WO1993019163A1 - Genes de recepteurs chimeres et cellules transformees au moyen desdits genes - Google Patents

Genes de recepteurs chimeres et cellules transformees au moyen desdits genes Download PDF

Info

Publication number
WO1993019163A1
WO1993019163A1 PCT/US1993/002506 US9302506W WO9319163A1 WO 1993019163 A1 WO1993019163 A1 WO 1993019163A1 US 9302506 W US9302506 W US 9302506W WO 9319163 A1 WO9319163 A1 WO 9319163A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
chimeric
cell
receptor
chain
Prior art date
Application number
PCT/US1993/002506
Other languages
English (en)
Inventor
Zelig Eshhar
Daniel Schindler
Tova Waks
Gideon Gross
Original Assignee
Yeda Research And Development Co, Ltd.
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
Priority claimed from IL101288A external-priority patent/IL101288A/en
Application filed by Yeda Research And Development Co, Ltd. filed Critical Yeda Research And Development Co, Ltd.
Priority to CA002132349A priority Critical patent/CA2132349C/fr
Priority to AU39243/93A priority patent/AU668156B2/en
Priority to EP93908414A priority patent/EP0638119B1/fr
Priority to DE69333038T priority patent/DE69333038T2/de
Priority to JP51673193A priority patent/JP3643590B2/ja
Priority to AT93908414T priority patent/ATE242800T1/de
Priority to US08/084,994 priority patent/US7741465B1/en
Publication of WO1993019163A1 publication Critical patent/WO1993019163A1/fr
Priority to US08/547,263 priority patent/US8211422B2/en
Priority to US13/281,560 priority patent/US20120093842A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70535Fc-receptors, e.g. CD16, CD32, CD64 (CD2314/705F)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7155Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for interleukins [IL]
    • 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
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/42Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins
    • C07K16/4283Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig
    • C07K16/4291Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig against IgE
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/44Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material not provided for elsewhere, e.g. haptens, metals, DNA, RNA, amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/033Fusion polypeptide containing a localisation/targetting motif containing a motif for targeting to the internal surface of the plasma membrane, e.g. containing a myristoylation motif

Definitions

  • the present invention relates to chimeric receptor genes suitable for endowing lymphocytes with antibody-type specificity, to expression vectors comprising said chimeric genes and to lymphocytes transformed with said expression vectors.
  • Various types of lymphocyte cells are suitable, for example, cytotoxic T cells, helper T cells, natural killer (NK) cells, etc.
  • the transformed lymphocytes are useful in therapeutic treatment methods.
  • TCR/CD3 antigen-specific T cell receptor complex
  • TCR T cell receptor for antigen
  • MHC major histocompatibility complex
  • CD3 CD3 is assumed to be responsible for incracellular signalling following occupancy of the TCR by ligand.
  • TCR/CD3 The T cell receptor for antigen-CD3 complex (TCR/CD3) recognizes antigenic peptides that are presented to it by the proteins of the major histocompatibility complex (MHC) . Complexes of MHC and pept-ide are expressed on the surface of antigen presenting cells and other T cell targets. Stimulation of the TCR/CD3 complex results in activation of the T cell and a consequent antigen-specific immune response.
  • the TCR/CD3 complex plays a central role in the effector function and regulation of the immune system.
  • Two forms of T cell receptor for antigen are expressed on the surface of T cells. These contain either a/ ⁇ heterodimers or - ⁇ / S heterodimers.
  • T cells are capable of rearranging the genes that encode the a, ⁇ , 7 and S chains of the T cell receptor.
  • T cell receptor gene rearrangements are analogous to those that produce functional immunoglobulins in ⁇ cells and the presence of multiple variable and joining regions in the genome allows the generation of T cell receptor with a diverse range of binding specificities.
  • Each a/ ⁇ or / ⁇ heterodimer is expressed on the surface of the T cell in association with four invariant peptides. These are the 7 , 5 and ⁇ subunits of the CD3 complex and the zeta chain.
  • the CD3 7. and ⁇ polypeptides are encoded by three members of the immunoglobulin supergene family and are found in a cluster on human chromosome 11 or murine chromosome 9.
  • the zeta chain gene is found separately" from other TCR and CD3 genes on chromosome 1 in both the mouse and human.
  • Murine T cells are able to generate a receptor-associated f chain through alternative splicing of the zeta m-RNA transcript.
  • the CD3 chains and the zeca subunic do not show variability, and are not involved directly in antigen recognitio .
  • T cell receptor All the components of the T cell receptor are membrane proteins and consist of a leader sequence, externally- disposed N-terminal extracellular domains, a single membrane- spanning domain, and cytoplasmic tails.
  • the a , ⁇ , 7 and 5 antigen-binding polypeptides are glycoproteins.
  • the zeta chain has a relatively short ectodomain of only nine amino acids and a long cytoplasmic tail of approximately 110 amino acids.
  • Most T cell receptor a ⁇ heterodimers are covalently linked through disulphi e binds, but many 7 ⁇ receptors associate with one another non-covalentl .
  • the zeta chain quantitatively forms either disulphide-linked ⁇ - ⁇ heterodimers or zeta-zeta homodimers.
  • Another example of a type of receptor on cells of the immune system is the Fc receptor.
  • the interaction of antibody- antigen complexes with cells of the immune system results in a wide array of responses, ranging from effector functions such as antibody-dependent cytotoxicity, mast cell degranulation, and phagocytosis to immunomodulatory signals such as regulating lymphocyte proliferation and antibody secretion. All these interactions are initiated through the binding of the Fc domain of antibodies or immune complexes to specialized cell surface receptors on hematopoietic cells. It is now well established that the diversity of cellular responses triggered by antibodies and immune complexes results from the structural heterogeneity of Fc receptors.
  • FcRs are defined by their specificity for immunoglobulin isotypes.
  • Fc receptors for IgG are referred to as FC7 , for IgE as Fc ⁇ R, for IgA as Fc ⁇ R, etc.
  • Structurally distinct receptors are distinguished by a Roman numeral, based on historical precedent.
  • FC7RS three groups of FC7RS, designated FC7RI. FC7RII, and FC7RIII.
  • Fc ⁇ RI and Fc ⁇ RlI Two groups of Fc ⁇ R have been defined; these are referred to as Fc ⁇ RI and Fc ⁇ RlI.
  • Structurally related although distinct genes within a group are denoted by A, E, C.
  • the protein subunit is given a Greek letter, such as Fc7RIIIA ⁇ , FC7RHIA7.
  • FC7R IgG and IgE Fc receptors
  • Fc receptors share structurally related ligand binding domains, but differ in their transmembrane and intracellular domains which presumably mediate intracellular signalling.
  • specific FC RS on different cells mediate different cellular responses upon interaction with an immune complex.
  • the structural analysis of the FC7RS and Fc ⁇ RI has also revealed at least one common subunit among some of these receptors. This common subunit is the 7 subunit, which is similar to the ! " or 11 chain of the TCR/CD3, and is involved in the signal transduction of the FC7RIH and Fc ⁇ RI.
  • the low affinity receptor for IgG (FC7RIIIA) is composed of the ligand binding CD16 ⁇ (Fc7RIIIA ⁇ -) polypeptide associated with the 7 chain (FC7RIHA7) .
  • the CD16 polypeptide appears as membrane anchored form in polymorphonuclear cells and as transmembrane form (CD16 IM ) in NK.
  • the FC7RIIIA serves as a triggering molecule for NK cells.
  • IL-2 receptor Another type of immune cell receptor is the IL-2 receptor. This receptor is composed of three chains, the a chain (p55) , the ⁇ chain (p75) and the 7 chain. When stimulated by IL-2, lymphocytes undergo proliferation and activation.
  • Antigen-specific effector lymphocytes such as tumor specific T cells (Tc)
  • Tc tumor specific T cells
  • Antibodies are readily obtainable, more easily derived, have wider spectrum and are not individual-specific.
  • the major problem of applying specific antibodies for cancer immunotherapy lies in the inability of sufficient amounts of monoclonal antibodies ( Ab) to reach large areas within solid tumors.
  • Ab monoclonal antibodies
  • many clinical attempts to recruit the humoral or cellular arms of the immune system for passive anti-tumor immunotherapy have not fulfilled expectations. While it has been possible to obtain anti-tumor antibodies, their therapeutic use has been limited so far to blood-borne tumors (1, 2) primarily because solid tumors are inaccessible to sufficient amounts of antibodies
  • genomi expression vectors have been constructed containing the rearranged gene segments coding for the V region domains of heavy (V H ) and light (V L ) chains of an anti-2,4,6- trinitrophenyl (TNP) antibody (Sp6) spliced to either one o the C-region gene segments of the a or ⁇ TCR chains.
  • TNP -2,4,6- trinitrophenyl
  • the transfectants specifically killed TNP-bearing target cells, and produced interleukin-2 (IL-2) in response thereto across strain and species barriers. Moreover, such transfectants responded t immobilized TNP-protein conjugates, bypassing the need for cellular processing and presentation.
  • the chimeric TCRs co provide T cells with an antibody-like specificity and, upon encountering antigen, were able to effectively transmit signals for T cell activation, secretion of lymphokines an specific target cell lysis in a MHC nonrestricted manner. Moreover, the cTCR bearing cells undergo stimulation by immobilized antigen, proving that receptor-mediated T-cell activation is not only nonrestricted but also independent o MHC expression on target cells (8, 9).
  • New expression cassettes were also developed based on reverse transcriptio mRNA and PCR amplification of rearranged V H and V L DNA, usi primers based on 3' and 5' consensus sequences (12) of these genes which allow rapid construction of cTCR genes from any mAb-producing hybridoma.
  • To determine the therapeutic potentia. 1 of the chimeric TCR approach we successfully constructed and functionally expressed cTCR genes composed of combining sites of anti-idiotypic antibody specific to the surface IgM of the 38C13 murine B lymphoma cell line.
  • retroviral vectors have been demonstrated to be effective for transgene expression in human T cells (13, 14), due to the fact that two genes have to be introduced in order to express functional cTCR (C ⁇ .V H +Q9V L or Co;V L ⁇ C ⁇ V E ) , and the very low efficiency of transduction of a single cell with two separate retroviral vectors, new vectors have to be tried which will allow the transduction of two genes in tandem (15) .
  • Another strategy which has recently been developed employs joining of the extracellular ligand binding domain of receptors such as CD4, CD8, the IL-2 receptor, or CD16, to the cytoplasmic tail of either one of the y/ ⁇ family members (26-28, 38) . It has been shown that crosslinking of such extracellular domains through a ligand or antibody results in T cell activation. Chimeric CD4 or CD16- ⁇ / ⁇ molecules expressed in cytotoxic lymphocytes could direct specific cytolysis against appropriate target cells (26, 38) .
  • receptors such as CD4, CD8, the IL-2 receptor, or CD16
  • scFv single-chain Fv domain
  • a chimeric gene which combines the antibody recognition site and the lymphocyte-signalling moiety into one continuous chain.
  • c-scFvR chimeric scFv-receptor
  • the present invention thus relates to chimeric genes suitable to endow lymphocyte cells with antibody-type specificity.
  • Various types of lymphocytes are suitable, for example, natural killer cells, helper T cells, suppressor T cells, cytotoxic T cells, lymphokine activated cells, subtypes thereof and any other cell type which can express chimeric receptor chain.
  • the chimeric gene comprises a first gene segment encoding the scFv of a specific antibody, i.e., DNA sequences encoding the variable regions of the heavy and light chains (V H and V L , respectively) of the specific antibody, linked by a flexible linker, and a second gene segment which comprises a DNA sequence encoding partially or entirely the transmembrane and cytoplasmic, and optionally the extracellular, domains of a lymphocyte-triggering molecule corresponding to a lymphocyte receptor or part thereof.
  • a first gene segment encoding the scFv of a specific antibody, i.e., DNA sequences encoding the variable regions of the heavy and light chains (V H and V L , respectively) of the specific antibody, linked by a flexible linker
  • V H and V L variable regions of the heavy and light chains
  • the present invention further relates to suitable vectors for transfecting cells of the type defined above with the chimeric gene.
  • the present invention further relates to cells of the type defined above into which such chimeric gene has been introduced so as to obtain its expression, and also to pharmaceutical prophylactic and curative compositions containing an effective quantity of such cells.
  • the present invention relates to a process for the generation of lymphocytes transfected with an expression vector containing a chimeric gene of the invention.
  • a model system which comprises an expression vector which was transfected into cytotoxic T cells and which was functionally expressed in said cells, i.e. r which directed the cellular response of the lymphocyte against a predefined target antigen in a MHC nonrestricted manner.
  • the genetically engineered lymphocyte cells of the present invention may be used in new therapeutic treatment processes.
  • T cells or NK cells isolated from a patient may be transfected with DNA encoding a chimeric gene including the variable region of an antibody directed toward a specific antigen, and then returned to the patient so that the cellular response generated by such cells will be triggered by and directed toward the specific antigen in a MHC nonrestricted manner.
  • peripheral blood cells of the patient are genetically engineered according to the invention and then administered to the patient.
  • the present invention allows us to confer antibody specificity using not only the TCR components, but other lymphocyte-signalling chains, such as the zeta/eta chains of CD3, 7 chain of the FC7R and Fc ⁇ R, a, ⁇ and 7 chains of the IL-2R or any other lymphokine receptor, CDl ⁇ a- chain, D2, CD28, and others.
  • lymphocyte-signalling chains such as the zeta/eta chains of CD3, 7 chain of the FC7R and Fc ⁇ R, a, ⁇ and 7 chains of the IL-2R or any other lymphokine receptor, CDl ⁇ a- chain, D2, CD28, and others.
  • Figure 1 depicts a scheme of the chimeric scFVR expression vector.
  • R represents any receptor chain, such as the zeta subunit of the CD3, gamma and CD16 ⁇ subunits of the FC7RIII, Co. and C ⁇ of the TCR, ⁇ chain of the IL-2 receptor or any other chain or part thereof described herein.
  • A depicts the preparation of the gene segments encoding the scFv of the V H and V L of a specific antibody linked by a flexible linker (hatched box) .
  • B represents the pRSV expression vector containing the kappa light chain leader (L j . ) , into which the receptor gene prepared from lymphocytes described in C and the gene segment of A are introduced.
  • LTR long terminal repeat
  • FIG. 2 illustrates the chimeric pRSVscFvR 7 expression vector obtained according to the scheme of Fig. 1.
  • the boxes from left to right represent DNA segments corresponding to the Rous sarcoma virus long terminal repeat promoter (LTR) , kappa light chain leader (L/c) and variable region (Vc) , the linker (hatched box) , heavy chain variable region (V H ) , the human gamma chain, the G418-resistance gene (neo r ) and the simian virus 40 origin of replication.
  • LTR Rous sarcoma virus long terminal repeat promoter
  • L/c kappa light chain leader
  • Vc variable region
  • Restriction sites indicated are EcoRI (RI) , SnaBI (Sn) , Ncol (N) , Xbal (Xb) , Sail (S) , BstEII (Bs) , and Xhol (X) .
  • the arrowheads numbered 1 to 6 represent the flanking regions amplified by using the oligonucleotide primers 4, 5, 6, 7, 14 and 15, respectively shown in Table I, infra.
  • Figure 3 shows the fluorescence-activated cell sorter (FACS) analysis of immunofluorescence staining of MD.45 hybridoma and its TCR o;-MD45.27J mutant, their corresponding scFvR7-transfected STA and STB clones, or STZ cells, which result from transfection of the scFvRf chimeric gene into MD45.27J.
  • Solid line staining with anti-Sp6 idiotypic antibody 20.5 or anti-CD3 mAb 145.2C11.
  • Broken line represents control irrelevant antibody.
  • Figure 4 shows immunoblotting analysis of lysates prepared from scFvR7 transfectants and parental hybridomas developed by anti-Sp6 idiotypic mAb 20.5 (panels A and C, respectively) and rabbit anti-human gamma chain (panels B and D, respectively) . Electrophoresis was on four separate gels. The molecular mass scales are related to B and D; the arrows point to the same bands in A and B or C and D.
  • Figure 5 shows the composition of the scFvR7 dimers.
  • Panel B Immunoprecipitation of lysates made of surface-iodinated STB ceils (scFvR7 transfectant cells) and their parent (MD45.27J hybridoma cells) .
  • FIG. 6 illustrates that transfectants expressing scFvR are stimulated to produce IL-2 after stimulation with TNP-A.20 (panel A) , or plastic immobilized TNP-F7G, without or with different concentrations of soluble TNP-F7G (panel B) .
  • GTAc.20 is an Sp6 double-chain cTCR transfectant described previously (9) .
  • the scFvR zeta-expressing STZ produced about 200 units (U) of IL-2 per ml after co-culture with TNP-A.20 at 8:1 stimulator-to-effector (S/E) cell ratio. Not shown are the responses of the transfectants to non-modified A.20 or F7G controls, which were completely negative, exactly like the background responses of the MD.45 and MD45.27 to TNP antigen.
  • Figure 7 shows specific 51 Cr release of TNP-A.20 cells after incubation with transfectants expressing scFvR. Effector cells were incubated with plastic-immobilized TNP- F7G for 8 hr before the killing assay. Kinetic assay was done at an effector-to-target (E/T) ceil ratio of 10:1 (panel A) ; dose response was determined in a 9 hr assay (panel B) . Control non-modified A.20 target cells incubated with the same effector cells in identical conditions did not release more 51 Cr than the spontaneous release (not shown) .
  • E/T effector-to-target
  • Figure 8 shows surface expression of chimeric scFvR7/ .
  • Figure 9 shows binding of detergent-solubilized scFvN29R7 and scFvN29Rf to Neu/HER2 antigen. The presence of chimeric receptors in cell lysates was evaluated by ELISA using HER2X-coated wells and anti-7 (A) or anti-f (B) antibodies. Functional molecules derived from hybridomas expressing the chimeric transgenes could bind to the immobilized antigen and expressed antigenic determinants specific to either 7 or f polypeptides.
  • Figure 10 shows antigen-specific activation of chimeric receptor expressing cells by HER2-bearing stimulator cells (A) or immobilized HER2X protein (B) plague T cell hybridomas expressing the chimeric scFvN29R7/f genes underwent antigen- specific, but MHC unrestricted stimulation for IL-2 production following co-culture with either HER2-expressing cells of different origins or with plastic-bound purified HER2/Neu receptor.
  • Stimulator cells used were human breast carcinoma cell lines SKBR3 and MDA 468, the human ovarian carcinoma cell line SK0V3 or HER2, a c-erbB-2 transfected 3T3-NIH fibroblasts (kindly provided by Dr. A. Ullrich) .
  • the Neu/HER2 protein is overexpressed in SKBR3, SK0V3 and HER2, while the MDA 468 cells have undetectable surface receptor.
  • untransfected parental cells MD45.27J did not produce any IL-2 following incubation with Neu/HER2 expressing cells.
  • B [filled square] - MD45.27J, untransfected cells; 0-N2971, transfectant expressing scFvN29R7.
  • Figure 11 shows that chimeric receptor expressing cells specifically lyse Neu/HER2 target cells.
  • Non-transfected CTL hybridomas and the scFvN29R7 expressing (N297I) or the scFvN29Rf expressing (MD45fl) transfectants were studied for their cytolytic potential either toward Neu/HER2 expressing NIH-3T3 murine fibroblasts or the human colon (N87) or breast (SKBR3) carcinoma cell lines. The percent 5 l Cr released by the parental cells at the same E:T were subtracted.
  • Figure 12 shows that chimeric receptor expressing cells specifically lyse HER2 target cells.
  • Non-transfected CTL hybridomas and the scFvN29R7 expressing (N2971) or the scFvN29Rr expressing (N29T18) transf ctants were studied for their cytolytic potential either toward Neu/HER2 expressing NIH-3T3 murine fibroblasts (filled symbols) or the non- transfected NIH-3T3 cells (open symbols) .
  • Substantial and specific lysis of HER2 target cells was demonstrated by N2971 at all effector to target (E:T) ratios.
  • cDNA is prepared from mRNA and amplification of the antibody light and heavy variable regions (V H and V L ) by PCR using a V L -5' (Xbal) , V L -3 (Sail), V H -5' (Sail) and V H -3' (BstEII) specific primers.
  • V H and V L antibody light and heavy variable regions
  • RNA from T lymphocytes was isolated and from the cDNA prepared the a , ⁇ chains of the TCR, , f subunits of the CD3, CDl6 ⁇ - of the FC7RIII, or IL-2 receptors (commonly denoted here as R) can be amplified using a specific set of primers for each chain. All the primers include a Xbal at their 5' end and a few bases downstream of the Xbal or the BstEII site. At the 3' end, all receptor chains contain a SnaBI site. Following introduction of the leader sequence into the pRSV 2 -neo plasmid the receptor was introduced at the Xbal site of the pRSVneoL*.
  • the amplified V L (digested with XBal-Sall) and V H (digested with Sall-BstEII) regions are introduced into the Xbal-BstEII digested pRSVneoL «-R plasmid in a three-piece ligation.
  • the resulting plasmid pRSVscFvR contains the complete chimeric single chain receptor.
  • the receptor (R) gene segment described in Figs. 12-18 is the human TCR C ⁇ .
  • Figure 13 shows transfer of the scFvR gene from the pRSVneo-scFvR to the pBJl-neo vector.
  • the scFvR was cut out from the pRSV vector using the SnaBI and introduced into the EcoRV site of the polylinker of the pBJl plasmid to drive the expression of the chimeric gene from the SR ⁇ promoter.
  • Figure 14 shows: A) Schematic representation of rosette formation by T cells expressing the anti-IgE scFvC ⁇ chimeric gene. Sheep red blood cells (SRBC) were coated with TNP and then with anti-TNP of the IgE class. The IgE-TNP-SRBC- complex was incubated with the T cells transfected with the scFvR comprising the scFv of the anti-IgE 84-lc mAb, and observed under microscope for rosette formation. B) Results of the rosette formation on scFvR-transfected JRT.T3.5 cells. Parental JRT.T3.5 cells were used as negative and the 84.1c as positive controls. Results are given in percentage of cells that form rosettes.
  • SRBC Sheep red blood cells
  • the transfectants were incubated with IgE, anti-Fc and anti-MYC, IgG (as negative control) and then with the SRBC-conjugate and counted.
  • Figure 15 shows: A) Schematic representation of the ELISA used to screen transfectants expressing scFvC3 chimeric gene (R is C ⁇ ) . Plates were covered with IgE and lysates of the transfectants were added, then anti-human a / ⁇ TCR antibodies were added and the reaction was developed with goat anti-mouse peroxidase. B) Results of some transfectants expressing the scFvR in the ELISA anti-human anti-3 TCR antibodies. Figure 16 shows stimulation of transfectants with immobilized IgE or anti-CD3 for IL-2 production.
  • Figure 17 shows stimulation for IL-2 production with IgE positive B cells.
  • the SPE-7 IgE secretor hybridoma was fixed with 0.25V glutaraldehyde for 10 min. at 0°C and mixed with the transfectants in different effector/stimulator (E/S) ratio. Cells were incubated for 20-24 hours and supernatants were collected and assayed for IL-2 production.
  • E/S effector/stimulator
  • Figure 18 shows specific inhibition of IgE production by cytotoxic hybridoma expressing the anti-IgE scFvR.
  • Spleen cells were stimulated with 20 ⁇ g/ml LPS and lOOU/ml IL-4 for four days.
  • spleen cells were washed and MD.45 cytotoxic hybridoma expressing the scFv was added and IgE and IgG concentrations were measured after 24, 48 and 72 hours.
  • 84.1c hybridoma cells were included as control as well as the MD.45.
  • Figure 19 is a schematic representation of the chimeric scFv-CD16 gene.
  • Figure 20 shows surface staining of rat basophilic leukemia (RBL) cells transfected with the scFvCD16 gene. Immunofluorescence staining was performed with anti-Sp6 idiotypic mAb 20.5 and irrelevant mouse antibody as negative control. The shift to the right in the FACS staining pattern is due to chimeric receptor expressing cells.
  • RBL basophilic leukemia
  • Figure 21 shows surface staining of RBL cells transfected with scFvR7 or scFvR ⁇ - chimeric genes. Immunofluorescence staining was performed with anti-Sp6 idiotypic mAb 20.5 and irrelevant mouse antibody as negative control.
  • Figure 22 shows surface staining of murine thymoma BW5147 cells transfected with the scFvCD16 gene. Immunofluorescence staining wSs performed with anti-Sp6 idiotypic mAb 20.5 and irrelevant mouse antibody as negative control.
  • Figure 23 shows stimulation of BW5147 cells co- transfected with scFvCD16 and normal 7 chain by TNP-labeled A.20 target cells.
  • BW-scFvCD16 clone 5 (A) or clone 50 (B) were co-cultured at different target: stimulation ratios with TNP modified irradiated A.20 cells.
  • IL-2 produced into the supernatant was determined following 24 hours by the MTT assay.
  • FIG 24 shows stimulation of BW5147 cells co- transfected with scFvCD16 and normal 7 chain by immobilized TNP-Fowl 7-globulin (TNP-F7G) .
  • TNP-F7G immobilized TNP-Fowl 7-globulin
  • Different concentrations of TNP-F7G at different TNP:F7G ratios were used to coat the wells of a microculture plate.
  • IL-2 was determined in the supernatant of 24 hr cultures of BW-scFvCD16 clone 5 (A) or clone 50 (B) . Incubation of either one of the cells with immobilized F7G by itself (filled squares) did not stimulate the cells.
  • the parental BW cells did not make any IL-2 in response to TNP-F7G under the same conditions (not shown) .
  • Figure 25 shows surface staining of RBL cells transfected with the scFvIL2R gene. Immunofluorescence staining was performed with anti-Sp6 idiotypic mAb 20.5 and irrelevant mouse antibody as negative control.
  • FIG. 26 shows that BW5147 cells transfected with scFvR express surface chimeric receptors.
  • BW5147 cells transfected with Sp6-scFvR were reacted with 1:200 dilution of ascites of 20.5 anti-Sp6 idiotypic antibody or anti-MOvl8 ascites in the same dilution as control, followed by FITC labeled anti-mouse Ig. Immunofluorescence was detected by FACS.
  • BW.Sp6-CD16 are cells co-transfected with scFvCD16 and 7 chain.
  • FIG. 27 shows stimulation of scFvR-BW5147 transfectants with TNP-A.20 cells. Different BW-scFvR transfectants were incubated with various amounts of TNP-A.20 cells for 24 hrs. IL-2 was determined by the MTT colorimetric assay. BWG are scFvR7 transfectants and BWZ are scFvR-r transfectants. Figure 28 shows that scFvR transfected BW5147 cells respond to immobilized TNP.
  • BWG SCFVR7 transfectants
  • BWZ scFvR transfectants
  • chimeric molecules were constructed composed of the scFv linked to receptor subunits that might serve to transduce the signal from the scFv and confer antibody specificity to T cells as well as other lymphocytes.
  • the new strategy according to the invention enables the use of other receptor molecules which might serve to transduce the signal from the scFv and confer antibody specificity to T cells as well as other immune cells.
  • it allows the expression of the scFv as the antigen recognition unit of chimeric molecules composed of the transmembrane and cytoplasmic domains of receptor molecules of immune cells, such as T cells and natural killer (NK) cells.
  • NK natural killer
  • Such receptors can be single or multi-chain in nature and not necessarily belong to the Ig gene superfamily.
  • Candidate molecules for this approach are receptor molecules which take part in signal transduction as an essential component of a receptor complex, such as receptors which trigger T cells and NK activation and/or proliferation,
  • Examples of triggers of T cells are subunits of the TCR, such as the ex, ⁇ , 7 or ⁇ 5 chain of the TCR, or any of the polypeptides constituting the CD3 complex which are involved in the signal transduction, e.g., the 7. and ⁇ CD3 chains.
  • polypeptides of the TCR/CD3 (the principal triggering receptor complex of T cells)
  • the zeta and its eta isoform chain which appear as either homo- or hetero-S-S-linked dimers, and are responsible for mediating at least a fraction of the cellular activation programs triggered by the TCR recognition of ligand (18, 19).
  • These polypeptides have very short extracellular domains which can serve for the attachment of the scFv.
  • immune cell trigger molecules are any one of the IL-2 receptor (IL-2R) p55 (a) ox p75 ( ⁇ ) chains, especially the p75 subunit which is responsible for signaling T cell and NK proliferation.
  • Further candidate receptor molecules for creation of scFv chimeras in accordance with the present invention include the subunit chains of Fc receptors.
  • FC7RIII the 7- and CD16 ⁇ -subunits of the low affinity receptor for IgG, FC7RIII. Occupancy or cross- linking of FC7RIII (either by anti-CD16 or through immune complexes) activates NK cells for cytokine production, expression of surface molecules and cytolytic aqtivity (20, 21) .
  • the FC ⁇ RIII appears as a heterooligomeric complex consisting of a ligand-binding a chain associated with a disulfide-linked 7 or zeta chain.
  • FC7RHIA signalling gamma chain (22) serves also as part of the Fc ⁇ RI complex, where it appears as a homodimer, is very similar to the CD3 zeta chain, and in fact can form heterodimers with it in some cytolytic T lymphocytes (CTL) and NK cells (23-25) .
  • CTL cytolytic T lymphocytes
  • NK cells NK cells
  • lymphocyte accessory and adhesion molecules such as CD2 and CD28, which transduce a co-stimulatory signal for T-cell activation.
  • co-stimulatory receptors can also be used in accordance with the present invention.
  • the single chain Fv chimeras can be made by joining the scFv domain with any receptor or co-receptor chain having a similar function to the disclosed molecules, e.g., derived from granulocytes, E lymphocytes, mast cells, macrophages, etc.
  • the distinguishing features of desirable immune cell trigger molecules comprise the ability to be expressed autonomously (i.e., as a single chain), the ability to be fused to an extracellular domain such that the resultant chimera is expressed on the surface of an immune cell into which the corresponding gene was genetically introduced, and the ability to take part in signal transduction programs secondary to encounter with a target ligand.
  • the ScFv domain must be joined to the immune cell triggering molecule such that the ScFv portion will be extracellular when the chimera is expressed. This is accomplished by joining the ScFv either to the very end of the transmembrane portion opposite the cytoplasmic domain of the trigger molecule or by using a spacer which is either part of the endogenous extracellular portion of the triggering molecule or from other sources.
  • the chimeric molecules of the present invention have the ability to confer on the immune cells on which they are expressed NHC nonrestricted antibody-type specificity. Thus, a continuous polypeptide of antigen binding and signal transducing properties can be produced and utilized as a targeting receptor on immune cells.
  • the target cells are tumor cells and the ScFv domain is derived from an antibody specific to an epitope expressed on the tumor cells. It is expected that such anti-tumor cytolysis can also be independent of exogenous supply of IL-2, thus providing a specific and safer means for adoptive immunotherapy.
  • the immune cells are T- cells or NK-cells.
  • the antibody scFvR design of the present invention will thus involve retargeting lymphocytes in vivo in an MHC- on-restricted manner.
  • the T-cells can be re ⁇ targeted in vivo to tumor cells or any other target of choice toward which antibodies can be raised.
  • the scFvR design is advantageous over the cTCR one. It requires the expression of only one gene instead of the gene pair required for the cTCR, thereby providing simpler construction and transfection.
  • single-chain Fv domain is intended to include not only the conventional single-chain antibodies as described in references 16 and 17, the entire contents of which are hereby incorporated herein by reference, but also any construct which provides the binding domain of an antibody in single-chain form as, for example, which may include only one or more of the complementarity determining regions (CDRs) , also known as the hypervariable regions, of an antibody.
  • CDRs complementarity determining regions
  • the gene encoding the transmembrane and cytoplasmic portions of the receptor molecule may correspond exactly to the natural gene or any gene which encodes the protein in its natural amino acid sequence.
  • the present invention comprehends muteins characterized by certain minor modifications to the aminc acid structure of the molecule, such that the mutant protein molecules are substantially similar in amino acid sequence and/or 3D structure, and possess a similar biological activity, relative to the native protein.
  • the transformed cells of the present invention may be used for the therapy of a number of diseases. Current methods of administering such transformed cells involve adoptive immunotherapy or cell-transfer therapy. These methods allow the return of the transformed immune system cells to the blood stream. Rosenberg, S.A., Scientific American 62 (May 1990); Rosenberg et al. , The New England Journal of Medicine 323 (9) :570 (1990) .
  • the transformed cells of the present invention may be administered in the form of a pharmaceutical composition with suitable pharmaceutically acceptable excipients. Such compositions may be administered to any animal which may experience the beneficial effects of the transformed cell of the present invention, including humans.
  • the antibodies which are used to make the ScFv portion of the present invention may be any antibody, the specificity of which is desired to be transferred to the immune cell.
  • Such antibody may be against tumor cells, cells expressing viral antigens, anti-idiotypic or anti-chromotypic antibodies in order to specifically eliminate certain B-cells and T-cells, or antibodies against the constant reach of immunoglobulin determinants.
  • the antibody is directed against the specific to the constant portion of IgE, it can serve to eliminate IgE-producing B-cell ⁇ in order to alleviate allergy, etc.
  • This list of possible antibodies is not intended to be exclusive and those of ordinary skill in the art will be aware of many additional antibodies for which important utilities exist upon combination with the receptor in accordance with the present invention.
  • genes of the present invention can be introduced into the immune cells by any manner known in the art, such as, for example, calcium phosphate transfection, electroporation, lipofection, transduction by retrovirus vector, use of a retroviral vector or a viral vector, etc.
  • the scFvR design can be employed to confer antibody specificity on a larger spectrum of signaling molecules composed of only one chain.
  • the scFv maintains both V H and V L together in one chain; thus, even upon mixed pairing of chimeric with endogenous chains, the antigen- binding properties of the molecule are conserved.
  • gamma and zeta constitute the signaling chains of the TCR/CD3, the FC7RIII and the Fc ⁇ RI expands the feasibility of exploiting the chimeric receptor for retargeting other hematopoietic cells, such as NK cells, basophils, or mast cells in addition to T cells.
  • This approach exploits the scFv as the antigen-recognition unit and the potent cytotoxic responses of NK cells and T cells and/or the ability of T cells to secrete lymphokines and cytokines upon activation at the target site, thus recruiting, regulating and amplifying other arms of the immune system.
  • the chimeric scFv receptors can confer on the lymphocytes the following functions: antibody-type specificity toward any predefined antigen; specific "homing" to their targets; specific recognition, activation, and execution of effector function as a result of encountering the target, and specific and controlled proliferation at the target site. Endowing the lymphocytes with an Fv from an antibody may also serve for controlled and selective blocking of the aforementioned functions using soluble haptens or Fab' of anti- idiotypic antibodies.
  • NK cells lymphokine- activated killer cells (LAK)
  • LAK lymphokine- activated killer cells
  • cytotoxic T cells helper T cells
  • helper T cells cytotoxic T cells
  • helper T cells cytotoxic T cells
  • helper T cells cytotoxic T cells
  • helper T cells cytotoxic T cells
  • helper T cells cytotoxic T cells
  • helper T cells cytotoxic T cells
  • helper T cells cytotoxic T cells
  • helper T cells can execute their authentic natural function and can serve, in addition, as carriers of foreign genes designated for gene therapy, and the chimeric receptor shall serve in this case to direct the cells to their target.
  • helper T cells expressing chimeric receptors made of Fv of antiidiotypic antibodies.
  • Such "designer lymphocytes” will interact and stimulate idiotype-bearing B cells to produce antigen-specific antibodies, thus bypassing the need for active immunization with toxic antigens.
  • the invention will now be illustrated by the following non-limiting examples.
  • MD.45 is a cytolytic T-lymphocyte (CTL) hybridoma of BALB/c mice allospecific to H- 2 b (29).
  • MD45.27J is a TCR ⁇ - mutant of MD.45.
  • A.20 is a B lymphoma of BALB/c origin (ATCC#T1B 208) .
  • Cells were cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal calf serum (FCS) .
  • Sp6, an anti-TNP mAb, and 20.5, an anti-Sp6 idiotype mAb were provided by G. Kohler (30) .
  • Anti- human FceRPy chain polyclonal and monoclonal (4D8) (31) antibodies were provided by J.-P. Kinet and J. Kochan, respectively, and rabbit antibodies to murine zeta chain by M. Baniyash.
  • the specific genes encoding the V E and V L of the Sp6 anti-TNP antibody were derived from the genomic constructs described for the preparation of the cTCR (12, 32) by PCR amplifications using oligodeoxynucleotide primers designed according to the 5' and 3 ⁇ consensus amino acid sequences of immunoglobulin V regions (33) introducing the Xba I and BstEII restriction sites at the ends of the scFv.
  • V L -linker-V H design containing a linker sequence similar to linker 212 described by Colcher et al. (34).
  • the V L -3' and the V H -5' primers include sequences comprising the 5' and 3' parts of the linker, introducing Sal I in their 3' and 5' ends, respectively.
  • Table I lists the oligonucleotide primers used in the different constructions. In the examples, reference is made to the number of the specific primer used.
  • Transfection of 20 ⁇ g of pRSVscFvR7/f DNA into 20xl0 6 MD.45 or MD.27J hybridoma cells was performed by electroporation using an ISCO power supply at 1.9 kV (32) .
  • Transfectants were selected in G418 at 2mg/ml.
  • Expression of scFvR7/r on the surface of transfected cells was evaluated by immunofluorescence staining using the 20.5 anti-Sp6 idiotype and fluorescein-isothiocyanate (FITC) -labelled anti-mouse Fab' antibody.
  • FITC fluorescein-isothiocyanate
  • Functional assays included an IL-2 production assay and a cytotoxicity assay in which the ability of transfectants to respond specifically to TNP-modified A.20 target cells was evaluated as detailed in Ref 9.
  • the amount of IL-2 was determined by using an IL-2-dependent CTL line and methyl tetrazolium acid (MTT) staining (36) .
  • Cytotoxicity assay was monitored by 5 x Cr release (29). All determinations were performed in triplicate.
  • cell lysates were mixed with sample buffer to a final concentration of 1% NaDodSO-, and either lOmM iodoacetamide (for non-reducing gels) or 15mM dithiothreitol (for reducing gels) .
  • the washed immunoprecipitates were dissociated in sample buffer under the same conditions.
  • the samples were incubated at 20°C for 30 min before NaDodSO-, /PAGE through 5-20% gel gradient.
  • Separated proteins were blotted onto nitrocellulose paper and allowed to react with anti-Sp6, anti- gamma or anti-zeta antibodies followed by peroxidase-labelled anti-immunoglobulin antibodies. Washed blots were developed by using a chemiluminescence kit (ECL, Amsterdam) according to the manuf cturer's recommendations, and exposed to film (Kodak, X- OMAT AR) .
  • oligonucleotide primers composed of sequences common to the majority of the 5' and 3' sequences of either V L or V H regions, flanked by relatively unique restriction sites, which allow both in-frame ligation of the different units and its removal to other vectors (see Table I) .
  • 5' -V L -linker-V E -3' design was found suitable for the expression of a variety of single-chain antibodies and their fragments in bacteria (17) , but the converse, 5' -V H -linker-V L -3' , alignment (16) can be used as well.
  • the surface expression of the scFvR7 or scFvRr molecule was independent of the TCR/CD3 complex; it did not restore surface expression of the CD3 in MD45.27J transfected STB or STZ cells , and some subclones of the STA that initially expressed both scFvR 7 and TCR/CD3 on their surface lost, upon a prolonged culture period, the TCR/CD3 expression without any apparent effect on the scFvR7 expression and function (not shown) .
  • the MD.45 T cell hybridoma can be triggered through its TCR to produce IL-2, IL-3 or GM-CSF. It specifically recognizes and responds to H-2 b target cells (29) , while its MD45.27J mutant cannot be stimulated through its TCR due to the absence of an a chain.
  • both of these cells could be specifically triggered to produce IL-2 following incubation with TNP-modified stimulator cells (Fig.
  • TNP-F7G plastic- immobilized TNP-fowl gamma globulin
  • Fig. 6B plastic- immobilized TNP-fowl gamma globulin
  • Non-modified A.20 cells or F7G did not activate the transfectants, demonstrating the specificity of the response toward TNP.
  • Stimulation of the various transfectants with immobilized antigen resulted in different degrees of reactivity.
  • STA responded to plastic-bound TNP-F7G in consistent manner, STB and STZ (transfected with the scFvR 7 and scFvRf, respectively) lost their ability to undergo stimulation with immobilized antigen but not with hapten- modified cells. Such behavior suggests the necessity of additional synergistic signals for these cells.
  • a single-chain Fv of an antibody molecule fused to the gamma chain of the immunoglobulin Fc receptor or to the zeta chain of the CD3 complex can be expressed in T cells as an antigen- specific receptor.
  • the chimeric scFvR7/ endowed T cells with antibody-type specificity transmitted a signal for IL-2 production and mediated target cell lysis.
  • the demonstration that the scFvR7/f fusion protein could mediate antigen- specific stimulation of T cells not expressing the TCR/CD3 receptor complex (as shown for the STB and STZ transfectants derived from the TCR-negative MD.27J mutant (Figs.
  • chimeric CD4 or CD16-gamma/zeta molecules expressed in cytotoxic lymphocytes could direct specific cytolysis against appropriate target cells (26, 38) .
  • This opens new possibilities in which the chimeric chain, composed of scFv and genetically modified zeta or gamma chains can be used not only to direct the specificity but also to dictate the selected reactivity of lymphocytes.
  • Neu/HER2 a human adenocarcinoma-associated growth factor receptor, were demonstrated to have tumor inhibitory capacity.
  • the Neu/HER2 (also calle c-erJbB-2) is a protooncogene product that encodes a growth factor receptor implicated in the malignancy of several human adenocarcinomas that overexpress it.
  • mAbs monoclonal antibodies specific to the extracellular portion of the Neu/HER2 protein (41)
  • mAb N29 whi significantly inhibited the tumorigenic growth of HER2/Neu transfected fibroblasts in nude mice, and induced phenotypic differentiation of various cultured breast cell lines (42) .
  • T cells equipped with anti-Neu/HER2 specificity as the ligand binding domain o the chimeric receptor respond specifically to Neu/HER2 beari target cells.
  • MD45 a murine allospecific CTL hybridoma (29) and MD45.27J, its mutant lacking the TCR chain, served as recipients for the chimeric genes.
  • Stimulator and target cells used were human breast carcinoma cell lines SKBR3 and MDA 468, the human ovarian carcinoma cell line SKOV3, or HER2, a c-erbB-2 transfected 3T3-NIH fibroblasts (kindly provided by Dr. A. Ullrich) .
  • Cells were cultured in DMEM containing 10% FCS.
  • N is a monoclonal anti-HER2 antibody (41) , deposited with the Collection Nationale de Cultures de Microorganismes, Institut Pasteur, Paris France, on August 19, 1992, under Registration No.
  • Chimeric scFvN29R7 or scFvN29R genes were constructed from single chain Fv DNA (in the V L -linker-V H alignment), amplified by PCR from cDNA prepared of hybridoma producing the N29 anti- HER2 mAb, and either ⁇ or f genes as described in Example 1 for the anti-TNP scFvR.
  • the MD45 or MD45.27J hybridomas were transfected by electroporation with 20 ⁇ g of DNA of pRSV2neo expression vectors harboring the chimeric genes and were selected for growth in the presence of 2 mg/ml G-418 (GIBCO) for 2-3 weeks as detailed in (9) .
  • Transfected cells were stained with either control serum or anti-N29 idiotypic antiserum (prepared by immunizing rabbits with purified N29 protein and adsorption of the immune serum on a normal mouse Ig-Agarose column) . Following incubation at 4°C with a 1:200 dilution of sera, the cells were washed and treated with fluorescein isothiocyanate-labeled goat anti-rabbit antibody (Jackson Labs, West Grove, PA, USA) for an additional hour at 4°C. Immunofluorescence of individual cells was determined with a FACSCAN (Becton Dickinson) . c.
  • Cell lysates were prepared from the transfectants by adding 100 ⁇ l of lysis buffer composed of 1% Triton X-100 in 0.15 M NaCl-lOmM Tris.HCl pH 7.4 buffer containing 10 mM EDTA, 1 mM phenylmethyi sulfonyl fluoride (Sigma) , 10 ⁇ g/ml aprotinin and 10 ⁇ g/ml leupeptin (Boehringer Mannheim, GmbH) to a pellet of 5xl0 6 cells. -After 30 min.
  • lysis buffer composed of 1% Triton X-100 in 0.15 M NaCl-lOmM Tris.HCl pH 7.4 buffer containing 10 mM EDTA, 1 mM phenylmethyi sulfonyl fluoride (Sigma) , 10 ⁇ g/ml aprotinin and 10 ⁇ g/ml leupeptin (Boehringer Mannheim, GmbH
  • HER2X is a recombinant extracellular domain of Neu/HER2 produced by CHO cells which were kindly provided by Dr. A. Ullrich. Following incubation for 2-4 hours at 4°C, plates were washed and incubated with I ⁇ g/ml of anti-human ⁇ or antibodies. After washing and the addition of horseradish peroxidase labeled anti-Ig antibodies (Jackson Labs) , peroxidase substrate was added and the degree of binding was determined by reading the OD 6 g 0 . D, IL-2 Production and Cytotoxic Assays.
  • Stimulator cells (3xl0 4 /well) were cultured in 96-well microculture plates for at least 6 hours until adherent.
  • wells of a microculture plate were coated with HER2X protein at the indicated concentrations for at least 3 h at 22°C and then washed twice with medium.
  • the transfected clones and their parental hybridoma were then added (10 5 /200 ⁇ l/well) in DMEM supplemented with 10% fetal calf serum and 10 " 5 M of 2-/9- mercaptoethanol.
  • the amount of IL-2 produced was evaluated by the proliferation of the IL-2 dependent CTL-L cell line by the MTT colorimetric assay as previously described (9) .
  • the transfectants and their parental hybridomas were co- incubated with 51 Cr labeled target cells at various effector to target ratios for 16 hrs.
  • the 51 Cr release assay was performed as described previously (16) .
  • fusion protein comprising the antigen binding and signal transducing moieties was verified by a receptor-specific, enzyme-linked immunosorbent assay (ELISA) , using a recombinant extracellular domain of Neu/HER2 (denoted HER2X) and anti-7 and ⁇ antibodies. As shown in Fig. 9, specific binding to HER2X was observed in whole cell lysates of the transfected but not of the untransfected parental cells.
  • ELISA receptor-specific, enzyme-linked immunosorbent assay
  • N29 ⁇ 1 and N29715 Three transfectants, N29 ⁇ 1 and N29715; both derived from MD45.27J cells transfected with the scFvR7 chimeric gene, and N29fM.l, a derivative of MD45 cells transfected with the scFvRf chimeric gene, were selected for functional studies.
  • the single-chain chimeric receptor was found to transduce specific signals for T cell activation.
  • This activation was mediated by the scFvR and was Neu/HER2- specific, since cells which do not overexpress Neu/HER2, like MDA-MB468 human breast carcinoma cells, did not stimulate the production of high levels of IL-2, whereas cells that display large amounts of Neu/HER2, like the breast carcinoma SKBR-3 cells, ovarian carcinoma SKOV-3 cells and an erJB-2 transfected murine fibroblast cell line, stimulated the hybridomas to produce high IL-2 levels. Soluble, purified HER2X partially blocked the activation by the breast carcinoma cells. However, upon immobilization, it served as a potent T cell activator, but only for the transfected cells (Fig. 10b) .
  • the T cell response to the immobilized antigen was in general weaker than to the cellular targets. Possibly, co-stimulatory signals provided by accessory and adhesion molecules during T cell interactions may amplify the intercellular interaction. Finally, the ability of the transfected cells to mediate specific target cell killing was determined by the 51 Cr release assay.
  • a variety of Neu/HER2 expressing cells were tested as targets (Fig. 11) , we found that the HER2 cell line, an NIH-3T3 fibroblast overexpressing Neu/HER2, could serve as an adequate target.
  • N297D Fig. 12
  • the scFvRf expressing hybridoma (N29_ * 18) gave only a marginal specific 51 Cr release signal when compared with the untransfected hybridomas.
  • the cytolytic effect was Neu/HER2- specific, since untransfected NIH-3T3 fibroblasts did not undergo killing.
  • the parental MD45.27J cells did not cause any significant 51 Cr release.
  • the high levels of spontaneous 51 Cr release from several candidate human tumor lines that we tested, did not allow us to determine the killing potency in a reproducible manner. Nevertheless, in all experiments, the transfected cells induced a significantly higher specific 51 Cr release from human tumors (such as SKBR-3 breast and N87 gastric carcinoma cell lines, Fig. 11) , than the parental hybridomas.
  • the production of IgE is regulated by antigen specific helper and suppressor T cells. T lymphocytes following activation, induce B cells to produce IgE.
  • the secreted IgE binds 0 preferentially to high affinity Fc ⁇ receptors (Fc ⁇ RI) on mast cells and basophils, thus sensitizing them.
  • Fc ⁇ RI-bound IgE is cross-linked and stimulates exocytosis of granule-associated preformed pharmacologic mediators such as histamine.
  • Elimination of IgE 5 producing cells can therefore terminate IgE production and thus prevent the onset of allergic responses.
  • IgE producing cells and their B-cell precursors express surface IgE and by employing the "T body” strategy using chimeric single-chain T cell Q receptor (scFvR) genes, made of an Fv of anti-IgE antibodies, we can specifically block IgE production.
  • scFvR chimeric single-chain T cell Q receptor
  • the basic strategy for construction of the chimeric genes encoding the 84.1c mAb V L and V H in a continuous single chain Fv linked to the constant region of the TCR a or ⁇ chains (C ⁇ or C ⁇ ) is similar to the one described for the preparation of the anti-TNP scFvR7/f chimeric genes and is schematically described in Figs. 1 and 2.
  • IILRNA was selected on oligo(dT) cellulose from the 84.1c hybridoma.
  • Single strand cDNA was synthesized using a 3'C ⁇ and 3'C H heavy primers employing "M-MLV-reverse transcriptase (BRL) .
  • V H and V ⁇ were amplified using the mouse consensus oligonucleotide primers similar to the ones described above for the Sp6 anti- TNP scFv (44) .
  • the V ⁇ -3' primer and the V H -5' primer (5 and 6 of Table I) included sequences comprising the 5' and the 3 ! parts of the linker, introducing a Sal I site in their 3 ! and 5' ends, respectively.
  • the fragments were ligated and introduced into the Xba I and BstE II sites of PRSVL J CQ!
  • C ⁇ expression cassettes have been originally designed to express the double chain chimeric TCR (cTCR) genes (45, 9, 32) and were constructed by cloning into the pRSV 2 neo the leader of the 38c.13 K-light chain 3' to the RSV LTR and downstream, either the Co. or C ⁇ of the human TCR.
  • the C ⁇ . and C ⁇ were PCR- amplified from human TCR clones using primers 9 and 12 from Table I for Co; and 11 and 13 for C ⁇ . Because we found previously (46) that the SR ⁇ promoter (47) drives transcription in T cells better than the RSV LTR, we adopted it here for the anti-IgE scFvR expression.
  • Fig. 13 describes the construction of the SR ⁇ based vector (pBJ-sc84. ⁇ ) .
  • the 84.1c based scFvQS chimeric gene was introduced into either the murine MD45 hybridoma (29) or the human Jurkat T cell leukemia ⁇ TCR negative JRT3.5 mutant (48), respectively. Transfection was carried out by electrophoresis and transfectants were selected in the presence of 2 mg/ml G418 as described in (9). JRT.T3.5 derived transfectants with the scFvR are denoted JSB and the MD.45 transfectants-JSMB.
  • chimeric receptors contain both the antigen- binding moiety and TCR determinants in the same complex.
  • lysates made of the transfectants were shown by analyzing lysates made of the transfectants. Incubation of such cell-free lysates on IgE-coated wells, followed by the addition of anti-TCR-S specific mAbs and peroxidase labeled anti-mouse Ig antibodies, yielded specific binding (Fig. 15) .
  • C. Functional expression Transfectants expressing chimeric surface receptors were tested for their ability to undergo specific activation for IL-2 production following stimulation with IgE, either immobilized by coating onto the plastic of the culture well or as a surface protein on IgE- producing hybridoma.
  • FIG 16 shows experiments in which the transfected cells were stimulated by plastic-bound IgE (or anti-CD3) . It is clear that the Jurkat-derived transfectants, generated by introduction of the scFvQS, specifically produced IL-2. When we tried to stimulate the transfectants with the SPE-hybridoma cells, we found that soluble IgE secreted by these hybridomas blocked stimulation (exactly like in the 38C.13 system) . We therefore fixed the IgE producing hybridoma cells and indeed, as evident in Fig. 17, such cells served as potent stimulators.
  • control 84.1c B cell hybridoma to cause such effect demonstrates that the lack of IgE accumulation in the culture medium is not because of passive absorption of IgE by the 84.1c anti-IgE antibodies.
  • This set of experiments clearly demonstrates that cytotoxic T cells equipped with chimeric scFv-TCR can specifically eliminate their target cells.
  • FC7RIHA low affinity receptor for IgG
  • FC7RIHA the low affinity receptor for IgG
  • FC7RIHA the low affinity receptor for IgG
  • Triggering of NK cells via FC7RIII includes cytokine production, expression of surface molecules and cytolytic activity (53, 21).
  • the CD16 polypeptide appears as membrane anchored form in polymorphonuclear cells and as transmembrane form (CD16 IM ) in NK (54).
  • FC7RHI- associated 7 chain serves also as part of the Fc ⁇ RI complex where it appears as homodimer, is very similar to the CD3 f chain and can form heterodimers with it in some CTL and NK cells (52, 21, 28, 23-25).
  • chimeras between 7 and CD4 directed CTL to recognize and kill cells expressing the HIV gpl20 (26) .
  • Similar chimeric receptors between either the extracellular domain of CDS (27) or Tac (28) in conjunctio with 7 , ⁇ or ⁇ have been recently reported in studies mapping the regions of these molecules which take part in the signaling process.
  • scFv-CD16 ⁇ design we have used the scFv of the Sp6 anti-TNP generated before.
  • the entire cytoplasmic and transmembrane and the immediate extracellular region (up to Gly206) of the CD16 ⁇ (see Fig. 19) were PCR amplified from a human CD16o; DNA clone (54) , using the primers 18 and 19 of Table I.
  • the truncated CD16 DNA was inserted instead of the 7 DNA in the pRSVneoscFvR7 vector previously described.
  • FC7RIII appears as a heterodimer complex consisted of CD16 and 7 chains, to check the expression of the chimeric scFvCDie gene, we transfected it into the rat basophilic leukemia (RBL) cell which is a mast cell expressing functional Fc ⁇ RI (56) . These cells produce excess of 7 chain as part of the Fc ⁇ RI and provide us with convenient function as the receptor-triggered degranulation assay.
  • RBL clones were obtained which could be surface-stained by the anti-Sp6 idiotypic antibody.
  • FIG. 20 shows the pattern of FACS analysis of scFvCD16 transfected RBL and Fig. 21 shows the staining of the scFvR7 and scFvRf transfectants.
  • Fig. 21 shows the staining of the scFvR7 and scFvRf transfectants.
  • RBL parental cells and transfectants expressing anti-TNP chimeric scFvR7 or scFvRf chains were stimulated with SPE anti-DNP IgE with or without its antigen DNP-BSA in order to assess the IgE mediated degranulation of the cells.
  • TNP-BSA served to induce a specific stimulus through the chimeric receptors. Degranulation was studied by measuring the enzymatic activity of ,5-hexoseaminidase released to the cell supernatant following degranulation as described before (43) .
  • BW5147 is a murine thymoma which does not express surface TCR/CD3 because it does not transcribe either 7 or r chains.
  • transfection of the BW5147 cells with chimeric scFvCDl ⁇ DNA did not yield any detectable surface receptor, yet intracellular receptor could be detected by immunoblotting of lysates (not shown) .
  • the chimeric scFvCDlo and normal 7 DNA were co- electroporated into BW5147 cells, significantly high level of the Sp6 idiotype could be detected on the surface of the transfectants as revealed by immunofluorescence staining and FACS analysis (Fig. 22) , The transfectants responded to specific stimulus and produced IL-2 following stimulation with TNP-modified A.20 cells or immobilized TNP-fowl 7-globulin (TNP-F 7 G) (Fig. 23, 24) .
  • the Sp6-IL-2-R chimeric gene was prepared by joining DNA containing the scFv of Sp6 to a 936bp DNA segment cloned from PCR amplified DNA (using primers 20 and 23 of Table I) containing the cytoplasmic and transmembrane regions (carboxy 312 amino acids) of the ⁇ -chains of the human IL-2 receptor.
  • Figure 25 shows the results of immunofluorescence staining of one such RBL transfectant with anti-Sp6 idiotypic antibodies.
  • BW5147 is a murine thymoma cell line which do not express the TCR or FC 7 R complexes (due to a defect in the f chain transcription (57) ) , and therefore served as a convenient cell-line to study the expression of the different chimeric scFv receptors. Because BW cells do not produce endogenous f or 7 chains, it is expected that following transfection, the chimeric receptors will be composed only of homodimers of the exogenous transgenes (in the case of scFvR 7 or scFvRf) . Also, it provides a system to study whether the chimeric scFvCD16 can be expressed independently of 7 or f chains.
  • the chimeric genes composed of an scFv of Sp6 anti- TNP mAb joined to either one of the f, ⁇ or CD16 chains were introduced by electroporation into the BW cells and selected transfectants which grew in the presence of G-418 were analyzed for surface expression of the Sp6 idiotope using the 20.5 anti-Sp6 idiotypic mAb.
  • a group of BW cells was co-transfected with a mixture of scFvCD16+7 chain DNA.
  • the immunofluorescence pattern of staining analysed by FACS is depicted in Fig. 26.
  • both BW.Sp6-7 and BW.Sp6-f transfectants (which received weither scFvR 7 or scFvRf DNA, respectively) could be specifically stained with anti-Sp6 idiotypic antibody and thus express a moderate level of the chimeric receptor chains on their surface.
  • specific anti-CD3 mAb we could not observe any surface staining of the scFvR7 or scFvR transfectants (not shown) , indicating that these chimeric genes are expressed on the cell surface independently of the CD3 complex. None of the transfectants which was electroporated with scFvCD16 alone did express surface receptor (unshown) .

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Oncology (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

L'invention se rapporte à des gènes chimères contenant un premier segment codant un domaine de Fv monocaténaire d'un anticorps spécifique, ainsi qu'un deuxième segment codant au moins les domaines de transmembrane et les domaines cytoplasmiques d'une molécule immune de déclenchement cellulaire, tel que des sous-ensembles d'un récepteur de lymphocytes T, un complexe CD3 et récepteur de lymphocytes T, un récepteur de Fc ou un récepteur de IL-2. L'invention décrit également un procédé de traitement d'une tumeur au moyen de cellules lymphocytaires transformées par des vecteurs d'expression contenant les gènes chimères.
PCT/US1993/002506 1992-03-18 1993-03-18 Genes de recepteurs chimeres et cellules transformees au moyen desdits genes WO1993019163A1 (fr)

Priority Applications (9)

Application Number Priority Date Filing Date Title
CA002132349A CA2132349C (fr) 1992-03-18 1993-03-18 Genes codant pour les recepteurs chimeriques et cellules transformees par ces genes
AU39243/93A AU668156B2 (en) 1992-03-18 1993-03-18 Chimeric receptor genes and cells transformed therewith
EP93908414A EP0638119B1 (fr) 1992-03-18 1993-03-18 Genes de recepteurs chimeres et cellules transformees au moyen desdits genes
DE69333038T DE69333038T2 (de) 1992-03-18 1993-03-18 Chimäre rezeptorgene und entsprechend damit transformierte zellen
JP51673193A JP3643590B2 (ja) 1992-03-18 1993-03-18 キメラ受容体遺伝子およびこれを用いて形質転換した細胞
AT93908414T ATE242800T1 (de) 1992-03-18 1993-03-18 Chimäre rezeptorgene und entsprechend damit transformierte zellen
US08/084,994 US7741465B1 (en) 1992-03-18 1993-07-02 Chimeric receptor genes and cells transformed therewith
US08/547,263 US8211422B2 (en) 1992-03-18 1995-10-24 Chimeric receptor genes and cells transformed therewith
US13/281,560 US20120093842A1 (en) 1992-03-18 2011-10-26 Chimeric receptor genes and cells transformed therewith

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
IL101288A IL101288A (en) 1992-03-18 1992-03-18 Chimeric genes suitable for endowing lymphocytes with antibody-type specificity, expression vectors, lymphocytes transformed therewith, and pharmaceutical compositions comprising them
IL101288 1992-03-18
IL10457092 1993-01-31
IL104570 1993-01-31

Publications (1)

Publication Number Publication Date
WO1993019163A1 true WO1993019163A1 (fr) 1993-09-30

Family

ID=26322411

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1993/002506 WO1993019163A1 (fr) 1992-03-18 1993-03-18 Genes de recepteurs chimeres et cellules transformees au moyen desdits genes

Country Status (1)

Country Link
WO (1) WO1993019163A1 (fr)

Cited By (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5359046A (en) * 1990-12-14 1994-10-25 Cell Genesys, Inc. Chimeric chains for receptor-associated signal transduction pathways
WO1995030014A1 (fr) * 1994-05-02 1995-11-09 Ciba-Geigy Ag Proteine a double fonction, sa preparation et son utilisation
WO1997015669A1 (fr) * 1995-10-24 1997-05-01 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Genes recepteurs chimeriques et cellules transformees au moyen de ces genes
WO1997020048A2 (fr) * 1995-11-30 1997-06-05 Bristol-Myers Squibb Company Molecules sfv modifiees mediatrices de l'adhesion entre cellules, et leurs utilisations
WO1997023613A2 (fr) * 1995-12-21 1997-07-03 Celltech Therapeutics Ltd. Procede d'activation de cellules et ses reactifs
WO1997035004A1 (fr) * 1996-03-15 1997-09-25 Medical Research Council Stimulation cellulaire
EP0821730A1 (fr) * 1995-02-02 1998-02-04 Cell Genesys, Inc. Recepteurs chimeres permettant la regularisation de la proliferation cellulaire et de la fonction effectrice
WO1998038324A2 (fr) * 1997-02-27 1998-09-03 Boehringer Mannheim Corporation Recepteurs de surface chimeres soumis a une association induite par un antigene
US5830755A (en) * 1995-03-27 1998-11-03 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services T-cell receptors and their use in therapeutic and diagnostic methods
US5843728A (en) * 1991-03-07 1998-12-01 The General Hospital Corporation Redirection of cellular immunity by receptor chimeras
US5851828A (en) * 1991-03-07 1998-12-22 The General Hospital Corporation Targeted cytolysis of HIV-infected cells by chimeric CD4 receptor-bearing cells
US5912170A (en) * 1991-03-07 1999-06-15 The General Hospital Corporation Redirection of cellular immunity by protein-tyrosine kinase chimeras
WO1999036437A1 (fr) * 1998-01-15 1999-07-22 Center For Molecular Medicine And Immunology Fraction ciblant des anticorps/recepteurs permettant d'ameliorer l'administration d'un ligand arme
WO1999057268A1 (fr) * 1998-05-06 1999-11-11 Celltech Therapeutics Limited Recepteurs chimeres
US6004811A (en) * 1991-03-07 1999-12-21 The Massachussetts General Hospital Redirection of cellular immunity by protein tyrosine kinase chimeras
WO2000018932A2 (fr) * 1998-09-25 2000-04-06 Regeneron Pharmaceuticals, Inc. Antagonistes a base de recepteurs, modes d'elaboration et d'utilisation
US6103521A (en) * 1995-02-06 2000-08-15 Cell Genesys, Inc. Multispecific chimeric receptors
US6132718A (en) * 1994-07-06 2000-10-17 Immunomedics, Inc. Multi-stage cascade boosting vaccine
WO2001062908A2 (fr) * 2000-02-22 2001-08-30 Ahuva Nissim Banques d'affichage de phages chimeriques et tcr, reactifs chimeriques et tcr et methodes d'utilisation correspondantes
US6319494B1 (en) 1990-12-14 2001-11-20 Cell Genesys, Inc. Chimeric chains for receptor-associated signal transduction pathways
US6339070B1 (en) 1997-05-10 2002-01-15 Zeneca Limited Gene construct encoding a heterologous prodrug-activating enzyme and a cell targeting moiety
US6407221B1 (en) 1990-12-14 2002-06-18 Cell Genesys, Inc. Chimeric chains for receptor-associated signal transduction pathways
US6451995B1 (en) * 1996-03-20 2002-09-17 Sloan-Kettering Institute For Cancer Research Single chain FV polynucleotide or peptide constructs of anti-ganglioside GD2 antibodies, cells expressing same and related methods
US6699715B1 (en) 1995-11-30 2004-03-02 Bristol-Myers Squibb Co. Modified sFv molecules which mediate adhesion between cells and uses thereof
US6703488B1 (en) 1998-01-15 2004-03-09 Center For Molecular Medicine And Immunology Antibody/receptor targeting moiety for enhanced delivery of armed ligand
US6753162B1 (en) 1991-03-07 2004-06-22 The General Hospital Corporation Targeted cytolysis of HIV-infected cells by chimeric CD4 receptor-bearing cells
US6927044B2 (en) 1998-09-25 2005-08-09 Regeneron Pharmaceuticals, Inc. IL-1 receptor based cytokine traps
US7049136B2 (en) 1991-03-07 2006-05-23 The General Hospital Corporation Redirection of cellular immunity by receptor chimeras
US7083949B2 (en) 1998-09-25 2006-08-01 Regeneron Pharmaceuticals, Inc. Receptor based antagonists and methods of making and using
US7354587B1 (en) 1994-07-06 2008-04-08 Immunomedics, Inc. Use of immunoconjugates to enhance the efficacy of multi-stage cascade boosting vaccines
US7723111B2 (en) 2001-03-09 2010-05-25 The United States Of America As Represented By The Department Of Health And Human Services Activated dual specificity lymphocytes and their methods of use
US7786269B2 (en) 2001-12-04 2010-08-31 Dana-Farber Cancer Institute, Inc. Antibody to latent membrane proteins and uses thereof
WO2014039523A1 (fr) * 2012-09-04 2014-03-13 Cellectis Récepteur d'antigène chimérique multicaténaire et utilisations de celui-ci
EP2126054B1 (fr) 2007-01-31 2016-07-06 Yeda Research And Development Company Limited Cellules régulatrices t redirigées, génétiquement modifiées et leur utilisation dans la suppression d'une maladie auto-immune et inflammatoire
WO2017123808A1 (fr) 2016-01-15 2017-07-20 The J. David Gladstone Institutes Méthodes de traitement de maladies par la régulation métabolique de la différenciation des lymphocytes t
US10342829B2 (en) 2012-05-25 2019-07-09 Cellectis Multi-chain chimeric antigen receptor and uses thereof
EP3298033B1 (fr) 2015-05-18 2020-08-05 TCR2 Therapeutics Inc. Compositions et usage medical de reprogrammation de tcr au moyen de protéines de fusion
US10780120B2 (en) 2018-03-06 2020-09-22 The Trustees Of The University Of Pennsylvania Prostate-specific membrane antigen cars and methods of use thereof
US10857181B2 (en) 2015-04-21 2020-12-08 Enlivex Therapeutics Ltd Therapeutic pooled blood apoptotic cell preparations and uses thereof
US11000548B2 (en) 2015-02-18 2021-05-11 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11304976B2 (en) 2015-02-18 2022-04-19 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11318163B2 (en) 2015-02-18 2022-05-03 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11466093B2 (en) 2015-07-27 2022-10-11 The General Hospital Corporation Antibody derivatives with conditionally enabled effector function
US11497767B2 (en) 2015-02-18 2022-11-15 Enlivex Therapeutics R&D Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11512289B2 (en) 2015-02-18 2022-11-29 Enlivex Therapeutics Rdo Ltd Combination immune therapy and cytokine control therapy for cancer treatment
WO2023022235A1 (fr) 2021-08-20 2023-02-23 大塚製薬株式会社 Association médicamenteuse
US11596652B2 (en) 2015-02-18 2023-03-07 Enlivex Therapeutics R&D Ltd Early apoptotic cells for use in treating sepsis
US11730761B2 (en) 2016-02-18 2023-08-22 Enlivex Therapeutics Rdo Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11851491B2 (en) 2016-11-22 2023-12-26 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1988009344A1 (fr) * 1987-05-21 1988-12-01 Creative Biomolecules, Inc. Proteines mutifonctionnelles a cible predeterminee

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1988009344A1 (fr) * 1987-05-21 1988-12-01 Creative Biomolecules, Inc. Proteines mutifonctionnelles a cible predeterminee

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
Cell, Vol. 58, issued 08 September 1989, BECKER et al., "Expression of a Hybrid Immunoglobulin-T Cell Receptor Protein in Transgenic Mice", pages 911-921, see entire document. *
Cell, Vol. 60, issued 23 March 1990, GOVERMAN et al., "Chimeric Immunoglobulin-T Cell Receptor Proteins from Functional Receptors: Implications for T Cell Receptor Complex Formation and Activation", pages 929-939, see entire document. *
Journal of Experimental Medicine, Vol. 170, issued August 1989, RAVETCH et al., "Alternative Membrane Forms of FcgammaRIII(CD16) on Human Natural Killer Cells and Neutrophils", pages 481-497, see entire document. *
Proceedings of the National Academy of Sciences, USA, Vol. 86, issued December 1989, GROSS et al., "Expression of Immunoglobulin-T-Cell Receptor Chimeric Molecules as Functional Receptors with Antibody-Type Specificity", pages 10024-10028, see entire document. *
Proceedings of the National Academy of Sciences, USA, Vol. 88, issued May 1991, BAUER et al., "Differential Signal Transduction via T-Cell Receptor CD3zeta2, CD3zeta-eta, and CD3eta2 Isoforms", pages 3842-3846, see entire document. *
Proceedings of the National Academy of Sciences, USA, Vol. 90, issued January 1993, ESHHAR et al., "Specific Activation and Targeting of Cytotoxic Lymphocytes Through Chimeric Single Chains Consisting of Antibody-Binding Domains and the gamma or zeta Subunits of the Immunoglobulin and T-Cell Receptors", pages 720-724, see *
See also references of EP0638119A4 *
The EMBO Journal, Vol. 8, No. 12, issued 1989, WEISSMAN et al., "Role of the Zeta Chain in the Expression of the T Cell Antigen Receptor: Genetic Reconstitution Studies", pages 3651-3656, see entire document. *
The New England Journal of Medicine, Vol. 323, No. 9, issued 30 August 1990, ROSENBERG et al., "Gene Transfer into Humans-Immunotherapy of Patients with Advanced Melanoma, using Tumor-Infiltrating Lymphocytes Modifed by Retroviral Gene Transduction", pages 570-578, see entire document. *

Cited By (82)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5359046A (en) * 1990-12-14 1994-10-25 Cell Genesys, Inc. Chimeric chains for receptor-associated signal transduction pathways
US6319494B1 (en) 1990-12-14 2001-11-20 Cell Genesys, Inc. Chimeric chains for receptor-associated signal transduction pathways
US6407221B1 (en) 1990-12-14 2002-06-18 Cell Genesys, Inc. Chimeric chains for receptor-associated signal transduction pathways
US7320787B2 (en) 1991-03-07 2008-01-22 The General Hospital Corporation Redirection of cellular immunity by protein tyrosine kinase chimeras
US5912170A (en) * 1991-03-07 1999-06-15 The General Hospital Corporation Redirection of cellular immunity by protein-tyrosine kinase chimeras
US6004811A (en) * 1991-03-07 1999-12-21 The Massachussetts General Hospital Redirection of cellular immunity by protein tyrosine kinase chimeras
US7094599B2 (en) 1991-03-07 2006-08-22 The General Hospital Corporation Targeted cytolysis of HIV-infected cells by chimeric CD4 receptor-bearing cells
US7049136B2 (en) 1991-03-07 2006-05-23 The General Hospital Corporation Redirection of cellular immunity by receptor chimeras
US6284240B1 (en) 1991-03-07 2001-09-04 The General Hospital Corporation Targeted cytolysis of HIV-infected cells by chimeric CD4 receptor-bearing cells
US6753162B1 (en) 1991-03-07 2004-06-22 The General Hospital Corporation Targeted cytolysis of HIV-infected cells by chimeric CD4 receptor-bearing cells
US6410014B1 (en) 1991-03-07 2002-06-25 The General Hospital Corporation Redirection of cellular immunity by protein-tyrosine kinase chimeras
US5851828A (en) * 1991-03-07 1998-12-22 The General Hospital Corporation Targeted cytolysis of HIV-infected cells by chimeric CD4 receptor-bearing cells
US6392013B1 (en) 1991-03-07 2002-05-21 The General Hospital Corporation Redirection of cellular immunity by protein tyrosine kinase chimeras
US5843728A (en) * 1991-03-07 1998-12-01 The General Hospital Corporation Redirection of cellular immunity by receptor chimeras
US8211422B2 (en) 1992-03-18 2012-07-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric receptor genes and cells transformed therewith
WO1995030014A1 (fr) * 1994-05-02 1995-11-09 Ciba-Geigy Ag Proteine a double fonction, sa preparation et son utilisation
US6984382B1 (en) 1994-05-02 2006-01-10 Bernd Groner Bifunctional protein, preparation and use
US7354587B1 (en) 1994-07-06 2008-04-08 Immunomedics, Inc. Use of immunoconjugates to enhance the efficacy of multi-stage cascade boosting vaccines
US6132718A (en) * 1994-07-06 2000-10-17 Immunomedics, Inc. Multi-stage cascade boosting vaccine
US8163887B2 (en) 1994-07-06 2012-04-24 Immunomedics, Inc. Use of immunoconjugates to enhance the efficacy of multi-stage cascade boosting vaccines
EP0821730A4 (fr) * 1995-02-02 2000-04-19 Cell Genesys Inc Recepteurs chimeres permettant la regularisation de la proliferation cellulaire et de la fonction effectrice
US5837544A (en) * 1995-02-02 1998-11-17 Cell Genesys, Inc. Method of inducing a cell to proliferate using a chimeric receptor comprising janus kinase
US6077947A (en) * 1995-02-02 2000-06-20 Cell Genesys, Inc. DNA encoding an intracellular chimeric receptor comprising Janus kinase
US5741899A (en) * 1995-02-02 1998-04-21 Cell Genesys, Inc. Chimeric receptors comprising janus kinase for regulating cellular pro liferation
EP0821730A1 (fr) * 1995-02-02 1998-02-04 Cell Genesys, Inc. Recepteurs chimeres permettant la regularisation de la proliferation cellulaire et de la fonction effectrice
US6103521A (en) * 1995-02-06 2000-08-15 Cell Genesys, Inc. Multispecific chimeric receptors
US5830755A (en) * 1995-03-27 1998-11-03 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services T-cell receptors and their use in therapeutic and diagnostic methods
WO1997015669A1 (fr) * 1995-10-24 1997-05-01 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Genes recepteurs chimeriques et cellules transformees au moyen de ces genes
WO1997020048A2 (fr) * 1995-11-30 1997-06-05 Bristol-Myers Squibb Company Molecules sfv modifiees mediatrices de l'adhesion entre cellules, et leurs utilisations
US6699715B1 (en) 1995-11-30 2004-03-02 Bristol-Myers Squibb Co. Modified sFv molecules which mediate adhesion between cells and uses thereof
WO1997020048A3 (fr) * 1995-11-30 1997-09-25 Bristol Myers Squibb Co Molecules sfv modifiees mediatrices de l'adhesion entre cellules, et leurs utilisations
WO1997023613A2 (fr) * 1995-12-21 1997-07-03 Celltech Therapeutics Ltd. Procede d'activation de cellules et ses reactifs
WO1997023613A3 (fr) * 1995-12-21 1997-08-21 Celltech Therapeutics Ltd Procede d'activation de cellules et ses reactifs
WO1997035004A1 (fr) * 1996-03-15 1997-09-25 Medical Research Council Stimulation cellulaire
US6451995B1 (en) * 1996-03-20 2002-09-17 Sloan-Kettering Institute For Cancer Research Single chain FV polynucleotide or peptide constructs of anti-ganglioside GD2 antibodies, cells expressing same and related methods
WO1998038324A3 (fr) * 1997-02-27 1999-01-28 Boehringer Mannheim Corp Recepteurs de surface chimeres soumis a une association induite par un antigene
WO1998038324A2 (fr) * 1997-02-27 1998-09-03 Boehringer Mannheim Corporation Recepteurs de surface chimeres soumis a une association induite par un antigene
US6339070B1 (en) 1997-05-10 2002-01-15 Zeneca Limited Gene construct encoding a heterologous prodrug-activating enzyme and a cell targeting moiety
WO1999036437A1 (fr) * 1998-01-15 1999-07-22 Center For Molecular Medicine And Immunology Fraction ciblant des anticorps/recepteurs permettant d'ameliorer l'administration d'un ligand arme
US6703488B1 (en) 1998-01-15 2004-03-09 Center For Molecular Medicine And Immunology Antibody/receptor targeting moiety for enhanced delivery of armed ligand
WO1999057268A1 (fr) * 1998-05-06 1999-11-11 Celltech Therapeutics Limited Recepteurs chimeres
US7927583B2 (en) 1998-09-25 2011-04-19 Regeneron Pharmaceuticals, Inc. Receptor based antagonists and methods of making and using
EP1229047A3 (fr) * 1998-09-25 2002-10-02 Regeneron Pharmaceuticals, Inc. Protéines de fusion du récepteur de IL-1 utilisées comme antagonistes et méthodes d'utilisation et de fabrication
US7083949B2 (en) 1998-09-25 2006-08-01 Regeneron Pharmaceuticals, Inc. Receptor based antagonists and methods of making and using
US6927044B2 (en) 1998-09-25 2005-08-09 Regeneron Pharmaceuticals, Inc. IL-1 receptor based cytokine traps
EP1229047A2 (fr) * 1998-09-25 2002-08-07 Regeneron Pharmaceuticals, Inc. Protéines de fusion du récepteur de IL-1 utilisées comme antagonistes et méthodes d'utilisation et de fabrication
US7417134B2 (en) 1998-09-25 2008-08-26 Regeneron Pharmaceuticals, Inc. IL-1 receptor based cytokine traps and method of using
WO2000018932A2 (fr) * 1998-09-25 2000-04-06 Regeneron Pharmaceuticals, Inc. Antagonistes a base de recepteurs, modes d'elaboration et d'utilisation
WO2000018932A3 (fr) * 1998-09-25 2000-11-02 Regeneron Pharma Antagonistes a base de recepteurs, modes d'elaboration et d'utilisation
WO2001062908A3 (fr) * 2000-02-22 2002-04-25 Ahuva Nissim Banques d'affichage de phages chimeriques et tcr, reactifs chimeriques et tcr et methodes d'utilisation correspondantes
WO2001062908A2 (fr) * 2000-02-22 2001-08-30 Ahuva Nissim Banques d'affichage de phages chimeriques et tcr, reactifs chimeriques et tcr et methodes d'utilisation correspondantes
US7723111B2 (en) 2001-03-09 2010-05-25 The United States Of America As Represented By The Department Of Health And Human Services Activated dual specificity lymphocytes and their methods of use
US7786269B2 (en) 2001-12-04 2010-08-31 Dana-Farber Cancer Institute, Inc. Antibody to latent membrane proteins and uses thereof
US7361350B2 (en) 2002-10-28 2008-04-22 Regeneron Pharmaceuticals, Inc. Use of an IL-1 antagonist for treating arthritis
EP2126054B1 (fr) 2007-01-31 2016-07-06 Yeda Research And Development Company Limited Cellules régulatrices t redirigées, génétiquement modifiées et leur utilisation dans la suppression d'une maladie auto-immune et inflammatoire
EP3097923B1 (fr) 2007-01-31 2022-07-27 Yeda Research And Development Co., Ltd. Redirigee, genetiquement modifiee cellule de type t et leur utilisation pour la suppression des maladies autimmunes et inflammatoires
EP3097923A1 (fr) 2007-01-31 2016-11-30 Yeda Research And Development Co., Ltd. Redirigee, genetiquement modifiee cellule de type t et leur utilisation pour la suppression des maladies autimmunes et inflammatoires
US10342829B2 (en) 2012-05-25 2019-07-09 Cellectis Multi-chain chimeric antigen receptor and uses thereof
CN104769103B (zh) * 2012-09-04 2018-06-08 塞勒克提斯公司 多链嵌合抗原受体和其用途
WO2014039523A1 (fr) * 2012-09-04 2014-03-13 Cellectis Récepteur d'antigène chimérique multicaténaire et utilisations de celui-ci
CN104769103A (zh) * 2012-09-04 2015-07-08 塞勒克提斯公司 多链嵌合抗原受体和其用途
US11318163B2 (en) 2015-02-18 2022-05-03 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11000548B2 (en) 2015-02-18 2021-05-11 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11304976B2 (en) 2015-02-18 2022-04-19 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11717539B2 (en) 2015-02-18 2023-08-08 Enlivex Therapeutics RDO Ltd. Combination immune therapy and cytokine control therapy for cancer treatment
US11596652B2 (en) 2015-02-18 2023-03-07 Enlivex Therapeutics R&D Ltd Early apoptotic cells for use in treating sepsis
US11497767B2 (en) 2015-02-18 2022-11-15 Enlivex Therapeutics R&D Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11512289B2 (en) 2015-02-18 2022-11-29 Enlivex Therapeutics Rdo Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US10857181B2 (en) 2015-04-21 2020-12-08 Enlivex Therapeutics Ltd Therapeutic pooled blood apoptotic cell preparations and uses thereof
US11883429B2 (en) 2015-04-21 2024-01-30 Enlivex Therapeutics Rdo Ltd Therapeutic pooled blood apoptotic cell preparations and uses thereof
EP3298033B1 (fr) 2015-05-18 2020-08-05 TCR2 Therapeutics Inc. Compositions et usage medical de reprogrammation de tcr au moyen de protéines de fusion
EP3298033B2 (fr) 2015-05-18 2023-07-12 TCR2 Therapeutics Inc. Compositions et usage medical de reprogrammation de tcr au moyen de protéines de fusion
US11965012B2 (en) 2015-05-18 2024-04-23 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US11466093B2 (en) 2015-07-27 2022-10-11 The General Hospital Corporation Antibody derivatives with conditionally enabled effector function
US11241455B2 (en) 2016-01-15 2022-02-08 The J. David Gladstone Institutes, A Testamentary Trust Established Under The Will Of J. David Gladstone Methods of treating disease by metabolic control of T-cell differentiation
WO2017123808A1 (fr) 2016-01-15 2017-07-20 The J. David Gladstone Institutes Méthodes de traitement de maladies par la régulation métabolique de la différenciation des lymphocytes t
US11730761B2 (en) 2016-02-18 2023-08-22 Enlivex Therapeutics Rdo Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11851491B2 (en) 2016-11-22 2023-12-26 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10780120B2 (en) 2018-03-06 2020-09-22 The Trustees Of The University Of Pennsylvania Prostate-specific membrane antigen cars and methods of use thereof
US11992503B2 (en) 2018-03-06 2024-05-28 The Trustees Of The University Of Pennsylvania Prostate-specific membrane antigen cars and methods of use thereof
KR20240051172A (ko) 2021-08-20 2024-04-19 오츠카 세이야쿠 가부시키가이샤 조합 의약
WO2023022235A1 (fr) 2021-08-20 2023-02-23 大塚製薬株式会社 Association médicamenteuse

Similar Documents

Publication Publication Date Title
EP0638119B1 (fr) Genes de recepteurs chimeres et cellules transformees au moyen desdits genes
US8211422B2 (en) Chimeric receptor genes and cells transformed therewith
WO1993019163A1 (fr) Genes de recepteurs chimeres et cellules transformees au moyen desdits genes
US11639387B2 (en) Bispecific chimeric antigen receptors, encoding polynucleotides thereof and methods of use thereof to treat disease
AU2020200751B2 (en) Universal chimeric antigen receptor expressing immune cells for targeting of diverse multiple antigens and method of manufacturing the same and use of the same for treatment of cancer, infections and autoimmune disorders
US5912172A (en) Endowing lymphocytes with antibody specificity
JP7428663B2 (ja) いくつかの多重抗原の標的用の逆ユニバーサルキメラ抗原受容体を発現する免疫細胞ならびにその製造方法ならびに癌、感染症および自己免疫疾患を治療するためのその使用
Stancovski et al. Targeting of T lymphocytes to Neu/HER2-expressing cells using chimeric single chain Fv receptors.
Eshhar et al. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors.
Yun et al. Targeting of T lymphocytes to melanoma cells through chimeric anti-GD3 immunoglobulin T-cell receptors
Moritz et al. Cytotoxic T lymphocytes with a grafted recognition specificity for ERBB2-expressing tumor cells.
EP0758394B1 (fr) Proteine a double fonction, sa preparation et son utilisation
CN106755107B (zh) 一种car新分子及其在肿瘤治疗中的应用
KR20180002604A (ko) 혈액 악성종양을 표적으로 하는 키메라 항원 수용체(car), 조성물 및 이의 용도
CN109280086B (zh) 一种肿瘤微环境特异性活化的缺氧诱导型嵌合抗原受体
JP2004537952A (ja) 多重特異的結合分子とその使用
JP2022522654A (ja) Pd-1外部ドメインを担持するキメラサイトカイン受容体
Sadelain T-cell engineering for cancer immunotherapy
US20210403885A1 (en) Chimeric adaptor and kinase signaling proteins and their use in immunotherapy
US20230348556A1 (en) Artificial immunosurveillance chimeric antigen receptor and cells expressing the same
US20230383009A1 (en) Anti-ror1 antibody and ror1-targeting engineered cells
US20050118185A1 (en) Recombinant immunoreceptors
WO1998041613A9 (fr) Cytolyse ciblee de cellules cancereuses
WO1998041613A1 (fr) Cytolyse ciblee de cellules cancereuses
US20220306719A1 (en) Ultramodular igg3-based spacer domain and multi-function site for implementation in chimeric antigen receptor design

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
COP Corrected version of pamphlet

Free format text: PAGES 1/26-26/26, DRAWINGS, REPLACED BY NEW PAGES 1/27-27/27

WWE Wipo information: entry into national phase

Ref document number: 2132349

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1993908414

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1993908414

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 1995 429498

Country of ref document: US

Date of ref document: 19950426

Kind code of ref document: A

WWG Wipo information: grant in national office

Ref document number: 1993908414

Country of ref document: EP