to a subject during the initial period of a treatment regimen.
  • An induction regimen may employ (in part or in whole) a "loading regimen", which may include administering a greater dose of the drug than a physician would employ during a maintenance regimen, administering a drug more frequently than a physician would administer the drug during a maintenance regimen, or both.
  • maintenance regimen refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the maintenance of a subject during treatment of an illness, e.g., to keep the subject in remission for long periods of time (months or years).
  • a maintenance regimen may employ continuous therapy (e.g., administering a drug at a regular intervals, e.g., weekly, monthly, yearly, etc.) or intermittent therapy (e.g., interrupted treatment, intermittent treatment, treatment at relapse, or treatment upon achievement of a particular predetermined criteria [e.g., disease manifestation, etc.]).
  • cancer has its general meaning in the art and includes, but is not limited to, solid tumors and blood borne tumors.
  • the term cancer includes diseases of the skin, tissues, organs, bone, cartilage, blood and vessels.
  • the term “cancer” further encompasses both primary and metastatic cancers. Examples of cancers that may be treated by methods and compositions of the present invention include, but are not limited to, cancer cells from the bladder, blood, bone, bone marrow, brain, breast, colon, esophagus, gastrointestine, gum, head, kidney, liver, lung, nasopharynx, neck, ovary, prostate, skin, stomach, testis, tongue, or uterus.
  • the cancer may specifically be of the following histological type, though it is not limited to these: neoplasm, malignant; carcinoma; carcinoma, undifferentiated; giant and spindle cell carcinoma; small cell carcinoma; papillary carcinoma; squamous cell carcinoma; lympho epithelial carcinoma; basal cell carcinoma; pilomatrix carcinoma; transitional cell carcinoma; papillary transitional cell carcinoma; adenocarcinoma; gastrinoma, malignant; cholangiocarcinoma; hepatocellular carcinoma; combined hepatocellular carcinoma and cholangiocarcinoma; trabecular adenocarcinoma; adenoid cystic carcinoma; adenocarcinoma in adenomatous polyp; adenocarcinoma, familial polyposis coli; solid carcinoma; carcinoid tumor, malignant; branchiolo-alveolar adenocarcinoma; papillary adenocarcinoma; chromophobe carcinoma; acid
  • the subject suffers from a cancer selected from the group consisting of prostate cancer, pancreatic cancer, colon cancer, rectal cancer, breast cancer, lung cancer, testicular cancer, brain cancer, skin cancer, gastric cancer, esophageal cancer, sarcomas, tracheal cancer, head and neck cancer, liver cancer, ovarian cancer, lymphoid cancer, cervical cancer, vulvar cancer, melanoma, mesothelioma, renal cancer, bladder cancer, thyroid cancer, bone cancers, carcinomas, sarcomas, and soft tissue cancers.
  • a cancer selected from the group consisting of prostate cancer, pancreatic cancer, colon cancer, rectal cancer, breast cancer, lung cancer, testicular cancer, brain cancer, skin cancer, gastric cancer, esophageal cancer, sarcomas, tracheal cancer, head and neck cancer, liver cancer, ovarian cancer, lymphoid cancer, cervical cancer, vulvar cancer, melanoma, mesothelioma,
  • the subject suffers from cancer resistant to anti-cancer treatment.
  • prostate cancer has its general meaning in the art and refers to prostate cancer such as revised in the World Health Organization Classification.
  • the term “prostate cancer” also refers to any type of prostate cancer and selected from the group of: Malignant neoplasm of prostate (C61); low and high grade dysplasia of prostate (N42.3, D07.5); Benign neoplasm of prostate (D21.1); Neoplasm of uncertain or unknown behavior of prostate (D40); localized prostate cancer; advanced prostate cancer; locally advanced prostate cancer; metastatic prostate cancer; hormone-sensitive prostate cancer (HSPCs); castration-resistant (CRPCs) prostate cancer.
  • the term“PD-l” has its general meaning in the art and refers to the programmed death- 1 receptor.
  • the term“PD-l” as used herein is intended to designate a type I transmembrane protein, belonging to the CD28-B7 signalling family of receptors that includes CD28, cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), inducible costimulator (ICOS), and B- and T- lymphocyte attenuator (BTLA) (Greenwald RJ et ah, 2005, Riley JL et aL., 2005).
  • CTLA-4 cytotoxic T-lymphocyte-associated antigen 4
  • ICOS inducible costimulator
  • BTLA B- and T- lymphocyte attenuator
  • BTLA has its general meaning in the art and refers to B and T lymphocyte attenuator.
  • the term“BTLA” also refers to CD272, a member of the CD28-B7 signalling family of receptors that includes CD28, cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), inducible costimulator (ICOS), and programmed death- 1 receptor (PD-l) (Greenwald RJ et al, 2005, Riley JL et aL., 2005).
  • CTLA-4 cytotoxic T-lymphocyte-associated antigen 4
  • ICOS inducible costimulator
  • PD-l programmed death- 1 receptor
  • BTN3 has its general meaning in the art and refers to butyrophilin (BT) belonging to the BT3 family (Williams and Barclay, 1988; Ruddy et al, 1997; Rhodes et al, 2001).
  • BT butyrophilin
  • BT3.1 is also called BTF5 (Rhodes et al., 2001), or BTN3A1 (Ruddy et al., 1997), or more recently CD277 (Bensussan and Olive, 2005);
  • BT3.2 is also called BTF4 (Rhodes et al, 2001), or BTN3A2 (Ruddy et al, 1997);
  • BT3.3 appears also as BTF3 (Rhodes et al., 2001) or BTN3A3 (Ruddy et al, 1997).
  • BT3 has two Ig-like extracellular domains that characterize the IgSF.
  • the present invention relates to a compound selected from the group consisting of PD-l antagonist or PD-l expression inhibitor in combination with a compound selected from the group consisting of BTLA antagonist or BTLA expression inhibitor for use in the treatment of cancer in a subject in need thereof
  • the present invention relates to a compound selected from the group consisting of PD-l antagonist or PD-l expression inhibitor in combination with a compound selected from the group consisting of BTN3 agonist or BTN3 expression activator for use in the treatment of cancer in a subject in need thereof.
  • the present invention relates to a compound selected from the group consisting of PD-l antagonist or PD-l expression inhibitor in combination with a compound selected from the group consisting of BTLA antagonist or BTLA expression inhibitor and compound selected from the group consisting of BTN3 agonist or BTN3 expression activator for use in the treatment of cancer in a subject in need thereof
  • the present invention relates to a compound selected from the group consisting of BTLA antagonist or BTLA expression inhibitor in combination with a compound selected from the group consisting of BTN3 agonist or BTN3 expression activator for use in the treatment of cancer in a subject in need thereof.
  • the invention relates to combined preparation of the compounds of the invention for simultaneous, separate or sequential use in the treatment of cancer in a subject in need thereof.
  • the term“administration simultaneously” refers to administration of at least 2 active ingredients by the same route and at the same time or at substantially the same time.
  • administration separately refers to administration of at least 2 active ingredients at the same time or at substantially the same time by different routes.
  • administration sequentially refers to an administration of 2 active ingredients at different times, the administration route being identical or different.
  • the invention relates to a i) PD-l antagonist or PD-l expression inhibitor and a ii) BTLA antagonist or BTLA expression inhibitor, as a combination for simultaneous, separate or sequential use in the treatment of a cancer in a subject in need thereof.
  • the invention relates to a i) PD-l antagonist or PD-l expression inhibitor and a ii) BTN3 agonist or BTN3 expression activator, as a combination for simultaneous, separate or sequential use in the treatment of a cancer in a subject in need thereof
  • the invention relates to a i) PD-l antagonist or PD-l expression inhibitor, a ii) BTLA antagonist or BTLA expression inhibitor and a iii) BTN3 agonist or BTN3 expression activator, as a combination for simultaneous, separate or sequential use in the treatment of a cancer in a subject in need thereof
  • the invention relates to a i) BTN3 agonist or BTN3 expression activator, a ii) BTLA antagonist or BTLA expression inhibitor, as a combination for simultaneous, separate or sequential use in the treatment of a cancer in a subject in need thereof
  • the invention provides the use of a combination as described herein, in the manufacture of a medicament for treating cancer.
  • the invention provides a medicament containing separately or together, a i) PD-l antagonist or PD-l expression inhibitor, a ii) BTLA antagonist or BTLA expression inhibitor and a iii) BTN3 agonist or BTN3 expression activator.
  • the invention provides a method of treating cancer, comprising administering a jointly therapeutically effective amount of a i) PD-l antagonist or PD-l expression inhibitor, a ii) BTLA antagonist or BTLA expression inhibitor and a iii) BTN3 agonist or BTN3 expression activator, to a subject in need thereof.
  • a gene product can be the direct transcriptional product of a gene (e.g., mRNA, tRNA, rRNA, antisense RNA, ribozyme, structural RNA or any other type of RNA) or a protein produced by translation of a mRNA.
  • Gene products also include messenger RNAs, which are modified, by processes such as capping, polyadenylation, methylation, and editing, and proteins modified by, for example, methylation, acetylation, phosphorylation, ubiquitination, SUMOylation, ADP- ribosylation, myristilation, and glycosylation.
  • An“inhibitor of expression” refers to a natural or synthetic compound that has a biological effect to inhibit the expression of a gene.
  • An“activator of expression” refers to a natural or synthetic compound that has a biological effect to activate the expression of a gene.
  • PD-l antagonist has its general meaning in the art and refers to a compound that selectively inactivates the PD-L
  • the term“PD-l antagonist” refers to any compound that can directly or indirectly inhibits the signal transduction cascade related to the PD-l and inhibits PD-l ligand binding (PD-L1; PD-L2).
  • the term“selectively inactivates” refers to a compound that preferentially inactivates PD-l with a greater affinity and potency, respectively, than its interaction with the other sub-types or iso forms of the CD28-B7 signalling family of receptors (CD28; CTLA-4; ICOS; BTLA).
  • PD-l antagonist also refers to a compound that decrease the PD-l activity level.
  • Compounds that prefer PD-l, but that may also inactivate other CD28-B7 signalling family of receptors sub-types, as partial or full antagonists are contemplated.
  • a PD-l antagonist is a small organic molecule, a polypeptide, an aptamer or an antibody.
  • Tests and assays for determining whether a compound is a PD-l antagonist are well known by the skilled person in the art such as described in Greenwald et ah, 2005; Riley et al, 2005.
  • the PD-l antagonist is selected from the group consisting of an anti-PDl antibody, PD-l ligand, an anti-PD-Ll antibody or an anti-PD-L2 antibody such as described in the International Patent Application W02009/024531 ; WO2010/0089411.
  • the anti-PDl antibody is a monoclonal antibody obtainable from a hybridoma deposited at the Collection Nationale de Cultures de Microorganismes (CNCM) on April 12, 2007 in accordance with the terms of the Budapest Treaty under the accession number CNCM 1-3745.
  • the anti-PDl antibody is a PD-l antibody (PD 1.3) which is obtainable from the hybridoma accessible under CNCM deposit number 1-4122 or a humanized variant thereof.
  • the PD-l antagonist is PD-L1 antibody which is obtainable from the hybridoma accessible under CNCM deposit number 1-4080 (PDL1.1) or a humanized variant thereof.
  • the PD-l antagonist is PD-L1 antibody which is obtainable from the hybridoma accessible under CNCM deposit number 1-4081 (PDL1.2) or a humanized variant thereof.
  • the antibodies of the present invention compete for binding to PD-l (and PD-L1) antibodies described above.
  • BTLA antagonist has its general meaning in the art and refers to a compound that selectively inactivates the BTLA.
  • the term“BTLA antagonist” refers to any compound that can directly or indirectly inhibits the signal transduction cascade related to the BTLA.
  • selectively inactivates refers to a compound that preferentially inactivates BTLA with a greater affinity and potency, respectively, than its interaction with the other sub-types or isoforms of the CD28-B7 signalling family of receptors (CD28; CTLA-4; ICOS; PD-l).
  • BTLA antagonist also refers to a compound that decrease the BTLA activity level.
  • a BTLA antagonist is a small organic molecule, a polypeptide, an aptamer or an antibody.
  • Tests and assays for determining whether a compound is a BTLA antagonist are well known by the skilled person in the art such as described in (Greenwald et ah, 2005; Riley et al, 2005).
  • the BTLA antagonist is selected from the group consisting of anti-BTLA antibody, or BTLA ligand such as described in the International Patent Application WO2010/106051; WO2011/014438; WO2017/144668.
  • the anti-BTLA antibody is the BTLA antibody (BTLA8.2) which is obtainable from the hybridoma accessible under CNCM deposit number 1-4123 such as disclosed in WO2010/106051.
  • the anti-BTLA antibody is the 4C7 mAb disclosed in WO2011/014438.
  • the anti-BTLA antibody is the mAb 629.3 mAb disclosed in WO2017/144668, or its humanized version.
  • the antibodies of the present invention compete for binding to BTLA antibodies described above.
  • the compound of the invention is an aptamer.
  • Aptamers are a class of molecule that represents an alternative to antibodies in term of molecular recognition.
  • Aptamers are oligonucleotide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity.
  • Such ligands may be isolated through Systematic Evolution of Ligands by Exponential enrichment (SELEX) of a random sequence library, as described in Tuerk C. and Gold L., 1990.
  • the random sequence library is obtainable by combinatorial chemical synthesis of DNA. In this library, each member is a linear oligomer, eventually chemically modified, of a unique sequence.
  • Peptide aptamers consists of a conformationally constrained antibody variable region displayed by a platform protein, such as E. coli Thioredoxin A that are selected from combinatorial libraries by two hybrid methods (Colas et al., 1996). Then after raising aptamers directed against the target of the invention as above described, the skilled man in the art can easily select those blocking or inactivating the target.
  • a platform protein such as E. coli Thioredoxin A
  • the compound of the invention is an antibody (the term including“antibody portion”) directed against the target and which is a PD-l antagonist or BTLA antagonist.
  • the antibody is a monoclonal antibody. In one embodiment of the antibodies or portions thereof described herein, the antibody is a polyclonal antibody. In one embodiment of the antibodies or portions thereof described herein, the antibody is a humanized antibody. In one embodiment of the antibodies or portions thereof described herein, the antibody is a chimeric antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a light chain of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a heavy chain of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a Fab portion of the antibody.
  • the portion of the antibody comprises a F(ab')2 portion of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a Fc portion of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a Fv portion of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises a variable domain of the antibody. In one embodiment of the antibodies or portions thereof described herein, the portion of the antibody comprises one or more CDR domains of the antibody.
  • antibody includes both naturally occurring and non-naturally occurring antibodies. Specifically, “antibody” includes polyclonal and monoclonal antibodies, and monovalent and divalent fragments thereof. Furthermore, “antibody” includes chimeric antibodies, wholly synthetic antibodies, single chain antibodies, and fragments thereof. The antibody may be a human or nonhuman antibody. A nonhuman antibody may be humanized by recombinant methods to reduce its immunogenicity in man.
  • Antibodies are prepared according to conventional methodology. Monoclonal antibodies may be generated using the method of Kohler and Milstein (Nature, 256:495, 1975). To prepare monoclonal antibodies useful in the invention, a mouse or other appropriate host animal is immunized at suitable intervals (e.g., twice-weekly, weekly, twice-monthly or monthly) with antigenic forms of the target. The animal may be administered a final "boost" of antigen within one week of sacrifice. It is often desirable to use an immunologic adjuvant during immunization.
  • Suitable immunologic adjuvants include Freund's complete adjuvant, Freund's incomplete adjuvant, alum, Ribi adjuvant, Hunter's Titermax, saponin adjuvants such as QS21 or Quil A, or CpG-containing immunostimulatory oligonucleotides.
  • Other suitable adjuvants are well-known in the field.
  • the animals may be immunized by subcutaneous, intraperitoneal, intramuscular, intravenous, intranasal or other routes. A given animal may be immunized with multiple forms of the antigen by multiple routes.
  • the antigen may be provided as synthetic peptides corresponding to antigenic regions of interest in the target.
  • lymphocytes are isolated from the spleen, lymph node or other organ of the animal and fused with a suitable myeloma cell line using an agent such as polyethylene glycol to form a hydridoma.
  • cells are placed in media permissive for growth of hybridomas but not the fusion partners using standard methods, as described (Coding, Monoclonal Antibodies: Principles and Practice: Production and Application of Monoclonal Antibodies in Cell Biology, Biochemistry and Immunology, 3rd edition, Academic Press, New York, 1996).
  • cell supernatants are analyzed for the presence of antibodies of the desired specificity, i.e., that selectively bind the antigen.
  • Suitable analytical techniques include ELISA, flow cytometry, immunoprecipitation, and western blotting. Other screening techniques are well-known in the field. Preferred techniques are those that confirm binding of antibodies to conformationally intact, natively folded antigen, such as non-denaturing ELISA, flow cytometry, and immunoprecipitation.
  • an antibody from which the pFc' region has been enzymatically cleaved, or which has been produced without the pFc' region designated an F(ab’)2 fragment, retains both of the antigen binding sites of an intact antibody.
  • an antibody from which the Fc region has been enzymatically cleaved, or which has been produced without the Fc region designated an Fab fragment, retains one of the antigen binding sites of an intact antibody molecule.
  • Fab fragments consist of a covalently bound antibody light chain and a portion of the antibody heavy chain denoted Fd.
  • the Fd fragments are the major determinant of antibody specificity (a single Fd fragment may be associated with up to ten different light chains without altering antibody specificity) and Fd fragments retain epitope-binding ability in isolation.
  • CDRs complementarity determining regions
  • FRs framework regions
  • CDR1 through CDRS complementarity determining regions
  • the residues in antibody variable domains are conventionally numbered according to a system devised by Rabat et al. This system is set forth in Rabat et al, 1987, in Sequences of Proteins of Immunological Interest, US Department of Health and Human Services, NIH, USA (Rabat et al, 1992, hereafter“Rabat et al.”). This numbering system is used in the present specification.
  • the Rabat residue designations do not always correspond directly with the linear numbering of the amino acid residues in SEQ ID sequences.
  • the actual linear amino acid sequence may contain fewer or additional amino acids than in the strict Rabat numbering corresponding to a shortening of, or insertion into, a structural component, whether framework or complementarity determining region (CDR), of the basic variable domain structure.
  • the correct Rabat numbering of residues may be determined for a given antibody by alignment of residues of homology in the sequence of the antibody with a“standard” Rabat numbered sequence.
  • the CDRs of the heavy chain variable domain are located at residues 31-35 (H- CDR1), residues 50-65 (H-CDR2) and residues 95-102 (H-CDR3) according to the Rabat numbering system.
  • the CDRs of the light chain variable domain are located at residues 24-34 (L-CDR1), residues 50-56 (L-CDR2) and residues 89-97 (L-CDR3) according to the Rabat numbering system.
  • the second proposal was that if an amino acid in the framework of the human immunoglobulin is unusual and the donor amino acid at that position is typical for human sequences, then the donor amino acid rather than the acceptor may be selected.
  • the third proposal was that in the positions immediately adjacent to the 3 CDRs in the humanized immunoglobulin chain, the donor amino acid rather than the acceptor amino acid may be selected.
  • the fourth proposal was to use the donor amino acid reside at the framework positions at which the amino acid is predicted to have a side chain atom within 3 A of the CDRs in a three dimensional model of the antibody and is predicted to be capable of interacting with the CDRs.
  • the above methods are merely illustrative of some of the methods that one skilled in the art could employ to make humanized antibodies. One of ordinary skill in the art will be familiar with other methods for antibody humanization.
  • humanized forms of the antibodies some, most or all of the amino acids outside the CDR regions have been replaced with amino acids from human immunoglobulin molecules but where some, most or all amino acids within one or more CDR regions are unchanged. Small additions, deletions, insertions, substitutions or modifications of amino acids are permissible as long as they would not abrogate the ability of the antibody to bind a given antigen.
  • Suitable human immunoglobulin molecules would include IgGl, IgG2, IgG3, IgG4, IgA and IgM molecules.
  • a "humanized" antibody retains a similar antigenic specificity as the original antibody.
  • the affinity and/or specificity of binding of the antibody may be increased using methods of "directed evolution", as described by Wu et ah, /. Mol. Biol. 294: 151, 1999, the contents of which are incorporated herein by reference.
  • Fully human monoclonal antibodies also can be prepared by immunizing mice transgenic for large portions of human immunoglobulin heavy and light chain loci. See, e.g., U.S. Pat. Nos. 5,591,669, 5,598,369, 5,545,806, 5,545,807, 6,150,584, and references cited therein, the contents of which are incorporated herein by reference. These animals have been genetically modified such that there is a functional deletion in the production of endogenous (e.g., murine) antibodies. The animals are further modified to contain all or a portion of the human germ-line immunoglobulin gene locus such that immunization of these animals will result in the production of fully human antibodies to the antigen of interest.
  • monoclonal antibodies can be prepared according to standard hybridoma technology. These monoclonal antibodies will have human immunoglobulin amino acid sequences and therefore will not provoke human anti-mouse antibody (KAMA) responses when administered to humans.
  • KAMA human anti-mouse antibody
  • the present invention also provides for F(ab') 2 Fab, Fv and Fd fragments; chimeric antibodies in which the Fc and/or FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; chimeric F(ab')2 fragment antibodies in which the FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; chimeric Fab fragment antibodies in which the FR and/or CDR1 and/or CDR2 and/or light chain CDR3 regions have been replaced by homologous human or non-human sequences; and chimeric Fd fragment antibodies in which the FR and/or CDR1 and/or CDR2 regions have been replaced by homologous human or non human sequences.
  • the present invention also includes so-called single chain antibodies.
  • the various antibody molecules and fragments may derive from any of the commonly known immunoglobulin classes, including but not limited to IgA, secretory IgA, IgE, IgG and IgM.
  • IgG subclasses are also well known to those in the art and include but are not limited to human IgGl, IgG2, IgG3 and IgG4.
  • the compound of the invention is a Human IgG4.
  • the antibody according to the invention is a single domain antibody.
  • the term“single domain antibody” (sdAb) or “VHH” refers to the single heavy chain variable domain of antibodies of the type that can be found in Camelid mammals which are naturally devoid of light chains. Such VHH are also called“nanobody®”. According to the invention, sdAb can particularly be llama sdAb.
  • the term“VHH” refers to the single heavy chain having 3 complementarity determining regions (CDRs): CDR1, CDR2 and CDR3.
  • CDRs complementarity determining region
  • CDR complementarity determining region
  • VHH according to the invention can readily be prepared by an ordinarily skilled artisan using routine experimentation.
  • VHH variants and modified form thereof may be produced under any known technique in the art such as in-vitro maturation.
  • VHHs or sdAbs are usually generated by PCR cloning of the V-domain repertoire from blood, lymph node, or spleen cDNA obtained from immunized animals into a phage display vector, such as pHEN2.
  • Antigen- specific VHHs are commonly selected by panning phage libraries on immobilized antigen, e.g., antigen coated onto the plastic surface of a test tube, biotinylated antigens immobilized on streptavidin beads, or membrane proteins expressed on the surface of cells.
  • immobilized antigen e.g., antigen coated onto the plastic surface of a test tube, biotinylated antigens immobilized on streptavidin beads, or membrane proteins expressed on the surface of cells.
  • VHHs often show lower affinities for their antigen than VHHs derived from animals that have received several immunizations.
  • VHHs from immune libraries are attributed to the natural selection of variant VHHs during clonal expansion of B-cells in the lymphoid organs of immunized animals.
  • the affinity of VHHs from non-immune libraries can often be improved by mimicking this strategy in vitro, i.e., by site directed mutagenesis of the CDR regions and further rounds of panning on immobilized antigen under conditions of increased stringency (higher temperature, high or low salt concentration, high or low pH, and low antigen concentrations).
  • VHHs derived from camelid are readily expressed in and purified from the E. coli periplasm at much higher levels than the corresponding domains of conventional antibodies.
  • VHHs generally display high solubility and stability and can also be readily produced in yeast, plant, and mammalian cells.
  • the“Hamers patents” describe methods and techniques for generating VHH against any desired target (see for example US 5,800,988; US 5,874, 541 and US 6,015,695).
  • The“Hamers patents” more particularly describe production of VHHs in bacterial hosts such as E. coli (see for example US 6,765,087) and in lower eukaryotic hosts such as moulds (for example Aspergillus or Trichoderma) or in yeast (for example Saccharomyces, Kluyveromyces, Hansenula or Pichia) (see for example US 6,838,254).
  • the invention provides an antibody that competes for binding to the target with the antibody of the invention.
  • binding in the context of the binding of an antibody to a predetermined antigen or epitope typically is a binding with an affinity corresponding to a KD of about 10-7 M or less, such as about 10-8 M or less, such as about 10-9 M or less, about 10- 10 M or less, or about 10-11 M or even less when determined by for instance surface plasmon resonance (SPR) technology in a BIAcore 3000 instrument using a soluble form of the antigen as the ligand and the antibody as the analyte.
  • SPR surface plasmon resonance
  • BIACORE® GE Healthcare, Piscaataway, NJ
  • BIACORE® is one of a variety of surface plasmon resonance assay formats that are routinely used to epitope bin panels of monoclonal antibodies.
  • an antibody binds to the predetermined antigen with an affinity corresponding to a KD that is at least ten- fold lower, such as at least lOO-fold lower, for instance at least 1, 000-fold lower, such as at least 10,000-fold lower, for instance at least 100,000-fold lower than its KD for binding to a non-specific antigen (e.g., BSA, casein), which is not identical or closely related to the predetermined antigen.
  • a non-specific antigen e.g., BSA, casein
  • An antibody is said to essentially not bind an antigen or epitope if such binding is either not detectable (using, for example, plasmon resonance (SPR) technology in a BIAcore 3000 instrument using a soluble form of the antigen as the ligand and the antibody as the analyte), or is 100 fold, 500 fold, 1000 fold or more than 1000 fold less than the binding detected by that antibody and an antigen or epitope having a different chemical structure or amino acid sequence.
  • SPR plasmon resonance
  • Additional antibodies can be identified based on their ability to cross-compete (e.g., to competitively inhibit the binding of, in a statistically significant manner) with other antibodies of the invention in standard antigen binding assays.
  • the ability of a test antibody to inhibit the binding of antibodies of the present invention to the target demonstrates that the test antibody can compete with that antibody for binding to the target; such an antibody may, according to non-limiting theory, bind to the same or a related (e.g., a structurally similar or spatially proximal) epitope on the target as the antibody with which it competes.
  • another aspect of the invention provides antibodies that bind to the same antigen as, and compete with, the antibodies disclosed herein.
  • the antibodies or antigen binding fragments of the invention bind to one or more epitopes of the target.
  • the epitopes to which the present antibodies or antigen binding fragments bind are linear epitopes. In other embodiments, the epitopes to which the present antibodies or antigen binding fragments bind are non-linear, conformational epitopes.
  • the compound of the invention is a PD-l expression inhibitor or a BTLA expression inhibitor.
  • An“inhibitor of expression” refers to a natural or synthetic compound that has a biological effect to inhibit the expression of a gene.
  • the target expression inhibitors for use in the present invention may be based on antisense oligonucleotide constructs.
  • Anti-sense oligonucleotides including anti-sense RNA molecules and anti-sense DNA molecules, would act to directly block the translation of the target mRNA by binding thereto and thus preventing protein translation or increasing mRNA degradation, thus decreasing the level of the target proteins, and thus activity, in a cell.
  • antisense oligonucleotides of at least about 15 bases and complementary to unique regions of the mRNA transcript sequence encoding the target can be synthesized, e.g., by conventional phosphodiester techniques and administered by e.g., intravenous injection or infusion.
  • Small inhibitory RNAs can also function as a target expression inhibitors for use in the present invention.
  • the target gene expression can be reduced by contacting the subject or cell with a small double stranded RNA (dsRNA), or a vector or construct causing the production of a small double stranded RNA, such that the target expression is specifically inhibited (i.e. RNA interference or RNAi).
  • dsRNA small double stranded RNA
  • RNAi RNA interference
  • Methods for selecting an appropriate dsRNA or dsRNA-encoding vector are well known in the art for genes whose sequence is known (e.g. see Tuschl, T. et al. (1999); Elbashir, S. M. et al. (2001); Hannon, GJ. (2002); McManus, MT. et al.
  • Ribozymes can also function as a target expression inhibitors for use in the present invention.
  • Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA.
  • the mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage.
  • Engineered hairpin or hammerhead motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of the target mRNA sequences are thereby useful within the scope of the present invention.
  • ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites, which typically include the following sequences, GUA, GUU, and GUC. Once identified, short RNA sequences of between about 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site can be evaluated for predicted structural features, such as secondary structure, that can render the oligonucleotide sequence unsuitable. The suitability of candidate targets can also be evaluated by testing their accessibility to hybridization with complementary oligonucleotides, using, e.g., ribonuclease protection assays.
  • antisense oligonucleotides and ribozymes useful a target inhibitors can be prepared by known methods. These include techniques for chemical synthesis such as, e.g., by solid phase phosphoramadite chemical synthesis. Alternatively, anti-sense RNA molecules can be generated by in vitro or in vivo transcription of DNA sequences encoding the RNA molecule. Such DNA sequences can be incorporated into a wide variety of vectors that incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters. Various modifications to the oligonucleotides of the invention can be introduced as a means of increasing intracellular stability and half-life.
  • Possible modifications include but are not limited to the addition of flanking sequences of ribonucleotides or deoxyribonucleotides to the 5' and/or 3’ ends of the molecule, or the use of phosphorothioate or 2’-0-methyl rather than phosphodiesterase linkages within the oligonucleotide backbone.
  • Antisense oligonucleotides siRNAs and ribozymes of the invention may be delivered in vivo alone or in association with a vector.
  • a "vector" is any vehicle capable of facilitating the transfer of the antisense oligonucleotide siRNA or ribozyme nucleic acid to the cells and preferably cells expressing the target.
  • the vector transports the nucleic acid to cells with reduced degradation relative to the extent of degradation that would result in the absence of the vector.
  • the vectors useful in the invention include, but are not limited to, plasmids, phagemids, viruses, other vehicles derived from viral or bacterial sources that have been manipulated by the insertion or incorporation of the antisense oligonucleotide siRNA or ribozyme nucleic acid sequences.
  • Viral vectors are a preferred type of vector and include, but are not limited to nucleic acid sequences from the following viruses: retrovirus, such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma virus; adenovirus, adeno-associated virus; SV40-type viruses; polyoma viruses; Epstein-Barr viruses; papilloma viruses; herpes virus; vaccinia virus; polio virus; and RNA virus such as a retrovirus.
  • retrovirus such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma virus
  • retrovirus such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma virus
  • adenovirus adeno
  • Non-cytopathic viral vectors are based on non-cytopathic eukaryotic viruses in which non- essential genes have been replaced with the gene of interest.
  • Non-cytopathic viruses include retroviruses (e.g., lentivirus), the life cycle of which involves reverse transcription of genomic viral RNA into DNA with subsequent proviral integration into host cellular DNA.
  • Retroviruses have been approved for human gene therapy trials. Most useful are those retroviruses that are replication-deficient (i.e., capable of directing synthesis of the desired proteins, but incapable of manufacturing an infectious particle).
  • retroviral expression vectors have general utility for the high-efficiency transduction of genes in vivo.
  • Standard protocols for producing replication-deficient retroviruses including the steps of incorporation of exogenous genetic material into a plasmid, transfection of a packaging cell lined with plasmid, production of recombinant retroviruses by the packaging cell line, collection of viral particles from tissue culture media, and infection of the target cells with viral particles
  • KRIEGLER A Laboratory Manual
  • MURRY Method of Recombinant retroviruses by the packaging cell line
  • Methods in Molecular Biology vol.7, Humana Press, Inc., Cliffton, N.J., 1991.
  • adeno-viruses and adeno-associated viruses are double-stranded DNA viruses that have already been approved for human use in gene therapy.
  • the adeno-associated virus can be engineered to be replication deficient and is capable of infecting a wide range of cell types and species. It further has advantages such as, heat and lipid solvent stability; high transduction frequencies in cells of diverse lineages, including hemopoietic cells; and lack of superinfection inhibition thus allowing multiple series of transductions.
  • the adeno-associated virus can integrate into human cellular DNA in a site-specific manner, thereby minimizing the possibility of insertional mutagenesis and variability of inserted gene expression characteristic of retroviral infection.
  • adeno-associated virus infections have been followed in tissue culture for greater than 100 passages in the absence of selective pressure, implying that the adeno-associated virus genomic integration is a relatively stable event.
  • the adeno-associated virus can also function in an extrachromosomal fashion.
  • Plasmid vectors have been extensively described in the art and are well known to those of skill in the art. See e.g., SANBROOK et al, "Molecular Cloning: A Laboratory Manual," Second Edition, Cold Spring Harbor Laboratory Press, 1989. In the last few years, plasmid vectors have been used as DNA vaccines for delivering antigen encoding genes to cells in vivo. They are particularly advantageous for this because they do not have the same safety concerns as with many of the viral vectors. These plasmids, however, having a promoter compatible with the host cell, can express a peptide from a gene operatively encoded within the plasmid.
  • Plasmids may be delivered by a variety of parenteral, mucosal and topical routes.
  • the DNA plasmid can be injected by intramuscular, intradermal, subcutaneous, or other routes. It may also be administered by intranasal sprays or drops, rectal suppository and orally.
  • the plasmids may be given in an aqueous solution, dried onto gold particles or in association with another DNA delivery system including but not limited to liposomes, dendrimers, cochleate and microencapsulation.
  • BTN3 agonist has its general meaning in the art and refers to a compound that selectively activates the BTN3.
  • the term“BTN3 agonist” refers to any compound that can directly or indirectly stimulate the signal transduction cascade related to the BTN3.
  • the term“selectively activates” refers to a compound that preferentially binds to and activates BTN3 with a greater affinity and potency, respectively, than its interaction with the other sub-types or isoforms of the butyrophilin (BT) family (BT2).
  • BT butyrophilin
  • Compounds that prefer BTN3, but that may also activate other butyrophilin (BT) sub-types, as partial or full agonists, and thus that may have multiple BTN3 activities are contemplated.
  • a BTN3 agonist is a small organic molecule, an antibody or a polypeptide.
  • Tests and assays for determining whether a compound is a BTN3 agonist are well known by the skilled person in the art such as described in Williams and Barclay, 1988; Ruddy et al, 1997; Rhodes et al., 2001.
  • the BTN3 agonist is an antibody directed against the target such as described above and which is a BTN3 agonist.
  • the BTN3 agonist is selected from the group consisting of antibodies such as described in the International Patent Application W02012/080769; W02012/080351.
  • the anti-BTN3 antibody is selected from mAh 20.1, mAh 7.2 and mAh 103.2 which is obtainable from one of the hybridomas accessible under CNCM deposit number 1-4401, 1-4402 and 1-4403 such as described in W02012/080769; W02012/080351.
  • the anti-BTN3 antibody is a humanized form of the antibodies 20.1, 7.2 or 103.2 and/or comprises the six CDRs of the antibodies 20.1, 7.2 or 103.2.
  • the antibodies of the present invention compete for binding to BTN3 antibodies described above.
  • the compound of the present invention is administered sequentially or concomitantly with one or more therapeutic active agent such as to anti-cancer compound, chemotherapeutic or radiotherapeutic.
  • anti-cancer compound has its general meaning in the art and refers to anti cancer compounds used in anti-cancer therapy such as tyrosine kinase inhibitors, tyrosine kinase receptor (TKR) inhibitors, EGFR tyrosine kinase inhibitors, anti-EGFR compounds, anti-HER2 compounds, Vascular Endothelial Growth Factor Receptors (VEGFRs) pathway inhibitors, interferon therapy, alkylating agents, anti-metabolites, immunotherapeutic agents, Interferons (IFNs), Interleukins, and chemotherapeutic agents such as described below.
  • TLR tyrosine kinase receptor
  • EGFR tyrosine kinase inhibitors anti-EGFR compounds
  • anti-HER2 compounds anti-HER2 compounds
  • VEGFRs Vascular Endothelial Growth Factor Receptors pathway inhibitors
  • interferon therapy alkylating agents
  • anti-metabolites anti-metabolites
  • tyrosine kinase inhibitor has its general meaning in the art and refers to any of a variety of therapeutic agents or drugs such as compounds inhibiting tyrosine kinase, tyrosine kinase receptor inhibitors (TKRI), EGFR tyrosine kinase inhibitors, EGFR antagonists.
  • TKRI tyrosine kinase receptor inhibitors
  • EGFR tyrosine kinase inhibitors
  • EGFR antagonists EGFR antagonists.
  • tyrosine kinase inhibitor has its general meaning in the art and refers to any of a variety of therapeutic agents or drugs that act as selective or non-selective inhibitors of receptor and/or non-receptor tyrosine kinases.
  • Tyrosine kinase inhibitors and related compounds are well known in the art and described in U.S Patent Publication 2007/0254295, which is incorporated by reference herein in its entirety. It will be appreciated by one of skill in the art that a compound related to a tyrosine kinase inhibitor will recapitulate the effect of the tyrosine kinase inhibitor, e.g., the related compound will act on a different member of the tyrosine kinase signaling pathway to produce the same effect as would a tyrosine kinase inhibitor of that tyrosine kinase.
  • tyrosine kinase inhibitors and related compounds suitable for use in methods of embodiments of the present invention include, but are not limited to Erlotinib, sunitinib (Sutent; SU11248), dasatinib (BMS-354825), PP2, BEZ235, saracatinib, gefitinib (Iressa), erlotinib (Tarceva; OSI-1774), lapatinib (GW572016; GW2016), canertinib (Cl 1033), semaxinib (SU5416), vatalanib (PTK787/ZK222584), sorafenib (BAY 43-9006), imatinib (Gleevec; STI571), leflunomide (SU101), vandetanib (Zactima; ZD6474), MK-2206 (8-[4-aminocyclobutyl)phenyl]-9-phenyl-l,2,4-tria
  • the tyrosine kinase inhibitor is a small molecule kinase inhibitor that has been orally administered and that has been the subject of at least one Phase I clinical trial, more preferably at least one Phase II clinical, even more preferably at least one Phase III clinical trial, and most preferably approved by the FDA for at least one hematological or oncological indication.
  • inhibitors include, but are not limited to Erlotinib, Gefitinib, Lapatinib, Canertinib, BMS-599626 (AC-480), Neratinib, KRN-633, CEP-11981, Imatinib, Nilotinib, Dasatinib, AZM-475271, CP-724714, TAK-165, Sunitinib, Vatalanib, CP- 547632, Vandetanib, Bosutinib, Lestaurtinib, Tandutinib, Midostaurin, Enzastaurin, AEE-788, Pazopanib, Axitinib, Motasenib, OSI-930, Cediranib, KRN-951, Dovitinib, Seliciclib, SNS- 032, PD-0332991, MKC-I (Ro-3 l7453; R-440), Sorafenib, ABT-8
  • EGFR tyrosine kinase inhibitors as used herein include, but are not limited to compounds selected from the group consisting of but not limited to Erlotinib, lapatinib, Rociletinib (CO- 1686), gefitinib, Dacomitinib (PF-00299804), Afatanib, Brigatinib (AP26113), WJTOG3405, NEJ002, AZD9291, HM61713, EGF816, ASP 8273, AC 0010.
  • antibody EGFR inhibitors examples include Cetuximab, panitumumab, matuzumab, zalutumumab, nimotuzumab, necitumumab, Imgatuzumab (GA201, RO5083945), and ABT- 806.
  • the compound of the present invention is administered with a chemotherapeutic agent.
  • chemotherapeutic agent refers to chemical compounds that are effective in inhibiting tumor growth.
  • examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolo melamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin,
  • calicheamicin especially calicheamicin (11 and calicheamicin 211, see, e.g., Agnew Chem Intl. Ed. Engl. 33: 183-186 (1994); dynemicin, including dynemicin A; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromomophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, canninomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including morpholino- doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrol
  • paclitaxel (TAXOL®, Bristol-Myers Squibb Oncology, Princeton, N.].) and doxetaxel (TAXOTERE®, Rhone-Poulenc Rorer, Antony, France); chlorambucil; gemcitabine; 6- thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP- 16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; CPT-l 1 ; topoisomerase inhibitor RFS 2000; difluoromethylomithine (DMFO); retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
  • anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
  • anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide,
  • the compound of the present invention is administered with a targeted cancer therapy.
  • Targeted cancer therapies are drugs or other substances that block the growth and spread of cancer by interfering with specific molecules ("molecular targets") that are involved in the growth, progression, and spread of cancer.
  • Targeted cancer therapies are sometimes called “molecularly targeted drugs", “molecularly targeted therapies”, “precision medicines”, or similar names.
  • the targeted therapy consists of administering the subject with a tyrosine kinase inhibitor as defined above.
  • compound of the present invention is administered with an immunotherapeutic agent.
  • immunotherapeutic agent refers to a compound, composition or treatment that indirectly or directly enhances, stimulates or increases the body's immune response against cancer cells and/or that decreases the side effects of other anticancer therapies. Immunotherapy is thus a therapy that directly or indirectly stimulates or enhances the immune system's responses to cancer cells and/or lessens the side effects that may have been caused by other anti-cancer agents. Immunotherapy is also referred to in the art as immunologic therapy, biological therapy biological response modifier therapy and biotherapy.
  • immunotherapeutic agents examples include, but are not limited to, cytokines, cancer vaccines, monoclonal antibodies and non-cytokine adjuvants.
  • the immunotherapeutic treatment may consist of administering the subject with an amount of immune cells (T cells, NK, cells, dendritic cells, B cells).
  • Immunotherapeutic agents can be non-specific, i.e. boost the immune system generally so that the human body becomes more effective in fighting the growth and/or spread of cancer cells, or they can be specific, i.e. targeted to the cancer cells themselves immunotherapy regimens may combine the use of non-specific and specific immunotherapeutic agents.
  • Non-specific immunotherapeutic agents are substances that stimulate or indirectly improve the immune system.
  • Non-specific immunotherapeutic agents have been used alone as a main therapy for the treatment of cancer, as well as in addition to a main therapy, in which case the non-specific immunotherapeutic agent functions as an adjuvant to enhance the effectiveness of other therapies (e.g. cancer vaccines).
  • Non-specific immunotherapeutic agents can also function in this latter context to reduce the side effects of other therapies, for example, bone marrow suppression induced by certain chemotherapeutic agents.
  • Non-specific immunotherapeutic agents can act on key immune system cells and cause secondary responses, such as increased production of cytokines and immunoglobulins. Alternatively, the agents can themselves comprise cytokines.
  • Non specific immunotherapeutic agents are generally classified as cytokines or non-cytokine adjuvants.
  • cytokines have found application in the treatment of cancer either as general non-specific immunotherapies designed to boost the immune system, or as adjuvants provided with other therapies.
  • Suitable cytokines include, but are not limited to, interferons, interleukins and colony- stimulating factors.
  • Interferons (IFNs) contemplated by the present invention include the common types of IFNs, IFN-alpha (IFN-a), IFN-beta (IFN-b) and IFN- gamma (IFN-g).
  • IFNs can act directly on cancer cells, for example, by slowing their growth, promoting their development into cells with more normal behaviour and/or increasing their production of antigens thus making the cancer cells easier for the immune system to recognise and destroy.
  • IFNs can also act indirectly on cancer cells, for example, by slowing down angiogenesis, boosting the immune system and/or stimulating natural killer (NK) cells, T cells and macrophages.
  • Recombinant IFN-alpha is available commercially as Roferon (Roche Pharmaceuticals) and Intron A (Schering Corporation).
  • Interleukins contemplated by the present invention include IL-2, IL-4, IL-l l and IL-12. Examples of commercially available recombinant interleukins include Proleukin® (IL-2; Chiron Corporation) and Neumega® (IL- 12; Wyeth Pharmaceuticals). Zymogenetics, Inc.
  • Colony-stimulating factors contemplated by the present invention include granulocyte colony stimulating factor (G-CSL or filgrastim), granulocyte-macrophage colony stimulating factor (GM-CSL or sargramostim) and erythropoietin (epoetin alfa, darbepoietin). Treatment with one or more growth factors can help to stimulate the generation of new blood cells in subjects undergoing traditional chemotherapy.
  • CSLs can be helpful in decreasing the side effects associated with chemotherapy and can allow for higher doses of chemotherapeutic agents to be used.
  • Various-recombinant colony stimulating factors are available commercially, for example, Neupogen® (G-CSL; Amgen), Neulasta (pelfilgrastim; Amgen), Leukine (GM-CSL; Berlex), Procrit (erythropoietin; Ortho Biotech), Epogen (erythropoietin; Amgen), Arnesp (erytropoietin).
  • immunotherapeutic agents can be active, i.e. stimulate the body's own immune response, or they can be passive, i.e.
  • Passive specific immunotherapy typically involves the use of one or more monoclonal antibodies that are specific for a particular antigen found on the surface of a cancer cell or that are specific for a particular cell growth factor.
  • Monoclonal antibodies may be used in the treatment of cancer in a number of ways, for example, to enhance a subject's immune response to a specific type of cancer, to interfere with the growth of cancer cells by targeting specific cell growth factors, such as those involved in angiogenesis, or by enhancing the delivery of other anticancer agents to cancer cells when linked or conjugated to agents such as chemotherapeutic agents, radioactive particles or toxins.
  • Monoclonal antibodies currently used as cancer immunotherapeutic agents that are suitable for inclusion in the combinations of the present invention include, but are not limited to, rituximab (Rituxan®), trastuzumab (Herceptin®), ibritumomab tiuxetan (Zevalin®), tositumomab (Bexxar®), cetuximab (C-225, Erbitux®), bevacizumab (Avastin®), gemtuzumab ozogamicin (Mylotarg®), alemtuzumab (Campath®), and BL22.
  • Other examples include anti-CTLA4 antibodies (e.g.
  • antibodies include B cell depleting antibodies.
  • Typical B cell depleting antibodies include but are not limited to anti-CD20 monoclonal antibodies [e.g.
  • the immunotherapeutic treatment may consist of allografting, in particular, allograft with hematopoietic stem cell HSC.
  • the immunotherapeutic treatment may also consist in an adoptive immunotherapy as described by Nicholas P. Restifo, Mark E.
  • the subject In adoptive immunotherapy, the subject’s circulating lymphocytes, NK cells, are isolated amplified in vitro and readministered to the subject.
  • the activated lymphocytes or NK cells are most preferably be the subject’s own cells that were earlier isolated from a blood or tumor sample and activated (or“expanded”) in vitro.
  • the compound of the present invention is administered with a radio therapeutic agent.
  • radiotherapeutic agent as used herein, is intended to refer to any radiotherapeutic agent known to one of skill in the art to be effective to treat or ameliorate cancer, without limitation.
  • the radiotherapeutic agent can be an agent such as those administered in brachytherapy or radionuclide therapy.
  • Such methods can optionally further comprise the administration of one or more additional cancer therapies, such as, but not limited to, chemotherapies, and/or another radiotherapy.
  • the term“radiotherapy” for“radiation therapy” has its general meaning in the art and refers the treatment of cancer with ionizing radiation.
  • Ionizing radiation deposits energy that injures or destroys cells in the area being treated (the target tissue) by damaging their genetic material, making it impossible for these cells to continue to grow.
  • One type of radiation therapy commonly used involves photons, e.g. X-rays.
  • the rays can be used to destroy cancer cells on the surface of or deeper in the body. The higher the energy of the x-ray beam, the deeper the x-rays can go into the target tissue. Linear accelerators and betatrons produce x-rays of increasingly greater energy.
  • Gamma rays are another form of photons used in radiation therapy. Gamma rays are produced spontaneously as certain elements (such as radium, uranium, and cobalt 60) release radiation as they decompose, or decay.
  • the radiation therapy is external radiation therapy.
  • external radiation therapy examples include, but are not limited to, conventional external beam radiation therapy; three-dimensional conformal radiation therapy (3D-CRT), which delivers shaped beams to closely fit the shape of a tumor from different directions; intensity modulated radiation therapy (IMRT), e.g., helical tomotherapy, which shapes the radiation beams to closely fit the shape of a tumor and also alters the radiation dose according to the shape of the tumor; conformal proton beam radiation therapy; image-guided radiation therapy (IGRT), which combines scanning and radiation technologies to provide real time images of a tumor to guide the radiation treatment; intraoperative radiation therapy (IORT), which delivers radiation directly to a tumor during surgery; stereotactic radiosurgery, which delivers a large, precise radiation dose to a small tumor area in a single session; hyperfractionated radiation therapy, e.g., continuous hyperfractionated accelerated radiation therapy (CHART), in which more than one treatment (fraction) of radiation therapy are given to a subject per day; and hypofractionated radiation therapy, in which larger doses of radiation therapy per fraction
  • the pharmaceutical composition of the invention relates to combined preparation for simultaneous, separate or sequential use in the treatment of cancer in a subject in need thereof
  • kits comprising the compound of the invention. Kits containing the compound of the invention find use in therapeutic methods.
  • the invention relates to a method of treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound selected from the group consisting of PD-l antagonist or PD-l expression inhibitor in combination with a compound selected from the group consisting of BTLA antagonist or BTLA expression inhibitor and/or compound selected from the group consisting of BTN3 agonist or BTN3 expression activator.
  • the compounds according to the invention as described above are administered to the patient in a therapeutically effective amount.
  • a “therapeutically effective amount” of the compound of the present invention as above described is meant a sufficient amount of the compound at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed, the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidential with the specific compound employed; and like factors well known in the medical arts.
  • the daily dosage of the products may be varied over a wide range from 0.01 to 1,000 mg per adult per day.
  • the compositions contain 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 250 and 500 mg of the compound of the present invention for the symptomatic adjustment of the dosage to the patient to be treated.
  • a medicament typically contains from about 0.01 mg to about 500 mg of the compound of the present invention, preferably from 1 mg to about 100 mg of the compound of the present invention.
  • An effective amount of the drug is ordinarily supplied at a dosage level from 0.0002 mg/kg to about 20 mg/kg of body weight per day, especially from about 0.001 mg/kg to 7 mg/kg of body weight per day.
  • the compound according to the invention may be used in a concentration between 0.01 mM and 20 mM, particularly, the compound of the invention may be used in a concentration of 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 20.0 mM.
  • the compound of the present invention is administered to the subject in the form of a pharmaceutical composition.
  • the compound of the present invention may be combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form therapeutic compositions.
  • pharmaceutically acceptable excipients or “pharmaceutically acceptable” refer to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to a mammal, especially a human, as appropriate.
  • a pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type.
  • the active principle in the pharmaceutical compositions of the present invention for oral, sublingual, subcutaneous, intramuscular, intravenous, transdermal, local or rectal administration, can be administered in a unit administration form, as a mixture with conventional pharmaceutical supports, to animals and human beings.
  • Suitable unit administration forms comprise oral-route forms such as tablets, gel capsules, powders, granules and oral suspensions or solutions, sublingual and buccal administration forms, aerosols, implants, subcutaneous, transdermal, topical, intraperitoneal, intramuscular, intravenous, subdermal, transdermal, intrathecal and intranasal administration forms and rectal administration forms.
  • the pharmaceutical compositions contain vehicles which are pharmaceutically acceptable for a formulation capable of being injected.
  • vehicles which are pharmaceutically acceptable for a formulation capable of being injected.
  • These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • Solutions comprising compounds of the invention as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the compound of the present invention can be formulated into a composition in a neutral or salt form.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like.
  • Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine,
  • the carrier can also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetables oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminium monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized agent of the present inventions into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • sterile powders for the preparation of sterile injectable solutions the typical methods of preparation are vacuum-drying and freeze- drying techniques which yield a powder of the compound of the present invention plus any additional desired ingredient from a previously sterile- filtered solution thereof.
  • the preparation of more, or highly concentrated solutions for direct injection is also contemplated, where the use of DMSO as solvent is envisioned to result in extremely rapid penetration, delivering high concentrations of the active agents to a small tumor area.
  • solutions Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • the formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like can also be employed.
  • aqueous solutions For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
  • sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject.
  • FIGURES are a diagrammatic representation of FIGURES.
  • FIG. 1 Resting Vy9V52 T cells reserve a significant intracellular pool of PD-1 located in early endosomes.
  • Multiparametric flow cytometry analysis ofintra- (white squares) and extra- (dark circles) cellular expression of co-signaling molecules CTLA-4, ICOS, BTLA, PD-l on peripheral gd T cells (CD3+TCRV52+) from healthy volunteers (n 14). Results were expressed as percentage of positive cells for given marker gated on gd T cells.
  • FIG. 1 PD-1 is quickly translocated to the cell surface of Vy9V52 T cells upon IPP stimulation.
  • FIG. 3 PD-1 engagement inhibits Vy9V52 T cell proliferative capacities as well as BTLA-HVEM interaction.
  • C CellTrace dilution in purified gd T cells from 8 HV stimulated 5 days with 200 Ul/ml IL-2 with (dark bars) or without (white bars) low-dose BrHPP (50 nM) with specified blocking mAbs (20 Lig/ml). Results were represented as the percentage of divided cells ⁇ SEM among gd T cells.
  • D Proliferation assessed by CFSE staining in purified gd T cells from 3 HV stimulated 5 days with specified Fc proteins of IgGl-Fc control protein (10 pg/ml). Results were expressed as the percentage of divided cells ⁇ SEM among gd-T cells.
  • E Effect of blocking anti-TCR mAb.
  • PD-1 differs from BTLA in their expression pattern in Vy9V52 T cells and combined blockade improves Vy9V52 T cell proliferation.
  • Results were expressed as MFI (mean fluorescence intensity) ⁇ SEM.
  • FIG. 6 Ex vivo and in vitro effect of PD-1 and BTLA blockade on Vy9V52 T cells co-cultured with PD-L1+ and HVEM+ tumor cells.
  • B Surface expression of PD-L1 and HVEM by flow cytometry on tumor and epithelial cells gated as CD45- cells isolated from tumor and control prostate tissues respectively. Results were expressed as % of positive cells ⁇ SEM.
  • C-D CellTrace dilution in purified gd T cells from 5 HV stimulated 5 days with IL-2 and low-dose BrHPP (50 nM), co-cultured with : (C) prostate tumor cell lines (PC3, DU145, LNCaP) or RL lymphoma cell line alone, (D) or with the addition of specified blocking mAbs (20 pg/ml). Results were expressed as % of divided cells ⁇ SEM, and statistical significance was established using the nonparametric paired Wilcoxon U test. *P ⁇ 0.05; **P ⁇ 0.01; ***P ⁇ 0.001.
  • Vy9V52 T cell antitumor functions toward tumor cells are enhanced by PD-1 and BTLA blockade.
  • Vy9V52 T cells were co-cultured with PC3 prostate tumor cell line (ratio 1 : 1) and treated with anti-BTN3A agonist (20.1) mAb or anti-BTN3A antagonist (103.2) mAb (10 pg/ml). After 4 hours, cytolytic degranulation of Vy9V52 T cells was assessed by CDl07a/b expression by flow cytometry. Data were normalized to condition with no pre-treatment (Vy9V52 T cells cultured 5 days with IgGl isotype control). Statistical significance was established using the nonparametric paired Wilcoxon U test. *P ⁇ 0.05; **P ⁇ 0.01; ***P ⁇ 0.001.
  • Blockade of the inhibitory receptors PD-1 and BTLA improves Vy9V62 T cell expansion in human tumors
  • Vy9V52 cells represent a major peripheral blood T-cell subset in humans with broad reactivity against microbial agents and tumors. Unlike abT cells, gdT cells do not require the help of conventional major histocompatibility complex (MHC) class I and class II molecules for recognizing the antigens. Strikingly, gdT cells have emerged as the most favorable prognostic immune population among many cancer types. However, the mechanisms of gd T- cell immune responses to tumors remain poorly understood.
  • MHC major histocompatibility complex
  • Vy9Vd2 cells reserve a significant intracellular pool of PD-l located in early endosomes, which is mobilized at the cell membrane after bisphosphonate activation.
  • PD-l is a major regulator of Vy9Vd2 T-cell homeostasis, similarly to BTLA, another member of the Ig superfamily, for which we previously demonstrated a key role in the regulation of TCR- independent and TCR-dependent Vy9Vd2 T-cell proliferation.
  • PD-l and BTLA differ in their expression pattern in Vy9Vd2 T cells and interestingly, combined blockade with monoclonal antibodies synergizes to improve Vy9Vd2 T cell expansion compared to single receptor blockade.
  • gdT cells infiltrate prostate tumors and overexpress PD-l, in addition to BTLA and other co-inhibitory molecules.
  • PD-l intracellular pool appears as a possible mechanism of immune escape by tumor cells, that upregulate PD-L1 and HVEM, by interfering with gdT cell antitumor activity.
  • PD-l and BTLA blockade improves Vy9Vd2 T cell proliferation and antitumor functions toward tumor cells.
  • Our data highlight a new role for PD-l and the potent combination with BTLA in the control of Vy9Vd2 cells.
  • Plasma samples were also collected from prostate cancer patients (n 20).
  • HV healthy volunteers
  • EFS local Blood Bank
  • PBMCs Peripheral Blood Mononuclear Cells
  • Bromohydrin pyrophosphate (BrHPP) was obtained from Innate Pharma (Marseille, France) and recombinant human (Rh) IF-2 (Proleukin) purchased from Novartis Pharma SAS.
  • the monoclonal antibodies (mAbs) and Fc-proteins used in this study are listed in Table 1.
  • HVEM and BTLA Abs mAbs recognizing human PD-l (clone 3.1), PD-F1 (clone 3.1), PD-F2 (clone 326.35), HVEM (clone 18.10) and BTFA (clone 8.2) were generated as previously described.
  • Effector gd-T cells were established as previously described. PBMCs from HV were stimulated with BtHPP (3 mM) and rhIF-2 (lOOIU/ml) at Day 0. From Day 5, rhIF-2 was renewed every two days and cells were kept at l .5xl0 6 /ml for 15 days. The purity of gd-T cells, assessed by flow cytometry, was greater than 80%.
  • Prostate (PC3, DU145 and FNCaP), follicular lymphoma (RE) and breast (MDA-MB- 134, MDA-MB-231, SKBR3) cancer cell lines were obtained from the American Type Culture Collection and cultured in RPMI 1640 or DMEM medium supplemented with 10% heat- inactivated fetal calf serum. Melanoma cell lines were established from patients’ tumors in our laboratory in the medium.
  • PBMCs peripheral blood mononuclear cells
  • PBMCs peripheral blood mononuclear cells
  • samples were alanyzed on a FSRFortessa cytometer (Becton Dickinson) and analyzed using FACSDiva (BD Biosciences) or FlowJo (TreeStar Inc.) softwares.
  • FSRFortessa cytometer Becton Dickinson
  • FACSDiva FACSDiva
  • FlowJo TeStar Inc.
  • gd-T cells were incubated at 37°C with anti-CD 107a and anti-CD 107b conjugates and Golgi stop, with or without BrHPP and specified mAbs. After 4 hours, cells were collected and analyzed by flow cytometry.
  • gd-T cells were isolated from PBMCs of HV using anti-TCR g/d microbead kit (Miltenyi Biotec). The purity of gd-T cells assessed by flow cytometry was greater than 80%. Purified gd- T cells were labeled with 2.5 mM CarboxyFluoroscein Succinimidyl Ester (CFSE) (Molecular Probes, FifeTech) for 10 minutes at 37°C or with 2 mM CellTrace Violet (Molecular Probes, FifeTech) for 20 minutes at 37°C. 5.
  • CFSE CarboxyFluoroscein Succinimidyl Ester
  • Proteins were resolved by sodium dodecyl sulfate polyacrylamide gel electrophoresis 10%, followed by Western blotting.
  • Primary antibodies used were from cell signaling: rabbit anti-phospho-Zap70 antibody, rabbit anti- phospho-Erkl/2 antibody, rabbit anti-ERKl/2 and rabbit anti-ZAP70 (all from Cell Signaling Technologies). Primary antibodies were detected with horseradish peroxidase-conjugated anti rabbit antibody (Jackson Laboratory). Immunoreactive bands were detected using enhanced chemiluminescent reagents (Pierce). Quantification of signals was performed using ImageJ software, and the signal of phosphorylation was normalized with that of the corresponding total protein.
  • Vy9V52 T cells Resting or lh-BrHPP (1 mM) preactivated Vy9V52 T cells were incubated for 30 minutes, on poly-L-lysine pretreated coverslips. Cells were then fixed in methanol at -20°C for 6 minutes and washed in PBS. After blocking in PBS 10% stromal vascular fraction, cells were incubated with primary antibodies: TCRVy9 mAb (mouse IgGl, 10 mg/mL), PD-l (mouse IgG2b, 10 mg/mL) mAb, syntaxin, TGN46 and EEA1 for 1 hour.
  • TCRVy9 mAb mouse IgGl, 10 mg/mL
  • PD-l mouse IgG2b, 10 mg/mL
  • PD-l staining was detected using a specific anti-IgG2b secondary antibody conjugated to cyanine 5 (Cy5) from Jackson Laboratories. DNA was stained with 250 ng/mL DAPI (Roche Diagnostics) during secondary staining. Cells were mounted in Prolong Gold antifade reagent (Invitrogen) and examined on a LSM-510 Carl Zeiss confocal microscope with a x63 NA1.4 Plan Apochromat objective. Five images of cell conjugates were taken among the cell conjugates observed.
  • Sections were revealed with the EnVision FLEX/HRP kit (Dako), DAB substrate kit (Dako), counterstained with the EnVision FLEX hematoxylin substrate buffer (Dako) and mounted using Pertex (Histolab). Controls were performed by incubating slides with second Ab alone.
  • Results are expressed as median ⁇ SEM. Statistical analysis was performed using Spearman correlation, Wilcoxon test and Mann- Whitney t test. P values ⁇ 0.05 were considered significant. Analyses were performed using GraphPad Prism program.
  • Vy9V52 T cells reserve a significant intracellular pool of PD-l observed by intracellular flow cytometry staining ( Figure 1). Immunofluorescence analysis by confocal microscopy showed that PD-l co-localized with EEA1 (early endosome marker) rather than syntaxin (which stains proteins implicated in the docking of synaptic vesicles), or TGN46 (Trans-Golgi Network 46), suggesting that PD-l intracellular pool is contained in early endosome compartment (Data not shown).
  • EEA1 eye endosome marker
  • syntaxin which stains proteins implicated in the docking of synaptic vesicles
  • TGN46 Trans-Golgi Network 46
  • PD-l blockade with an antagonist antibody enhanced Vy9V52 T cell proliferation; as well as BTLA blockade with an antagonist antibody (clone 8.2), measured as CFSE staining ( Figure 3A and B); or CellTrace Violet staining ( Figure 3C and Data not shown).
  • Blockade with antagonist antibodies against their respective ligands i.e anti-PD-Ll (clone 3.1), PD-L2 (clone 326.35), anti-HVEM (clone 18.10) also significantly enhanced Vy9V52 T cell proliferation.
  • Vy9V52 T cells were then pre-incubated with a blocking anti-TCR antibody before addition of either anti-PD-l, anti-PD-Fl, anti-PD-F2, anti-BTFA, or anti-HVEM antagonist antibody.
  • Results show that PD-l and BTFA effects on Vy9V52 T cell proliferation are partly dependent of TCR stimulation (Data not shown).
  • Figure 4 shows that BTFA is highly expressed on naive gd T cells and is progressively downregulated in memory and differentiated effector-type cells.
  • Figure 6A shows that tumor cells lines (derived from breast cancer, prostate cancer, or melanoma) highly expressed PD-F1 and HVEM, respectively ligands for PD- 1 and BTFA.
  • Figure 6B shows that PD-F1 and HVEM were expressed on tumor cells gated as CD45- cells isolated from prostate tumors.
  • Prostate tumor cell lines inhibited Vy9Vd2 T cell proliferation (Figure 6C), and blockade with anti-PD-l or anti-BTFA enhanced Vy9Vd2 T cell proliferation co-cultured with tumor cell lines ( Figure 6D).
  • Figure 7 shows that pre-treatment of Vy9Vd2 T cells with anti-PD-l or anti-BTFA antibody during 5 days, potentiates Vy9Vd2 T cell anti-tumor effect against tumor cells, measured as Vy9Vd2 T cell degranulation (CD 107 staining by flow cytometry) induced by BTN3 agonist mAh.

Abstract

La présente invention concerne des méthodes et des compositions pharmaceutiques pour le traitement du cancer chez un sujet le nécessitant. Les inventeurs montrent que les cellules Vγ9Vδ2 ont en réserve un pool intracellulaire significatif de PD-1 situé dans des endosomes précoces, qui est mobilisé au niveau de la membrane cellulaire après activation du bisphosphonate. Les inventeurs montrent que PD-1 est un régulateur majeur de l'homéostasie des lymphocytes T Vγ9Vδ2, de manière similaire à BTLA, un autre membre de la superfamille des Ig, pour lequel les inventeurs ont précédemment mis en évidence un rôle clé dans la régulation de la prolifération des lymphocytes T Vγ9Vδ2 dépendant de TCR et indépendant de TCR. PD-1 et BTLA diffèrent dans leur motif d'expression dans les lymphocytes T Vγ9Vδ2 et, de manière intéressante, un blocage combiné avec des anticorps monoclonaux fonctionne en synergie pour améliorer l'expansion des lymphocytes T Vγ9Vδ2 par rapport à un blocage de récepteur unique. Les inventeurs ont montré que les cellules γδT s'infiltrent dans les tumeurs de la prostate et surexpriment PD-1, en plus de BTLA et d'autres molécules co-inhibitrices. Ainsi, la régulation positive du pool intracellulaire de PD-1 apparaît comme un mécanisme possible d'échappement à la réponse immune par des cellules tumorales, qui régulent positivement PD-L1 et HVEM, en interférant avec l'activité antitumorale des cellules γδT. Les inventeurs ont montré que le blocage PD-1 et BTLA améliore la prolifération des lymphocytes T Vγ9Vδ2 et les fonctions antitumorales envers des cellules tumorales. La présente invention met en évidence un nouveau rôle pour PD-1 et la combinaison efficace avec BTLA dans la régulation des cellules Vγ9Vδ2. Ainsi, l'invention concerne un antagoniste de PD-1 en combinaison avec un antagoniste de BTLA et/ou un agoniste de BTN3 pour une utilisation dans le traitement du cancer.
PCT/EP2019/061228 2018-05-03 2019-05-02 Méthodes et compositions pharmaceutiques pour le traitement du cancer WO2019211370A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP18305558.1 2018-05-03
EP18305558 2018-05-03
EP18305692 2018-06-07
EP18305692.8 2018-06-07

Publications (1)

Publication Number Publication Date
WO2019211370A1 true WO2019211370A1 (fr) 2019-11-07

Family

ID=66542205

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2019/061228 WO2019211370A1 (fr) 2018-05-03 2019-05-02 Méthodes et compositions pharmaceutiques pour le traitement du cancer

Country Status (1)

Country Link
WO (1) WO2019211370A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114457025A (zh) * 2020-10-30 2022-05-10 未来智人再生医学研究院(广州)有限公司 一种表达btla阻断物的多能干细胞或其衍生物及应用
WO2023161457A1 (fr) 2022-02-27 2023-08-31 Evobright Gmbh Anticorps bispécifiques dirigés contre cd277 et un antigène tumoral

Citations (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1990007861A1 (fr) 1988-12-28 1990-07-26 Protein Design Labs, Inc. IMMUNOGLOBULINES CHIMERIQUES SPECIFIQUES CONTRE LA PROTEINE TAC p55 DU RECEPTEUR D'IL-2
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5229275A (en) 1990-04-26 1993-07-20 Akzo N.V. In-vitro method for producing antigen-specific human monoclonal antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5545807A (en) 1988-10-12 1996-08-13 The Babraham Institute Production of antibodies from transgenic animals
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5567610A (en) 1986-09-04 1996-10-22 Bioinvent International Ab Method of producing human monoclonal antibodies and kit therefor
US5573905A (en) 1992-03-30 1996-11-12 The Scripps Research Institute Encoded combinatorial chemical libraries
US5585089A (en) 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US5591669A (en) 1988-12-05 1997-01-07 Genpharm International, Inc. Transgenic mice depleted in a mature lymphocytic cell-type
US5598369A (en) 1994-06-28 1997-01-28 Advanced Micro Devices, Inc. Flash EEPROM array with floating substrate erase operation
US5618829A (en) 1993-01-28 1997-04-08 Mitsubishi Chemical Corporation Tyrosine kinase inhibitors and benzoylacrylamide derivatives
US5639757A (en) 1995-05-23 1997-06-17 Pfizer Inc. 4-aminopyrrolo[2,3-d]pyrimidines as tyrosine kinase inhibitors
US5728868A (en) 1993-07-15 1998-03-17 Cancer Research Campaign Technology Limited Prodrugs of protein tyrosine kinase inhibitors
US5800988A (en) 1992-08-21 1998-09-01 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US5804396A (en) 1994-10-12 1998-09-08 Sugen, Inc. Assay for agents active in proliferative disorders
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
WO1999032619A1 (fr) 1997-12-23 1999-07-01 The Carnegie Institution Of Washington Inhibition genetique par de l'arn double brin
US5981732A (en) 1998-12-04 1999-11-09 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-13 expression
US6046321A (en) 1999-04-09 2000-04-04 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-i1 expression
US6100254A (en) 1997-10-10 2000-08-08 Board Of Regents, The University Of Texas System Inhibitors of protein tyrosine kinases
US6107091A (en) 1998-12-03 2000-08-22 Isis Pharmaceuticals Inc. Antisense inhibition of G-alpha-16 expression
US6127374A (en) 1997-07-29 2000-10-03 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO2001036646A1 (fr) 1999-11-19 2001-05-25 Cancer Research Ventures Limited Inhibition d"expression genique a l"aide d"arn bicatenaire
US6245759B1 (en) 1999-03-11 2001-06-12 Merck & Co., Inc. Tyrosine kinase inhibitors
WO2001068836A2 (fr) 2000-03-16 2001-09-20 Genetica, Inc. Procedes et compositions d'interference d'arn
US6306874B1 (en) 1999-10-19 2001-10-23 Merck & Co., Inc. Tyrosine kinase inhibitors
US6313138B1 (en) 2000-02-25 2001-11-06 Merck & Co., Inc. Tyrosine kinase inhibitors
US6316444B1 (en) 1999-06-30 2001-11-13 Merck & Co., Inc. SRC kinase inhibitor compounds
US6329380B1 (en) 1999-06-30 2001-12-11 Merck & Co., Inc. SRC kinase inhibitor compounds
US6344459B1 (en) 1996-04-12 2002-02-05 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6365354B1 (en) 2000-07-31 2002-04-02 Isis Pharmaceuticals, Inc. Antisense modulation of lysophospholipase I expression
US6410323B1 (en) 1999-08-31 2002-06-25 Isis Pharmaceuticals, Inc. Antisense modulation of human Rho family gene expression
US6420382B2 (en) 2000-02-25 2002-07-16 Merck & Co., Inc. Tyrosine kinase inhibitors
US6479512B1 (en) 1999-10-19 2002-11-12 Merck & Co., Inc. Tyrosine kinase inhibitors
US6498165B1 (en) 1999-06-30 2002-12-24 Merck & Co., Inc. Src kinase inhibitor compounds
US6566135B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of caspase 6 expression
US6566131B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of Smad6 expression
US6573099B2 (en) 1998-03-20 2003-06-03 Benitec Australia, Ltd. Genetic constructs for delaying or repressing the expression of a target gene
US6586423B2 (en) 1999-09-10 2003-07-01 Merck & Co., Inc. Tyrosine kinase inhibitors
US6740665B1 (en) 1999-02-10 2004-05-25 Ramachandran Murali Tyrosine kinase inhibitors and methods of using the same
US6765087B1 (en) 1992-08-21 2004-07-20 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US6794393B1 (en) 1999-10-19 2004-09-21 Merck & Co., Inc. Tyrosine kinase inhibitors
US6838254B1 (en) 1993-04-29 2005-01-04 Conopco, Inc. Production of antibodies or (functionalized) fragments thereof derived from heavy chain immunoglobulins of camelidae
US6875767B2 (en) 2001-06-22 2005-04-05 Merck & Co., Inc. (5-cyano-2-thiazolyl)amino-4-pyridine tyrosine kinase inhibitors
US6927293B2 (en) 2001-08-30 2005-08-09 Merck & Co., Inc. Tyrosine kinase inhibitors
US6958340B2 (en) 2001-08-01 2005-10-25 Merck & Co., Inc. Tyrosine kinase inhibitors
US7109304B2 (en) 2003-07-31 2006-09-19 Immunomedics, Inc. Humanized anti-CD19 antibodies
US20070254295A1 (en) 2006-03-17 2007-11-01 Prometheus Laboratories Inc. Methods of predicting and monitoring tyrosine kinase inhibitor therapy
WO2009024531A1 (fr) 2007-08-17 2009-02-26 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthode de traitement et de diagnostic de malignités hématologiques
WO2010089411A2 (fr) 2009-02-09 2010-08-12 Universite De La Mediterranee Anticorps pd-1 et anticorps pd-l1 et leurs utilisations
WO2010106051A1 (fr) 2009-03-17 2010-09-23 Universite De La Mediterranee Anticorps anti-btla et leurs utilisations
WO2011014438A1 (fr) 2009-07-31 2011-02-03 N.V. Organon Anticorps totalement humains dirigés contre le btla
WO2012080769A1 (fr) 2010-12-15 2012-06-21 INSERM (Institut National de la Santé et de la Recherche Médicale) Anticorps anti-cd277 et leurs utilisations
WO2014183885A1 (fr) * 2013-05-17 2014-11-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Antagoniste de l'interaction btla/hvem pour une utilisation en thérapie
WO2017144668A1 (fr) 2016-02-26 2017-08-31 INSERM (Institut National de la Santé et de la Recherche Médicale) Anticorps présentant une spécificité pour btla et leurs utilisations
WO2018068756A1 (fr) * 2016-10-14 2018-04-19 Beijing Biocytogen Co., Ltd Animal non humain génétiquement modifié à btla humaine ou chimérique

Patent Citations (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5567610A (en) 1986-09-04 1996-10-22 Bioinvent International Ab Method of producing human monoclonal antibodies and kit therefor
US5545807A (en) 1988-10-12 1996-08-13 The Babraham Institute Production of antibodies from transgenic animals
US5591669A (en) 1988-12-05 1997-01-07 Genpharm International, Inc. Transgenic mice depleted in a mature lymphocytic cell-type
US5693761A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Polynucleotides encoding improved humanized immunoglobulins
US5585089A (en) 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
WO1990007861A1 (fr) 1988-12-28 1990-07-26 Protein Design Labs, Inc. IMMUNOGLOBULINES CHIMERIQUES SPECIFIQUES CONTRE LA PROTEINE TAC p55 DU RECEPTEUR D'IL-2
US5693762A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Humanized immunoglobulins
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5229275A (en) 1990-04-26 1993-07-20 Akzo N.V. In-vitro method for producing antigen-specific human monoclonal antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5573905A (en) 1992-03-30 1996-11-12 The Scripps Research Institute Encoded combinatorial chemical libraries
US6765087B1 (en) 1992-08-21 2004-07-20 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US5800988A (en) 1992-08-21 1998-09-01 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US5874541A (en) 1992-08-21 1999-02-23 Vrije Universiteit Immunoglobulins devoid of light chains
US6015695A (en) 1992-08-21 2000-01-18 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
US5618829A (en) 1993-01-28 1997-04-08 Mitsubishi Chemical Corporation Tyrosine kinase inhibitors and benzoylacrylamide derivatives
US6838254B1 (en) 1993-04-29 2005-01-04 Conopco, Inc. Production of antibodies or (functionalized) fragments thereof derived from heavy chain immunoglobulins of camelidae
US5728868A (en) 1993-07-15 1998-03-17 Cancer Research Campaign Technology Limited Prodrugs of protein tyrosine kinase inhibitors
US5598369A (en) 1994-06-28 1997-01-28 Advanced Micro Devices, Inc. Flash EEPROM array with floating substrate erase operation
US5804396A (en) 1994-10-12 1998-09-08 Sugen, Inc. Assay for agents active in proliferative disorders
US5639757A (en) 1995-05-23 1997-06-17 Pfizer Inc. 4-aminopyrrolo[2,3-d]pyrimidines as tyrosine kinase inhibitors
US6344459B1 (en) 1996-04-12 2002-02-05 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6127374A (en) 1997-07-29 2000-10-03 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6562818B1 (en) 1997-07-29 2003-05-13 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6100254A (en) 1997-10-10 2000-08-08 Board Of Regents, The University Of Texas System Inhibitors of protein tyrosine kinases
WO1999032619A1 (fr) 1997-12-23 1999-07-01 The Carnegie Institution Of Washington Inhibition genetique par de l'arn double brin
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
US6573099B2 (en) 1998-03-20 2003-06-03 Benitec Australia, Ltd. Genetic constructs for delaying or repressing the expression of a target gene
US6107091A (en) 1998-12-03 2000-08-22 Isis Pharmaceuticals Inc. Antisense inhibition of G-alpha-16 expression
US5981732A (en) 1998-12-04 1999-11-09 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-13 expression
US6740665B1 (en) 1999-02-10 2004-05-25 Ramachandran Murali Tyrosine kinase inhibitors and methods of using the same
US6245759B1 (en) 1999-03-11 2001-06-12 Merck & Co., Inc. Tyrosine kinase inhibitors
US6544988B1 (en) 1999-03-11 2003-04-08 Merck & Co., Inc. Tyrosine kinase inhibitors
US6046321A (en) 1999-04-09 2000-04-04 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-i1 expression
US6329380B1 (en) 1999-06-30 2001-12-11 Merck & Co., Inc. SRC kinase inhibitor compounds
US6316444B1 (en) 1999-06-30 2001-11-13 Merck & Co., Inc. SRC kinase inhibitor compounds
US6498165B1 (en) 1999-06-30 2002-12-24 Merck & Co., Inc. Src kinase inhibitor compounds
US6410323B1 (en) 1999-08-31 2002-06-25 Isis Pharmaceuticals, Inc. Antisense modulation of human Rho family gene expression
US6586423B2 (en) 1999-09-10 2003-07-01 Merck & Co., Inc. Tyrosine kinase inhibitors
US6586424B2 (en) 1999-09-10 2003-07-01 Merck & Co., Inc. Tyrosine kinase inhibitors
US6794393B1 (en) 1999-10-19 2004-09-21 Merck & Co., Inc. Tyrosine kinase inhibitors
US6306874B1 (en) 1999-10-19 2001-10-23 Merck & Co., Inc. Tyrosine kinase inhibitors
US6479512B1 (en) 1999-10-19 2002-11-12 Merck & Co., Inc. Tyrosine kinase inhibitors
WO2001036646A1 (fr) 1999-11-19 2001-05-25 Cancer Research Ventures Limited Inhibition d"expression genique a l"aide d"arn bicatenaire
US6313138B1 (en) 2000-02-25 2001-11-06 Merck & Co., Inc. Tyrosine kinase inhibitors
US6420382B2 (en) 2000-02-25 2002-07-16 Merck & Co., Inc. Tyrosine kinase inhibitors
WO2001068836A2 (fr) 2000-03-16 2001-09-20 Genetica, Inc. Procedes et compositions d'interference d'arn
US6365354B1 (en) 2000-07-31 2002-04-02 Isis Pharmaceuticals, Inc. Antisense modulation of lysophospholipase I expression
US6566135B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of caspase 6 expression
US6566131B1 (en) 2000-10-04 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of Smad6 expression
US6875767B2 (en) 2001-06-22 2005-04-05 Merck & Co., Inc. (5-cyano-2-thiazolyl)amino-4-pyridine tyrosine kinase inhibitors
US6958340B2 (en) 2001-08-01 2005-10-25 Merck & Co., Inc. Tyrosine kinase inhibitors
US6927293B2 (en) 2001-08-30 2005-08-09 Merck & Co., Inc. Tyrosine kinase inhibitors
US7109304B2 (en) 2003-07-31 2006-09-19 Immunomedics, Inc. Humanized anti-CD19 antibodies
US20070254295A1 (en) 2006-03-17 2007-11-01 Prometheus Laboratories Inc. Methods of predicting and monitoring tyrosine kinase inhibitor therapy
WO2009024531A1 (fr) 2007-08-17 2009-02-26 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthode de traitement et de diagnostic de malignités hématologiques
WO2010089411A2 (fr) 2009-02-09 2010-08-12 Universite De La Mediterranee Anticorps pd-1 et anticorps pd-l1 et leurs utilisations
WO2010106051A1 (fr) 2009-03-17 2010-09-23 Universite De La Mediterranee Anticorps anti-btla et leurs utilisations
WO2011014438A1 (fr) 2009-07-31 2011-02-03 N.V. Organon Anticorps totalement humains dirigés contre le btla
WO2012080769A1 (fr) 2010-12-15 2012-06-21 INSERM (Institut National de la Santé et de la Recherche Médicale) Anticorps anti-cd277 et leurs utilisations
WO2012080351A1 (fr) 2010-12-15 2012-06-21 INSERM (Institut National de la Santé et de la Recherche Médicale) Anticorps anti-cd277 et leurs utilisations
WO2014183885A1 (fr) * 2013-05-17 2014-11-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Antagoniste de l'interaction btla/hvem pour une utilisation en thérapie
WO2017144668A1 (fr) 2016-02-26 2017-08-31 INSERM (Institut National de la Santé et de la Recherche Médicale) Anticorps présentant une spécificité pour btla et leurs utilisations
WO2018068756A1 (fr) * 2016-10-14 2018-04-19 Beijing Biocytogen Co., Ltd Animal non humain génétiquement modifié à btla humaine ou chimérique

Non-Patent Citations (16)

* Cited by examiner, † Cited by third party
Title
"Biochemistry and Immunology", 1996, ACADEMIC PRESS, article "Coding, Monoclonal Antibodies: Principles and Practice: Production and Application of Monoclonal Antibodies in Cell Biology"
AGNEW CHEM INTL. ED. ENGL., vol. 33, 1994, pages 183 - 186
CANCER RESEARCH 20170701 AMERICAN ASSOCIATION FOR CANCER RESEARCH INC. NLD, vol. 77, no. 13, Supplement 1, 1 July 2017 (2017-07-01), ISSN: 1538-7445 *
CLARK, W. R.: "The Experimental Foundations of Modern Immunology", 1986, WILEY & SONS, INC.
DATABASE EMBASE [online] ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL; 1 July 2017 (2017-07-01), MISELIS N R ET AL: "Antagonism of the co-inhibitory receptor BTLA enhances efficacy of anti-PD-1 treatment in murine syngeneic tumor models", XP002785522, Database accession no. EMB-619155924 *
DE BENEDETTI ET AL., J IMMUNOL, vol. 166, 2001, pages 4334 - 4340
EPRATUZUMAB, LEONARD ET AL., CLINICAL CANCER RESEARCH, vol. 10, pages 53Z7 - 5334
GERTNER-DARDENNE ET AL., BLOOD, 2013
KOHLERMILSTEIN, NATURE, vol. 256, 1975, pages 495
KRIEGLER: "A Laboratory Manual", 1990, W.H. FREEMAN C.O.
MURRY: "Methods in Molecular Biology", vol. 7, 1991, HUMANA PRESS, INC.
NICHOLAS P. RESTIFOMARK E. DUDLEYSTEVEN A. ROSENBERG: "Adoptive immunotherapy for cancer: harnessing the T cell response", NATURE REVIEWS IMMUNOLOGY, vol. 12, April 2012 (2012-04-01), XP055034896, DOI: doi:10.1038/nri3191
ROITT, I.: "Essential Immunology", 1991, BLACKWELL SCIENTIFIC PUBLICATIONS
SANBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY PRESS
SUZUKI ET AL., J OF IMMUNOL, vol. 22, no. 8, 1992, pages 1989 - 1993
WU ET AL., MOL. BIOL., vol. 294, 1999, pages 151

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114457025A (zh) * 2020-10-30 2022-05-10 未来智人再生医学研究院(广州)有限公司 一种表达btla阻断物的多能干细胞或其衍生物及应用
WO2023161457A1 (fr) 2022-02-27 2023-08-31 Evobright Gmbh Anticorps bispécifiques dirigés contre cd277 et un antigène tumoral

Similar Documents

Publication Publication Date Title
EP3419999B1 (fr) Anticorps ayant une spécificité pour le btla et leurs utilisations
Pai et al. Clonal deletion of tumor-specific T cells by interferon-γ confers therapeutic resistance to combination immune checkpoint blockade
KR102636539B1 (ko) 암에 대한 조합 요법
US10259875B2 (en) Methods for treating cancer in patients with elevated levels of BIM
US20110293629A1 (en) Methods of Treating and/or Preventing Cell Proliferation Disorders with IL-17 Antagonists
EP3490568A1 (fr) Traitement d'une tumeur solide par ciblage de signalisation de dectine 1
WO2018185516A1 (fr) Méthodes et compositions pharmaceutiques destinées à traiter une toxicité cardiovasculaire induite par une thérapie anticancéreuse
WO2019211370A1 (fr) Méthodes et compositions pharmaceutiques pour le traitement du cancer
TW201444868A (zh) Hgf抗體及其組成物
US20230045791A1 (en) Anti-tnfr2 antibody and uses thereof
WO2019106126A1 (fr) Modulateurs mdm2 pour le diagnostic et le traitement du liposarcome
WO2019234221A1 (fr) Procédés de stratification et de traitement d'un patient souffrant de leucémie lymphoïde chronique
WO2018189403A1 (fr) Méthodes et compositions pharmaceutiques destinées au traitement du cancer
US20180028566A1 (en) Gamma delta t cells as a target for treatment of solid tumors
US20210047696A1 (en) Methods and pharmaceutical compositions for treating cancer
US20210164984A1 (en) Methods for predicting outcome and treatment of patients suffering from prostate cancer or breast cancer
EP3800201A1 (fr) Stimulation cd28h améliorant des activités de destruction de cellules nk
US20220290151A1 (en) Use of müllerian inhibiting substance inhibitors for treating cancer
WO2019211369A1 (fr) Procédés et compositions pharmaceutiques pour le traitement du cancer
WO2023041744A1 (fr) Inhibiteurs de bet pour le traitement du cancer à déficit en bap1
US20230227565A1 (en) Method for Treating CD127-Positive Cancers by Administering an Anti-CD127 Agent
WO2024056668A1 (fr) Nouveaux anticorps anti-itgb8 et leurs utilisations
US20210340232A1 (en) Monoclonal antibodies against human dickkopf3 and uses thereof
WO2023012343A1 (fr) Procédés de traitement du cancer
EP4347043A1 (fr) Méthode de traitement de cancers cd1276-positifs par administration d'un agent anti-cd127

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19724108

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19724108

Country of ref document: EP

Kind code of ref document: A1