WO2016059220A1 - Tcr-activating agents for use in the treatment of t-all - Google Patents

Tcr-activating agents for use in the treatment of t-all Download PDF

Info

Publication number
WO2016059220A1
WO2016059220A1 PCT/EP2015/074033 EP2015074033W WO2016059220A1 WO 2016059220 A1 WO2016059220 A1 WO 2016059220A1 EP 2015074033 W EP2015074033 W EP 2015074033W WO 2016059220 A1 WO2016059220 A1 WO 2016059220A1
Authority
WO
WIPO (PCT)
Prior art keywords
tcr
antibody
cell
cells
mice
Prior art date
Application number
PCT/EP2015/074033
Other languages
French (fr)
Inventor
Vahid Asnafi
Jacques Ghysdael
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Université Paris Descartes
Assistance Publique-Hôpitaux De Paris (Aphp)
Centre National De La Recherche Scientifique (Cnrs)
Institut Curie
Université Paris-Sud
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), Université Paris Descartes, Assistance Publique-Hôpitaux De Paris (Aphp), Centre National De La Recherche Scientifique (Cnrs), Institut Curie, Université Paris-Sud filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2016059220A1 publication Critical patent/WO2016059220A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57426Specifically defined cancers leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70503Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
    • G01N2333/7051T-cell receptor (TcR)-CD3 complex
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the invention is in the field of oncology and more particularly treatment of T-cell acute lymphoblastic leukemia (T-ALL).
  • T-ALL T-cell acute lymphoblastic leukemia
  • T-cell acute lymphoblastic leukemias are malignant proliferations of thymocytes by multi-step oncogenic processes leading to T-cell differentiation arrest.
  • Targeted therapy including abrogation of a block to cell maturation, is a promising therapeutic approach in cancer.
  • the mechanism underlying the therapeutic effect of OKT3 monoclonal antibody in said T-ALL patient is based on the stimulation of the immune system as suggested by the induction of IL-2 responsiveness of the malignant cells leading to propose a sequential application of OKT3 and IL-2 to render certain T cell tumours more susceptible to chemotherapy.
  • the invention relates to a non-mitogenic anti-CD3 antibody for use in a method for treating T-cell acute lymphoblastic leukemia (T-ALL).
  • T-ALL T-cell acute lymphoblastic leukemia
  • the invention in a second aspect, relates to a non-mitogenic anti-CD3 antibody for use in a method for inducing cell death of T-ALL cells.
  • the invention in a third aspect, relates to a pharmaceutical composition comprising a non-mitogenic anti-CD3 antibody for use in a method for treating T-ALL.
  • the invention in a fourth aspect, relates to an in vitro method for predicting the responsiveness of a patient affected with a T-ALL to a treatment with a TCR-activating agent comprising a step of determining the expression of TCR/CD3 complex at the surface of T-ALL cells obtained from a patient, wherein the absence of the TCR/CD3 complex is indicative of the non-responsiveness to said treatment.
  • the invention relates to a TCR-activating agent for use in a method for treating T-ALL in a patient classified as responder according to the method of the invention.
  • the invention relates to an anti-TCR antibody for use in a method for treating T-ALL.
  • OKT3 monoclonal antibody acts directly on T-ALL blasts. They demonstrated indeed that targeting CD3 both in vitro and in vivo, induces human primary T-ALL cell death and a molecular program that mimics thymic negative selection. Importantly, the inventors identified a new method for stratifying T-ALL patients and predicting their responsiveness to a treatment with an anti-CD3 antibody since they observed massive T-ALL cell death and TCR signaling activation in TCR-expressing T-ALL (also referred herein as CD3 -positive primary T-ALL cells) regardless of their molecular oncogenic subtype, but not in TCR-negative cases.
  • the present data provide strong and unexpected rationale for targeted therapy based on non-mitogenic anti-CD3 treatment of T-ALLs in patients identified as responder.
  • the invention provides methods and compositions (such as pharmaceutical compositions) for use in a method for treating T-ALL.
  • the invention also provides methods and compositions for use in a method for inhibiting proliferation and/or inducing cell death of T-ALL cells.
  • the invention also provides methods and compositions for use in a method for inducing irreversible differentiation of T-ALL cells.
  • the invention relates to an anti-CD3 antibody for use in a method for treating T-cell acute lymphoblastic leukemia (T-ALL).
  • T-ALL T-cell acute lymphoblastic leukemia
  • CD3 Cluster of Differentiation 3 refers to an antigen which is expressed on T cells as part of the multimolecular T cell receptor (TCR) and which consists of a homodimer or heterodimer formed from the association of two of four receptor chains: CD3-epsilon, CD3 -delta, CD3-zeta, and CD3 -gamma.
  • antibody that binds CD3 or "anti-CD3 antibody” include antibodies and antigen-binding fragments thereof that specifically recognize a single CD3 subunit (e.g., epsilon, delta, gamma or zeta), as well as antibodies and antigen-binding fragments thereof that specifically recognize a dimeric complex of two CD3 subunits (e.g., gamma/epsilon, delta/epsilon, and zeta/zeta CD3 dimers).
  • the antibodies and antigen-binding fragments of the invention may bind soluble CD3 and/or cell surface expressed CD3.
  • anti-CD3 antibody includes both monovalent antibodies with a single specificity, as well as bispecific antibodies comprising a first arm that binds CD3 and a second arm that binds a second (target) antigen.
  • the invention relates to an anti-TCR antibody for use in a method for treating T-cell acute lymphoblastic leukemia (T-ALL).
  • T-ALL T-cell acute lymphoblastic leukemia
  • TCR T Cell Receptor
  • MHC major histocompatibility complex
  • antibody that binds TCR or “anti-TCR antibody” include antibodies and antigen-binding fragments thereof that specifically recognize the constant region of the a chain of the TCR or otherwise specifically binds the a chain regardless of clonal origin of the T-cell as well as antibodies, or antigen binding fragment thereof, that specifically binds the constant region of the ⁇ chain of the TCR or otherwise specifically binds the ⁇ chain regardless of clonal origin of the T-cell.
  • antibody or “immunoglobulin” have the same meaning, and will be used equally in the present invention.
  • the term “antibody” as used herein refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that immunospecifically binds an antigen.
  • the term antibody encompasses not only whole antibody molecules, but also antibody fragments as well as variants (including derivatives) of antibodies and antibody fragments.
  • two heavy chains are linked to each other by disulfide bonds and each heavy chain is linked to a light chain by a disulfide bond. There are two types of light chain, lambda (1) and kappa (k).
  • the heavy chain includes two domains, a variable domain (VL) and a constant domain (CL).
  • the heavy chain includes four domains, a variable domain (VH) and three constant domains (CHI, CH2 and CH3, collectively referred to as CH).
  • VL variable domain
  • VH variable domain
  • CH constant domain
  • the constant region domains of the light (CL) and heavy (CH) chains confer important biological properties such as antibody chain association, secretion, trans-placental mobility, complement binding, and binding to Fc receptors (FcR).
  • the Fv fragment is the N-terminal part of the Fab fragment of an immunoglobulin and consists of the variable portions of one light chain and one heavy chain.
  • the specificity of the antibody resides in the structural complementarity between the antibody combining site and the antigenic determinant.
  • Antibody combining sites are made up of residues that are primarily from the hypervariable or complementarity determining regions (CDRs). Occasionally, residues from non hypervariable or framework regions (FR) influence the overall domain structure and hence the combining site.
  • Complementarity Determining Regions or CDRs refer to amino acid sequences which together define the binding affinity and specificity of the natural Fv region of a native immunoglobulin binding site.
  • the light and heavy chains of an immunoglobulin each have three CDRs, designated L-CDR1, L-CDR2, L- CDR3 and H-CDR1, H-CDR2, H-CDR3, respectively.
  • An antigen-binding site therefore, includes six CDRs, comprising the CDR set from each of a heavy and a light chain V region.
  • Framework Regions (FRs) refer to amino acid sequences interposed between CDRs.
  • Fab denotes an antibody fragment having a molecular weight of about
  • F(ab')2 refers to an antibody fragment having a molecular weight of about 100,000 and antigen binding activity, which is slightly larger than the Fab bound via a disulfide bond of the hinge region, among fragments obtained by treating IgG with a protease, pepsin.
  • Fab refers to an antibody fragment having a molecular weight of about 50,000 and antigen binding activity, which is obtained by cutting a disulfide bond of the hinge region of the F(ab')2.
  • a single chain Fv (“scFv”) polypeptide is a covalently linked VH: : VL heterodimer which is usually expressed from a gene fusion including VH and VL encoding genes linked by a peptide-encoding linker.
  • dsFv is a VH:: VL heterodimer stabilised by a disulfide bond.
  • Divalent and multivalent antibody fragments can form either spontaneously by association of monovalent scFvs, or can be generated by coupling monovalent scFvs by a peptide linker, such as divalent sc(Fv)2.
  • a peptide linker such as divalent sc(Fv)2.
  • the term "diabodies” refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy- chain variable domain (VH) connected to a light- chain variable domain (VL) in the same polypeptide chain (VH-VL).
  • Antibodies directed against an antigen of interest can be raised according to known methods by administering the appropriate antigen or epitope (e.g. a full- length CD3 protein or antigenic peptide fragment of CD3) to a host animal selected, e.g., from pigs, cows, horses, rabbits, goats, sheep, Camelidae (camel, dromedary, llama) and mice, among others.
  • a host animal selected, e.g., from pigs, cows, horses, rabbits, goats, sheep, Camelidae (camel, dromedary, llama) and mice, among others.
  • a host animal selected, e.g., from pigs, cows, horses, rabbits, goats, sheep, Camelidae (camel, dromedary, llama) and mice, among others.
  • Various adjuvants known in the art can be used to enhance antibody production.
  • antibodies useful in practicing the invention can be
  • Techniques for production and isolation include but are not limited to the hybridoma technique, the human B-cell hybridoma technique and the EBV-hybridoma technique.
  • techniques described for the production of single chain antibodies can be adapted to produce single chain antibodies against the CD3.
  • Useful antibodies according to the invention also include antibody fragments including but not limited to F(ab')2 fragments, which can be generated by pepsin digestion of an intact antibody molecule, and Fab fragments, which can be generated by reducing the disulfide bridges of the F(ab')2 fragments.
  • Fab and/or scFv expression libraries can be constructed to allow rapid identification of fragments having the desired specificity to the CD3.
  • the anti-CD3 or the anti-TCR antibodies can be polyclonal or monoclonal.
  • the antibodies can also be chimeric (i.e., a combination of sequences from more than one species, for example, a chimeric mouse-human immunoglobulin), humanized or fully-human.
  • Human antibodies avoid certain of the problems associated with antibodies that possess murine or rat (or other species) variable and/or constant regions. The presence of such murine or rat derived proteins can lead to the rapid clearance of the antibodies or can lead to the generation of an immune response against the antibody by a patient.
  • murine or rat derived antibodies In order to avoid the utilization of murine or rat derived antibodies, one can develop humanized antibodies or generate fully human antibodies through the introduction of human antibody function into a rodent so that the rodent would produce antibodies having fully human sequences.
  • chimeric antibody refers to a genetically engineered fusion of parts of an animal antibody, typically a mouse antibody, with parts of a human antibody. Generally, chimeric antibodies contain approximately 33% mouse protein and 67% human protein. Developed to reduce the Human Anti-animal Antibodies response elicited by animal antibodies, they combine the specificity of the animal antibody with the efficient human immune system interaction of a human antibody.
  • Humanized antibodies and antibody fragments therefrom can also be prepared according to known techniques.
  • “Humanized antibodies” are forms of non-human (e.g., rodent) chimeric antibodies that contain minimal sequence derived from non- human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region (CDRs) of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity and capacity.
  • donor antibody such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity and capacity.
  • framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • the human chimeric antibody of the invention can be produced by obtaining nucleic sequences encoding VL and VH domains as previously described, constructing a human chimeric antibody expression vector by inserting them into an expression vector for animal cell having genes encoding human antibody CH and human antibody CL, and expressing the coding sequence by introducing the expression vector into an animal cell.
  • the CH domain of a human chimeric antibody it may be any region which belongs to human immunoglobulin, but those of IgG class are suitable and any one of subclasses belonging to IgG class, such as IgGl, IgG2, IgG3 and IgG4, can also be used.
  • the CL of a human chimeric antibody may be any region which belongs to Ig, and those of kappa class or lambda class can be used.
  • Methods for producing chimeric antibodies involve conventional recombinant DNA and gene transfection techniques are well known in the art (See patent documents US5,202,238; and US5,204,244).
  • the humanized antibody of the invention may be produced by obtaining nucleic acid sequences encoding CDR domains, as previously described, constructing a humanized antibody expression vector by inserting them into an expression vector for animal cell having genes encoding (i) a heavy chain constant region identical to that of a human antibody and (ii) a light chain constant region identical to that of a human antibody, and expressing the genes by introducing the expression vector into an animal cell.
  • the humanized antibody expression vector may be either of a type in which a gene encoding an antibody heavy chain and a gene encoding an antibody light chain exists on separate vectors or of a type in which both genes exist on the same vector (tandem type).
  • humanized antibody expression vector of the tandem type In respect of easiness of construction of a humanized antibody expression vector, easiness of introduction into animal cells, and balance between the expression levels of antibody H and L chains in animal cells, humanized antibody expression vector of the tandem type is preferred.
  • tandem type humanized antibody expression vector include pKANTEX93 (WO 97/10354), pEE18 and the like.
  • the antibodies are fully human monoclonal antibodies.
  • Such fully human monoclonal antibodies directed against human CD3 or human TCR can be generated using transgenic or transchromosomic mice carrying parts of the human immune system rather than the mouse system.
  • transgenic and transchromosomic mice include mice referred to herein as the HuMAb Mouse® and KM Mouse®, respectively, and referred as "human Ig mice.”
  • the HuMAb Mouse® (Medarex®, Inc.) contains human immunoglobulin gene miniloci that encode unrearranged human heavy ( ⁇ and ⁇ ) and ⁇ light chain immunoglobulin sequences, together with targeted mutations that inactivate the endogenous ⁇ and ⁇ chain loci (see e.g., Lonberg et al. (1994) Nature 368(6474): 856-859). Accordingly, the mice exhibit reduced expression of mouse IgM or ⁇ , and in response to immunization, the introduced human heavy and light chain transgenes undergo class switching and somatic mutation to generate high affinity human IgGK monoclonal antibodies (Lonberg et al. (1994), supra; reviewed in Lonberg
  • Fully human antibodies of the invention can also be raised using a mouse that carries human immunoglobulin sequences on transgenes and transchomosomes, such as a mouse that carries a human heavy chain transgene and a human light chain transchromosome.
  • This mouse is referred to herein as a "KM mouse” and is described in detail in PCT Publication WO 02/43478.
  • a modified form of this mouse, which further comprises a homozygous disruption of the endogenous FcyRIIB receptor gene, is also described in PCT Publication WO 02/43478 and referred to herein as a "KM/FCGR2D mouse®.”
  • mice with either the HCo7 or HCol2 heavy chain transgenes or both can be used.
  • transgenic animal embodiments include the Xenomouse (Abgenix, Inc., U.S. Patent Nos. 5,939,598; 6,075, 181; 6,114,598; 6,150,584 and 6,162,963). Further embodiments include "TC mice” (Tomizuka et al. (2000) Proc. Natl. Acad. Set USA 97:722-727) and cows carrying human heavy and light chain transchromo somes (Kuroiwa et al. (2002) Nature Biotechnology 20:889-894; PCT Publication WO 02/092812).
  • a number of anti-CD3 antibodies are known, including but not limited to, OKT3 (muromonab/Orthoclone OKT3TM, Ortho Biotech, Raritan, NJ; U. S. Patent No. 4,361, 549); hOKT3Yl (teplizumab) (MGA031) (Herald et al, NEJM 346 (22): 1692-1698 (2002); TRX4 (otelixizumab); HuM291 (NuvionTM, Protein Design Labs, Fremont, CA); gOKT3-5 (Alegre et al. , J. Immunol. 148 (11): 3461-8 (1992); 1F4 (Tanaka et al, J. Immunol.
  • the anti-CD3 antibody is a non-mitogenic anti-CD3 antibody or a fragment thereof.
  • the non-mitogenic anti-CD3 antibody is a non- mitogenic anti-CD3 monoclonal antibody.
  • the present invention relates to a non- mitogenic anti-CD3 antibody for use in a method for treating T-cell acute lymphoblastic leukemia (T-ALL).
  • T-ALL T-cell acute lymphoblastic leukemia
  • the non-mitogenic anti-CD3 antibody is used in a method for inducing cell death of T-ALL cells, in particular TCR-expressing T-ALL cells or blasts (also referred herein as CD3 -positive primary T-ALL cells).
  • non-mitogenic has its general meaning in the art and has properties to bind to the CD3 antigen but without inducing cytokine production, as described in US7041289 Bl .
  • a non-mitogenic anti-CD3 antibody delivers for instance a partial T cell signal that renders activated T cells unresponsive.
  • the non-mitogenic anti-CD3 antibody is TRX4 (otelixizumab) or MGA031 (teplizumab).
  • the present invention relates to an anti-TCR antibody for use in a method for treating T-cell acute lymphoblastic leukemia (T-ALL).
  • T-ALL T-cell acute lymphoblastic leukemia
  • the anti-TCR antibody is used in a method for inducing cell death of T-ALL cells, in particular TCR- expressing T-ALL cells or blasts (also referred herein as CD3-positive primary T-ALL cells).
  • the antibody of the invention recognizes TCR a chains, and, thus the ⁇ / ⁇ TCR, generally (i.e., is a pan-specific TCR antibody, and, in particular, a pan-specific TCR a chain antibody).
  • the antibody of the invention recognizes TCR ⁇ chains, and, thus the ⁇ / ⁇ TCR, generally (i.e., is a pan-specific TCR antibody, and, in particular, a pan-specific TCR ⁇ chain antibody).
  • anti-TCR antibodies in particular anti-c$-TCR antibodies
  • anti-c$-TCR antibodies are known, including but not limited to those described in the international patent applications n° WO2013037484 and WO2015066379 which disclose humanized anti-c ⁇ TCR antibodies, derived from the murine monoclonal antibody BMA031 (see European patent application EP0403156) such as GL1BM VH31, GL1BM VH28, HEBE1 H66, and HEBE1 H71.
  • 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.]).
  • T-cell acute lymphoblastic leukemia refers a malignancy accounting for about 15% and 25% of ALL in pediatric and adult cohorts respectively.
  • T-ALLs are a heterogeneous group of diseases with regard to immunophenotype, cytogenetics, molecular genetic abnormalities and clinical features, including response to therapy as described in Graux et al. 2006.
  • EGIL European Group for the Immunological Characterization of Leukemias
  • the presence of cytoplasmic or membrane expression of CD3 defines T-ALL.
  • T-ALL the immature subgroup or pro-T-ALL is defined by the expression of only CD7;
  • pre-T-ALL also expresses CD2 and/or CD5 and/or CD8;
  • Till or cortical T-ALL shows CD la positivity;
  • TIV finally, mature T-ALL is characterized by the presence of surface CD3 and CD la negativity.
  • T-ALL present genetic abnormalities (involving the T- cell receptor (TCR), involving known oncogenes such as Notchl, HOXA cluster, TLXl(HOXl l) and TLX3 (HOX11L2), TALI and LYL1, involving tyrosine kinases such as ABL1, JAK3 and JAKl).
  • TCR T- cell receptor
  • the patients are refractory to other treatments.
  • refractory represents previously treated patients which were either non responsive to treatment with the agent or had a response to treatment and then relapsed.
  • sequential polychemotherapy are used for treating T-ALL according to different protocols.
  • said sequential polychemotherapy comprises sequentially administering to said patients corticosteroids (e.g. prednisone) and L-asparaginase (GRAALL regimen).
  • corticosteroids e.g. prednisone
  • GAAALL regimen L-asparaginase
  • the invention relates to an in vitro method for predicting the responsiveness of a patient affected with a T-ALL to a treatment with a TCR-activating agent.
  • the invention relates to an in vitro method for predicting the responsiveness of a patient affected with a T-ALL to a treatment with a TCR-activating agent comprising a step of determining the expression of TCR/CD3 complex at the surface of T-ALL cells obtained from a patient, wherein the absence of the TCR/CD3 complex is indicative of non-responsiveness to said treatment.
  • the term "predicting” refers to a probability or likelihood for a patient to respond to the treatment with a TCR-activating agent.
  • the term “responsiveness” refers to ability to assess the likelihood that treatment will or will not be clinically effective.
  • a patient denotes a mammal, typically a human.
  • a patient refers to any subject (typically human) afflicted with T-ALL.
  • TCR-activating agent is intended to encompass any refers to a natural or synthetic compound that has a biological effect which stimulate TCR/CD3 complex intracellular signaling activity.
  • the TCR activating agent stimulates TCR/CD3 complex presents on T-ALL cells.
  • stimulation of the TCR/CD3 complex is associated with a series of biochemical events initiated by TCR engagement (e.g., by antigen-MHC complex or anti-CD3 cross-linking) and that typically triggers (or contributes to triggering) one or more pathways (by, for example, activating transcription of certain genes such as, e.g., BCL2L1-BIM, GIMAP4, LiPA4 or Nur77 also referred as NR4A1).
  • the TCR-activating agents include but are not limited to antibodies, synthetic or native sequence peptides and small molecules which bind to the TCR/CD3 complex.
  • the TCR-activating agent is an anti-CD3 antibody as above- described.
  • the anti-CD3 antibody is a non-mitogenic anti-CD3 antibody such as for instance TRX4 (otelixizumab) or MGA031 (teplizumab).
  • the TCR-activating agent is an anti-TCR antibody as above- described.
  • the anti-TCR antibody is an anti-aP-TCR antibody such as for instance GL1BM VH31, GL1BM VH28, HEBE1 H66, and HEBE1 H71.
  • the TCR-activating agent is an epitope obtained from a T-ALL cells of the patient, more particularly TCR idiotypes which may function as antigen and activate T cells.
  • the TCR-activating agent is an inhibitor of TCR regulating tyrosine phosphatases, e.g. CD45, SHPl, SHP2 and PTPN22.
  • the term "inhibitor of TCR regulating tyrosine phosphatases” refers to any compound, natural or synthetic, which results in a decreased phosphorylation of the tyrosine present on the intracellular domain of TCR.
  • TCR regulating tyrosine phosphatases inhibitors such as inhibitors of CD45 and/or SHPl are described in the international patent applications n° W09946268 and W09946236.
  • the inhibitor of TCR regulating tyrosine phosphatases is an inhibitor of CD45, SHPl, SHP2 or PTPN22 gene expression.
  • inhibitor of gene expression refers to a natural or synthetic compound that has a biological effect to inhibit the expression of a gene is meant a CD45, SHP1, SHP2 or PTPN22 mRNA, protein, peptide, or polypeptide.
  • Inhibitors of gene expression for use in the 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 targeted mRNA by binding thereto and thus preventing protein translation or increasing mRNA degradation, thus decreasing the level of the targeted protein (i.e. CD45), 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 protein can be synthesized, e.g., by conventional phosphodiester techniques and administered by e.g., intravenous injection or infusion.
  • RNA interference small double stranded RNA
  • dsRNA or dsRNA-encoding vector are well known in the art for genes whose sequence is known (e.g. see Tuschi, T. et al. (1999); Elbashir, S. M. et al. (2001); Hannon, GJ. (2002); McManus, MT. et al. (2002); Brummelkamp, TR. et al. (2002); U.S. Pat. Nos. 6,573,099 and 6,506,559; and International Patent Publication Nos. WO 01/36646, WO 99/32619, and WO 01/68836).
  • Short hairpin RNAs can also function as inhibitors of gene expression for use in the invention.
  • determining encompasses detecting or quantifying. Indeed, an expression level can be qualitative or quantitative with or without reference to a control or a predetermined value. Thus, a determination of whether a polypeptide is present or absent (e.g., detectable or undetectable) constitutes determining its expression level in various embodiments while in other embodiments, a quantitative level is determined.
  • detecting means determining if TCR/CD3 complex is present or not at the surface of T-ALL cells obtained from a patient and "quantifying" means determining the amount of TCR/CD3 complex present at the surface of T-ALL cells.
  • TCR/CD3 T cell receptor-CD3 complex
  • MHC major histocompatibility complex
  • the method of the invention comprises a step of determining the expression TCR/CD3 complex at the surface of T-ALL cells obtained from a patient, wherein the absence of TCR/CD3 complex is indicative of non-responsiveness to said treatment.
  • Determination of the expression level of TCR/CD3 complex at the surface of T-ALL cells may be performed by a variety of techniques.
  • the expression level as determined is a relative expression level.
  • the determination comprises contacting the biological sample with selective reagents ligands, and thereby detecting the presence, or measuring the amount, of polypeptides of interest originally in said biological sample. Contacting may be performed in any suitable device, such as a plate, microtiter dish, test tube, well, glass, column, and so forth. In specific embodiments, the contacting is performed on a substrate coated with the reagent, such as a specific ligand array.
  • the substrate may be a solid or semi-solid substrate such as any suitable support comprising glass, plastic, nylon, paper, metal, polymers and the like.
  • the substrate may be of various forms and sizes, such as a slide, a membrane, a bead, a column, a gel, etc.
  • the contacting may be made under any condition suitable for a detectable complex, such as an antibody-antigen complex, to be formed between the reagent and the polypeptides of the biological sample.
  • Such methods comprise contacting the biological sample of a patient containing T-ALL cells with a binding partner capable of selectively interacting with the protein present in said sample.
  • the binding partner is generally an antibody that may be polyclonal or monoclonal, preferably monoclonal.
  • the binding partners of the invention such as antibodies, may be labelled with a detectable molecule or substance, such as a fluorescent molecule, a radioactive molecule or any others labels known in the art.
  • Labels are known in the art that generally provide (either directly or indirectly) a signal.
  • the term "labelled" with regard to the antibody or aptamer is intended to encompass direct labeling of the antibody by coupling (i.e., physically linking) a detectable substance, such as a radioactive agent or a fluorophore (e.g.
  • FITC fluorescein isothiocyanate
  • PE phycoerythrin
  • Cy5 lndocyanine
  • the aforementioned assays generally involve the coating of the binding partner (ie. antibody) in a solid support.
  • Solid supports which can be used in the practice of the invention include substrates such as nitrocellulose (e. g., in membrane or microtiter well form); polyvinylchloride (e.
  • polystyrene latex e.g., beads or microtiter plates
  • polyvinylidine fluoride e.g., diazotized paper
  • nylon membranes e.g., nylon membranes
  • activated beads e.g., magnetically responsive beads, and the like.
  • the step of determining the expression of TCR/CD3 complex at the surface of T-ALL cells is performed by immunoassays such as for instance immunophenotyping as described in Asnafi et al., 2004 or by immunohistochemistry (IHC).
  • immunoassays such as for instance immunophenotyping as described in Asnafi et al., 2004 or by immunohistochemistry (IHC).
  • the step of determining the expression of a target protein such as TCR/CD3 complex by a cell population such as T-ALL cells may be carried out by a variety of methods for detecting a particular immune cell population available for a skilled artisan, including immunoselection techniques, such as high-throughput cell sorting using flow cytometric methods, affinity methods with antibodies labeled to magnetic beads, biodegradable beads, non-biodegradable beads, and combination of such methods.
  • immunoselection techniques such as high-throughput cell sorting using flow cytometric methods, affinity methods with antibodies labeled to magnetic beads, biodegradable beads, non-biodegradable beads, and combination of such methods.
  • flow cytometric methods refers to a technique for counting cells of interest, by suspending them in a stream of fluid and passing them through an electronic detection apparatus.
  • Flow cytometric methods allow simultaneous multiparametric analysis of the physical and/or chemical parameters of up to thousands of particles per second, such as fluorescent parameters.
  • Modern flow cytometric instruments usually have multiple lasers and fluorescence detectors.
  • a common variation of flow cytometric techniques is to physically sort particles based on their properties, so as to purify or detect populations of interest, using "fluorescence-activated cell sorting".
  • FACS fluorescence-activated cell sorting
  • T-ALL cells may be stained with a FITC-labeled anti-CD38 mAb (such as the clone 145-2C11 purchased by Pharmingen).
  • a FITC-labeled anti-CD38 mAb such as the clone 145-2C11 purchased by Pharmingen.
  • the invention relates to an in vitro method for predicting the responsiveness of a patient affected with a T-ALL to a treatment with a TCR-activating agent comprising a step of determining the presence of TCR- expressing T-ALL cells (also referred as CD3-positive T-ALL cells) in a biological sample obtained from a patient, wherein the absence of the TCR-expressing T-ALL cells (or CD3- positive T-ALL cells) is indicative of non-responsiveness to said treatment.
  • TCR-expressing T-ALL cells also referred as CD3-positive T-ALL cells
  • IHC specifically provides a method of determining the expression of a target protein in a biological sample or tissue specimen in situ.
  • the overall cellular integrity of the sample is maintained in IHC, thus allowing detection of both the presence and location of the target of interest.
  • a biological sample is fixed with formalin, embedded in paraffin and cut into sections for staining and subsequent inspection by light microscopy.
  • Current methods of IHC use either direct labelling or secondary antibody-based or hapten-based labelling. Examples of known IHC systems include, for example, EnVision TM (DakoCytomation), Powervision® (Immunovision, Springdale, AZ), the NBATM kit (Zymed Laboratories Inc., South San Francisco, CA), HistoFine ® (Nichirei Corp, Tokyo, Japan).
  • the method further comprises a step of analysing TCR clustering by performing flow cytometry analysis.
  • the invention relates to a TCR-activating agent for use in a method for treating T-ALL in a patient classified as responder according to the method above-described.
  • the TCR-activating agent is a non-mitogenic anti-CD3 antibody.
  • the invention also relates to a method for treating T-ALL in a patient in need thereof and identified as responder according to a method of the invention comprising a step of administrating to said patient a therapeutically amount of a TCR-activating agent.
  • responder refers to a patient who may respond to a treatment with a TCR-activating agent such as an anti-CD3 antibody, i.e. least one of his symptoms is expected to be alleviated, or the development of the disease is stopped, or slowed down.
  • the invention also relates to pharmaceutical compositions comprising a TCR-activating agent for use in a method for treating T-cell acute lymphoblastic leukemia (T-ALL).
  • T-ALL T-cell acute lymphoblastic leukemia
  • the invention also relates to pharmaceutical compositions comprising an anti-CD3 antibody for use in a method for treating T-cell acute lymphoblastic leukemia (T-ALL).
  • T-ALL T-cell acute lymphoblastic leukemia
  • said anti-CD3 antibody is a non-mitogenic anti-CD3 antibody such as for instance TRX4 (otelixizumab) or MGA031 (teplizumab).
  • the invention also relates to pharmaceutical compositions comprising an anti-TCR antibody for use in a method for treating T-cell acute lymphoblastic leukemia (T-ALL).
  • T-ALL T-cell acute lymphoblastic leukemia
  • the anti-TCR antibody is an anti-aP-TCR antibody such as for instance GL1BM VH31, GL1BM VH28, HEBE1 H66, and HEBE1 H71.
  • an antibody of the invention may be combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form therapeutic compositions.
  • pharmaceutically acceptable excipients such as a carboxylate, aminoethylcellulose, aminoethyl cellulose, aminoethyl cellulose, aminoethyl cellulose, aminoethyl sulfate, a pharmaceutically acceptable graft copolymer, or adiluent, encapsulating material or formulation auxiliary of any type.
  • compositions of the invention can be formulated for a topical, oral, parenteral, intranasal, intravenous, intramuscular, subcutaneous or intraocular administration and the like.
  • 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 doses used for the administration can be adapted as a function of various parameters, and in particular as a function of the mode of administration used, of the relevant pathology, or alternatively of the desired duration of treatment.
  • an effective amount of the antibody may be dissolved or dispersed in a pharmaceutically acceptable carrier or aqueous medium.
  • 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 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.
  • Dosages to be administered depend on individual needs, on the desired effect and the chosen route of administration. It is understood that the dosage administered will be dependent upon the age, sex, health, and weight of the recipient, concurrent treatment, if any, frequency of treatment, and the nature of the effect desired.
  • the total dose required for each treatment may be administered by multiple doses or in a single dose.
  • the doses used for the administration can be adapted as a function of various parameters, and in particular as a function of the mode of administration used, of the relevant pathology, or alternatively of the desired duration of treatment.
  • the daily dosage of the antibodies 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 active ingredient for the symptomatic adjustment of the dosage to the subject to be treated.
  • a medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, preferably from 1 mg to about 100 mg of the active ingredient.
  • 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 10 mg/kg of body weight per day.
  • the pharmaceutical composition of the invention may further comprise an additional drug, in particular a chemotherapeutic drug useful in T-ALL.
  • chemotherapeutic drugs useful in T-ALL include but not limited to, corticosteroids (e.g. prednisone), L-asparaginase, non-selective inhibitors of NOTCH 1, ABLl inhibitors and JAK3 inhibitors and PBK-AKT-mTOR pathway inhibitors as described in Roti et al, 2014.
  • Non-selective inhibitors of NOTCH1 include soluble or cell-associated Notch decoys, [gamma] -secretase inhibitors, intracellular MAML1 decoys, and Ras signaling inhibitors.
  • [gamma]-Secretase inhibitors (GSI) have the most immediate therapeutic potential.
  • Blocking specific E3 ligases responsible for ubiquitination of Notch ligands or mono-ubiquitination of Notch receptors can also be used.
  • Still other NOTCH 1 inhibitors that can be used are described in WO 2003/013527 and paragraphs 0016-0019 of US 2006/205666.
  • Non-limiting examples of ABLl inhibitors include imatinib (STI571) (Novartis), nilotinib AMN107 (Novartis), dasatinib (BMS-354825) (BMS), ponatinib (AP24534), bosutinib (SKI-606) (Pfizer) and bafetinib.
  • Exemplary ABLl inhibitors include but are not limited to, those as described in the following patent applications: N-phenyl-2-pyrimidine- amine derivatives (EP0564409), pyrimidinylaminobenzamide derivatives (WO2004/005281), cyclic compounds (WO00/62778), bicyclic heteroaryl compounds (WO2007/075869), substituted 3-cyano quinoline derivatives (US6002008), 4-anilo-3-quinolinecarbonitrile derivatives (WO2005/04669) and amide derivatives (US7728131 and WO2005/063709).
  • Non- limiting examples of PI3K inhibitors include: NVP-BEZ235 (BEZ235) (Novartis); LY294002 (Cell Signaling #9901); GDC-0941 (Genentech/Roche); GDC-0980 (Genentech); PI- 103 (Piramed); XL147 (Exilixis/Sanofi-Aventis); XL418 (Exilixis); XL665 (Exelixis); LY29002 (Eli Lilly); ZSTK474 (Zenyaku Kogyo); BGT226 (Novartis); wortmannin; quercetin; tetrodotoxin citrate (Wex Pharmaceuticals); thioperamide maleate; IC87114; PIK93; TGX-115; deguelin; NU 7026; OSU03012; tandutinib (Millennium Pharmaceuticals); MK-2206 (Merck); OSU-03012;
  • Exemplary PI3K inhibitors include but are not limited to those as described in the following patent applications: WO2008/027584, WO2008070150, 2,3-dihydroimidazo[l,2-c]quinazolines (WO2008/125833), 2-morpholin-4- yl-pyrimidines (WO2008/125835), pyrimidines (WO2008/125839), bicyclic heteroaryls (WO2009/010530), thiazolidinones (WO2009/026345), pyrrolothiazoles (WO2009/071888), tricyclic thiazole and thiophene derivatives (WO2009/071890), fused bicyclic thiazole and thiophene derivatives (WO2009/071895) and oxazole substituted indazoles (WO2010/125082).
  • the invention also relates to a method for treating T-cell acute lymphoblastic leukemia (T-ALL) in a patient in need thereof, comprising a step of administering to said patient a therapeutically effective amount of an anti-CD3 antibody or an anti TCR-antibody.
  • T-ALL T-cell acute lymphoblastic leukemia
  • terapéuticaally effective amount is meant an amount sufficient to achieve a concentration of antibodies which is capable of preventing, treating or slowing down the disease to be treated. Such concentrations can be routinely determined by those of skilled in the art.
  • the amount of the polypeptide actually administered will typically be determined by a physician or a veterinarian, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the patient, the severity of the subject's symptoms, and the like. It will also be appreciated by those of skilled in the art that the dosage may be dependent on the stability of the administered polypeptide.
  • the treatment with an anti-CD3 antibody is administered in more than one cycle, i.e. the administration of an anti-CD3 antibody (such as an OKT3, TRX4 or MGA031 monoclonal antibodies) is repeated at least once.
  • the intervals i.e. the time between the start of two subsequent cycles, are typically several days.
  • Kit-of-parts compositions
  • the anti-CD3 antibody such as the OKT3 or the anti-TCR antibody as above-described and the chemotherapeutic agent may be combined within one formulation and administered simultaneously. However, they may also be administered separately, using separate compositions. It is further noted that they may be administered at different times.
  • the invention relates to a kit-of-parts composition
  • a kit-of-parts composition comprising an anti-CD3 antibody as above-defined and a chemotherapeutic agent.
  • the invention relates to a kit-of-parts composition
  • a kit-of-parts composition comprising an anti-TCR antibody as above-defined and a chemotherapeutic agent.
  • the invention relates to a kit-of-parts composition
  • a kit-of-parts composition comprising an anti-CD3 antibody as above-defined and a chemotherapeutic agent for use in a method for treating T-ALL.
  • the invention relates to a kit-of-parts composition
  • a kit-of-parts composition comprising an anti-TCR antibody as above-defined and a chemotherapeutic agent for use in a method for treating T-ALL.
  • kit means especially a "kit-of-parts" in the sense that the combination partners as defined above can be dosed independently or by use of different fixed combinations with distinguished amounts of the combination partners, i.e. simultaneously or at different time points.
  • the parts of the kit of parts can then, e.g., be administered simultaneously or chronologically staggered, that is at different time points and with equal or different time intervals for any part of the kit of parts.
  • the ratio of the total amounts of the combination partners to be administered in the combined preparation can be varied.
  • the combination partners can be administered by the same route or by different routes. When the administration is sequential, the first partner may be for instance administered 1, 2, 3, 4, 5, 6, 7, days before the second partner.
  • FIGURES are a diagrammatic representation of FIGURES.
  • Figure 1 In vivo TCR stimulation with an agonistic monoclonal antibody inhibits T-cell leukemogenesis in the mouse TEL-JAK2 model.
  • A Percentage of leukemic cells within peripheral blood leukocytes of female mice treated with 5 daily doses of 20 ⁇ g 145- 2C11 mAb or control hamster IgG (arrows), starting 1 day after female TJM leukemic cell inoculation.
  • B Kaplan-Meier leukemia- free survival curves of mice transplanted with TJM leukemic cells and treated with antibody before leukemia onset (arrows) (Log-rank (Mantel- Cox) test, P value ⁇ 0.01).
  • C Percentage of leukemic cells within peripheral blood leukocytes of mice treated with 5 daily doses of 20 ⁇ g 145-2C11 mAb or control hamster IgG, starting 2 days after TEL-JAK2 leukemic cell inoculation (arrows).
  • D Kaplan-Meier leukemia-free survival curves for mice transplanted with TEL-JAK2 leukemic cells and treated with antibody before leukemia onset (P value ⁇ 0.01).
  • E Percentage of leukemic cells within peripheral blood leukocytes of female TEL-JAK2:Marylin double transgenic mice (TJM) secondary leukemia mice treated with 5 daily doses of 20 ⁇ g 145-2C11 mAb or control hamster IgG (arrows), after leukemia detection.
  • F Kaplan-Meier leukemia- free survival curves for mice transplanted with TEL-JAK2 leukemic cells and treated with antibody after leukemia onset (arrows) (P value ⁇ 0.05).
  • FIG. 2 In vivo TCR stimulation with an agonistic monoclonal antibody inhibits T-cell leukemogenesis in human T-ALL cells xenografted in NSG mice.
  • B NSG mice were injected with 10 6 T-ALL cells.
  • Figure 3 Anti-CD3 stimulation of ALL-SIL TCR-HY cells induces TCR signaling and cell death.
  • A Percentage of annexin V-positive ALL-SIL or ALL-SIL TCR-HY cells either left unstimulated or stimulated by CD3/CD28 coated-beads for the indicated time.
  • B Western Blot analysis of caspase3 and caspase7 cleavage in ALL-SIL and ALL-SIL TCR-HY cells either left unstimulatedor stimulated by CD3/CD28 coated-beads for 48 and 72 h. Actin was used as a loading control.
  • C Growth curve of unstimulated and CD3/CD28-stimulated ALL-SIL and ALL-SIL TCR-HY cells.
  • E Phospho-ERK, phospho-AKT and phospho- STAT3 expression detected by flow cytometry in non- stimulated (top line in each panel) or 30 min anti-CD3/CD28-stimulated (bottom line) ALL-SIL TCR-HY cells.
  • FIG. 4 Anti-CD3 induces human primary T-ALL cell death in vitro and mimics thymic negative selection. Comparison of cell death induction in CD3-positive primary, diagnostic T-ALL cells (9 cases) versus CD3 -negative primary T-ALL cells (4 cases) after 3 days of co-culture on OP9-DL1 in the presence or absence of CD3/CD28 stimulation.
  • mice The E ⁇ SR ⁇ -7 ⁇ X-J4AT2 transgenic mice (line 71) (Carron et al, 2000), Marylin transgenic mice (Lantz et al, 2000), and Rag2 knock-out, all on the C57BL/6 background, and Swiss Nude were maintained under specific-pathogen- free conditions in the animal facilities of the Institut Curie (Orsay, France) and University of Algarve (Faro, Portugal). All experimental procedures were performed in strict accordance with the recommendations of the European Commission (Directive 86/609/EEC), French National Committee (87/848) and Portuguese authorities (Decreto-Lei n°129/92) for the care and use of laboratory animals.
  • European Commission Directive 86/609/EEC
  • French National Committee 87/848
  • Portuguese authorities Decreto-Lei n°129/92
  • mice were euthanized by CO2 inhalation when terminally ill, due to either severe dyspnea caused by massive expansion of leukemic cells in the thymus, or weakness caused by leukemic dissemination to vital organs such as bone marrow, lung, and liver. Blood was collected from the submandibular vein.
  • leukemia transplantation assays 0.5-2 x 10 6 leukemic T cells collected from diseased female TEL-JAK2: Marylin double transgenic mice (TJM) mice were intravenously injected in the tail vein of recipient mice of the indicated gender and strain and regularly monitored for external signs of disease development or through blood analysis.
  • TJM Marylin double transgenic mice
  • Anti- CD3s 145-2C11 monoclonal antibody or control ChromPureSyrian Hamster IgG (Jackson Immunoresearch) was diluted in sterile PBS and intravenously administered on a regimen of 5 daily doses of 20 ⁇ g for C57BL6 mice or 50 ⁇ g for SwissNudemice). Statistical analyses and survival curves were calculated using Prism 5 (GraphPad).
  • the anti-CD3 OKT3 MAb BioXCell; 2 rounds of treatment with 40 ⁇ g/mice/day for 5 consecutive days separated by a 2 days interval
  • IgG2a iso type-matched
  • mice were treated when leukemic cells reached 1-4% of blood nucleated cells. Mice were monitored weekly by flow cytometry for leukemic load in peripheral blood, T-ALL cells being identified as FSC hl , hCD7+, hCD45+ cells.
  • T-ALL patient samples and cell lines Immunophenotypic and oncogenic features of T-ALL patients were identified as described (6, 47, 48). Fresh or thawed primary T-ALL samples (peripheral blood or bone marrow) obtained at diagnosis from adult and pediatric patients were used, as well as T-ALL cells xenografted in NSG mice. Informed consent was obtained according to the Declaration of Helsinki. All samples used contained >80% blasts. CD4+CD8+ (DP) human thymocytes were obtained and processed as described in Supplementary methods. The ALL-SIL cell line (DSMZ, Braunschweig, Germany, ACC511) were grown in vitro and transduced as described in Supplementary methods.
  • Flow cytometry Single cell suspensions prepared from lymphoid organs, bone marrow flushed from tibias or blood were stained with fluorochrome-labeled antibodies, and detected on a FACSCalibur cytometer (BD Biosciences), as previously described (Carron et al, 2000). FITC-, PE-, PE-Cy5-, PerCP/Cy5.5- or APC-conjugated antibodies specific for CD4 (H129.19), CD8aD(53-6.7), CD3s (145-2C11), TCRp (H57-597), V 6 TCR (RR4-7), (GL3), CD25 (7D4), CD45.1 (A20), and CD45.2 (104) (BD Biosciences) were used.
  • Antibodies used to identify human T-ALL cells were FITC-conjugated anti- human CD7 cl 4H9 and APC- conjugated anti-human CD45 (eBiosciences). Data analysis was performed using CellQuest (BD Biosciences) software.
  • Splenocytes were obtained from C57BL/6 female mice, subsequently irradiated (2000 rad) and preincubated with antigenic peptides at 37°C for 1 h (peptide pulsing).
  • Peptides used were DBY, the male antigen epitope specifically recognized by Marilyn transgenic TCR-HY, at 10, 1, 0.1, 0.01, 0.001 and 0.0001 ⁇ and OVA, a non-specific peptide, at 10 ⁇ .
  • Splenocytes were then plated out in 48-well plates, without removal of peptides. ALL-SIL TCR-HY cells were added to the co-culture system at a 1 : 100 ratio.
  • Antibodies used for human cells CD3, TCRVb6, Antibodies against phosphorylated ERK1/2, phosphorylated p38, phosphorylatedS6, were from BD, phosphorylated AKT from Cell Signaling.
  • Anti CD3/CD28 cell stimulation Short-term stimulation was performed on cells cultured in serum- free medium for at least 15min at 37°C prior to stimulation.
  • Murine anti- hCD3 mAb (OKT3, Bio legend) (20 ⁇ g/ml) was added to the cell culture on ice for 10 min and mAbs were then cross-linked by addition of goat-anti-mouse antibody to a final concentration of 50 ⁇ g/ml for 15 min.
  • Stimulation (0-10 min) was triggered by warming cells at 37°C and terminated either by cooling cells in cold PBS or fixing cells for Phosphokinase array and intracellular phosphoprotein (Phosflow) assays, respectively.
  • a bead-based assay was used in which cell lines were cultured in 96- or 24-well-plates and exposed to 4.5 ⁇ diameter superparamagnetic beads covalently coupled to anti-CD3 and anti- CD28 antibodies (Dynabeads® Human T-Activator CD3/CD28, Invitrogen) (duration of stimulation from 1 to 12 days).
  • Prolonged stimulation of primary T-ALL cells was performed in a co-culture assay with confluent OP9-DL1 in a a-MEM media supplemented with 20% FBS (Hyclone; ThermoFisherScientific), 50 ⁇ g/ml streptomycin and 50 IU penicillin and recombinant human cytokines hFLT3-L (5 ng/mL), hIL-7 (2 ng/mL) and hSCF (10 ng/mL) (Miltenyi).
  • FBS Hyclone; ThermoFisherScientific
  • 50 ⁇ g/ml streptomycin 50 ⁇ g/ml streptomycin and 50 IU penicillin and recombinant human cytokines hFLT3-L (5 ng/mL), hIL-7 (2 ng/mL) and hSCF (10 ng/mL) (Miltenyi).
  • CD3/CD28 coated-beads were
  • Cell stimulation and intracellular flow cytometry with phosphorylation specific antibodies Before being stimulated, cells were deprived of serum for at least 15min at 37°C. Cells were stimulated in serum-free RPMI medium at 37°C. For bead-based stimulation, 4.5 ⁇ diameter, superparamagnetic beads covalently coupled to anti-CD3 and anti-CD28 antibodies (Dynabeads® Human T-Activator CD3/CD28, Invitrogen) were used. The stimulation was terminated by addition of prewarmed formaldehyde (Cytofix Buffer, BD Biosciences) at 37°C for lOmin.
  • Cell death analysis Cells were incubated with CD3/CD28 beads according to the manufacturer's instruction, in a 96-well plate. A different time of culture, cell death was analysed by flow cytometry using AnnexinV-APC and propidium iodide (BD Biosciences) Western Blot: Cells were stimulated as previously describe with OKT3 or beads.
  • Proteins were separated on tris-glycine gels under reducing conditions and transferred to nitrocellulose membranes (Biorad). Immunob lotting was performed with following antibodies: phosphor-ERKl/2 (ref, cell Signaling), ERK1/2 (cell Signaling), NFAT1 (cell Signaling), Caspase3 (BD), Caspase7 (BD). Flow cytometry analysis of TCR clustering.
  • ALL-SIL TCR-HY cells were stained with TCR V 6-PE mAb (BD Biosciences) for 30 min at 4°C and DAPI for 5 min at room temperature prior stimulation and split into two samples; one sample remained unstimulated while the other one was stimulated by styrene beads (Polyscience) pre-coated with human anti- CD3s antibody (OKT3, Biolegend) for 10 min at 37°C. Cells were fixed with 1% formaldehyde. Staining was analyzed usinglmageStream X mkll (Amnis Merck-Millipore). Data analysis was performed using the IDEAS image analysis software (Amnis).
  • ALL-SIL TCR-HY cells and human DP thymocytes were either left unstimulated or were stimulated with CD3/CD28 mAbs for 5 min. Each stimulated condition was performed in replicate.
  • Cell lysates and Proteome Profiler Human phosphokinase array were performed according to the manufacturer's protocol. Chemiluminescence was detected by ChemiDoc XRS+ (Bio-Rad). The average signal (pixel density) of duplicate spots representing each phosphorylated kinase protein was normalized with the average signal of reference positive duplicate spots of each membrane using Image Lab software (Bio-Rad). Signal ratios for selected phosphoproteins were displayed in a heat-map using Treeview software. Cyclosporine A (Novartis) and PD184352 (Selleckchem) were used at ⁇ M. Effects were determined by analysis of cell death (Annexin V/PI assay) and expression of CD25 and CD69 activation markers.
  • RNA extraction was performed for the following cells: ALL-SIL-TCRa -GFP co-cultured on OP9-DL1, ALL-SIL-TCRa -GFP without co-culture, ALL-SIL transduced with TLX shRNA (sh-TLX), and ALL-SIL transduced with sh-control vectors. These samples were obtained as previously described (14) in duplicate, and RNA extraction was performed at an early time-point before cell death onset (48 h of co- culture for ALL-SIL-TCRa -GFP and 48h after puromycin selection for TLX shRNA/sh- control ALL-SIL). RNA hybridization was performed on Affymetrix U133 plus 2.0 microarrays.
  • the statistical data analysis was performed with R version2.9.0 using the "Affy" package from Bioconductor.
  • the probe intensities were log2 transformed and normalized using RMA.
  • Identification of differentially expressed genes was performed by Significance Analysis of Microarrays (SAM), using 500 permutations and a false discovery rate threshold of 5%.
  • Functional analysis of differentially expressed genes was performed using the Ingenuity Pathway Analysis (IP A; www.ingenuity.com) and the Database for Annotation, Visualization, and Integrated Discovery (DAVID; http://david.abcc.ncifcrf.gov)softwares.
  • Transgenic Marylin mice express in thymocytes an MHC class II -restricted TCR ( ⁇ . ⁇ ) specific for the H-Y male antigen (DBY), which in males induces thymocyte negative selection and maturation arrest (Lantz et al, 2000).
  • DBY H-Y male antigen
  • TEL-JAK2 transgenic mice were crossed with Marilyn mice to generate TEL-JAK2; Marylin (TJM) double transgenic mice.
  • TJM males developed leukemia with significantly delayed latency.
  • Flow cytometry analysis showed that of five diseased mice analyzed, one developed an IgM-positive immature-type B-cell lymphoma affecting the thymus and spleen, while four mice developed Thyl .2 + CD4 ⁇ CD8 + CD25 + T-cell leukemia infiltrating the thymus, spleen, lymph nodes and bone marrow.
  • transgenic TCR- ⁇ cell surface expression was low or absent.
  • mice expressing the transgenic Marilyn TCR present reduced thymocyte cellularity due to self-antigen-induced negative selection (Lantz et al, 2000).
  • female TJM CD45.1+ leukemic cells were transplanted to either female or male syngenic CD45.2 recipients.
  • the majority of recipient males presented on average much lower numbers of donor malignant cells in blood and survived considerably longer.
  • female recipients presented higher infiltration of CD45.1+ leukemic cells in spleen, lymph nodes, bone marrow and other organs than males sacrificed simultaneously.
  • TJM leukemic mice treated TJM leukemic mice with an anti-CD3smAb (145-2C11).
  • anti-CD3smAb 145-2C11
  • mice were transplanted to female mice, to avoid Marilyn TCR antigenic stimulation, and then treated 24 hours later and for 5 consecutive days with anti-CD3smAb, peripheral blood leukemia and fatal disease was prevented, as compared to control antibody- treated mice ( Figure 1 A,B).
  • anti-CD3streatment delayed the increase in peripheral blood leukemia and prolonged the survival of mice transplanted with either TJM ( Figure IE) or TEL-JAK2 ( Figure IF) leukemic cells.
  • the CD3santibody also delayed peripheral blood leukemia in Swiss Nude mice transplanted with TEL-JAK2 leukemic cells, indicating that the anti-leukemogenic effect of this antibody is due to a direct action on TCR/CD3 -positive leukemic T cells and not on host-derived T cells.
  • TALL cells obtained from a TCR+ T-ALL diagnostic cases xenotransplanted into NOD/SCID/y c -/- (NSG) mice.
  • NSG mice were injected with T-ALL cells and, 24 hours later, divided into 2 groups. The first was treated (2 rounds of 5 consecutive days) with the CD3-specific OKT3 monoclonal antibody and the second with an isotype control antibody.
  • Leukemic cells hCD45 + hCD7 + ) expansion was followed over time in the blood of recipient mice.
  • Anti-CD3 stimulation of ALL-SIL TCR-HY cells induces TCR signaling and cell death.
  • TCR-expressing T-ALL To test whether anti-CD3 stimulation of TCR-expressing T-ALL diagnostic samples also induced cell death, primary human T-ALL cells belonging to the major molecular oncogenic T-ALL subtypes(10, 18) were treated with anti-CD3/anti-CD28. Massive T-ALL cell death and TCR signaling activation were observed in TCR-expressing T-ALL but not in TCR-negative cases, regardless of their molecular oncogenic subtype (Fig. 4). As silencing of the TLX1 driving oncogene in ALL-SIL cells was previously shown to induce both differentiation and cell death(14), we compared the transcriptional profiles of ALL-SIL cells in response to either TCR stimulation or small hairpin RNA (shRNA)-mediated knock-down of TLX1.
  • shRNA small hairpin RNA
  • T-ALL therapies involve complex, often toxic chemotherapeutic regimens. Although T-ALL outcome has improved with current therapy, survival rates remain only around 50 and 70% at 5 years in adult and pediatric T-ALL, respectively(20, 21).
  • the genetic bases of T-ALL progression and maintenance are well characterized but have not translated so far into targeted therapies(lO). There is thus unmet need for new treatments to offer therapeutic options for refractory disease and to prevent relapse.
  • chronic/strong TCR signalling causes massive T-ALL cell death and shows potent tumor suppressive function in vivo.
  • Muromonab-CD3 (OKT3) was approved by the U.S. Food and Drug Administration (FDA) in 1985 for therapy of acute, glucocorticoid-resistant rejection of allogeneic renal, heart and liver transplants (22) and was in fact the first monoclonal antibody introduced in the clinic. Since then, a number of other monoclonal antibodies to CD3 were developed (23) that may prove superior to OKT3 in T-ALL treatment. Of note, encouraging response to OKT3 therapy was reported in an adult patient with an aggressive and chemotherapy-resistant T-ALL, but the basis of this response was not studied (24). A major drawback of current chemotherapeutic regimens in T-ALL is the frequent resistance to treatment and relapse.
  • FDA U.S. Food and Drug Administration
  • LICs Leukemia initiating cells from residual disease are thought to be responsible for relapsing cases (25, 26).
  • resistance of T-ALL to chemotherapy is in part linked to the recurrent genetic abnormalities selected during disease progression, e.g. inactivation of the PTEN tumor suppressor gene and the resulting activation of PI3kinase/AKT signaling (27, 28).
  • Whether administration of anti-CD3 therapy during the remission phase or its association with conventional chemotherapy regimens could target LICs and/or bypass molecular mechanisms of primitive resistance represent promising directions to be explored in future prospective studies.
  • T cells mature in the thymus following a highly orchestrated process controlled both by cell intrinsic (e.g. transcription factors) and extrinsic (e.g. stroma-derived cytokines/chemokines) molecular cues (29, 30).
  • Cell surface TCRD ⁇ expression in DP thymocytes allows recognition of specific self-MHC/peptide to transduce a positive selection signal and maturation into SP thymocytes.
  • DP thymocytes not receiving this signal die through lack of stimulation; whereas those whose TCR binds too strongly to self-MHC/peptide undergo activation- induced cell death and negative selection (5, 31).
  • TCR binding to pMHC is the triggering event but how TCR engagement leads to such divergent outcomes (survival and proliferation versus death) remains unclear (32).
  • these two contrasting processes are driven by a TCR signaling machinery of qualitatively similar composition (31).
  • the difference lies in the molecular interpretation of signals of different strength, which may rely on compartmentalization of key signaling players. Indeed, it has been shown that a small increase in ligand affinity for the TCR leads to a marked change in the subcellular localization (plasma membrane for negative selecting ligands versus Golgi complex for positively selecting ones) of essential adaptors of the Ras signaling pathway.
  • TCR B-cell receptor
  • pre-BCR B-cell malignancies
  • T-ALL often arises from immature T-cell precursors before the stage of negative selection (6, 42). Consequently, contingent expression of a non-negatively selected TCR will render T-ALL cells sensitive to TCR-activating apoptotic signals that mimic negative selection, as demonstrated here by anti- CD3 treatment in vitro and in vivo.
  • selective events enable cells to escape post-malignancy negative selection, such as loss of TCR surface expression, as found in a subset of T-ALL patients (6) and as demonstrated here in TJM double transgenic male mice.
  • Gene expression signatures define novel oncogenic pathways in T cell acute lymphoblastic leukemia. Cancer cell. 2002 Feb;l(l):75-87.
  • TEL-JAK2 transgenic mice develop T-cell leukemia. Blood. 2000 Jun 15;95(12):3891-9.
  • Yui MA Rothenberg EV. Developmental gene networks: a triathlon on the course to T cell identity. Nature reviews. 2014 Aug;14(8):529-45.
  • Pre-TCR expression cooperates with TEL-JAK2 to transform immature thymocytes and induce T-cell leukemia. Blood. 2007 May 1;109(9):3972-81.
  • T-ALL T-cell acute lymphoblastic leukemia
  • GAAALL Ratio ALL

Abstract

The present invention relates to an anti-CD3 antibody for use in a method for treating T-cell acute lymphoblastic leukemia (T-ALL).The present invention also relates to an in vitro method for predicting the responsiveness of a patient affected with a T-ALL to a treatment with a TCR-activating agent comprising a step of determining the expression of TCR/CD3 complex at the surface of T-ALL cells obtained from a patient, wherein the absence of the TCR/CD3 complex is indicative of the non-responsiveness to said treatment.

Description

TCR- ACTIVATING AGENTS FOR USE IN THE TREATMENT OF T-ALL
FIELD OF THE INVENTION:
The invention is in the field of oncology and more particularly treatment of T-cell acute lymphoblastic leukemia (T-ALL).
BACKGROUND OF THE INVENTION:
T-cell acute lymphoblastic leukemias (T-ALL) are malignant proliferations of thymocytes by multi-step oncogenic processes leading to T-cell differentiation arrest. Targeted therapy, including abrogation of a block to cell maturation, is a promising therapeutic approach in cancer. It has been recently reported that the cortical differentiation block observed within the TLX overexpressing subtype of T-ALL is due to failure to rearrange TCRa and that this blockage can be overcome by TLX abrogation or, more importantly, by downstream TCRaP expression despite the acquisition of a variety of additional genetic abnormalities (involving the T-cell receptor (TCR), involving known oncogenes such as Notchl, HOXA cluster, TLXl(HOXl l) and TLX3 (HOX11L2), TALI and LYL1, involving tyrosine kinases such as ABL1, JAK3 and JAKl). Therefore, despite advances in treatment of T-ALL, such pathology remains associated with high morbidity and mortality and there is a desperate need for new therapeutic strategies independently of the genetic abnormalities leading to said pathology as described in Roti et al., 2014. Gramatzki et al., 1995 treated a single patient with refractory T-ALL with OKT3 monoclonal antibody and observed a dramatic but transient decrease of lymphoblasts in the blood of said patient. OKT3 monoclonal antibody is known as a mitogenic anti-CD3 antibody. Indeed, OKT3 monoclonal antibody induces T cell activation leading to a massive release of cytokines before the suppression of T cell responses. According to this study, the mechanism underlying the therapeutic effect of OKT3 monoclonal antibody in said T-ALL patient is based on the stimulation of the immune system as suggested by the induction of IL-2 responsiveness of the malignant cells leading to propose a sequential application of OKT3 and IL-2 to render certain T cell tumours more susceptible to chemotherapy. SUMMARY OF THE INVENTION:
In a first aspect, the invention relates to a non-mitogenic anti-CD3 antibody for use in a method for treating T-cell acute lymphoblastic leukemia (T-ALL).
In a second aspect, the invention relates to a non-mitogenic anti-CD3 antibody for use in a method for inducing cell death of T-ALL cells.
In a third aspect, the invention relates to a pharmaceutical composition comprising a non-mitogenic anti-CD3 antibody for use in a method for treating T-ALL.
In a fourth aspect, the invention relates to an in vitro method for predicting the responsiveness of a patient affected with a T-ALL to a treatment with a TCR-activating agent comprising a step of determining the expression of TCR/CD3 complex at the surface of T-ALL cells obtained from a patient, wherein the absence of the TCR/CD3 complex is indicative of the non-responsiveness to said treatment.
In a fifth aspect, the invention relates to a TCR-activating agent for use in a method for treating T-ALL in a patient classified as responder according to the method of the invention.
In a sixth aspect, the invention relates to an anti-TCR antibody for use in a method for treating T-ALL.
DETAILED DESCRIPTION OF THE INVENTION:
Now, the inventors showed that OKT3 monoclonal antibody acts directly on T-ALL blasts. They demonstrated indeed that targeting CD3 both in vitro and in vivo, induces human primary T-ALL cell death and a molecular program that mimics thymic negative selection. Importantly, the inventors identified a new method for stratifying T-ALL patients and predicting their responsiveness to a treatment with an anti-CD3 antibody since they observed massive T-ALL cell death and TCR signaling activation in TCR-expressing T-ALL (also referred herein as CD3 -positive primary T-ALL cells) regardless of their molecular oncogenic subtype, but not in TCR-negative cases.
Thus, the present data provide strong and unexpected rationale for targeted therapy based on non-mitogenic anti-CD3 treatment of T-ALLs in patients identified as responder. Therapeutic methods and uses:
The invention provides methods and compositions (such as pharmaceutical compositions) for use in a method for treating T-ALL. The invention also provides methods and compositions for use in a method for inhibiting proliferation and/or inducing cell death of T-ALL cells.
The invention also provides methods and compositions for use in a method for inducing irreversible differentiation of T-ALL cells.
In a first aspect, the invention relates to an anti-CD3 antibody for use in a method for treating T-cell acute lymphoblastic leukemia (T-ALL).
As used herein, the term "Cluster of Differentiation 3" (CD3) refers to an antigen which is expressed on T cells as part of the multimolecular T cell receptor (TCR) and which consists of a homodimer or heterodimer formed from the association of two of four receptor chains: CD3-epsilon, CD3 -delta, CD3-zeta, and CD3 -gamma.
As used herein, the terms "antibody that binds CD3" or "anti-CD3 antibody" include antibodies and antigen-binding fragments thereof that specifically recognize a single CD3 subunit (e.g., epsilon, delta, gamma or zeta), as well as antibodies and antigen-binding fragments thereof that specifically recognize a dimeric complex of two CD3 subunits (e.g., gamma/epsilon, delta/epsilon, and zeta/zeta CD3 dimers). The antibodies and antigen-binding fragments of the invention may bind soluble CD3 and/or cell surface expressed CD3. The term "anti-CD3 antibody" includes both monovalent antibodies with a single specificity, as well as bispecific antibodies comprising a first arm that binds CD3 and a second arm that binds a second (target) antigen.
In another aspect, the invention relates to an anti-TCR antibody for use in a method for treating T-cell acute lymphoblastic leukemia (T-ALL).
As used herein, the term "T Cell Receptor" (TCR) refers to a molecule found on the surface of T lymphocytes (or T cells) that is responsible for recognizing antigens bound to major histocompatibility complex (MHC) molecules. The TCR is composed of two different protein chains (that is, it is a heterodimer). In humans 95% of T cells the TCR consists of an alpha (a) and beta (β) chain. As used herein, the terms "antibody that binds TCR" or "anti-TCR antibody" include antibodies and antigen-binding fragments thereof that specifically recognize the constant region of the a chain of the TCR or otherwise specifically binds the a chain regardless of clonal origin of the T-cell as well as antibodies, or antigen binding fragment thereof, that specifically binds the constant region of the β chain of the TCR or otherwise specifically binds the β chain regardless of clonal origin of the T-cell.
According to the invention, "antibody" or "immunoglobulin" have the same meaning, and will be used equally in the present invention. The term "antibody" as used herein refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that immunospecifically binds an antigen. As such, the term antibody encompasses not only whole antibody molecules, but also antibody fragments as well as variants (including derivatives) of antibodies and antibody fragments. In natural antibodies, two heavy chains are linked to each other by disulfide bonds and each heavy chain is linked to a light chain by a disulfide bond. There are two types of light chain, lambda (1) and kappa (k). There are five main heavy chain classes (or isotypes) which determine the functional activity of an antibody molecule: IgM, IgD, IgG, IgA and IgE. Each chain contains distinct sequence domains. The light chain includes two domains, a variable domain (VL) and a constant domain (CL). The heavy chain includes four domains, a variable domain (VH) and three constant domains (CHI, CH2 and CH3, collectively referred to as CH). The variable regions of both light (VL) and heavy (VH) chains determine binding recognition and specificity to the antigen. The constant region domains of the light (CL) and heavy (CH) chains confer important biological properties such as antibody chain association, secretion, trans-placental mobility, complement binding, and binding to Fc receptors (FcR). The Fv fragment is the N-terminal part of the Fab fragment of an immunoglobulin and consists of the variable portions of one light chain and one heavy chain. The specificity of the antibody resides in the structural complementarity between the antibody combining site and the antigenic determinant. Antibody combining sites are made up of residues that are primarily from the hypervariable or complementarity determining regions (CDRs). Occasionally, residues from non hypervariable or framework regions (FR) influence the overall domain structure and hence the combining site. Complementarity Determining Regions or CDRs refer to amino acid sequences which together define the binding affinity and specificity of the natural Fv region of a native immunoglobulin binding site. The light and heavy chains of an immunoglobulin each have three CDRs, designated L-CDR1, L-CDR2, L- CDR3 and H-CDR1, H-CDR2, H-CDR3, respectively. An antigen-binding site, therefore, includes six CDRs, comprising the CDR set from each of a heavy and a light chain V region. Framework Regions (FRs) refer to amino acid sequences interposed between CDRs. The term "Fab" denotes an antibody fragment having a molecular weight of about
50,000 and antigen binding activity, in which about a half of the N-terminal side of H chain and the entire L chain, among fragments obtained by treating IgG with a protease, papaine, are bound together through a disulfide bond. The term "F(ab')2" refers to an antibody fragment having a molecular weight of about 100,000 and antigen binding activity, which is slightly larger than the Fab bound via a disulfide bond of the hinge region, among fragments obtained by treating IgG with a protease, pepsin. The term "Fab"' refers to an antibody fragment having a molecular weight of about 50,000 and antigen binding activity, which is obtained by cutting a disulfide bond of the hinge region of the F(ab')2. A single chain Fv ("scFv") polypeptide is a covalently linked VH: : VL heterodimer which is usually expressed from a gene fusion including VH and VL encoding genes linked by a peptide-encoding linker. "dsFv" is a VH:: VL heterodimer stabilised by a disulfide bond. Divalent and multivalent antibody fragments can form either spontaneously by association of monovalent scFvs, or can be generated by coupling monovalent scFvs by a peptide linker, such as divalent sc(Fv)2. The term "diabodies" refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy- chain variable domain (VH) connected to a light- chain variable domain (VL) in the same polypeptide chain (VH-VL). By using a linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites. Antibodies directed against an antigen of interest (such as CD3 or TCR) can be raised according to known methods by administering the appropriate antigen or epitope (e.g. a full- length CD3 protein or antigenic peptide fragment of CD3) to a host animal selected, e.g., from pigs, cows, horses, rabbits, goats, sheep, Camelidae (camel, dromedary, llama) and mice, among others. Various adjuvants known in the art can be used to enhance antibody production. Although antibodies useful in practicing the invention can be polyclonal, monoclonal antibodies are preferred. Monoclonal antibodies can be prepared and isolated using any technique that provides for the production of antibody molecules by continuous cell lines in culture. Techniques for production and isolation include but are not limited to the hybridoma technique, the human B-cell hybridoma technique and the EBV-hybridoma technique. Alternatively, techniques described for the production of single chain antibodies (see, e.g., U.S. Pat. No. 4,946,778) can be adapted to produce single chain antibodies against the CD3. Useful antibodies according to the invention also include antibody fragments including but not limited to F(ab')2 fragments, which can be generated by pepsin digestion of an intact antibody molecule, and Fab fragments, which can be generated by reducing the disulfide bridges of the F(ab')2 fragments. Alternatively, Fab and/or scFv expression libraries can be constructed to allow rapid identification of fragments having the desired specificity to the CD3.
The anti-CD3 or the anti-TCR antibodies can be polyclonal or monoclonal. The antibodies can also be chimeric (i.e., a combination of sequences from more than one species, for example, a chimeric mouse-human immunoglobulin), humanized or fully-human. Human antibodies avoid certain of the problems associated with antibodies that possess murine or rat (or other species) variable and/or constant regions. The presence of such murine or rat derived proteins can lead to the rapid clearance of the antibodies or can lead to the generation of an immune response against the antibody by a patient. In order to avoid the utilization of murine or rat derived antibodies, one can develop humanized antibodies or generate fully human antibodies through the introduction of human antibody function into a rodent so that the rodent would produce antibodies having fully human sequences.
The term "chimeric antibody" refers to a genetically engineered fusion of parts of an animal antibody, typically a mouse antibody, with parts of a human antibody. Generally, chimeric antibodies contain approximately 33% mouse protein and 67% human protein. Developed to reduce the Human Anti-animal Antibodies response elicited by animal antibodies, they combine the specificity of the animal antibody with the efficient human immune system interaction of a human antibody.
Humanized antibodies and antibody fragments therefrom can also be prepared according to known techniques. "Humanized antibodies" are forms of non-human (e.g., rodent) chimeric antibodies that contain minimal sequence derived from non- human immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region (CDRs) of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity and capacity. In some instances, framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence. The humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. Methods for making humanized antibodies are described, for example, by Winter (U.S. Pat. No. 5,225,539) and Boss (Celltech, U.S. Pat. No. 4,816,397).
The human chimeric antibody of the invention can be produced by obtaining nucleic sequences encoding VL and VH domains as previously described, constructing a human chimeric antibody expression vector by inserting them into an expression vector for animal cell having genes encoding human antibody CH and human antibody CL, and expressing the coding sequence by introducing the expression vector into an animal cell. As the CH domain of a human chimeric antibody, it may be any region which belongs to human immunoglobulin, but those of IgG class are suitable and any one of subclasses belonging to IgG class, such as IgGl, IgG2, IgG3 and IgG4, can also be used. Also, as the CL of a human chimeric antibody, it may be any region which belongs to Ig, and those of kappa class or lambda class can be used. Methods for producing chimeric antibodies involve conventional recombinant DNA and gene transfection techniques are well known in the art (See patent documents US5,202,238; and US5,204,244).
The humanized antibody of the invention may be produced by obtaining nucleic acid sequences encoding CDR domains, as previously described, constructing a humanized antibody expression vector by inserting them into an expression vector for animal cell having genes encoding (i) a heavy chain constant region identical to that of a human antibody and (ii) a light chain constant region identical to that of a human antibody, and expressing the genes by introducing the expression vector into an animal cell. The humanized antibody expression vector may be either of a type in which a gene encoding an antibody heavy chain and a gene encoding an antibody light chain exists on separate vectors or of a type in which both genes exist on the same vector (tandem type). In respect of easiness of construction of a humanized antibody expression vector, easiness of introduction into animal cells, and balance between the expression levels of antibody H and L chains in animal cells, humanized antibody expression vector of the tandem type is preferred. Examples of tandem type humanized antibody expression vector include pKANTEX93 (WO 97/10354), pEE18 and the like.
Methods for identifying CDRs of a monoclonal antibody are well known in the art (See Antibody Engineering: Methods and Protocols, 2004).
In one embodiment, the antibodies are fully human monoclonal antibodies. Such fully human monoclonal antibodies directed against human CD3 or human TCR can be generated using transgenic or transchromosomic mice carrying parts of the human immune system rather than the mouse system. These transgenic and transchromosomic mice include mice referred to herein as the HuMAb Mouse® and KM Mouse®, respectively, and referred as "human Ig mice."
The HuMAb Mouse® (Medarex®, Inc.) contains human immunoglobulin gene miniloci that encode unrearranged human heavy (μ and γ) and κ light chain immunoglobulin sequences, together with targeted mutations that inactivate the endogenous μ and κ chain loci (see e.g., Lonberg et al. (1994) Nature 368(6474): 856-859). Accordingly, the mice exhibit reduced expression of mouse IgM or κ, and in response to immunization, the introduced human heavy and light chain transgenes undergo class switching and somatic mutation to generate high affinity human IgGK monoclonal antibodies (Lonberg et al. (1994), supra; reviewed in Lonberg
(1994) Handbook of Experimental Pharmacology 113:49-101 : Lonberg, N. and Huszar, D.
(1995) Intern. Rev. Immunol. 13: 65-93, and Harding and Lonberg (1995) Ann. N Y. Acad. Sci. 764:536-546). Preparation and use of the HuMAb Mouse®, and the genomic modifications carried by such mice, is further described in Taylor et al. (1992) Nucleic Acids Research 20:6287-6295; Chen et al. (1993) International Immunology 5: 647-656; Tuaillon et al. (1993) Proc. Natl. Acad. Sci. USA 90:3720-3724; Choi et al. (1993) Nature Genetics 4: 117 -123; Chen et al. (1993) EMBO J. 12: 821-830; Tuaillon et al. (1994) J. Immunol. 152:2912-2920; Taylor et al. (1994) International Immunology 6: 579-591 ; and Fishwild et al. (1996) Nature Biotechnology 14: 845-851, the contents of all of which are hereby specifically incorporated by reference in their entirety. See further, U.S. Patent Nos. 5,545,806; 5,569,825; 5,625, 126; 5,633,425; 5,789,650; 877,397; 5,661,016; 5,814,318; 5,874,299; 5,770,429; and 5,545,807; PCT Publication Nos. WO 92/03918; WO 93/12227; WO 94/25585; WO 97/13852; WO 98/24884; WO 99/45962 and WO 01/14424, the contents of which are incorporated herein by reference in their entirety.
Fully human antibodies of the invention can also be raised using a mouse that carries human immunoglobulin sequences on transgenes and transchomosomes, such as a mouse that carries a human heavy chain transgene and a human light chain transchromosome. This mouse is referred to herein as a "KM mouse" and is described in detail in PCT Publication WO 02/43478. A modified form of this mouse, which further comprises a homozygous disruption of the endogenous FcyRIIB receptor gene, is also described in PCT Publication WO 02/43478 and referred to herein as a "KM/FCGR2D mouse®." In addition, mice with either the HCo7 or HCol2 heavy chain transgenes or both can be used.
Additional transgenic animal embodiments include the Xenomouse (Abgenix, Inc., U.S. Patent Nos. 5,939,598; 6,075, 181; 6,114,598; 6,150,584 and 6,162,963). Further embodiments include "TC mice" (Tomizuka et al. (2000) Proc. Natl. Acad. Set USA 97:722-727) and cows carrying human heavy and light chain transchromo somes (Kuroiwa et al. (2002) Nature Biotechnology 20:889-894; PCT Publication WO 02/092812).
A number of anti-CD3 antibodies are known, including but not limited to, OKT3 (muromonab/Orthoclone OKT3TM, Ortho Biotech, Raritan, NJ; U. S. Patent No. 4,361, 549); hOKT3Yl (teplizumab) (MGA031) (Herald et al, NEJM 346 (22): 1692-1698 (2002); TRX4 (otelixizumab); HuM291 (Nuvion™, Protein Design Labs, Fremont, CA); gOKT3-5 (Alegre et al. , J. Immunol. 148 (11): 3461-8 (1992); 1F4 (Tanaka et al, J. Immunol. 142: 2791-2795 (1989) ) ; G4.18 (Nicolls et al, Transplantation 55: 459-468 (1993)) ; 145-2C11 (Davignon et al, J. Immunol. 141 (6): 1848-54 (1988)); and as described in Frenken et al, Transplantation 51 (4): 881-7 (1991); U. S. Patent Nos. 6,491, 9116, 6,406, 696, and 6,143, 297).
In a particular embodiment, the anti-CD3 antibody is a non-mitogenic anti-CD3 antibody or a fragment thereof. Typically, the non-mitogenic anti-CD3 antibody is a non- mitogenic anti-CD3 monoclonal antibody. Thus, the present invention relates to a non- mitogenic anti-CD3 antibody for use in a method for treating T-cell acute lymphoblastic leukemia (T-ALL). In some embodiment, the non-mitogenic anti-CD3 antibody is used in a method for inducing cell death of T-ALL cells, in particular TCR-expressing T-ALL cells or blasts (also referred herein as CD3 -positive primary T-ALL cells).
As used herein, the term "non-mitogenic" has its general meaning in the art and has properties to bind to the CD3 antigen but without inducing cytokine production, as described in US7041289 Bl . Thus, a non-mitogenic anti-CD3 antibody delivers for instance a partial T cell signal that renders activated T cells unresponsive.
In one embodiment, the non-mitogenic anti-CD3 antibody is TRX4 (otelixizumab) or MGA031 (teplizumab).
Alternatively, the present invention relates to an anti-TCR antibody for use in a method for treating T-cell acute lymphoblastic leukemia (T-ALL). In some embodiment, the anti-TCR antibody is used in a method for inducing cell death of T-ALL cells, in particular TCR- expressing T-ALL cells or blasts (also referred herein as CD3-positive primary T-ALL cells).
In one embodiment, the antibody of the invention recognizes TCR a chains, and, thus the α/β TCR, generally (i.e., is a pan-specific TCR antibody, and, in particular, a pan-specific TCR a chain antibody). In another embodiment, the antibody of the invention recognizes TCR β chains, and, thus the α/β TCR, generally (i.e., is a pan-specific TCR antibody, and, in particular, a pan- specific TCR β chain antibody).
A number of anti-TCR antibodies, in particular anti-c$-TCR antibodies, are known, including but not limited to those described in the international patent applications n° WO2013037484 and WO2015066379 which disclose humanized anti-c^TCR antibodies, derived from the murine monoclonal antibody BMA031 (see European patent application EP0403156) such as GL1BM VH31, GL1BM VH28, HEBE1 H66, and HEBE1 H71.
As used herein, the term "treatment" or "treat" refer 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. By "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. The phrase "maintenance regimen" or "maintenance period" 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.]).
As used herein, the term "T-cell acute lymphoblastic leukemia" (T-ALL) refers a malignancy accounting for about 15% and 25% of ALL in pediatric and adult cohorts respectively. T-ALLs are a heterogeneous group of diseases with regard to immunophenotype, cytogenetics, molecular genetic abnormalities and clinical features, including response to therapy as described in Graux et al. 2006. Several immunophenotypic classifications have been proposed. Among these, the classification proposed by the European Group for the Immunological Characterization of Leukemias (EGIL) is commonly used in Europe. According to EGIL, the presence of cytoplasmic or membrane expression of CD3 defines T-ALL. Four subgroups are proposed: (TI) the immature subgroup or pro-T-ALL is defined by the expression of only CD7; (Til) pre-T-ALL also expresses CD2 and/or CD5 and/or CD8; (Till) or cortical T-ALL shows CD la positivity; (TIV) finally, mature T-ALL is characterized by the presence of surface CD3 and CD la negativity. T-ALL present genetic abnormalities (involving the T- cell receptor (TCR), involving known oncogenes such as Notchl, HOXA cluster, TLXl(HOXl l) and TLX3 (HOX11L2), TALI and LYL1, involving tyrosine kinases such as ABL1, JAK3 and JAKl).
In one embodiment, the patients are refractory to other treatments.
The term "refractory" represents previously treated patients which were either non responsive to treatment with the agent or had a response to treatment and then relapsed.
Today, sequential polychemotherapy are used for treating T-ALL according to different protocols. Typically, in adult T-ALL patients said sequential polychemotherapy comprises sequentially administering to said patients corticosteroids (e.g. prednisone) and L-asparaginase (GRAALL regimen).
Methods for predicting the responsiveness to a treatment with a TCR-activating agent In a another aspect, the invention relates to an in vitro method for predicting the responsiveness of a patient affected with a T-ALL to a treatment with a TCR-activating agent.
The invention relates to an in vitro method for predicting the responsiveness of a patient affected with a T-ALL to a treatment with a TCR-activating agent comprising a step of determining the expression of TCR/CD3 complex at the surface of T-ALL cells obtained from a patient, wherein the absence of the TCR/CD3 complex is indicative of non-responsiveness to said treatment.
As used herein, the term "predicting" refers to a probability or likelihood for a patient to respond to the treatment with a TCR-activating agent. As used herein, the term "responsiveness" refers to ability to assess the likelihood that treatment will or will not be clinically effective.
As used herein the term "patient" denotes a mammal, typically a human. In a preferred embodiment, a patient refers to any subject (typically human) afflicted with T-ALL.
As used herein, the term "TCR-activating agent" is intended to encompass any refers to a natural or synthetic compound that has a biological effect which stimulate TCR/CD3 complex intracellular signaling activity. In the context of the invention, the TCR activating agent stimulates TCR/CD3 complex presents on T-ALL cells. Typically, stimulation of the TCR/CD3 complex is associated with a series of biochemical events initiated by TCR engagement (e.g., by antigen-MHC complex or anti-CD3 cross-linking) and that typically triggers (or contributes to triggering) one or more pathways (by, for example, activating transcription of certain genes such as, e.g., BCL2L1-BIM, GIMAP4, LiPA4 or Nur77 also referred as NR4A1).
Within the context of the invention, the TCR-activating agents include but are not limited to antibodies, synthetic or native sequence peptides and small molecules which bind to the TCR/CD3 complex.
In some embodiments, the TCR-activating agent is an anti-CD3 antibody as above- described. In a particular embodiment, the anti-CD3 antibody is a non-mitogenic anti-CD3 antibody such as for instance TRX4 (otelixizumab) or MGA031 (teplizumab). In some embodiments, the TCR-activating agent is an anti-TCR antibody as above- described. In a particular embodiment, the anti-TCR antibody is an anti-aP-TCR antibody such as for instance GL1BM VH31, GL1BM VH28, HEBE1 H66, and HEBE1 H71.
In some embodiments, the TCR-activating agent is an epitope obtained from a T-ALL cells of the patient, more particularly TCR idiotypes which may function as antigen and activate T cells.
In some embodiments, the TCR-activating agent is an inhibitor of TCR regulating tyrosine phosphatases, e.g. CD45, SHPl, SHP2 and PTPN22.
As used herein, the term "inhibitor of TCR regulating tyrosine phosphatases " refers to any compound, natural or synthetic, which results in a decreased phosphorylation of the tyrosine present on the intracellular domain of TCR. A number of TCR regulating tyrosine phosphatases inhibitors such as inhibitors of CD45 and/or SHPl are described in the international patent applications n° W09946268 and W09946236. In a particular embodiment, the inhibitor of TCR regulating tyrosine phosphatases is an inhibitor of CD45, SHPl, SHP2 or PTPN22 gene expression. As used herein, the term "inhibitor of gene expression" refers to a natural or synthetic compound that has a biological effect to inhibit the expression of a gene is meant a CD45, SHP1, SHP2 or PTPN22 mRNA, protein, peptide, or polypeptide.
Inhibitors of gene expression for use in the 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 targeted mRNA by binding thereto and thus preventing protein translation or increasing mRNA degradation, thus decreasing the level of the targeted protein (i.e. CD45), and thus activity, in a cell. For example, antisense oligonucleotides of at least about 15 bases and complementary to unique regions of the mRNA transcript sequence encoding the target protein can be synthesized, e.g., by conventional phosphodiester techniques and administered by e.g., intravenous injection or infusion. Methods for using antisense techniques for specifically inhibiting gene expression of genes whose sequence is known are well known in the art (e.g. see U.S. Pat. Nos. 6,566,135; 6,566,131; 6,365,354; 6,410,323; 6,107,091; 6,046,321; and 5,981,732). Small inhibitory RNAs (siRNAs) can also function as inhibitors of gene expression for use in the present invention. 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 gene expression is specifically inhibited (i.e. RNA interference or RNAi). 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 Tuschi, T. et al. (1999); Elbashir, S. M. et al. (2001); Hannon, GJ. (2002); McManus, MT. et al. (2002); Brummelkamp, TR. et al. (2002); U.S. Pat. Nos. 6,573,099 and 6,506,559; and International Patent Publication Nos. WO 01/36646, WO 99/32619, and WO 01/68836). Short hairpin RNAs (siRNAs) can also function as inhibitors of gene expression for use in the invention.
As used herein, "determining" encompasses detecting or quantifying. Indeed, an expression level can be qualitative or quantitative with or without reference to a control or a predetermined value. Thus, a determination of whether a polypeptide is present or absent (e.g., detectable or undetectable) constitutes determining its expression level in various embodiments while in other embodiments, a quantitative level is determined. As used herein, "detecting" means determining if TCR/CD3 complex is present or not at the surface of T-ALL cells obtained from a patient and "quantifying" means determining the amount of TCR/CD3 complex present at the surface of T-ALL cells. In the context of the invention, the terms "TCR/CD3", "CD3/TCR", "TCR/CD3 complex" or "CD3/TCR complex" have the same meaning and refer to the T cell receptor-CD3 complex (TCR-CD3) which is responsible for recognizing antigens bound to major histocompatibility complex (MHC) molecules.
In one embodiment, the method of the invention comprises a step of determining the expression TCR/CD3 complex at the surface of T-ALL cells obtained from a patient, wherein the absence of TCR/CD3 complex is indicative of non-responsiveness to said treatment. Methods for determining the expression level of the biomarkers of the invention:
Determination of the expression level of TCR/CD3 complex at the surface of T-ALL cells may be performed by a variety of techniques. Generally, the expression level as determined is a relative expression level. For example, the determination comprises contacting the biological sample with selective reagents ligands, and thereby detecting the presence, or measuring the amount, of polypeptides of interest originally in said biological sample. Contacting may be performed in any suitable device, such as a plate, microtiter dish, test tube, well, glass, column, and so forth. In specific embodiments, the contacting is performed on a substrate coated with the reagent, such as a specific ligand array. The substrate may be a solid or semi-solid substrate such as any suitable support comprising glass, plastic, nylon, paper, metal, polymers and the like. The substrate may be of various forms and sizes, such as a slide, a membrane, a bead, a column, a gel, etc. The contacting may be made under any condition suitable for a detectable complex, such as an antibody-antigen complex, to be formed between the reagent and the polypeptides of the biological sample. Such methods comprise contacting the biological sample of a patient containing T-ALL cells with a binding partner capable of selectively interacting with the protein present in said sample. The binding partner is generally an antibody that may be polyclonal or monoclonal, preferably monoclonal. The binding partners of the invention such as antibodies, may be labelled with a detectable molecule or substance, such as a fluorescent molecule, a radioactive molecule or any others labels known in the art. Labels are known in the art that generally provide (either directly or indirectly) a signal. As used herein, the term "labelled", with regard to the antibody or aptamer, is intended to encompass direct labeling of the antibody by coupling (i.e., physically linking) a detectable substance, such as a radioactive agent or a fluorophore (e.g. fluorescein isothiocyanate (FITC) or phycoerythrin (PE) or lndocyanine (Cy5)) to the antibody, as well as indirect labelling of the antibody by reactivity with a detectable substance. The aforementioned assays generally involve the coating of the binding partner (ie. antibody) in a solid support. Solid supports which can be used in the practice of the invention include substrates such as nitrocellulose (e. g., in membrane or microtiter well form); polyvinylchloride (e. g., sheets or microtiter wells); polystyrene latex (e.g., beads or microtiter plates); polyvinylidine fluoride; diazotized paper; nylon membranes; activated beads, magnetically responsive beads, and the like.
In one embodiment, the step of determining the expression of TCR/CD3 complex at the surface of T-ALL cells is performed by immunoassays such as for instance immunophenotyping as described in Asnafi et al., 2004 or by immunohistochemistry (IHC).
The step of determining the expression of a target protein such as TCR/CD3 complex by a cell population such as T-ALL cells may be carried out by a variety of methods for detecting a particular immune cell population available for a skilled artisan, including immunoselection techniques, such as high-throughput cell sorting using flow cytometric methods, affinity methods with antibodies labeled to magnetic beads, biodegradable beads, non-biodegradable beads, and combination of such methods.
As used herein, the term "flow cytometric methods" refers to a technique for counting cells of interest, by suspending them in a stream of fluid and passing them through an electronic detection apparatus. Flow cytometric methods allow simultaneous multiparametric analysis of the physical and/or chemical parameters of up to thousands of particles per second, such as fluorescent parameters. Modern flow cytometric instruments usually have multiple lasers and fluorescence detectors. A common variation of flow cytometric techniques is to physically sort particles based on their properties, so as to purify or detect populations of interest, using "fluorescence-activated cell sorting". As used herein, "fluorescence-activated cell sorting" (FACS) refers to a flow cytometric method for sorting a heterogeneous mixture of cells from a biological sample into two or more containers, one cell at a time, based upon the specific light scattering and fluorescent characteristics of each cell and provides fast, objective and quantitative recording of fluorescent signals from individual cells as well as physical separation of cells of particular interest.
For instance, T-ALL cells may be stained with a FITC-labeled anti-CD38 mAb (such as the clone 145-2C11 purchased by Pharmingen).
Using such methods, cells can be separated and detected positively or negatively with respect to the particular cell-surface markers such as for instance CD3. The invention relates to an in vitro method for predicting the responsiveness of a patient affected with a T-ALL to a treatment with a TCR-activating agent comprising a step of determining the presence of TCR- expressing T-ALL cells (also referred as CD3-positive T-ALL cells) in a biological sample obtained from a patient, wherein the absence of the TCR-expressing T-ALL cells (or CD3- positive T-ALL cells) is indicative of non-responsiveness to said treatment. Alternatively, IHC specifically provides a method of determining the expression of a target protein in a biological sample or tissue specimen in situ. The overall cellular integrity of the sample is maintained in IHC, thus allowing detection of both the presence and location of the target of interest. Typically a biological sample is fixed with formalin, embedded in paraffin and cut into sections for staining and subsequent inspection by light microscopy. Current methods of IHC use either direct labelling or secondary antibody-based or hapten-based labelling. Examples of known IHC systems include, for example, EnVision (DakoCytomation), Powervision® (Immunovision, Springdale, AZ), the NBA™ kit (Zymed Laboratories Inc., South San Francisco, CA), HistoFine® (Nichirei Corp, Tokyo, Japan). In a particular embodiment, the method further comprises a step of analysing TCR clustering by performing flow cytometry analysis.
In another aspect, the invention relates to a TCR-activating agent for use in a method for treating T-ALL in a patient classified as responder according to the method above-described.
In one embodiment, the TCR-activating agent is a non-mitogenic anti-CD3 antibody. The invention also relates to a method for treating T-ALL in a patient in need thereof and identified as responder according to a method of the invention comprising a step of administrating to said patient a therapeutically amount of a TCR-activating agent. As used herein, the term "responder" refers to a patient who may respond to a treatment with a TCR-activating agent such as an anti-CD3 antibody, i.e. least one of his symptoms is expected to be alleviated, or the development of the disease is stopped, or slowed down.
Pharmaceutical compositions
The invention also relates to pharmaceutical compositions comprising a TCR-activating agent for use in a method for treating T-cell acute lymphoblastic leukemia (T-ALL).
The invention also relates to pharmaceutical compositions comprising an anti-CD3 antibody for use in a method for treating T-cell acute lymphoblastic leukemia (T-ALL).
In a particular embodiment, said anti-CD3 antibody is a non-mitogenic anti-CD3 antibody such as for instance TRX4 (otelixizumab) or MGA031 (teplizumab).
The invention also relates to pharmaceutical compositions comprising an anti-TCR antibody for use in a method for treating T-cell acute lymphoblastic leukemia (T-ALL).
In a particular embodiment, the anti-TCR antibody is an anti-aP-TCR antibody such as for instance GL1BM VH31, GL1BM VH28, HEBE1 H66, and HEBE1 H71.
Therefore, an antibody of the invention may be combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form therapeutic compositions. "Pharmaceutically" or "pharmaceutically acceptable" refers 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 form of the pharmaceutical compositions, the route of administration, the dosage and the regimen naturally depend upon the condition to be treated, the severity of the illness, the age, weight, and sex of the patient, etc. The pharmaceutical compositions of the invention can be formulated for a topical, oral, parenteral, intranasal, intravenous, intramuscular, subcutaneous or intraocular administration and the like.
Preferably, the pharmaceutical compositions contain 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 doses used for the administration can be adapted as a function of various parameters, and in particular as a function of the mode of administration used, of the relevant pathology, or alternatively of the desired duration of treatment. To prepare pharmaceutical compositions, an effective amount of the antibody may be dissolved or dispersed in a pharmaceutically acceptable carrier or aqueous medium. 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. 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.
Dosages to be administered depend on individual needs, on the desired effect and the chosen route of administration. It is understood that the dosage administered will be dependent upon the age, sex, health, and weight of the recipient, concurrent treatment, if any, frequency of treatment, and the nature of the effect desired. The total dose required for each treatment may be administered by multiple doses or in a single dose.
The doses used for the administration can be adapted as a function of various parameters, and in particular as a function of the mode of administration used, of the relevant pathology, or alternatively of the desired duration of treatment. For example, it is well within the skill of the art to start doses of the antibodies at levels lower than those required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved. However, the daily dosage of the antibodies may be varied over a wide range from 0.01 to 1,000 mg per adult per day. Preferably, 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 active ingredient for the symptomatic adjustment of the dosage to the subject to be treated. A medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, preferably from 1 mg to about 100 mg of the active ingredient. 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 10 mg/kg of body weight per day.
The pharmaceutical composition of the invention may further comprise an additional drug, in particular a chemotherapeutic drug useful in T-ALL.
A number of chemotherapeutic drugs useful in T-ALL are known, including but not limited to, corticosteroids (e.g. prednisone), L-asparaginase, non-selective inhibitors of NOTCH 1, ABLl inhibitors and JAK3 inhibitors and PBK-AKT-mTOR pathway inhibitors as described in Roti et al, 2014.
Non-selective inhibitors of NOTCH1 include soluble or cell-associated Notch decoys, [gamma] -secretase inhibitors, intracellular MAML1 decoys, and Ras signaling inhibitors. [gamma]-Secretase inhibitors (GSI) have the most immediate therapeutic potential. Blocking specific E3 ligases responsible for ubiquitination of Notch ligands or mono-ubiquitination of Notch receptors can also be used. Still other NOTCH 1 inhibitors that can be used are described in WO 2003/013527 and paragraphs 0016-0019 of US 2006/205666.
Non-limiting examples of ABLl inhibitors include imatinib (STI571) (Novartis), nilotinib AMN107 (Novartis), dasatinib (BMS-354825) (BMS), ponatinib (AP24534), bosutinib (SKI-606) (Pfizer) and bafetinib. Exemplary ABLl inhibitors include but are not limited to, those as described in the following patent applications: N-phenyl-2-pyrimidine- amine derivatives (EP0564409), pyrimidinylaminobenzamide derivatives (WO2004/005281), cyclic compounds (WO00/62778), bicyclic heteroaryl compounds (WO2007/075869), substituted 3-cyano quinoline derivatives (US6002008), 4-anilo-3-quinolinecarbonitrile derivatives (WO2005/04669) and amide derivatives (US7728131 and WO2005/063709).
Non- limiting examples of PI3K inhibitors include: NVP-BEZ235 (BEZ235) (Novartis); LY294002 (Cell Signaling #9901); GDC-0941 (Genentech/Roche); GDC-0980 (Genentech); PI- 103 (Piramed); XL147 (Exilixis/Sanofi-Aventis); XL418 (Exilixis); XL665 (Exelixis); LY29002 (Eli Lilly); ZSTK474 (Zenyaku Kogyo); BGT226 (Novartis); wortmannin; quercetin; tetrodotoxin citrate (Wex Pharmaceuticals); thioperamide maleate; IC87114; PIK93; TGX-115; deguelin; NU 7026; OSU03012; tandutinib (Millennium Pharmaceuticals); MK-2206 (Merck); OSU-03012; triciribine (M.D. Anderson Cancer Center); PIK75; TGX-221; NU 7441 ; PI 828; WHI-P 154; AS-604850; AS-041164 (Merck Serono); AS-252424; AS-605240; AS-604850; compound 15e;17-P-hydroxywortmannin; PP121; WAY-266176; WAY-266175; BKM120 (Novartis); PKI-587 (Pfizer); BYL719 (Novartis) ; XL765 (Sanofi-Aventis); GSK1059615 or GSK615 (Glaxo SmithKline); IC486068; SF1126 (Semafore Pharmaceuticals); CAL-101 (Gilead Sciences); LME00084; PX-478 (Oncothyreon); PX-866 (Oncothyreon); PX-867 (Oncothyreon), BAY 80-6946 (Bayer), GSK2126458 (GlaxoSmithKline), INK1117 (Intellikine), IPI-145 (Infinity Pharmaceuticals) Palomid 529 (Paloma Pharmaceuticals); ZSTK474 (Zenyaku Kogyo); PWT33597 (Pathway Therapeutics); TG100-115 (TargeGen); CAL263 (Gilead Sciences); SAR245408 (Sanofi-Aventis); SAR245409 (Sanofi-Aventis); GNE-477; CUDC-907; and BMK120 (Novartis). Exemplary PI3K inhibitors include but are not limited to those as described in the following patent applications: WO2008/027584, WO2008070150, 2,3-dihydroimidazo[l,2-c]quinazolines (WO2008/125833), 2-morpholin-4- yl-pyrimidines (WO2008/125835), pyrimidines (WO2008/125839), bicyclic heteroaryls (WO2009/010530), thiazolidinones (WO2009/026345), pyrrolothiazoles (WO2009/071888), tricyclic thiazole and thiophene derivatives (WO2009/071890), fused bicyclic thiazole and thiophene derivatives (WO2009/071895) and oxazole substituted indazoles (WO2010/125082).
The invention also relates to a method for treating T-cell acute lymphoblastic leukemia (T-ALL) in a patient in need thereof, comprising a step of administering to said patient a therapeutically effective amount of an anti-CD3 antibody or an anti TCR-antibody.
By "therapeutically effective amount" is meant an amount sufficient to achieve a concentration of antibodies which is capable of preventing, treating or slowing down the disease to be treated. Such concentrations can be routinely determined by those of skilled in the art. The amount of the polypeptide actually administered will typically be determined by a physician or a veterinarian, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the patient, the severity of the subject's symptoms, and the like. It will also be appreciated by those of skilled in the art that the dosage may be dependent on the stability of the administered polypeptide. In one embodiment, the treatment with an anti-CD3 antibody (such as OKT3, TRX4 or MGA031 monoclonal antibodies) is administered in more than one cycle, i.e. the administration of an anti-CD3 antibody (such as an OKT3, TRX4 or MGA031 monoclonal antibodies) is repeated at least once.
For example, 2 to 10 cycles or even more, depending on the specific patient status and response, may be administered. The intervals, i.e. the time between the start of two subsequent cycles, are typically several days.
Kit-of-parts compositions:
The anti-CD3 antibody such as the OKT3 or the anti-TCR antibody as above-described and the chemotherapeutic agent may be combined within one formulation and administered simultaneously. However, they may also be administered separately, using separate compositions. It is further noted that they may be administered at different times.
The invention relates to a kit-of-parts composition comprising an anti-CD3 antibody as above-defined and a chemotherapeutic agent.
The invention relates to a kit-of-parts composition comprising an anti-TCR antibody as above-defined and a chemotherapeutic agent.
The invention relates to a kit-of-parts composition comprising an anti-CD3 antibody as above-defined and a chemotherapeutic agent for use in a method for treating T-ALL.
The invention relates to a kit-of-parts composition comprising an anti-TCR antibody as above-defined and a chemotherapeutic agent for use in a method for treating T-ALL.
The terms "kit", "product" or "combined preparation", as used herein, define especially a "kit-of-parts" in the sense that the combination partners as defined above can be dosed independently or by use of different fixed combinations with distinguished amounts of the combination partners, i.e. simultaneously or at different time points. The parts of the kit of parts can then, e.g., be administered simultaneously or chronologically staggered, that is at different time points and with equal or different time intervals for any part of the kit of parts. The ratio of the total amounts of the combination partners to be administered in the combined preparation can be varied. The combination partners can be administered by the same route or by different routes. When the administration is sequential, the first partner may be for instance administered 1, 2, 3, 4, 5, 6, 7, days before the second partner. The invention will be further illustrated by the following figures and examples.
However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention.
FIGURES:
Figure 1: In vivo TCR stimulation with an agonistic monoclonal antibody inhibits T-cell leukemogenesis in the mouse TEL-JAK2 model. A, Percentage of leukemic cells within peripheral blood leukocytes of female mice treated with 5 daily doses of 20 μg 145- 2C11 mAb or control hamster IgG (arrows), starting 1 day after female TJM leukemic cell inoculation. B, Kaplan-Meier leukemia- free survival curves of mice transplanted with TJM leukemic cells and treated with antibody before leukemia onset (arrows) (Log-rank (Mantel- Cox) test, P value < 0.01). C, Percentage of leukemic cells within peripheral blood leukocytes of mice treated with 5 daily doses of 20 μg 145-2C11 mAb or control hamster IgG, starting 2 days after TEL-JAK2 leukemic cell inoculation (arrows). D, Kaplan-Meier leukemia-free survival curves for mice transplanted with TEL-JAK2 leukemic cells and treated with antibody before leukemia onset (P value < 0.01). E, Percentage of leukemic cells within peripheral blood leukocytes of female TEL-JAK2:Marylin double transgenic mice (TJM) secondary leukemia mice treated with 5 daily doses of 20 μg 145-2C11 mAb or control hamster IgG (arrows), after leukemia detection. F, Kaplan-Meier leukemia- free survival curves for mice transplanted with TEL-JAK2 leukemic cells and treated with antibody after leukemia onset (arrows) (P value < 0.05). G, Percentage of leukemic cells within peripheral blood leukocytes of TEL- JAK2secondary leukemiaNude mice treated with 5 daily doses of 50 μg 145-2C11 mAb or control hamster IgG after leukemia detection and analyzed 5 days after end of treatment.*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001
Figure 2: In vivo TCR stimulation with an agonistic monoclonal antibody inhibits T-cell leukemogenesis in human T-ALL cells xenografted in NSG mice. (A) NSG mice were injected with 106 T-ALL cells. 24 hours later, mice were treated with either 40μg/mouse of the anti-CD3 OKT3 antibody (n=5), or an isotype control antibody (n=5) for 5 consecutive days. The same treatment schedule was applied 2 days later. Leukemia burden in peripheral blood was followed over time. (B) NSG mice were injected with 106 T-ALL cells. When leukemic burden reached 1-4 % of blood nucleated cells mice were treated with either the OKT3 (n=5) or control (n=5) antibody for 3 rounds of the 4(^g/mouse on a 5 days on /2days off schedule. Leukemia burden in peripheral blood was followed over time.
Figure 3: Anti-CD3 stimulation of ALL-SIL TCR-HY cells induces TCR signaling and cell death. (A) Percentage of annexin V-positive ALL-SIL or ALL-SIL TCR-HY cells either left unstimulated or stimulated by CD3/CD28 coated-beads for the indicated time. (B) Western Blot analysis of caspase3 and caspase7 cleavage in ALL-SIL and ALL-SIL TCR-HY cells either left unstimulatedor stimulated by CD3/CD28 coated-beads for 48 and 72 h. Actin was used as a loading control. (C) Growth curve of unstimulated and CD3/CD28-stimulated ALL-SIL and ALL-SIL TCR-HY cells. (D) Percentage of unstimulated or CD3/CD28- stimulated ALL-SIL TCR-HY cells in S-phase of the cell cycle as measured by EdU incorporation and 7-AAD staining of DNA. (E) Phospho-ERK, phospho-AKT and phospho- STAT3 expression detected by flow cytometry in non- stimulated (top line in each panel) or 30 min anti-CD3/CD28-stimulated (bottom line) ALL-SIL TCR-HY cells. (F) Percentage of annexin V-positive unstimulated (left) or CD3/CD28-stimulated (right) ALL-SIL TCR-HY cells at day 5, in the presence of indicated signaling pathway inhibitors.
Figure 4: Anti-CD3 induces human primary T-ALL cell death in vitro and mimics thymic negative selection. Comparison of cell death induction in CD3-positive primary, diagnostic T-ALL cells (9 cases) versus CD3 -negative primary T-ALL cells (4 cases) after 3 days of co-culture on OP9-DL1 in the presence or absence of CD3/CD28 stimulation.
EXAMPLE:
Material & Methods
Mice: The EμSRα-7ΐX-J4AT2 transgenic mice (line 71) (Carron et al, 2000), Marylin transgenic mice (Lantz et al, 2000), and Rag2 knock-out, all on the C57BL/6 background, and Swiss Nude were maintained under specific-pathogen- free conditions in the animal facilities of the Institut Curie (Orsay, France) and University of Algarve (Faro, Portugal). All experimental procedures were performed in strict accordance with the recommendations of the European Commission (Directive 86/609/EEC), French National Committee (87/848) and Portuguese authorities (Decreto-Lei n°129/92) for the care and use of laboratory animals. Mice were euthanized by CO2 inhalation when terminally ill, due to either severe dyspnea caused by massive expansion of leukemic cells in the thymus, or weakness caused by leukemic dissemination to vital organs such as bone marrow, lung, and liver. Blood was collected from the submandibular vein. For leukemia transplantation assays, 0.5-2 x 106 leukemic T cells collected from diseased female TEL-JAK2: Marylin double transgenic mice (TJM) mice were intravenously injected in the tail vein of recipient mice of the indicated gender and strain and regularly monitored for external signs of disease development or through blood analysis. Anti- CD3s (145-2C11) monoclonal antibody or control ChromPureSyrian Hamster IgG (Jackson Immunoresearch) was diluted in sterile PBS and intravenously administered on a regimen of 5 daily doses of 20 μg for C57BL6 mice or 50 μg for SwissNudemice). Statistical analyses and survival curves were calculated using Prism 5 (GraphPad).
NOD/SCID/yc-/- (NSG) mice were purchased from Charles River Laboratories and maintained under constant antibiotic treatment (Baytril 0.01% in drinking water). 2 months-old mice were intravenously injected with 106 fresh leukemic cells obtained from a primary NSG mice (n= 20) engrafted with patient material from T-ALL. The mice were injected intravenously with either the anti-CD3 OKT3 MAb (BioXCell; 2 rounds of treatment with 40 μg/mice/day for 5 consecutive days separated by a 2 days interval) or the iso type-matched (IgG2a) CI .18.4 MAb, both diluted in PBS. In the curative setting, mice were treated when leukemic cells reached 1-4% of blood nucleated cells. Mice were monitored weekly by flow cytometry for leukemic load in peripheral blood, T-ALL cells being identified as FSChl, hCD7+, hCD45+ cells.
T-ALL patient samples and cell lines. Immunophenotypic and oncogenic features of T-ALL patients were identified as described (6, 47, 48). Fresh or thawed primary T-ALL samples (peripheral blood or bone marrow) obtained at diagnosis from adult and pediatric patients were used, as well as T-ALL cells xenografted in NSG mice. Informed consent was obtained according to the Declaration of Helsinki. All samples used contained >80% blasts. CD4+CD8+ (DP) human thymocytes were obtained and processed as described in Supplementary methods. The ALL-SIL cell line (DSMZ, Braunschweig, Germany, ACC511) were grown in vitro and transduced as described in Supplementary methods.
Flow cytometry: Single cell suspensions prepared from lymphoid organs, bone marrow flushed from tibias or blood were stained with fluorochrome-labeled antibodies, and detected on a FACSCalibur cytometer (BD Biosciences), as previously described (Carron et al, 2000). FITC-, PE-, PE-Cy5-, PerCP/Cy5.5- or APC-conjugated antibodies specific for CD4 (H129.19), CD8aD(53-6.7), CD3s (145-2C11), TCRp (H57-597), V 6 TCR (RR4-7), (GL3), CD25 (7D4), CD45.1 (A20), and CD45.2 (104) (BD Biosciences) were used. Antibodies used to identify human T-ALL cells were FITC-conjugated anti- human CD7 cl 4H9 and APC- conjugated anti-human CD45 (eBiosciences). Data analysis was performed using CellQuest (BD Biosciences) software.
In vitro TCR stimulation by antigen presenting cells. Splenocytes were obtained from C57BL/6 female mice, subsequently irradiated (2000 rad) and preincubated with antigenic peptides at 37°C for 1 h (peptide pulsing). Peptides used were DBY, the male antigen epitope specifically recognized by Marilyn transgenic TCR-HY, at 10, 1, 0.1, 0.01, 0.001 and 0.0001 μΜ and OVA, a non-specific peptide, at 10μΜ. Splenocytes were then plated out in 48-well plates, without removal of peptides. ALL-SIL TCR-HY cells were added to the co-culture system at a 1 : 100 ratio. Control conditions were performed in parallel with non-pulsed splenocytes, DBY without splenocytes and ALL-SIL without transgenic TCR co-cultured on DBY-pulsed splenocytes. Cell death was analyzed by flow cytometry at day 3 of culture, as described in Supplementary methods. Antibodies: Antibodies used for human cells: CD3, TCRVb6, Antibodies against phosphorylated ERK1/2, phosphorylated p38, phosphorylatedS6, were from BD, phosphorylated AKT from Cell Signaling.
Anti CD3/CD28 cell stimulation. Short-term stimulation was performed on cells cultured in serum- free medium for at least 15min at 37°C prior to stimulation. Murine anti- hCD3 mAb (OKT3, Bio legend) (20μg/ml) was added to the cell culture on ice for 10 min and mAbs were then cross-linked by addition of goat-anti-mouse antibody to a final concentration of 50μg/ml for 15 min. Stimulation (0-10 min) was triggered by warming cells at 37°C and terminated either by cooling cells in cold PBS or fixing cells for Phosphokinase array and intracellular phosphoprotein (Phosflow) assays, respectively. For prolonged stimulation, a bead-based assay was used in which cell lines were cultured in 96- or 24-well-plates and exposed to 4.5 μιη diameter superparamagnetic beads covalently coupled to anti-CD3 and anti- CD28 antibodies (Dynabeads® Human T-Activator CD3/CD28, Invitrogen) (duration of stimulation from 1 to 12 days). Prolonged stimulation of primary T-ALL cells was performed in a co-culture assay with confluent OP9-DL1 in a a-MEM media supplemented with 20% FBS (Hyclone; ThermoFisherScientific), 50 μg/ml streptomycin and 50 IU penicillin and recombinant human cytokines hFLT3-L (5 ng/mL), hIL-7 (2 ng/mL) and hSCF (10 ng/mL) (Miltenyi). Similarly, CD3/CD28 coated-beads were added to the co-culture assay following the manufacturer's instructions.
Cell stimulation and intracellular flow cytometry with phosphorylation specific antibodies: Before being stimulated, cells were deprived of serum for at least 15min at 37°C. Cells were stimulated in serum- free RPMI medium at 37°C. For bead-based stimulation, 4.5 μιη diameter, superparamagnetic beads covalently coupled to anti-CD3 and anti-CD28 antibodies (Dynabeads® Human T-Activator CD3/CD28, Invitrogen) were used. The stimulation was terminated by addition of prewarmed formaldehyde (Cytofix Buffer, BD Biosciences) at 37°C for lOmin. After stimulation, cells were placed on ice for 1 minute and pelleted at 4°C and resuspended in 1 ml of ice-old methanol (Perm Buffer III, BD Biosciences). Cells were incubated on ice for 30 minutes, then washed 3 times with 3ml of PBS and resuspended in 50μ1 for 20 minutes intracellular staining at room temperature and resuspended in PBS for flow cytometric analysis on Canto II (BD). Data were analysed using DIVA software (BD).
Cell death analysis: Cells were incubated with CD3/CD28 beads according to the manufacturer's instruction, in a 96-well plate. A different time of culture, cell death was analysed by flow cytometry using AnnexinV-APC and propidium iodide (BD Biosciences) Western Blot: Cells were stimulated as previously describe with OKT3 or beads.
Proteins were separated on tris-glycine gels under reducing conditions and transferred to nitrocellulose membranes (Biorad). Immunob lotting was performed with following antibodies: phosphor-ERKl/2 (ref, cell Signaling), ERK1/2 (cell Signaling), NFAT1 (cell Signaling), Caspase3 (BD), Caspase7 (BD). Flow cytometry analysis of TCR clustering. ALL-SIL TCR-HY cells were stained with TCR V 6-PE mAb (BD Biosciences) for 30 min at 4°C and DAPI for 5 min at room temperature prior stimulation and split into two samples; one sample remained unstimulated while the other one was stimulated by styrene beads (Polyscience) pre-coated with human anti- CD3s antibody (OKT3, Biolegend) for 10 min at 37°C. Cells were fixed with 1% formaldehyde. Staining was analyzed usinglmageStream X mkll (Amnis Merck-Millipore). Data analysis was performed using the IDEAS image analysis software (Amnis).
Phosphokinase antibody array analysis and pharmacological inhibition. ALL-SIL,
ALL-SIL TCR-HY cells and human DP thymocytes were either left unstimulated or were stimulated with CD3/CD28 mAbs for 5 min. Each stimulated condition was performed in replicate. Cell lysates and Proteome Profiler Human phosphokinase array (R&D Systems) were performed according to the manufacturer's protocol. Chemiluminescence was detected by ChemiDoc XRS+ (Bio-Rad). The average signal (pixel density) of duplicate spots representing each phosphorylated kinase protein was normalized with the average signal of reference positive duplicate spots of each membrane using Image Lab software (Bio-Rad). Signal ratios for selected phosphoproteins were displayed in a heat-map using Treeview software. Cyclosporine A (Novartis) and PD184352 (Selleckchem) were used at^M. Effects were determined by analysis of cell death (Annexin V/PI assay) and expression of CD25 and CD69 activation markers.
Microarray gene expression profiling. RNA extraction was performed for the following cells: ALL-SIL-TCRa -GFP co-cultured on OP9-DL1, ALL-SIL-TCRa -GFP without co-culture, ALL-SIL transduced with TLX shRNA (sh-TLX), and ALL-SIL transduced with sh-control vectors. These samples were obtained as previously described (14) in duplicate, and RNA extraction was performed at an early time-point before cell death onset (48 h of co- culture for ALL-SIL-TCRa -GFP and 48h after puromycin selection for TLX shRNA/sh- control ALL-SIL). RNA hybridization was performed on Affymetrix U133 plus 2.0 microarrays. The statistical data analysis was performed with R version2.9.0 using the "Affy" package from Bioconductor. The probe intensities were log2 transformed and normalized using RMA. Identification of differentially expressed genes was performed by Significance Analysis of Microarrays (SAM), using 500 permutations and a false discovery rate threshold of 5%. Functional analysis of differentially expressed genes was performed using the Ingenuity Pathway Analysis (IP A; www.ingenuity.com) and the Database for Annotation, Visualization, and Integrated Discovery (DAVID; http://david.abcc.ncifcrf.gov)softwares. Heat map construction, used GENE-E software (http://www.broadinstitute.org/cancer/software/GENE- E/).Gene Set Enrichment Analysis (GSEA) was performed using the negative selection signature described by Baldwin et al, 2007(19) as gene set. GSEA was run using signal-to-noise for the ranking gene metric and 1000 permutations. All microarray data have been submitted to the Gene Expression Omnibus database (http://www.ncbi.nlm.nih.gov/geo) under accession numbers: GSE65496.
Luciferase xenograft studies. lxlO6 GFP-Luciferase sorted ALL-SIL TCR-HY cells were intravenously injected into 2 month-old NSG mice (n=14). Isofluorane-anesthesized mice were intraperitoneally injected with 2.5 mg of D-luciferin (CALIPER Life Sciences) and monitored on a weekly basis to detect luciferase activity using an IVIS Spectrum (Perkin- Elmer). When leukemic cells were detected (21 days post-injection), mice were treated with either OKT3 or control mAb as described above.
Results
Stimulation of TCR/CD3 induced cell death in T-ALL
Here we sought to gain insights into the TCR induced cell death in T-ALL. We first took advantage from our previously reported SIL-ALL-GFP-TCRa (TLX+ T-ALL cell line transducer by a tristronic GFP-TCRa-TCR vector) which undergo massive cell death when co-cultured on OP9-DLlto test whether Notchl pathway activation, mutated in more than 70% of T-ALL could be implicated. When these cells were co-cultured on OP9 without DL1, the same cell death occurred excluding a critical role for Notch pathway. In addition co-culture by transwell do not lead to cell cell death stressing a cell-cell contact need. In order to test whether cell death by TCRa-β expression could be similar to specific oncogene deprivation we compared transcriptomic data from SIL-ALL TLX 1 Knock-downed by Sh-RNA transfection (which leads also to massive cell death) to those obtained by SIL-ALL-GFP-TCRa cocultured on OP9-DL1. Importantly this demonstrated that two distinct apoptotic pathways drive cell dead suggesting that TCR-induced cell death is independent from underlying oncogene (TLX) deregulation. Transgenic Marylin mice express in thymocytes an MHC class II -restricted TCR (ναΐ.ΐνββ) specific for the H-Y male antigen (DBY), which in males induces thymocyte negative selection and maturation arrest (Lantz et al, 2000). In vitro data using co-cultured ALL-SIL-TCRH-Y on splenocytes pulsed by various DBY quantities demonstrated that this TCR-induced cell death could be specifically related to the TCR activation. To determine the effect of persistent TCR signaling on T-cell leukemogenesis, TEL-JAK2 transgenic mice were crossed with Marilyn mice to generate TEL-JAK2; Marylin (TJM) double transgenic mice. As compared to TJM double transgenic females, that all developed T-ALL, TJM males developed leukemia with significantly delayed latency. Flow cytometry analysis showed that of five diseased mice analyzed, one developed an IgM-positive immature-type B-cell lymphoma affecting the thymus and spleen, while four mice developed Thyl .2+CD4±CD8+ CD25+ T-cell leukemia infiltrating the thymus, spleen, lymph nodes and bone marrow. Interestingly, in these 4 T-cell leukemia cases transgenic TCR-νββ cell surface expression was low or absent. Two cases expressed non transgenic TCR chains, suggesting that in these mice expression of a non- H-Y-specific TCR complex substituting for the Marilyn TCR was selected during leukemogenesis. These results indicate that self-antigen TCR stimulation exerts a negative pressure on thymocyte malignant transformation, thus delaying leukemogenesis in TJM male mice.
Male mice expressing the transgenic Marilyn TCR present reduced thymocyte cellularity due to self-antigen-induced negative selection (Lantz et al, 2000). To investigate whether delayed leukemogenesis in TJM male mice involved H-Y antigenic stimulation of TCR rather than a reduction in cellular targets for malignant transformation, female TJM CD45.1+ leukemic cells were transplanted to either female or male syngenic CD45.2 recipients. As compared to female recipients, which rapidly developed fatal T-ALL, the majority of recipient males presented on average much lower numbers of donor malignant cells in blood and survived considerably longer. In addition, female recipients presented higher infiltration of CD45.1+ leukemic cells in spleen, lymph nodes, bone marrow and other organs than males sacrificed simultaneously. The protracted or prevented leukemogenesis in males receiving syngenic TJM leukemic cells depended on H-Y antigen MHC presentation but not upon an adaptive immune response, since leukemogenesis developed equally efficiently in female and male allogenic Swiss Nude recipient mice, but was hampered in male Rag2-deficient syngenic recipients.
We then hypothesized than anti-CD3 could mimic the TCR signaling and induce cell cell death. In vitro experiments using the OKT3-CD3 coupled to CD28 clearly demonstrated that ALL-SILTCR-HY undergo massive and rapid cell death when treated by CD3/28 beads compared to ALL-SIL in flow cytometry and confirmed in western blot. Test of proliferation of ALL-SIL and ALL-SIL-TCR-HY shows no effect of beads on ALL-SIL without TCR. To illustrate TCR activation, we examined some of the main key second messengers downstream from TCR signaling apparatus, and as expected CD3 crosslinking induced phosphorylation of ER , p38, AKT and its downstream target S6 ribosomal protein and translocation of NFAT1. Kinetic of phosphorylation of ERK was performed in flow cytometry confirmed in western blot and prove the functionality of this TCR transgene in our cell line ALL-SIL.
To explore whether stimulation of TCR/CD3 signaling could suppress in vivo leukemogenesis, we treated TJM leukemic mice with an anti-CD3smAb (145-2C11). When female TJM leukemic cells were transplanted to female mice, to avoid Marilyn TCR antigenic stimulation, and then treated 24 hours later and for 5 consecutive days with anti-CD3smAb, peripheral blood leukemia and fatal disease was prevented, as compared to control antibody- treated mice (Figure 1 A,B). Such effect of anti-CD3s in preventing leukemogenesis was also observed in mice transplanted with TEL-JAK2 transgenic leukemic cells (Figure 1 C,D), which express an endogenously generated surface TCR/CD3 complex (dos Santos et al, 2007). These results indicate that stimulation of transgenic as well as endogenous TCR signaling prevents leukemogenesis in secondary transplanted mice. To investigate whether the anti-CD3smAb could also have a curative effect, antibody treatment was initiated only when leukemic cells were detected in blood of secondary transplanted mice. Strikingly, anti-CD3streatment delayed the increase in peripheral blood leukemia and prolonged the survival of mice transplanted with either TJM (Figure IE) or TEL-JAK2 (Figure IF) leukemic cells. The CD3santibody also delayed peripheral blood leukemia in Swiss Nude mice transplanted with TEL-JAK2 leukemic cells, indicating that the anti-leukemogenic effect of this antibody is due to a direct action on TCR/CD3 -positive leukemic T cells and not on host-derived T cells.
We next investigated whether acute stimulation of TCR/CD3 signaling also shows antileukemic activity on TALL cells obtained from a TCR+ T-ALL diagnostic cases xenotransplanted into NOD/SCID/yc-/- (NSG) mice. NSG mice were injected with T-ALL cells and, 24 hours later, divided into 2 groups. The first was treated (2 rounds of 5 consecutive days) with the CD3-specific OKT3 monoclonal antibody and the second with an isotype control antibody. Leukemic cells (hCD45+hCD7+) expansion was followed over time in the blood of recipient mice. The results (Figure 2C) show that while fast expansion of T-ALL cells began at day 22 in the control group and rapidly enhanced thereafter to reach >90% blood nucleated cells at day 42, T-ALL cells did to expand in mice treated with the OKT3 antibody during 2 weeks, and very slowly expanded thereafter. We next investigated whether OKT3 treatment could also result in anti-leukemic effects in a curative setting. Mice were xenotransplanted with the same T-ALL but treatment with either the OKT3 or the control antibody only started when leukemic cells reached 1-4% of nucleated blood cells. The results (Figure 2D) show while T- ALL cells rapidly expanded in the control group, rapid clearance of leukemic cells in the blood of recipient mice was observed in blood of OKT3-treated mice. Thus, acute stimulation of TCR signaling clearly results in striking anti-leukemic effects on a xenotransplanted T-ALL both in a preventive or curative setting.
Anti-CD3 stimulation of ALL-SIL TCR-HY cells induces TCR signaling and cell death.
We next investigated whether anti-CD3 treatment could mimic TCR signaling to induce leukemic cell cell death. Unlike control ALL-SIL cells, TCR-HY-expressing cells underwent massive cell death when treated with anti-CD3/anti-CD28, as revealed by increased levels of cell surface AnnexinV, increased cleavage of caspase 3 and 7 and reduced cell expansion(Fig. 3A-D). Importantly, anti-CD3/anti-CD28 treatment did not impact on cell cycle progression per se (Fig. 3E and data not shown). To investigate the signaling properties of the mouse TCR- HY complex in human leukemic ALL-SIL cells, we first demonstrated that CD3 crosslinking induced TCR clustering at the ALL-SIL cell surface. Next, phosphokinase-array analysis of anti-CD3/CD28-stimulated ALL-SIL-TCR-HY and normal human DP thymocytesshowed increased phosphorylation of overlapping TCR signalling components, includingthe key downstream second messengers ERK, JNKand AKT. Anti-CD3 -induced phosphorylation of these proteins, and not of the cytokine signaling STAT3 protein, was confirmed by flow cytometry (Fig. 3E). Of note, inhibition of TCR signaling by cyclosporine or the MEK kinase inhibitor PD 184352 impaired anti-CD3 -induced cell death and expression of the CD69 and CD25 T-cell activation markers (Fig. 3F). Together these results indicatethat the TCR/CD3 signaling module is functional in T-ALL cells, and that its activation leads to cell death. Targeting CD3m vitro induces human primary T-ALL cell death and mimics thymic negative selection
To test whether anti-CD3 stimulation of TCR-expressing T-ALL diagnostic samples also induced cell death, primary human T-ALL cells belonging to the major molecular oncogenic T-ALL subtypes(10, 18) were treated with anti-CD3/anti-CD28. Massive T-ALL cell death and TCR signaling activation were observed in TCR-expressing T-ALL but not in TCR-negative cases, regardless of their molecular oncogenic subtype (Fig. 4). As silencing of the TLX1 driving oncogene in ALL-SIL cells was previously shown to induce both differentiation and cell death(14), we compared the transcriptional profiles of ALL-SIL cells in response to either TCR stimulation or small hairpin RNA (shRNA)-mediated knock-down of TLX1. Interestingly we found only a small overlap in differential gene expression between these signatures. Indeed, although both signatures were enriched for cell death-related GO terms, deregulated genes were generally distinct in the two conditions, suggesting that cell death induced in response to either TCR signaling or TLX1 knock-down are not mediated through the same pathways. Importantly, we found in the ALL-SIL TCR- induced transcriptional profile a significant enrichment of the TCR-induced negative selection signature of normal thymic progenitors(19). We conclude that T-ALL cell death induced by TCR stimulation is distinct from that induced by inhibition of underlying oncogenic pathways and rather involves signaling cues resembling those of TCR-induced negative selection in normal T cell progenitors.
Discussion
Current T-ALL therapies involve complex, often toxic chemotherapeutic regimens. Although T-ALL outcome has improved with current therapy, survival rates remain only around 50 and 70% at 5 years in adult and pediatric T-ALL, respectively(20, 21).The genetic bases of T-ALL progression and maintenance are well characterized but have not translated so far into targeted therapies(lO). There is thus unmet need for new treatments to offer therapeutic options for refractory disease and to prevent relapse. We report here that chronic/strong TCR signalling causes massive T-ALL cell cell death and shows potent tumor suppressive function in vivo. These findings call for the incorporation of TCR-directed therapies, as shown here for OKT3 treatment, in current treatment regimens of CD3 -expressing T-ALL. Muromonab-CD3 (OKT3) was approved by the U.S. Food and Drug Administration (FDA) in 1985 for therapy of acute, glucocorticoid-resistant rejection of allogeneic renal, heart and liver transplants (22) and was in fact the first monoclonal antibody introduced in the clinic. Since then, a number of other monoclonal antibodies to CD3 were developed (23) that may prove superior to OKT3 in T-ALL treatment. Of note, encouraging response to OKT3 therapy was reported in an adult patient with an aggressive and chemotherapy-resistant T-ALL, but the basis of this response was not studied (24). A major drawback of current chemotherapeutic regimens in T-ALL is the frequent resistance to treatment and relapse. Leukemia initiating cells (LICs) from residual disease are thought to be responsible for relapsing cases (25, 26). In addition, resistance of T-ALL to chemotherapy is in part linked to the recurrent genetic abnormalities selected during disease progression, e.g. inactivation of the PTEN tumor suppressor gene and the resulting activation of PI3kinase/AKT signaling (27, 28). Whether administration of anti-CD3 therapy during the remission phase or its association with conventional chemotherapy regimens could target LICs and/or bypass molecular mechanisms of primitive resistance represent promising directions to be explored in future prospective studies.
T cells mature in the thymus following a highly orchestrated process controlled both by cell intrinsic (e.g. transcription factors) and extrinsic (e.g. stroma-derived cytokines/chemokines) molecular cues (29, 30). Cell surface TCRD□ expression in DP thymocytes allows recognition of specific self-MHC/peptide to transduce a positive selection signal and maturation into SP thymocytes. DP thymocytes not receiving this signal die through lack of stimulation; whereas those whose TCR binds too strongly to self-MHC/peptide undergo activation- induced cell death and negative selection (5, 31). In both situations TCR binding to pMHC is the triggering event but how TCR engagement leads to such divergent outcomes (survival and proliferation versus death) remains unclear (32). Remarkably, these two contrasting processes are driven by a TCR signaling machinery of qualitatively similar composition (31). The difference lies in the molecular interpretation of signals of different strength, which may rely on compartmentalization of key signaling players. Indeed, it has been shown that a small increase in ligand affinity for the TCR leads to a marked change in the subcellular localization (plasma membrane for negative selecting ligands versus Golgi complex for positively selecting ones) of essential adaptors of the Ras signaling pathway. This compartmentalization induces the conversion of a small change in analogue input (affinity for ligand) into a digital output (positive versus negative selection)(33, 34). More extensive studies on the composition and function of TCR- induced signalosomes formed in T-ALL stimulated by anti-CD3 and the identification of critical components in T-ALL of the transcriptomic signature akin to that of thymic negative selection (19, 35, 36) should provide information on the molecular pathways involved in anti-CD3 induced cell death in leukemic cells. These pathways could in turn constitute new pharmaceutical targets to treat T-ALL. Although signaling from the B-cell receptor (BCR) and pre-BCR was shown to promote B-cell malignancies (37, 38), the role of TCR signaling in T-ALL has thus far remained controversial. Studies in T-ALL mouse models indicated a pro-oncogenic role for TCR signalling (39-42). However, these data were based on transgenic TCR systems, which do not reflect the expression levels and receptor diversity found among T-ALL patients where TCR expression is heterogeneous or absent (6). Moreover, gene inactivation studies in other mouse T-cell leukemia models have shown that TCR expression is not essential for T-ALL development (43, 44). The current work demonstrates that anti-CD3 -mediated activation of endogenous TCR in murine or human T-ALL has an anti-leukemic function. T-ALL often arises from immature T-cell precursors before the stage of negative selection (6, 42). Consequently, contingent expression of a non-negatively selected TCR will render T-ALL cells sensitive to TCR-activating apoptotic signals that mimic negative selection, as demonstrated here by anti- CD3 treatment in vitro and in vivo. During T-ALL development, it is possible that selective events enable cells to escape post-malignancy negative selection, such as loss of TCR surface expression, as found in a subset of T-ALL patients (6) and as demonstrated here in TJM double transgenic male mice.
The dual role of developmental molecular pathways in organogenesis and tumorigenesisis increasingly recognised, the modulation of which may provide potential therapeutic opportunities (1). In this study, we found that reactivation in T-ALL blasts of a lineage-specific checkpoint control normally set by TCR signalling during T-cell development,displays anti-tumoral functions. Importantly, despite the multiple and complex oncogenic mechanisms driving T-ALL, which include anti-apoptotic (45) and pro- proliferative(46) signaling cues, this TCR-dependent checkpoint remains switchable to induce massive tumor cell cell death. Reactivation of similar lineage-specific developmental checkpoints in malignancies originating from other lineages and tissues could provide a novel class of therapeutic targets in cancer.
REFERENCES:
Throughout this application, various references describe the state of the art to which this invention pertains. The disclosures of these references are hereby incorporated by reference into the present disclosure. 1. Garraway LA, Sellers WR. Lineage dependency and lineage-survival oncogenes in human cancer. Nat Rev Cancer. 2006 Aug;6(8):593-602.
2. Dik WA, Pike-Overzet K, Weerkamp F, de Ridder D, de Haas EF, Baert MR, et al. New insights on human T cell development by quantitative T cell receptor gene rearrangement studies and gene expression profiling. The Journal of experimental medicine. 2005 Jun 6;201(l l): 1715-23.
3. Von Boehmer H. Deciphering thymic development. Front Immunol. 2014;5:424.
4. Gascoigne NR, Palmer E. Signaling in thymic selection. Curr Opin Immunol. 2011 Apr;23(2):207-12.
5. Klein L, Kyewski B, Allen PM, Hogquist KA. Positive and negative selection of the T cell repertoire: what thymocytes see (and don't see). Nature reviews. 2014 Jun;14(6):377- 91.
6. Asnafi V, Beldjord K, Boulanger E, Comba B, Le Tutour P, Estienne MH, et al. Analysis of TCR, pT alpha, and RAG-1 in T-acute lymphoblastic leukemias improves understanding of early human T-lymphoid lineage commitment. Blood. 2003 Apr l;101(7):2693-703.
7. Asnafi V, Beldjord K, Libura M, Villarese P, Millien C, Ballerini P, et al. Age- related phenotypic and oncogenic differences in T-cell acute lymphoblastic leukemias may reflect thymic atrophy. Blood. 2004 Dec 15;104(13):4173-80.
8. Ferrando AA, Neuberg DS, Staunton J, Loh ML, Huard C, Raimondi SC, et al.
Gene expression signatures define novel oncogenic pathways in T cell acute lymphoblastic leukemia. Cancer cell. 2002 Feb;l(l):75-87.
9. Ntziachristos P, Tsirigos A, Van Vlierberghe P, Nedjic J, Trimarchi T, Flaherty MS, et al. Genetic inactivation of the polycomb repressive complex 2 in T cell acute lymphoblastic leukemia. Nature medicine. 2012 Feb;18(2):298-301.
10. Van Vlierberghe P, Ferrando A. The molecular basis of T cell acute lymphoblastic leukemia. The Journal of clinical investigation. 2012 Oct;122(10):3398-406.
11. Weng AP, Ferrando AA, Lee W, Morris JPt, Silverman LB, Sanchez-Irizarry C, et al. Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia. Science (New York, NY. 2004 Oct 8;306(5694):269-71.
12. Zhang J, Ding L, Holmfeldt L, Wu G, Heatley SL, Payne-Turner D, et al. The genetic basis of early T-cell precursor acute lymphoblastic leukaemia. Nature. 2012 Jan 12;481(7380): 157-63. 13. Soulier J, Clappier E, Cayuela JM, Regnault A, Garcia-Peydro M, Dombret H, et al. HOXA genes are included in genetic and biologic networks defining human acute T-cell leukemia (T-ALL). Blood. 2005 Jul l;106(l):274-86.
14. Dadi S, Le Noir S, Payet-Bornet D, Lhermitte L, Zacarias-Cabeza J, Bergeron J, et al. TLX homeodomain oncogenes mediate T cell maturation arrest in T-ALL via interaction with ETSl and suppression of TCRalpha gene expression. Cancer cell. 2012 Apr 17;21(4):563- 76.
15. De Keersmaecker K. Novel insights on TLX1 function in T-ALL pave the way towards differentiation therapy. Haematologica. 2012 Jun;97(6):795.
16. Carron C, Cormier F, Janin A, Lacronique V, Giovannini M, Daniel MT, et al.
TEL-JAK2 transgenic mice develop T-cell leukemia. Blood. 2000 Jun 15;95(12):3891-9.
17. Lantz O, Grandjean I, Matzinger P, Di Santo JP. Gamma chain required for naive CD4+ T cell survival but not for antigen proliferation. Nature immunology. 2000 Jul;l(l):54- 8.
18. Aifantis I, Raetz E, Buonamici S. Molecular pathogenesis of T-cell leukaemia and lymphoma. Nature reviews. 2008 May;8(5):380-90.
19. Baldwin TA, Hogquist KA. Transcriptional analysis of clonal deletion in vivo. J Immunol. 2007 Jul 15;179(2):837-44.
20. Huguet F, Leguay T, Raffoux E, Thomas X, Beldjord K, Delabesse E, et al. Pediatric-inspired therapy in adults with Philadelphia chromosome-negative acute lymphoblastic leukemia: the GRAALL-2003 study. J Clin Oncol. 2009 Feb 20;27(6):911-8.
21. Pui CH, Evans WE. Treatment of acute lymphoblastic leukemia. The New England journal of medicine. 2006 Jan 12;354(2): 166-78.
22. Smith SL. Ten years of Orthoclone OKT3 (muromonab-CD3): a review. J Transpl Coord. 1996 Sep;6(3): 109-19; quiz 20-1.
23. Martin A, Tisch RM, Getts DR. Manipulating T cell-mediated pathology: targets and functions of monoclonal antibody immunotherapy. Clinical immunology (Orlando, Fla. 2013 Jul;148(l): 136-47.
24. Gramatzki M, Burger R, Strobel G, Trautmann U, Bartram CR, Helm G, et al. Therapy with OKT3 monoclonal antibody in refractory T cell acute lymphoblastic leukemia induces interleukin-2 responsiveness. Leukemia. 1995 Mar;9(3):382-90.
25. Clappier E, Gerby B, Sigaux F, Delord M, Touzri F, Hernandez L, et al. Clonal selection in xenografted human T cell acute lymphoblastic leukemia recapitulates gain of malignancy at relapse. The Journal of experimental medicine. 2011 Apr 11;208(4):653-61. 26. King B, Trimarchi T, Reavie L, Xu L, Mullenders J, Ntziachristos P, et al. The ubiquitin ligase FBXW7 modulates leukemia-initiating cell activity by regulating MYC stability. Cell. 2013 Jun 20;153(7): 1552-66.
27. Piovan E, Yu J, Tosello V, Herranz D, Ambesi-Impiombato A, Da Silva AC, et al. Direct reversal of glucocorticoid resistance by AKT inhibition in acute lymphoblastic leukemia. Cancer cell. 2013 Dec 9;24(6):766-76.
28. Trinquand A, Tanguy-Schmidt A, Ben Abdelali R, Lambert J, Beldjord K, Lengline E, et al. Toward a NOTCH l/FBXW7/RAS/PTEN-based oncogenetic risk classification of adult T-cell acute lymphoblastic leukemia: a Group for Research in Adult Acute Lymphoblastic Leukemia study. J Clin Oncol. 2013 Dec l;31(34):4333-42.
29. Schwarz BA, Sambandam A, Maillard I, Harman BC, Love PE, Bhandoola A. Selective thymus settling regulated by cytokine and chemokine receptors. J Immunol. 2007 Feb 15;178(4):2008-17.
30. Yui MA, Rothenberg EV. Developmental gene networks: a triathlon on the course to T cell identity. Nature reviews. 2014 Aug;14(8):529-45.
31. Morris GP, Allen PM. How the TCR balances sensitivity and specificity for the recognition of self and pathogens. Nature immunology. 2012 Feb;13(2): 121-8.
32. Hogquist KA, Jameson SC. The self-obsession of T cells: how TCR signaling thresholds affect fate 'decisions' and effector function. Nature immunology. 2014 Sep;15(9):815-23.
33. Daniels MA, Teixeiro E, Gill J, Hausmann B, Roubaty D, Holmberg K, et al. Thymic selection threshold defined by compartmentalization of Ras/MAPK signalling. Nature. 2006 Dec 7;444(7120):724-9.
34. Prasad A, Zikherman J, Das J, Roose JP, Weiss A, Chakraborty AK. Origin of the sharp boundary that discriminates positive and negative selection of thymocytes.
Proceedings of the National Academy of Sciences of the United States of America. 2009 Jan 13;106(2):528-33.
35. Liston A, Hardy K, Pittelkow Y, Wilson SR, Makaroff LE, Fahrer AM, et al. Impairment of organ-specific T cell negative selection by diabetes susceptibility genes: genomic analysis by mRNA profiling. Genome biology. 2007;8(1):R12.
36. Schmitz I, Clayton LK, Reinherz EL. Gene expression analysis of thymocyte selection in vivo. International immunology. 2003 Oct;15(10): 1237-48. 37. Davis RE, Ngo VN, Lenz G, Tolar P, Young RM, Romesser PB, et al. Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature. 2010 Jan 7;463(7277):88-92.
38. Geng H, Hurtz C, Lenz KB, Chen Z, Baumjohann D, Thompson S, et al. Self- Enforcing Feedback Activation between BCL6 and Pre-B Cell Receptor Signaling Defines a
Distinct Subtype of Acute Lymphoblastic Leukemia. Cancer cell. 2015 Mar 9;27(3):409-25.
39. Brabb T, Huseby ES, Morgan TM, SantAngelo DB, Kirchner J, Farr AG, et al. Thymic stromal organization is regulated by the specificity of T cell receptor/major histocompatibility complex interactions. European journal of immunology. 1997 Jan;27(l): 136-46.
40. Cui Y, Onozawa M, Garber HR, Samsel L, Wang Z, McCoy JP, et al. Thymic expression of a T cell receptor targeting a tumor associated antigen co-expressed in the thymus induces T-ALL. Blood. 2015 Mar 26.
41. Kelly JA, Spolski R, Kovanen PE, Suzuki T, Bollenbacher J, Pise-Masison CA, et al. Stat5 synergizes with T cell receptor/antigen stimulation in the development of lymphoblastic lymphoma. The Journal of experimental medicine. 2003 Jul 7;198(l):79-89.
42. Strzadala L, Miazek A, Matuszyk J, Kisielow P. Role of thymic selection in the development of thymic lymphomas in TCR transgenic mice. International immunology. 1997 Jan;9(l): 127-38.
43. dos Santos NR, Rickman DS, de Reynies A, Cormier F, Williame M, Blanchard
C, et al. Pre-TCR expression cooperates with TEL-JAK2 to transform immature thymocytes and induce T-cell leukemia. Blood. 2007 May 1;109(9):3972-81.
44. Klinger MB, Guilbault B, Goulding RE, Kay RJ. Deregulated expression of RasGRPl initiates thymic lymphomagenesis independently of T-cell receptors. Oncogene. 2005 Apr 14;24(16):2695-704.
45. Sanda T, Tyner JW, Gutierrez A, Ngo VN, Glover J, Chang BH, et al. TYK2- STAT1-BCL2 pathway dependence in T-cell acute lymphoblastic leukemia. Cancer Discov. 2013 May;3(5):564-77.
46. Dail M, Wong J, Lawrence J, O'Connor D, Nakitandwe J, Chen SC, et al. Loss of oncogenic Notchl with resistance to a PI3K inhibitor in T-cell leukaemia. Nature. 2014 Sep
25;513(7519):512-6.
47. Asnafi V, Buzyn A, Le Noir S, Baleydier F, Simon A, Beldjord K, et al. NOTCH 1/FBXW7 mutation identifies a large subgroup with favorable outcome in adult T-cell acute lymphoblastic leukemia (T-ALL): a Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL) study. Blood. 2009 Apr 23; 113(17):3918-24.
48. Bergeron J, Clappier E, Radford I, Buzyn A, Millien C, Soler G, et al. Prognostic and oncogenic relevance of TLX1/HOX11 expression level in T-ALLs. Blood. 2007 Oct l;110(7):2324-30.
49. Carron C, Cormier F, Janin A, et al. TEL-JAK2 transgenic mice develop T-cell leukemia. Blood. 2000;95:3891-3899.50. dos Santos NR, Rickman DS, de Reynies A, Cormier F, Williame M, Blanchard C, Stern MH, Ghysdael J. Pre-TCR expression cooperates with TEL- JAK2 to transform immature thymocytes and induce T-cell leukemia. Blood. 2007;109:3972- 81.
51. Graux C, Cools J, Michaux L, Vandenberghe P, Hagemeijer A. Cytogenetics and molecular genetics of T-cell acute lymphoblastic leukemia: from thymocyte to lymphoblast. Leukemia. 2006 Sep;20(9): 1496-510.
52. Lantz O, Grandjean I, Matzinger P, Di Santo JP. Gamma chain required for naive CD4T cell survival but not for antigen proliferation. Nat Immunol. 2000;1 :54-58.
53. Roti G, Stegmaier K. New Approaches to Target T-ALL. Front Oncol. 2014 Jul 8;4:170.
54. Vahid Asnafi, Kheira Beldjord, Marta Libura, Patrick Villarese, Corrine Millien, Paola Ballerini, Emilienne Kuhlein, Marina Lafage-Pochitaloff, Eric Delabesse, Olivier Bernard, and Elizabeth Macintyre. Age-related phenotypic and oncogenic differences in T-cell acute lymphoblastic leukemias may reflect thymic atrophy. 2004 by The American Society of Hematology.
55. Gramatzki M, Burger R, Strobel G, Trautmann U, Bartram CR, Helm G, Horneff G, Alsalameh S, Jonker M, Gebhart E, et al. Therapy with OKT3 monoclonal antibody in refractory T cell acute lymphoblastic leukemia induces interleukin-2 responsiveness. Leukemia 1995.

Claims

CLAIMS:
1. A non-mitogenic anti-CD3 antibody for use in a method for treating T-cell acute lymphoblastic leukemia (T-ALL).
2. A non-mitogenic anti-CD3 antibody for use in a method for inducing cell death of T- ALL cells.
3. The non-mitogenic anti-CD3 antibody for use according to claim 1 or 2, wherein said anti-CD3 is an anti-CD3 monoclonal antibody (mAb).
5. The non-mitogenic anti-CD3 antibody for use according to any one claims 1 to 3, wherein the anti-CD3 mAb is TRX4 (otelixizumab) or MGA031 (teplizumab).
6. A pharmaceutical composition comprising a non-mitogenic anti-CD3 antibody for use in a method for treating T-ALL.
7. An in vitro method for predicting the responsiveness of a patient affected with a T-ALL to a treatment with a TCR-activating agent comprising a step of determining the expression of TCR/CD3 complex at the surface of T-ALL cells obtained from a patient, wherein the absence of the TCR/CD3 complex is indicative of the non-responsiveness to said treatment.
8. The method of claim 7, wherein the step of detecting the expression of TCR/CD3 complex at the surface of T-ALL cells is performed by immunoassay.
9. The method of claim 7 or 8, further comprising a step of analysing TCR clustering.
10. The method according to any one claims 7 to 9, wherein the TCR-activating agent is a non-mitogenic anti-CD3 antibody.
11. A TCR-activating agent for use in a method for treating T-ALL in a patient classified as responder according to the method defined in any one claims 7 to 10.
12. The TCR-activating agent for use according to claim 11, wherein the TCR-activating agent is a non-mitogenic anti-CD3 antibody.
13. The TCR-activating agent for use according to claim 11, wherein the TCR-activating activating agent is anti-TCR antibody.
14. The TCR-activating agent for use according to claim 11, wherein the TCR-activating activating agent is inhibitor of TCR regulating tyrosine phosphatase. 15. An anti-TCR antibody for use in a method for treating T-ALL.
16. A method for treating T-ALL in a patient in need thereof and identified as responder in the method for predicting comprising a step of administrating to said patient a therapeutically amount of a TCR-activating agent.
PCT/EP2015/074033 2014-10-16 2015-10-16 Tcr-activating agents for use in the treatment of t-all WO2016059220A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP14306643.9 2014-10-16
EP14306643 2014-10-16

Publications (1)

Publication Number Publication Date
WO2016059220A1 true WO2016059220A1 (en) 2016-04-21

Family

ID=51799061

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2015/074033 WO2016059220A1 (en) 2014-10-16 2015-10-16 Tcr-activating agents for use in the treatment of t-all

Country Status (1)

Country Link
WO (1) WO2016059220A1 (en)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018045238A1 (en) * 2016-09-01 2018-03-08 Mayo Foundation For Medical Education And Research Methods and compositions for targeting t-cell cancers
US10213513B2 (en) 2014-06-16 2019-02-26 Mayo Foundation For Medical Education And Research Treating myelomas
US10279035B2 (en) 2012-10-01 2019-05-07 Mayo Foundation For Medical Education And Research Nanoparticle complexes of paclitaxel, trastuzumab, and albumin
US10300016B2 (en) 2014-10-06 2019-05-28 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10561726B2 (en) 2015-10-06 2020-02-18 Vavotar Life Sciences LLC Methods of treating cancer using compositions of antibodies and carrier proteins with antibody pretreatment
US10618969B2 (en) 2016-04-06 2020-04-14 Mayo Foundation For Medical Education And Research Carrier-binding agent compositions and methods of making and using the same
US10765741B2 (en) 2011-05-09 2020-09-08 Mayo Foundation For Medical Education And Research Methods for treating VEGF-expressing cancer using preformed nanoparticle complexes comprising albumin-bound paclitaxel and bevacizumab
WO2021064069A1 (en) * 2019-10-02 2021-04-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of adult t-cell leukemia/lymphoma
US11241387B2 (en) 2015-08-18 2022-02-08 Mayo Foundation For Medical Education And Research Carrier-binding agent compositions and methods of making and using the same
US11305020B2 (en) 2016-03-21 2022-04-19 Mayo Foundation For Medical Education And Research Methods for reducing toxicity of a chemotherapeutic drug
US11311631B2 (en) 2016-09-06 2022-04-26 Mayo Foundation For Medical Education And Research Paclitaxel-albumin-binding agent compositions and methods for using and making the same
US11427637B2 (en) 2016-09-06 2022-08-30 Mayo Foundation For Medical Education And Research Methods of treating PD-L1 expressing cancer
US20220389104A1 (en) * 2021-05-28 2022-12-08 Ose Immunotherapeutics Method for Treating CD127-Positive Cancers by Administering an Anti-CD127 Agent
US11548946B2 (en) 2016-09-01 2023-01-10 Mayo Foundation For Medical Education And Research Carrier-PD-L1 binding agent compositions for treating cancers
US11571469B2 (en) 2016-01-07 2023-02-07 Mayo Foundation For Medical Education And Research Methods of treating cancer with interferon wherein the cancer cells are HLA negative or have reduced HLA expression
US11590098B2 (en) 2016-09-06 2023-02-28 Mayo Foundation For Medical Education And Research Methods of treating triple-negative breast cancer using compositions of antibodies and carrier proteins
US11878061B2 (en) 2016-03-21 2024-01-23 Mayo Foundation For Medical Education And Research Methods for improving the therapeutic index for a chemotherapeutic drug

Citations (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US877397A (en) 1907-07-15 1908-01-21 American Pulley Co Reel.
US4361549A (en) 1979-04-26 1982-11-30 Ortho Pharmaceutical Corporation Complement-fixing monoclonal antibody to human T cells, and methods of preparing same
US4816397A (en) 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
EP0403156A1 (en) 1989-06-07 1990-12-19 Genzyme Corporation Improved monoclonal antibodies against the human alpha/beta t-cell receptor, their production and use
WO1992003918A1 (en) 1990-08-29 1992-03-19 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5202238A (en) 1987-10-27 1993-04-13 Oncogen Production of chimeric antibodies by homologous recombination
US5204244A (en) 1987-10-27 1993-04-20 Oncogen Production of chimeric antibodies by homologous recombination
WO1993012227A1 (en) 1991-12-17 1993-06-24 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
EP0564409A1 (en) 1992-04-03 1993-10-06 Ciba-Geigy Ag Pyrimidin derivatives and process for their preparation
WO1994025585A1 (en) 1993-04-26 1994-11-10 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5545807A (en) 1988-10-12 1996-08-13 The Babraham Institute Production of antibodies from transgenic animals
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
WO1997010354A1 (en) 1995-09-11 1997-03-20 Kyowa Hakko Kogyo Co., Ltd. ANTIBODY AGAINTS α-CHAIN OF HUMAN INTERLEUKIN 5 RECEPTOR
WO1997013852A1 (en) 1995-10-10 1997-04-17 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
WO1998024884A1 (en) 1996-12-02 1998-06-11 Genpharm International Transgenic non-human animals capable of producing heterologous antibodies
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US5874299A (en) 1990-08-29 1999-02-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
WO1999032619A1 (en) 1997-12-23 1999-07-01 The Carnegie Institution Of Washington Genetic inhibition by double-stranded rna
US5939598A (en) 1990-01-12 1999-08-17 Abgenix, Inc. Method of making transgenic mice lacking endogenous heavy chains
WO1999045962A1 (en) 1998-03-13 1999-09-16 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
WO1999046268A1 (en) 1998-03-12 1999-09-16 Novo Nordisk A/S MODULATORS OF PROTEIN TYROSINE PHOSPHATASES (PTPases)
WO1999046236A1 (en) 1998-03-12 1999-09-16 Novo Nordisk A/S Modulators of protein tyrosine phosphatases (ptpases)
US5981732A (en) 1998-12-04 1999-11-09 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-13 expression
US6002008A (en) 1997-04-03 1999-12-14 American Cyanamid Company Substituted 3-cyano quinolines
US6046321A (en) 1999-04-09 2000-04-04 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-i1 expression
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6107091A (en) 1998-12-03 2000-08-22 Isis Pharmaceuticals Inc. Antisense inhibition of G-alpha-16 expression
WO2000062778A1 (en) 1999-04-15 2000-10-26 Bristol-Myers Squibb Co. Cyclic protein tyrosine kinase inhibitors
US6143297A (en) 1989-10-27 2000-11-07 Arch Development Corporation Methods of promoting immunopotentiation and preparing antibodies with anti-CD3 antibodies
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6162963A (en) 1990-01-12 2000-12-19 Abgenix, Inc. Generation of Xenogenetic antibodies
WO2001014424A2 (en) 1999-08-24 2001-03-01 Medarex, Inc. Human ctla-4 antibodies and their uses
WO2001036646A1 (en) 1999-11-19 2001-05-25 Cancer Research Ventures Limited Inhibiting gene expression with dsrna
WO2001068836A2 (en) 2000-03-16 2001-09-20 Genetica, Inc. Methods and compositions for rna interference
US6365354B1 (en) 2000-07-31 2002-04-02 Isis Pharmaceuticals, Inc. Antisense modulation of lysophospholipase I expression
WO2002043478A2 (en) 2000-11-30 2002-06-06 Medarex, Inc. Transgenic transchromosomal rodents for making human antibodies
US6406696B1 (en) 1989-10-27 2002-06-18 Tolerance Therapeutics, Inc. Methods of stimulating the immune system with anti-CD3 antibodies
US6410323B1 (en) 1999-08-31 2002-06-25 Isis Pharmaceuticals, Inc. Antisense modulation of human Rho family gene expression
WO2002092812A1 (en) 2001-05-11 2002-11-21 Kirin Beer Kabushiki Kaisha ARTIFICIAL HUMAN CHROMOSOME CONTAINING HUMAN ANTIBODY μ LIGHT CHAIN GENE
US6491916B1 (en) 1994-06-01 2002-12-10 Tolerance Therapeutics, Inc. Methods and materials for modulation of the immunosuppresive activity and toxicity of monoclonal antibodies
WO2003013527A1 (en) 2001-08-03 2003-02-20 Schering Corporation Sulfonamide derivatives as gamma secretase inhibitors
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
WO2004005281A1 (en) 2002-07-05 2004-01-15 Novartis Ag Inhibitors of tyrosine kinases
WO2005004669A1 (en) 2003-07-10 2005-01-20 Kai Li An adjustable toothbrush
WO2005063709A1 (en) 2003-12-25 2005-07-14 Nippon Shinyaku Co., Ltd. Amide derivative and medicine
US7041289B1 (en) 1997-12-05 2006-05-09 Institut National De La Sante Et De La Recherche Medicale (Inserm) Method for treating established spontaneous auto-immune diseases in mammals
US20060205666A1 (en) 2002-04-24 2006-09-14 Hiroshi Mori Gamma-secretase inhibitors
WO2007075869A2 (en) 2005-12-23 2007-07-05 Ariad Pharmaceuticals, Inc. Bicyclic heteroaryl compounds
WO2008027584A2 (en) 2006-09-01 2008-03-06 Vertex Pharmaceuticals Incorporated 5- (2-furyl)-1, 3-thiazole derivatives useful as inhibitors of phosphatidylinositol 3-kinase
WO2008070150A1 (en) 2006-12-05 2008-06-12 Bayer Schering Pharma Aktiengesellschaft Substituted 2,3-dihydroimidazo[1,2-c]quinazoline derivatives useful for treating hyper-proliferative disorders and diseases associated with angiogenesis
WO2008125833A1 (en) 2007-04-12 2008-10-23 Piramed Limited Pharmaceutical compounds
WO2008125839A2 (en) 2007-04-12 2008-10-23 Piramed Limited Pyrimidine derivatives as inhibitors of phosphatidylinositol-3-kinase
WO2008125835A1 (en) 2007-04-12 2008-10-23 Piramed Limited 2-morpholin-4-yl-pyrimidines as pi3k inhibitors
WO2009010530A1 (en) 2007-07-18 2009-01-22 Novartis Ag Bicyclic heteroaryl compounds and their use as kinase inhibitors
WO2009026345A1 (en) 2007-08-20 2009-02-26 Targegen Inc. Thiazolidinone compounds, and methods of making and using same
WO2009071888A1 (en) 2007-12-04 2009-06-11 Ucb Pharma S.A. Pyrrolothiazoles as pi3-kinase inhibitors
WO2009071895A1 (en) 2007-12-04 2009-06-11 Ucb Pharma S.A. Fused thiazole and thiophene derivatives as kinase inhibitors
WO2009071890A1 (en) 2007-12-04 2009-06-11 Ucb Pharma S.A. Tricyclic kinase inhibitors
WO2010125082A1 (en) 2009-04-30 2010-11-04 Glaxo Group Limited Oxazole substituted indazoles as pi3-kinase inhibitors
WO2013037484A2 (en) 2011-09-12 2013-03-21 Genzyme Corporation Anti-aplhabetatcr antibody
US8512967B2 (en) * 2002-04-05 2013-08-20 Ventana Medical Systems, Inc. Method for predicting the response to HER2-directed therapy
WO2015066379A2 (en) 2013-10-30 2015-05-07 Genzyme Corporation Methods for enhancing immunosuppressive therapy by multiple administration of alpha beta tcr-binding polypeptide

Patent Citations (75)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US877397A (en) 1907-07-15 1908-01-21 American Pulley Co Reel.
US4361549A (en) 1979-04-26 1982-11-30 Ortho Pharmaceutical Corporation Complement-fixing monoclonal antibody to human T cells, and methods of preparing same
US4816397A (en) 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5202238A (en) 1987-10-27 1993-04-13 Oncogen Production of chimeric antibodies by homologous recombination
US5204244A (en) 1987-10-27 1993-04-20 Oncogen Production of chimeric antibodies by homologous recombination
US5545807A (en) 1988-10-12 1996-08-13 The Babraham Institute Production of antibodies from transgenic animals
EP0403156A1 (en) 1989-06-07 1990-12-19 Genzyme Corporation Improved monoclonal antibodies against the human alpha/beta t-cell receptor, their production and use
US6143297A (en) 1989-10-27 2000-11-07 Arch Development Corporation Methods of promoting immunopotentiation and preparing antibodies with anti-CD3 antibodies
US6406696B1 (en) 1989-10-27 2002-06-18 Tolerance Therapeutics, Inc. Methods of stimulating the immune system with anti-CD3 antibodies
US6162963A (en) 1990-01-12 2000-12-19 Abgenix, Inc. Generation of Xenogenetic antibodies
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6114598A (en) 1990-01-12 2000-09-05 Abgenix, Inc. Generation of xenogeneic antibodies
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5939598A (en) 1990-01-12 1999-08-17 Abgenix, Inc. Method of making transgenic mice lacking endogenous heavy chains
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5569825A (en) 1990-08-29 1996-10-29 Genpharm International Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
WO1992003918A1 (en) 1990-08-29 1992-03-19 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US5874299A (en) 1990-08-29 1999-02-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
WO1993012227A1 (en) 1991-12-17 1993-06-24 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
EP0564409A1 (en) 1992-04-03 1993-10-06 Ciba-Geigy Ag Pyrimidin derivatives and process for their preparation
WO1994025585A1 (en) 1993-04-26 1994-11-10 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US6491916B1 (en) 1994-06-01 2002-12-10 Tolerance Therapeutics, Inc. Methods and materials for modulation of the immunosuppresive activity and toxicity of monoclonal antibodies
WO1997010354A1 (en) 1995-09-11 1997-03-20 Kyowa Hakko Kogyo Co., Ltd. ANTIBODY AGAINTS α-CHAIN OF HUMAN INTERLEUKIN 5 RECEPTOR
WO1997013852A1 (en) 1995-10-10 1997-04-17 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
WO1998024884A1 (en) 1996-12-02 1998-06-11 Genpharm International Transgenic non-human animals capable of producing heterologous antibodies
US6002008A (en) 1997-04-03 1999-12-14 American Cyanamid Company Substituted 3-cyano quinolines
US7041289B1 (en) 1997-12-05 2006-05-09 Institut National De La Sante Et De La Recherche Medicale (Inserm) Method for treating established spontaneous auto-immune diseases in mammals
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
WO1999032619A1 (en) 1997-12-23 1999-07-01 The Carnegie Institution Of Washington Genetic inhibition by double-stranded rna
WO1999046268A1 (en) 1998-03-12 1999-09-16 Novo Nordisk A/S MODULATORS OF PROTEIN TYROSINE PHOSPHATASES (PTPases)
WO1999046236A1 (en) 1998-03-12 1999-09-16 Novo Nordisk A/S Modulators of protein tyrosine phosphatases (ptpases)
WO1999045962A1 (en) 1998-03-13 1999-09-16 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
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
US6046321A (en) 1999-04-09 2000-04-04 Isis Pharmaceuticals Inc. Antisense modulation of G-alpha-i1 expression
WO2000062778A1 (en) 1999-04-15 2000-10-26 Bristol-Myers Squibb Co. Cyclic protein tyrosine kinase inhibitors
WO2001014424A2 (en) 1999-08-24 2001-03-01 Medarex, Inc. Human ctla-4 antibodies and their uses
US6410323B1 (en) 1999-08-31 2002-06-25 Isis Pharmaceuticals, Inc. Antisense modulation of human Rho family gene expression
WO2001036646A1 (en) 1999-11-19 2001-05-25 Cancer Research Ventures Limited Inhibiting gene expression with dsrna
WO2001068836A2 (en) 2000-03-16 2001-09-20 Genetica, Inc. Methods and compositions for rna interference
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
WO2002043478A2 (en) 2000-11-30 2002-06-06 Medarex, Inc. Transgenic transchromosomal rodents for making human antibodies
WO2002092812A1 (en) 2001-05-11 2002-11-21 Kirin Beer Kabushiki Kaisha ARTIFICIAL HUMAN CHROMOSOME CONTAINING HUMAN ANTIBODY μ LIGHT CHAIN GENE
WO2003013527A1 (en) 2001-08-03 2003-02-20 Schering Corporation Sulfonamide derivatives as gamma secretase inhibitors
US8512967B2 (en) * 2002-04-05 2013-08-20 Ventana Medical Systems, Inc. Method for predicting the response to HER2-directed therapy
US20060205666A1 (en) 2002-04-24 2006-09-14 Hiroshi Mori Gamma-secretase inhibitors
WO2004005281A1 (en) 2002-07-05 2004-01-15 Novartis Ag Inhibitors of tyrosine kinases
WO2005004669A1 (en) 2003-07-10 2005-01-20 Kai Li An adjustable toothbrush
WO2005063709A1 (en) 2003-12-25 2005-07-14 Nippon Shinyaku Co., Ltd. Amide derivative and medicine
US7728131B2 (en) 2003-12-25 2010-06-01 Nippon Shinyaku Co., Ltd. Amide derivative and medicine
WO2007075869A2 (en) 2005-12-23 2007-07-05 Ariad Pharmaceuticals, Inc. Bicyclic heteroaryl compounds
WO2008027584A2 (en) 2006-09-01 2008-03-06 Vertex Pharmaceuticals Incorporated 5- (2-furyl)-1, 3-thiazole derivatives useful as inhibitors of phosphatidylinositol 3-kinase
WO2008070150A1 (en) 2006-12-05 2008-06-12 Bayer Schering Pharma Aktiengesellschaft Substituted 2,3-dihydroimidazo[1,2-c]quinazoline derivatives useful for treating hyper-proliferative disorders and diseases associated with angiogenesis
WO2008125835A1 (en) 2007-04-12 2008-10-23 Piramed Limited 2-morpholin-4-yl-pyrimidines as pi3k inhibitors
WO2008125839A2 (en) 2007-04-12 2008-10-23 Piramed Limited Pyrimidine derivatives as inhibitors of phosphatidylinositol-3-kinase
WO2008125833A1 (en) 2007-04-12 2008-10-23 Piramed Limited Pharmaceutical compounds
WO2009010530A1 (en) 2007-07-18 2009-01-22 Novartis Ag Bicyclic heteroaryl compounds and their use as kinase inhibitors
WO2009026345A1 (en) 2007-08-20 2009-02-26 Targegen Inc. Thiazolidinone compounds, and methods of making and using same
WO2009071888A1 (en) 2007-12-04 2009-06-11 Ucb Pharma S.A. Pyrrolothiazoles as pi3-kinase inhibitors
WO2009071895A1 (en) 2007-12-04 2009-06-11 Ucb Pharma S.A. Fused thiazole and thiophene derivatives as kinase inhibitors
WO2009071890A1 (en) 2007-12-04 2009-06-11 Ucb Pharma S.A. Tricyclic kinase inhibitors
WO2010125082A1 (en) 2009-04-30 2010-11-04 Glaxo Group Limited Oxazole substituted indazoles as pi3-kinase inhibitors
WO2013037484A2 (en) 2011-09-12 2013-03-21 Genzyme Corporation Anti-aplhabetatcr antibody
WO2015066379A2 (en) 2013-10-30 2015-05-07 Genzyme Corporation Methods for enhancing immunosuppressive therapy by multiple administration of alpha beta tcr-binding polypeptide

Non-Patent Citations (83)

* Cited by examiner, † Cited by third party
Title
AIFANTIS I; RAETZ E; BUONAMICI S.: "Molecular pathogenesis of T-cell leukaemia and lymphoma", NATURE REVIEWS, vol. 8, no. 5, May 2008 (2008-05-01), pages 380 - 90
ALEGRE ET AL., J. IMMUNOL., vol. 148, no. 11, 1992, pages 3461 - 8
ANTIBODY ENGINEERING: METHODS AND PROTOCOLS, 2004
ASNAFI V; BELDJORD K; BOULANGER E; COMBA B; LE TUTOUR P; ESTIENNE MH ET AL.: "Analysis of TCR, pT alpha, and RAG-1 in T-acute lymphoblastic leukemias improves understanding of early human T-lymphoid lineage commitment.", BLOOD, vol. 101, no. 7, 1 April 2003 (2003-04-01), pages 2693 - 703
ASNAFI V; BELDJORD K; LIBURA M; VILLARESE P; MILLIEN C; BALLERINI P ET AL.: "Age-related phenotypic and oncogenic differences in T-cell acute lymphoblastic leukemias may reflect thymic atrophy", BLOOD, vol. 104, no. 13, 15 December 2004 (2004-12-15), pages 4173 - 80
ASNAFI V; BUZYN A; LE NOIR S; BALEYDIER F; SIMON A; BELDJORD K ET AL.: "NOTCH 1/FBXW7 mutation identifies a large subgroup with favorable outcome in adult T-cell acute lymphoblastic leukemia (T-ALL): a Group for Research on Adult Acute Lymphoblastic Leukemia (GRAALL) study", BLOOD, vol. 113, no. 17, 23 April 2009 (2009-04-23), pages 3918 - 24
BALDWIN TA; HOGQUIST KA.: "Transcriptional analysis of clonal deletion in vivo", J IMMUNOL., vol. 179, no. 2, 15 July 2007 (2007-07-15), pages 837 - 44
BERGERON J; CLAPPIER E; RADFORD I; BUZYN A; MILLIEN C; SOLER G ET AL.: "Prognostic and oncogenic relevance of TLX1/HOX11 expression level in T-ALLs", BLOOD, vol. 110, no. 7, 1 October 2007 (2007-10-01), pages 2324 - 30
BRABB T; HUSEBY ES; MORGAN TM; SANT'ANGELO DB; KIRCHNER J; FARR AG ET AL.: "Thymic stromal organization is regulated by the specificity of T cell receptor/major histocompatibility complex interactions", EUROPEAN JOURNAL OF IMMUNOLOGY, vol. 27, no. 1, January 1997 (1997-01-01), pages 136 - 46
CAMPANA D ET AL: "Stages of T-cell receptor protein expression in T-cell acute lymphoblastic leukemia.", BLOOD 1 APR 1991, vol. 77, no. 7, 1 April 1991 (1991-04-01), pages 1546 - 1554, XP002755663, ISSN: 0006-4971 *
CARRON C; CORMIER F; JANIN A ET AL.: "TEL-JAK2 transgenic mice develop T-cell leukemia", BLOOD, vol. 95, 2000, pages 3891 - 3899
CARRON C; CORMIER F; JANIN A; LACRONIQUE V; GIOVANNINI M; DANIEL MT ET AL.: "TEL-JAK2 transgenic mice develop T-cell leukemia", BLOOD, vol. 95, no. 12, 15 June 2000 (2000-06-15), pages 3891 - 9
CHEN ET AL., EMBO J., vol. 12, 1993, pages 821 - 830
CHEN ET AL., INTERNATIONAL IMMUNOLOGY, vol. 5, 1993, pages 647 - 656
CHOI ET AL., NATURE GENETICS, vol. 4, 1993, pages 117 - 123
CLAPPIER E; GERBY B; SIGAUX F; DELORD M; TOUZRI F; HERNANDEZ L ET AL.: "Clonal selection in xenografted human T cell acute lymphoblastic leukemia recapitulates gain of malignancy at relapse", THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 208, no. 4, 11 April 2011 (2011-04-11), pages 653 - 61
CUI Y; ONOZAWA M; GARBER HR; SAMSEL L; WANG Z; MCCOY JP ET AL.: "Thymic expression of a T cell receptor targeting a tumor associated antigen co-expressed in the thymus induces T-ALL", BLOOD, 26 March 2015 (2015-03-26)
DADI S; LE NOIR S; PAYET-BORNET D; LHERMITTE L; ZACARIAS-CABEZA J; BERGERON J ET AL.: "TLX homeodomain oncogenes mediate T cell maturation arrest in T-ALL via interaction with ETS and suppression of TCRalpha gene expression", CANCER CELL., vol. 21, no. 4, 17 April 2012 (2012-04-17), pages 563 - 76
DAIL M; WONG J; LAWRENCE J; O'CONNOR D; NAKITANDWE J; CHEN SC ET AL.: "Loss of oncogenic Notchl with resistance to a PI3K inhibitor in T-cell leukaemia.", NATURE, vol. 513, no. 7519, 25 September 2014 (2014-09-25), pages 512 - 6
DANIELS MA; TEIXEIRO E; GILL J; HAUSMANN B; ROUBATY D; HOLMBERG K ET AL.: "Thymic selection threshold defined by compartmentalization of Ras/MAPK signalling", NATURE, vol. 444, no. 7120, 7 December 2006 (2006-12-07), pages 724 - 9
DATABASE EMBASE [online] ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL; January 2011 (2011-01-01), REICHERT J M: "Antibody-based therapeutics to watch in 2011", XP002752847, Database accession no. EMB-2011033057 *
DATABASE MEDLINE [online] US NATIONAL LIBRARY OF MEDICINE (NLM), BETHESDA, MD, US; June 2014 (2014-06-01), SHIHEIDO HIROKAZU ET AL: "Novel CD3-specific antibody induces immunosuppression via impaired phosphorylation of LAT and PLC[gamma]1 following T-cell stimulation.", XP002738303, Database accession no. NLM24595757 *
DAVIGNON ET AL., J. IMMUNOL, vol. 141, no. 6, 1988, pages 1848 - 54
DAVIS RE; NGO VN; LENZ G; TOLAR P; YOUNG RM; ROMESSER PB ET AL.: "Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma", NATURE, vol. 463, no. 7277, 7 January 2010 (2010-01-07), pages 88 - 92
DE KEERSMAECKER K.: "Novel insights on TLX1 function in T-ALL pave the way towards differentiation therapy", HAEMATOLOGICA, vol. 97, no. 6, June 2012 (2012-06-01), pages 795
DIK WA; PIKE-OVERZET K; WEERKAMP F; DE RIDDER D; DE HAAS EF; BAERT MR ET AL.: "New insights on human T cell development by quantitative T cell receptor gene rearrangement studies and gene expression profiling", THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 201, no. 11, 6 June 2005 (2005-06-06), pages 1715 - 23
DOS SANTOS NR; RICKMAN DS; DE REYNIES A; CORMIER F; WILLIAME M; BLANCHARD C ET AL.: "Pre-TCR expression cooperates with TEL-JAK2 to transform immature thymocytes and induce T-cell leukemia", BLOOD, vol. 109, no. 9, 1 May 2007 (2007-05-01), pages 3972 - 81
DOS SANTOS NR; RICKMAN DS; DE REYNIES A; CORMIER F; WILLIAME M; BLANCHARD C; STERN MH; GHYSDAEL J.: "Pre-TCR expression cooperates with TEL-JAK2 to transform immature thymocytes and induce T-cell leukemia", BLOOD, vol. 109, 2007, pages 3972 - 81
FERRANDO AA; NEUBERG DS; STAUNTON J; LOH ML; HUARD C; RAIMONDI SC ET AL.: "Gene expression signatures define novel oncogenic pathways in T cell acute lymphoblastic leukemia.", CANCER CELL, vol. 1, no. 1, February 2002 (2002-02-01), pages 75 - 87
FISHWILD ET AL., NATURE BIOTECHNOLOGY, vol. 14, 1996, pages 845 - 851
FRENKEN ET AL., TRANSPLANTATION, vol. 51, no. 4, 1991, pages 881 - 7
GARRAWAY LA; SELLERS WR.: "Lineage dependency and lineage-survival oncogenes in human cancer", NAT REV CANCER, vol. 6, no. 8, August 2006 (2006-08-01), pages 593 - 602
GASCOIGNE NR; PALMER E.: "Signaling in thymic selection.", CURR OPIN IMMUNOL., vol. 23, no. 2, April 2011 (2011-04-01), pages 207 - 12
GENG H; HURTZ C; LENZ KB; CHEN Z; BAUMJOHANN D; THOMPSON S ET AL.: "Self-Enforcing Feedback Activation between BCL6 and Pre-B Cell Receptor Signaling Defines a Distinct Subtype of Acute Lymphoblastic Leukemia", CANCER CELL, vol. 27, no. 3, 9 March 2015 (2015-03-09), pages 409 - 25
GRAMATZKI M ET AL: "Therapy with OKT3 monoclonalantibody in refractory T cell acutelymphoblastic leukemia induces interleukin-2 responsiveness", LEUKEMIA, MACMILLAN PRESS LTD, US, vol. 9, no. 3, 1 March 1995 (1995-03-01), pages 382 - 390, XP008175816, ISSN: 0887-6924 *
GRAMATZKI M; BURGER R; STROBEL G; TRAUTMANN U; BARTRAM CR; HELM G ET AL.: "Therapy with OKT3 monoclonal antibody in refractory T cell acute lymphoblastic leukemia induces interleukin-2 responsiveness", LEUKEMIA, vol. 9, no. 3, March 1995 (1995-03-01), pages 382 - 90
GRAMATZKI M; BURGER R; STROBEL G; TRAUTMANN U; BARTRAM CR; HELM G; HORNEFFG; ALSALAMEH S; JONKER M; GEBHART E ET AL.: "Therapy with OKT3 monoclonal antibody in refractory T cell acute lymphoblastic leukemia induces interleukin-2 responsiveness", LEUKEMIA, 1995
GRAUX C ET AL: "Cytogenetics and molecular genetics of T-cell acute lymphoblastic leukemia: from thymocyte to lymphoblast.", LEUKEMIA SEP 2006, vol. 20, no. 9, September 2006 (2006-09-01), pages 1496 - 1510, XP002738304, ISSN: 0887-6924 *
GRAUX C; COOLS J; MICHAUX L; VANDENBERGHE P; HAGEMEIJER A.: "Cytogenetics and molecular genetics of T-cell acute lymphoblastic leukemia: from thymocyte to lymphoblast", LEUKEMIA, vol. 20, no. 9, September 2006 (2006-09-01), pages 1496 - 510
HARDING; LONBERG, ANN. N Y. ACAD. SCI, vol. 764, 1995, pages 536 - 546
HEROLD ET AL., NEJM, vol. 346, no. 22, 2002, pages 1692 - 1698
HOGQUIST KA; JAMESON SC.: "The self-obsession of T cells: how TCR signaling thresholds affect fate 'decisions' and effector function", NATURE IMMUNOLOGY, vol. 15, no. 9, September 2014 (2014-09-01), pages 815 - 23
HUGUET F; LEGUAY T; RAFFOUX E; THOMAS X; BELDJORD K; DELABESSE E ET AL.: "Pediatric-inspired therapy in adults with Philadelphia chromosome-negative acute lymphoblastic leukemia: the GRAALL-2003 study", J CLIN ONCOL., vol. 27, no. 6, 20 February 2009 (2009-02-20), pages 911 - 8
KELLY JA; SPOLSKI R; KOVANEN PE; SUZUKI T; BOLLENBACHER J; PISE-MASISON CA ET AL.: "Stat5 synergizes with T cell receptor/antigen stimulation in the development of lymphoblastic lymphoma", THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 198, no. 1, 7 July 2003 (2003-07-07), pages 79 - 89
KING B; TRIMARCHI T; REAVIE L; XU L; MULLENDERS J; NTZIACHRISTOS P ET AL.: "The ubiquitin ligase FBXW7 modulates leukemia-initiating cell activity by regulating MYC stability", CELL, vol. 153, no. 7, 20 June 2013 (2013-06-20), pages 1552 - 66
KLEIN L; KYEWSKI B; ALLEN PM; HOGQUIST KA.: "Positive and negative selection of the T cell repertoire: what thymocytes see (and don't see", NATURE REVIEWS, vol. 14, no. 6, June 2014 (2014-06-01), pages 377 - 91
KLINGER MB; GUILBAULT B; GOULDING RE; KAY RJ.: "Deregulated expression of RasGRP1 initiates thymic lymphomagenesis independently of T-cell receptors", ONCOGENE, vol. 24, no. 16, 14 April 2005 (2005-04-14), pages 2695 - 704
KUROIWA ET AL., NATURE BIOTECHNOLOGY, vol. 20, 2002, pages 889 - 894
LANTZ 0; GRANDJEAN I; MATZINGER P; DI SANTO JP: "Gamma chain required for naive CD4+ T cell survival but not for antigen proliferation.", NATURE IMMUNOLOGY, vol. L, no. L, July 2000 (2000-07-01), pages 54 - 8
LANTZ 0; GRANDJEAN I; MATZINGER P; DI SANTO JP: "Gamma chain required for naive CD4T cell survival but not for antigen proliferation", NAT IMMUNOL, vol. 1, 2000, pages 54 - 58
LISTON A; HARDY K; PITTELKOW Y; WILSON SR; MAKAROFF LE; FAHRER AM ET AL.: "Impairment of organ-specific T cell negative selection by diabetes susceptibility genes: genomic analysis by mRNA profiling", GENOME BIOLOGY., vol. 8, no. 1, 2007, pages R12
LONBERG ET AL., NATURE, vol. 368, no. 6474, 1994, pages 856 - 859
LONBERG, N.; HUSZAR, D., INTERN. REV. IMMUNOL, vol. 13, 1995, pages 65 - 93
LONBERG: "Handbook of Experimental Pharmacology", vol. 113, 1994, pages: 49 - 101
MARTIN A; TISCH RM; GETTS DR.: "Manipulating T cell-mediated pathology: targets and functions of monoclonal antibody immunotherapy", CLINICAL IMMUNOLOGY, vol. 148, no. 1, July 2013 (2013-07-01), pages 136 - 47
MORRIS GP; ALLEN PM.: "How the TCR balances sensitivity and specificity for the recognition of self and pathogens", NATURE IMMUNOLOGY, vol. 13, no. 2, February 2012 (2012-02-01), pages 121 - 8
NICOLLS ET AL., TRANSPLANTATION, vol. 55, 1993, pages 459 - 468
NTZIACHRISTOS P; TSIRIGOS A; VAN VLIERBERGHE P; NEDJIC J; TRIMARCHI T; FLAHERTY MS ET AL.: "Genetic inactivation of the polycomb repressive complex 2 in T cell acute lymphoblastic leukemia", NATURE MEDICINE, vol. 18, no. 2, February 2012 (2012-02-01), pages 298 - 301
PIOVAN E; YU J; TOSELLO V; HERRANZ D; AMBESI-IMPIOMBATO A; DA SILVA AC ET AL.: "Direct reversal of glucocorticoid resistance by AKT inhibition in acute lymphoblastic leukemia", CANCER CELL, vol. 24, no. 6, 9 December 2013 (2013-12-09), pages 766 - 76
PRASAD A; ZIKHERMAN J; DAS J; ROOSE JP; WEISS A; CHAKRABORTY AK.: "Origin of the sharp boundary that discriminates positive and negative selection of thymocytes", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 106, no. 2, 13 January 2009 (2009-01-13), pages 528 - 33
PUI CH; EVANS WE.: "Treatment of acute lymphoblastic leukemia.", THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 354, no. 2, 12 January 2006 (2006-01-12), pages 166 - 78
REICHERT J M: "Antibody-based therapeutics to watch in 2011", MABS 2011 LANDES BIOSCIENCE USA, vol. 3, no. 1, January 2011 (2011-01-01), pages 76 - 99, ISSN: 1942-0862 *
ROTI G; STEGMAIER K.: "New Approaches to Target T-ALL.", FRONT ONCOL, vol. 4, 8 July 2014 (2014-07-08), pages 170
SANDA T; TYNER JW; GUTIERREZ A; NGO VN; GLOVER J; CHANG BH ET AL.: "TYK2-STAT1-BCL2 pathway dependence in T-cell acute lymphoblastic leukemia", CANCER DISCOV, vol. 3, no. 5, May 2013 (2013-05-01), pages 564 - 77
SCHMITZ I; CLAYTON LK; REINHERZ EL.: "Gene expression analysis of thymocyte selection in vivo.", INTERNATIONAL IMMUNOLOGY., vol. 15, no. 10, October 2003 (2003-10-01), pages 1237 - 48
SCHWARZ BA; SAMBANDAM A; MAILLARD I; HARMAN BC; LOVE PE; BHANDOOLA A: "Selective thymus settling regulated by cytokine and chemokine receptors", J IMMUNOL., vol. 178, no. 4, 15 February 2007 (2007-02-15), pages 2008 - 17
SHIHEIDO HIROKAZU ET AL: "Novel CD3-specific antibody induces immunosuppression via impaired phosphorylation of LAT and PLC[gamma]1 following T-cell stimulation.", EUROPEAN JOURNAL OF IMMUNOLOGY JUN 2014, vol. 44, no. 6, June 2014 (2014-06-01), pages 1770 - 1780, ISSN: 1521-4141 *
SMITH SL.: "Ten years of Orthoclone OKT3 (muromonab-CD3): a review", J TRANSPL COORD., vol. 6, no. 3, September 1996 (1996-09-01), pages 109 - 19,20-1
SOULIER J; CLAPPIER E; CAYUELA JM; REGNAULT A; GARCIA-PEYDRO M; DOMBRET H ET AL.: "HOXA genes are included in genetic and biologic networks defining human acute T-cell leukemia (T-ALL).", BLOOD, vol. 106, no. 1, 1 July 2005 (2005-07-01), pages 274 - 86
STRZADALA L; MIAZEK A; MATUSZYK J; KISIELOW P.: "Role of thymic selection in the development of thymic lymphomas in TCR transgenic mice.", INTERNATIONAL IMMUNOLOGY, vol. 9, no. 1, January 1997 (1997-01-01), pages 127 - 38
TANAKA ET AL., J. IMMUNOL., vol. 142, 1989, pages 2791 - 2795
TAYLOR ET AL., INTERNATIONAL IMMUNOLOGY, vol. 6, 1994, pages 579 - 591
TAYLOR ET AL., NUCLEIC ACIDS RESEARCH, vol. 20, 1992, pages 6287 - 6295
TOMIZUKA ET AL., PROC. NATL. ACAD. SET USA, vol. 97, 2000, pages 722 - 727
TRINQUAND A; TANGUY-SCHMIDT A; BEN ABDELALI R; LAMBERT J; BELDJORD K; LENGLINE E ET AL.: "Toward a NOTCHl/FBXW7/RAS/PTEN-based oncogenetic risk classification of adult T-cell acute lymphoblastic leukemia: a Group for Research in Adult Acute Lymphoblastic Leukemia study", J CLIN ONCOL., vol. 31, no. 34, 1 December 2013 (2013-12-01), pages 4333 - 42
TUAILLON ET AL., J. IMMUNOL., vol. 152, 1994, pages 2912 - 2920
TUAILLON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 3720 - 3724
VAHID ASNAFI; KHEIRA BELDJORD; MARTA LIBURA; PATRICK VILLARESE; CORRINE MILLIEN; PAOLA BALLERINI; EMILIENNE KUHLEIN; MARINA LAFAGE: "Age-related phenotypic and oncogenic differences in T-cell acute lymphoblastic leukemias may reflect thymic atrophy", 2004, THE AMERICAN SOCIETY OF HEMATOLOGY
VAN VLIERBERGHE P; FERRANDO A.: "The molecular basis of T cell acute lymphoblastic leukemia", THE JOURNAL OF CLINICAL INVESTIGATION., vol. 122, no. 10, October 2012 (2012-10-01), pages 3398 - 406
VON BOEHMER H.: "Deciphering thymic development.", FRONT IMMUNOL., vol. 5, 2014, pages 424
WENG AP; FERRANDO AA; LEE W; MORRIS JPT; SILVERMAN LB; SANCHEZ-IRIZARRY C ET AL.: "Activating mutations of NOTCH in human T cell acute lymphoblastic leukemia", SCIENCE (NEW YORK, NY., vol. 306, no. 5694, 8 October 2004 (2004-10-08), pages 269 - 71
YUI MA; ROTHENBERG EV.: "Developmental gene networks: a triathlon on the course to T cell identity", NATURE REVIEWS., vol. 14, no. 8, August 2014 (2014-08-01), pages 529 - 45
ZHANG J; DING L; HOLMFELDT L; WU G; HEATLEY SL; PAYNE-TURNER D ET AL.: "The genetic basis of early T-cell precursor acute lymphoblastic leukaemia", NATURE, vol. 481, no. 7380, 12 January 2012 (2012-01-12), pages 157 - 63

Cited By (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10765741B2 (en) 2011-05-09 2020-09-08 Mayo Foundation For Medical Education And Research Methods for treating VEGF-expressing cancer using preformed nanoparticle complexes comprising albumin-bound paclitaxel and bevacizumab
US10376580B2 (en) 2012-10-01 2019-08-13 Mayo Foundation For Medical Education And Research Methods of treating cancer with antibody-albumin nanoparticle complexes comprising albumin, trastuzumab, and paclitaxel
US10420839B2 (en) 2012-10-01 2019-09-24 Mayo Foundation For Medical Education And Research Methods for treating CD52-expressing cancers using compositions comprising nanoparticle complexes of paclitaxel, alemtuzumab, and albumin
US10376579B2 (en) 2012-10-01 2019-08-13 Mayo Foundation For Medical Education And Research Nanoparticle complexes of albumin, paclitaxel, and anti-VEGF antibody for treatment of cancer
US11648311B2 (en) 2012-10-01 2023-05-16 Mayo Foundation For Medical Education And Research Cancer treatments
US10668151B2 (en) 2012-10-01 2020-06-02 Mayo Foundation For Medical Education And Research Nanoparticle complexes of albumin, paclitaxel, and panitumumab for treatment of cancer
US10478495B2 (en) 2012-10-01 2019-11-19 Mayo Foundation For Medical Education And Research Methods for treating cancer using nanoparticle complexes of paclitaxel, cetuximab, and albumin
US10471145B2 (en) 2012-10-01 2019-11-12 Mayo Foundation For Medical Education And Research Methods for treating cancer using nanoparticle complexes of paclitaxel, rituximab, and albumin
US10406224B2 (en) 2012-10-01 2019-09-10 Mayo Foundation For Medical Education And Research Nanoparticle complexes of paclitaxel, trastuzumab, and albumin
US10279036B2 (en) 2012-10-01 2019-05-07 Mayo Foundation For Medical Education And Research Antibody-albumin nanoparticle complexes comprising albumin, bevacizumab, and paclitaxel, and methods of making and using the same
US10307482B2 (en) 2012-10-01 2019-06-04 Mayo Foundation For Medical Education And Research Nanoparticle complexes of paclitaxel, cetuximab, and albumin
US10493150B2 (en) 2012-10-01 2019-12-03 Mayo Foundation For Medical Education And Research Nanoparticle complexes of paclitaxel, alemtuzumab, and albumin
US10413606B2 (en) 2012-10-01 2019-09-17 Mayo Foundation For Medical Education And Research Methods for treating cancer with nanoparticle complexes of albumin-bound paclitaxel and anti-VEGF antibodies
US10507243B2 (en) 2012-10-01 2019-12-17 Mayo Foundation For Medical Education And Research Nanoparticle complexes of rituximab, albumin and pacltaxel
US10279035B2 (en) 2012-10-01 2019-05-07 Mayo Foundation For Medical Education And Research Nanoparticle complexes of paclitaxel, trastuzumab, and albumin
US10441656B2 (en) 2012-10-01 2019-10-15 Mayo Foundation For Medical Education And Research Nanoparticle complexes of paclitaxel, cetuximab, and albumin, and methods of making the same
US10213513B2 (en) 2014-06-16 2019-02-26 Mayo Foundation For Medical Education And Research Treating myelomas
US11285221B2 (en) 2014-06-16 2022-03-29 Mayo Foundation For Medical Education And Research Treating myelomas
US10624846B2 (en) 2014-10-06 2020-04-21 Mayo Foundation For Medical Education And Research Lyophilized compositions comprising albumin-antibody paclitaxel nanoparticle complexes
US10993912B2 (en) 2014-10-06 2021-05-04 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10300016B2 (en) 2014-10-06 2019-05-28 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10596112B2 (en) 2014-10-06 2020-03-24 Mayo Foundation For Medical Education And Research Methods of using albumin-antibody nanoparticle complex compositions for treating cancer
US10596111B2 (en) 2014-10-06 2020-03-24 Mayo Foundation For Medical Education And Research Methods of making lyophilized compositions comprising albumin-VEGF paclitaxel nanoparticle complexes
US11433023B2 (en) 2014-10-06 2022-09-06 Mayo Foundation For Medical Education And Research Albumin-PD-1 paclitaxel nanoparticle complex compositions and methods of making and using the same
US10993911B2 (en) 2014-10-06 2021-05-04 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10391055B2 (en) 2014-10-06 2019-08-27 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10322084B2 (en) 2014-10-06 2019-06-18 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10772833B2 (en) 2014-10-06 2020-09-15 Mayo Foundation For Medical Education And Research Albumin-CTLA-4 paclitaxel nanopartilce complex compositions and methods of making and using the same
US10780049B2 (en) 2014-10-06 2020-09-22 Mayo Foundation For Medical Education And Research Lyophilized compositions comprising albumin-rankl or albumin-GD2 paclitaxel nanoparticle complexes
US10780050B2 (en) 2014-10-06 2020-09-22 Mayo Foundation For Medical Education And Research Lyophilized compositions comprosing albumin-EGFR paclitaxel nanoparticle complexes
US10966923B2 (en) 2014-10-06 2021-04-06 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US11241387B2 (en) 2015-08-18 2022-02-08 Mayo Foundation For Medical Education And Research Carrier-binding agent compositions and methods of making and using the same
US11660339B2 (en) 2015-10-06 2023-05-30 Mayo Foundation For Medical Education And Research Methods of treating cancer using compositions of antibodies and carrier proteins with antibody pretreatment
US10561726B2 (en) 2015-10-06 2020-02-18 Vavotar Life Sciences LLC Methods of treating cancer using compositions of antibodies and carrier proteins with antibody pretreatment
US11571469B2 (en) 2016-01-07 2023-02-07 Mayo Foundation For Medical Education And Research Methods of treating cancer with interferon wherein the cancer cells are HLA negative or have reduced HLA expression
US11305020B2 (en) 2016-03-21 2022-04-19 Mayo Foundation For Medical Education And Research Methods for reducing toxicity of a chemotherapeutic drug
US11878061B2 (en) 2016-03-21 2024-01-23 Mayo Foundation For Medical Education And Research Methods for improving the therapeutic index for a chemotherapeutic drug
US10618969B2 (en) 2016-04-06 2020-04-14 Mayo Foundation For Medical Education And Research Carrier-binding agent compositions and methods of making and using the same
US11655301B2 (en) 2016-04-06 2023-05-23 Mayo Foundation For Medical Education And Research Carrier-binding agent compositions and methods of making and using the same
US11160876B2 (en) 2016-09-01 2021-11-02 Mayo Foundation For Medical Education And Research Methods and compositions for targeting t-cell cancers
CN109843336A (en) * 2016-09-01 2019-06-04 梅约医学教育与研究基金会 Method and composition for targeting T-cells cancer
WO2018045238A1 (en) * 2016-09-01 2018-03-08 Mayo Foundation For Medical Education And Research Methods and compositions for targeting t-cell cancers
US11548946B2 (en) 2016-09-01 2023-01-10 Mayo Foundation For Medical Education And Research Carrier-PD-L1 binding agent compositions for treating cancers
JP2019526579A (en) * 2016-09-01 2019-09-19 マヨ ファウンデーション フォー メディカル エデュケーション アンド リサーチMayo Foundation For Medical Education And Research Methods and compositions for targeting T cell cancer
US11590098B2 (en) 2016-09-06 2023-02-28 Mayo Foundation For Medical Education And Research Methods of treating triple-negative breast cancer using compositions of antibodies and carrier proteins
US11427637B2 (en) 2016-09-06 2022-08-30 Mayo Foundation For Medical Education And Research Methods of treating PD-L1 expressing cancer
US11872205B2 (en) 2016-09-06 2024-01-16 Mayo Foundation For Medical Education And Research Methods of treating triple-negative breast cancer using compositions of antibodies and carrier proteins
US11311631B2 (en) 2016-09-06 2022-04-26 Mayo Foundation For Medical Education And Research Paclitaxel-albumin-binding agent compositions and methods for using and making the same
WO2021064069A1 (en) * 2019-10-02 2021-04-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of adult t-cell leukemia/lymphoma
US20220389104A1 (en) * 2021-05-28 2022-12-08 Ose Immunotherapeutics Method for Treating CD127-Positive Cancers by Administering an Anti-CD127 Agent

Similar Documents

Publication Publication Date Title
WO2016059220A1 (en) Tcr-activating agents for use in the treatment of t-all
US11207393B2 (en) Regulatory T cell PD-1 modulation for regulating T cell effector immune responses
JP2021103174A (en) Biomarkers and combination therapies using oncolytic virus and immunomodulation
JP2019214589A (en) Combination of PD-1 antagonist and IDO1 inhibitor for treating cancer
US20150110779A1 (en) Methods for predicting gastrointestinal immune-related adverse events (gi-irae) in patients treated with modulation of the co-stimulatory pathway
CN115569192A (en) Treatment of cancer with dual targeting of CD47 and EGFR
JP2022513685A (en) Methods for Treatment with Adoptive Cell Therapy
JP7308190B2 (en) Treatment of ovarian cancer with anti-CD47 and anti-PD-L1
JP2019506400A (en) Treatment of cancer using an immunomodulator
CN112703012A (en) Methods of treating cancer
KR102416144B1 (en) Methods of Predicting Therapeutic Benefit of Anti-CD19 Therapy in Patients
US20210115127A1 (en) Anti-il-27 antibodies and uses thereof
EP3779447B1 (en) Method to activate the anti-tumoral cd8+t cell response of a patient affected with a cancer
Stone et al. TNF blockade uncouples toxicity from antitumor efficacy induced with CD40 chemoimmunotherapy
US10746726B2 (en) Method for assessing therapeutic effect of anti-cancer agent having anti-CD4 antibody as active ingredient
JP2022500378A (en) Treatment of cancer by inhibiting the ubiquitin-conjugating enzyme E2K (UBE2K)
WO2019234221A1 (en) Methods for stratification and treatment of a patient suffering from chronic lymphocytic leukemia
US20180028566A1 (en) Gamma delta t cells as a target for treatment of solid tumors
US11168134B2 (en) Methods of treating androgen deprivation therapy resistant prostate cancer
US20230037414A1 (en) Inhibitors of adrenomedullin for the treatment of acute myeloid leukemia by eradicating leukemic stem cells
US20220290151A1 (en) Use of müllerian inhibiting substance inhibitors for treating cancer
US9868792B2 (en) Methods of enhancing anti-tumor immunity by administering antibodies to the CCRL2 chemerin receptor
Jimenez et al. CD40 is an immune checkpoint regulator that potentiates myocardial inflammation through activation and expansion of CCR2+ macrophages and CD8 T-cells
KR20230044315A (en) Methods and compositions for evaluating and treating pancreatic cancer

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: 15784322

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: 15784322

Country of ref document: EP

Kind code of ref document: A1