WO1993019163A1 - Chimeric receptor genes and cells transformed therewith - Google Patents

Chimeric receptor genes and cells transformed therewith 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
French (fr)
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 AU39243/93A priority Critical patent/AU668156B2/en
Priority to AT93908414T priority patent/ATE242800T1/en
Priority to DE69333038T priority patent/DE69333038T2/en
Priority to JP51673193A priority patent/JP3643590B2/en
Priority to CA002132349A priority patent/CA2132349C/en
Priority to EP93908414A priority patent/EP0638119B1/en
Priority to US08/084,994 priority patent/US7741465B1/en
Publication of WO1993019163A1 publication Critical patent/WO1993019163A1/en
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) .

Abstract

The disclosed invention relates to chimeric genes which contain a first segment encoding a single chain Fv domain of a specific antibody and a second segment encoding at least the transmembrane and cytoplasmic domains of an immune cell-triggering molecule such as subunits of either a T cell receptor, a T cell receptor-CD3 complex, a Fc receptor or an IL-2 receptor. Also disclosed is a method of treatment of a tumor using lymphocyte cells transformed with expression vectors containing the chimeric genes.

Description

CHIMERIC RECEPTOR GENES AND CELLS TRANSFORMED THEREWITH
Field of the Invention
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.
Background of the Invention
Cells of the immune system are known to recognize and interact with specific molecules by means of receptors or receptor complexes which, upon recognition of an interaction with such molecules, causes activation of the cell to preform various functions. An example of such a receptor is the antigen-specific T cell receptor complex (TCR/CD3) .
The T cell receptor for antigen (TCR) is responsible for the recognition of antigen associated with the major histocompatibility complex (MHC) . The TCR expressed on the surface of T cells is associated with an invariant structure, CD3. CD3 is assumed to be responsible for incracellular signalling following occupancy of the TCR by ligand.
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/β heterodimers or -γ/ S heterodimers. T cells are capable of rearranging the genes that encode the a, β , 7 and S chains of the T cell receptor. T cell receptor gene rearrangements are analogous to those that produce functional immunoglobulins in β 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/β or /δ heterodimer is expressed on the surface of the T cell in association with four invariant peptides. These are the 7 , 5 and ε subunits of the CD3 complex and the zeta chain. The CD3 7. and ε 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 .
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 , β , 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 β heterodimers are covalently linked through disulphi e binds, but many 7 δ receptors associate with one another non-covalentl . The zeta chain quantitatively forms either disulphide-linked ς -η 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 FcεR, for IgA as FcαR, etc. Structurally distinct receptors are distinguished by a Roman numeral, based on historical precedent. We now recognize three groups of FC7RS, designated FC7RI. FC7RII, and FC7RIII. Two groups of FcεR have been defined; these are referred to as FcεRI and FcεRlI. Structurally related although distinct genes within a group are denoted by A, E, C. Finally.. the protein subunit is given a Greek letter, such as Fc7RIIIAα, FC7RHIA7.
Considerable progress has been made in the last three years in defining the heterogeneity for IgG and IgE Fc receptors (FC7R, FcεR) through their molecular cloning. Those studies make it apparent that Fc receptors share structurally related ligand binding domains, but differ in their transmembrane and intracellular domains which presumably mediate intracellular signalling. Thus, specific FC RS on different cells mediate different cellular responses upon interaction with an immune complex. The structural analysis of the FC7RS and FcεRI 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 FcεRI.
The low affinity receptor for IgG (FC7RIIIA) , is composed of the ligand binding CD16α (Fc7RIIIAα-) polypeptide associated with the 7 chain (FC7RIHA7) . The CD16 polypeptide appears as membrane anchored form in polymorphonuclear cells and as transmembrane form (CD16IM) in NK. The FC7RIIIA serves as a triggering molecule for NK cells.
Another type of immune cell receptor is the IL-2 receptor. This receptor is composed of three chains, the a chain (p55) , the β 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) , are very rare, individual-specific, limited in their recognition spectrum and difficult to obtain against most malignancies. Antibodies, on the other hand, 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. In practice, 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
(3) . The use of effector lymphocytes in adoptive immunotherapy. although effective in selected solid tumors, suffers on the other handr from a lack of specificity (such as in the case of lymphokine-activated killer cells (LA cells) (4) which are mainly NK cells) or from the difficulty in recruiting tumor- infiltrating lymphocytes (TILs) and expanding such specific T cells for most malignancies (5) . Yet, the observations that TILs can be obtained in melanoma and renal cell carcinoma tumors, that they can be effective in selected patients and that foreign genes can function in these cells (6) demonstrates the therapeutic potential embodied in these cells. A strategy which has been recently developed (European Published Patent Application No. 0340793, Ref. 7- allows one to combine the advantage of the antibody's specificity with the homing, tissue penetration, cytokine production and target-cell destruction of T lymphocytes and extend, by ex vivo genetic manipulations, the spectrum of a tu or specificity of T cells. In this approach the laborat of the present inventors succeeded to functionally express cells chimeric T cell receptor (cTCR) genes composed of the variable region domain (Fv) of an antibody molecule and the constant region domain of the antigen-binding TCR chains, i the a/β or /δ chains. In this gene-pairs approach, genomi expression vectors have been constructed containing the rearranged gene segments coding for the V region domains of heavy (VH ) and light (VL ) 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 β TCR chains. Following transfection into a cytotoxic T-cell hybridoma, expression of a functional TCR was detected. The chimeric T exhibited the idiotope of the Sp6 anti-TNP antibody and end the T cells with a major histocompatibility complex (MHC) n restricted response to the hapten TNP. 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 VH and VL 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.
Broad application of the cTCR approach is dependent on efficient expression of the cTCR genes in primary T cells. So far, utilizing protoplast fusion, lipofection or electroporation, we succeeded in expressing the cTCR in T cell hybridomas (8, 9) or human T cell tumors, such as Jurkat, but like others, achieved only limited and transient expression in non-transformed murine T cell lines. Although 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 (Cα.VH+Q9VL or Co;VL ÷CβVE ) , 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/ζ 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-Ύ/Γ molecules expressed in cytotoxic lymphocytes could direct specific cytolysis against appropriate target cells (26, 38) . In PCT W092/15322 it is suggested that the formation of chimeras consisting of the intracellular portion of T cell/Fc receptor f, ε or chains joined to the extracellular portion of a suitably engineered antibody molecule will allow the target recognition potential of an immune system cell to be specifically redirected to the antigen recognized by the extracellular antibody portion. However, while specific examples are present showing that such activation is possible when the extracellular portion of receptors such as the CD4 receptor are joined to such ζ , ε or 7 chains, no proof was presented that when a portion of an antibody is joined to such chains one can obtain expression in lymphocytes or activation of lymphocytes.
Summary of the Invention
It has now been found according to the present invention that by fusing a single-chain Fv domain (scFv) gene of a specific antibody, composed of VL linked to VH by a flexible linker, with a gene segment encoding a short extracellular and the entire transmembrane and cytoplasmic domains of a lymphocyte-activation molecule, a chimeric gene is obtained which combines the antibody recognition site and the lymphocyte-signalling moiety into one continuous chain. Upon transfection of such chimeric scFv-receptor (c-scFvR)gene into lymphocytes, it is expressed in the cell as a functional receptor and endows the cells with antibody-type specificity. 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 (VH and VL , 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.
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.
In general terms, the present invention relates to a process for the generation of lymphocytes transfected with an expression vector containing a chimeric gene of the invention. As set out in the following, there was constructed 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. For example, 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. In another embodiment, peripheral blood cells of the patient are genetically engineered according to the invention and then administered to the patient.
Because of the restrictions imposed by corecognition of self MHC plus antigen, the acquisition of new specificity by grafting of TCR genes is limited to inbred combinations. Such manipulations are practically impossible in an outbred population. However, 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 FcεR, a, β and 7 chains of the IL-2R or any other lymphokine receptor, CDlβ a- chain, D2, CD28, and others. Thus, grafting the chimeric genes into NK cells which are not antigen-specific will endow them with antibody specificity.
Description of the Drawings 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 CD16α subunits of the FC7RIII, Co. and Cβ of the TCR, β 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 VH and VL of a specific antibody linked by a flexible linker (hatched box) . B represents the pRSV expression vector containing the kappa light chain leader (Lj. ) , into which the receptor gene prepared from lymphocytes described in C and the gene segment of A are introduced.
Expression of the chimeric gene is driven by the long terminal repeat (LTR) promoter of the Rous sarcoma virus.
Figure 2 illustrates the chimeric pRSVscFvR7 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 (VH ) , the human gamma chain, the G418-resistance gene (neor ) and the simian virus 40 origin of replication.
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.
These primers were designed to match the consensus sequences of VH and VL . The relevant restriction sites are in bold letters.
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 A-Immunoblot analysis of anti-Sp6 precipitates prepared from STB (scFvR7 transfectant cells) , and their parent (MD45.27J hybridoma cells) . After electrophoresis under non-reducing conditions and blotting, the blot was allowed to react with anti-Sp6f anti-human gamma, or anti-mouse ζ antibodies. Panel B - Immunoprecipitation of lysates made of surface-iodinated STB ceils (scFvR7 transfectant cells) and their parent (MD45.27J hybridoma cells) .
Figure 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 51Cr 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 51Cr than the spontaneous release (not shown) .
Figure 8 shows surface expression of chimeric scFvR7/ . T cell hybridoma transfected with the scFvR7 (N2971, N29715) or scFvRr (N29fM.l) chimeric genes composed of the variable region of N29 anti-HER2 mAb, were stained with anti-N29 idiotypic antibodies or control serum (broken lines) and analyzed by FACS. 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) „ 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. As shown, untransfected parental cells MD45.27J did not produce any IL-2 following incubation with Neu/HER2 expressing cells. In 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 lCr 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. Weak lysis of HER2 as compared to the untransfected fibroblasts was observed for N29fl8, while the MD45 and MD45.27 , non-transfected hybridomas did not cause any significant 51Cr release. [filled triangle] , [empty triangle] , -N297l; [filled circle] - N29r 18; [filled square], [empty square] -MD45.27 . This construction is preferably accomplished in the manner shown in Fig. 1 at A,- DNA or RNA from antibody forming cells is isolated. cDNA is prepared from mRNA and amplification of the antibody light and heavy variable regions (VH and VL ) by PCR using a VL-5' (Xbal) , VL -3 (Sail), VH-5' (Sail) and VH-3' (BstEII) specific primers. As shown at B, Tc the pRSV2 -neo plasmid a leader sequence from the 38c.13 kappa chain was introduced down stream from the LTR promoter. At C, RNA from T lymphocytes was isolated and from the cDNA prepared the a , β chains of the TCR, , f subunits of the CD3, CDl6α- 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 pRSV2 -neo plasmid the receptor was introduced at the Xbal site of the pRSVneoL*. vector obtaining pRSVneoLκ-R. The amplified VL (digested with XBal-Sall) and VH (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 Cβ .
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 SRα promoter.
Figure 14 shows: A) Schematic representation of rosette formation by T cells expressing the anti-IgE scFvCβ 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. C) Inhibition of rosette formation of transfectants expressing scFvR. The transfectants were incubated with IgE, anti-Fc and anti-MYC, IgG (as negative control) and then with the SRBC-conjugate and counted. D) Rosette formation of the JSB.15 transfectant. E) Rosette formation of the MD.45 derived transfectants expressing the scFvR.MD.45 was used as negative control.
Figure 15 shows: A) Schematic representation of the ELISA used to screen transfectants expressing scFvC3 chimeric gene (R is Cβ ) . Plates were covered with IgE and lysates of the transfectants were added, then anti-human a /β 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. Plates were coated with 2.5 μg/ml of either IgE or anti-CD3 purified antibodies and transfectant cells were incubated in the presence of phorbol 12-mirystate 13-acetate (PMA) , (10 ng/ml) for 20-24 hours. Supernatants were collected and IL-2 production was determined using the IL-2 dependent cell line CTLL. Untransfected JRT.T3.5 cell was used as negative control and controls for the different media were also included in the CTLL assay.
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.
Figure 18 shows specific inhibition of IgE production by cytotoxic hybridoma expressing the anti-IgE scFvR. Spleen cells were stimulated with 20 μg/ml LPS and lOOU/ml IL-4 for four days. At day 4 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.
Figure 21 shows surface staining of RBL cells transfected with scFvR7 or scFvRζ- 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.
Figure 24 shows stimulation of BW5147 cells co- transfected with scFvCD16 and normal 7 chain by immobilized TNP-Fowl 7-globulin (TNP-F7G) . 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.
Figure 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. Cells transfected with scFvCDlδ alone did not stain above the untransfected BW cells- Figure 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. Different BW-scFvR transfectants were incubated with TNP15-F7G coated wells for 24 hrs. IL-2 was determined by the MTT colorimetric assay. The abscissa describes the concentrations of TNP-F7G used to coat the wells of a microtitre plate. BWG are SCFVR7 transfectants and BWZ are scFvR transfectants.
Detailed Description of the Invention
To overcome the difficulties of the prior method involving the gene-pairs approach (the "T-body" approach) and to extend its applicability to other cells and receptor molecules, a new alternative design was developed according to the invention. It is based on a single-chain approach to the cTCR and on the demonstrated ability to express in bacteria an antibody single-chain Fv domain (scFv) (16, 17) . Such scFv domains, which join the antibody's heavy and light variable (VΞ and VL ) gene segments with a flexible linker, have proven to exhibit the same specificity and affinity as the natural Fabf fragment. Thus, one immediate application of the scFv is to construct chimeric molecules composed of scFv linked to one of the TCR constant domains,
According to the invention, 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.
Thus, 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. In fact, 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. 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, β , 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 ε CD3 chains. Among the polypeptides of the TCR/CD3 (the principal triggering receptor complex of T cells) , especially promising are 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.
Additional examples of immune cell trigger molecules are any one of the IL-2 receptor (IL-2R) p55 (a) ox p75 (β ) 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.
In the group of NK-stimulatory receptors the most attractive candidates are the 7- and CD16α-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) . In NK cells, macrophages, and B and T cells, the FCΎRIII appears as a heterooligomeric complex consisting of a ligand-binding a chain associated with a disulfide-linked 7 or zeta chain. The FC7RHIA signalling gamma chain (22) serves also as part of the FcεRI 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) . Most recently prepared chimeras between these polypeptides and the CD4 (25) , the CD8 (26) , IL-2 receptor chain (27) or CD16 extracellular domains, proved to be active in signalling T cell stimulation even in the absence of other TCR/CD3 components.
In addition to the receptor molecules discussed above, there are lymphocyte accessory and adhesion molecules such as CD2 and CD28, which transduce a co-stimulatory signal for T-cell activation. These co-stimulatory receptors can also be used in accordance with the present invention.
Besides the specific receptor chains specifically mentioned herein, 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. In vivo, cells expressing these genetically engineered chimeric receptors will home to their target, will be stimulated by it to attract other effector cells, where, by itself, will mediate specific distraction of the target cells. In a preferred embodiment, 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.
In preferred embodiments, 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. Thus, the T-cells can be re¬ targeted in vivo to tumor cells or any other target of choice toward which antibodies can be raised. In this regard, 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.
The term "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. 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. Furthermore, 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.
Those of ordinary skill in the art will further understand that 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. Thus, for example, if the antibody is directed against the specific to the constant portion of IgE, it can serve to eliminate IgE-producing B-cellε 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.
The 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. Second, the scFvR design can be employed to confer antibody specificity on a larger spectrum of signaling molecules composed of only one chain. Third, the scFv maintains both VH and VL together in one chain; thus, even upon mixed pairing of chimeric with endogenous chains, the antigen- binding properties of the molecule are conserved. Finally, the fact that gamma and zeta constitute the signaling chains of the TCR/CD3, the FC7RIII and the FcεRI 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.
The chimeric scFvR7 of the invention or any of the simple modifications thereof described below, that combines the specificity of an antibody as a continuous single-chain and the effector function of cytotoxic T cells and NK cells or regulatory function of helper T cells, constitute an important consequential development for targeted immunotherapy. 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.
Candidate immune cells to be endowed with antibody specificity using this approach are: NK cells, lymphokine- activated killer cells (LAK) , cytotoxic T cells, helper T cells, and the various subtypes of the above. These 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. This approach can be applied also to anti-idiotypic vaccination by using 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.
EXAMPLES Example 1Ϊ Constructions aπf. Repression of the Chimeric Sc vR-y/C chain genes
In this example, the following materials and methods were used.
A. Cell lines and antibodies. MD.45 is a cytolytic T-lymphocyte (CTL) hybridoma of BALB/c mice allospecific to H- 2b (29). MD45.27J is a TCR α- 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.
B. Constructions of c-b±meric genes. All the recombinant DNA manipulations were carrier out as described in updated editions of Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, New York, and Ausubel et al. (1987) Current Protocols in Molecular Biology. John Wiley & Sons. The specific genes encoding the VE and VL 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. In constiructing the scFv we used the VL -linker-VH design containing a linker sequence similar to linker 212 described by Colcher et al. (34). Accordingly, the VL-3' and the VH-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. Following digestion of the purified PCR products with Xba I and Sal I (VL ) and Sal I and BstEII (VH ) , the fragments were ligated into the Xba I and BstEII sites of a pRSV2neo-based expression vector containing the leader of SC15 kappa light chain (provided by S. Levy) and TCR constant region β chain {Cβ ) , prepared for the expression of anti-38C13 cDNA cTCR genes (12) . The Cβ of this plasmid was then replaced with either the gamma chain amplified from a human cDNA clone (35) or the zeta chain amplified from Jurkat cDNA by using primers introducing BstEII and Xho I at the 5' and 3' ends. A schematic diagram of the final scFvR7 expression vector is depicted in Fig. 2. The sequences of the oligodeoxynucleotide primers used for the construction of the chimeric scFvR7 and scFvRf are delineated in the legend to Fig. 2.
TABLE I
Figure imgf000026_0001
R-A or G S=C or G TACGTA SnaBI GTCGAC Sail GGATCC BamHI Y=C or T K=G or T CTCGAG Xhol GGTGACC BstEII AAGCTT HindiII =A or T M=A or C TGTAGA Xbal GAATTC EcoRI GGTACC Kpnl
Primers for cDNA Reverse Transcription
SEQ. ID.NO. 1 mouse Ccl 5' GGCCAGTGGATAGAC 3' SEQ.ID.NO. 2 mouse kappa 5' GATGGTGGGAAGATG 3' SEQ.ID.NO. 3 rat Cc3 5' CCATGRYGTATACCTGTGG 3'
Primers for Single chain Fv
5' CCCGTCTAGAGGAGAYATYGTWATGACCCAGTCTCCA 3'
5' CCCGTCGACCCTTT ATTTCCAGCTTWGTSCC 3'
5' CGGGTCGACTTCCGGTAGCGGCAAATCCTCTGAAGGCAAAGGTSAGGTSCAGCTGSAGSAGTCTGG
Figure imgf000026_0002
5' TGMRGAGACGGTGACCGTRGTYCCTTGGCCCCAG 3'
Receptor primers
GCTCGAGATAAAAAATATCCAGAACCCTGACCCTGCC 3' CGGTCACCGTCTCCTCAAATATCCAGAACCCTGACCCTGCC 3' CCTCGAGATAAAAGAGGACCTGAAAAACGTGTTCCCA 3' CGGTGACCGTCTCCTCAGAGGACCTGAAAAACGTGTTCCCA 3' TACGTATCAGCTGGACCACAGCCGCAGCGTCAT 3' TACGTATCAGCCTCTGGAATCCTTTCTCTTGAC 3' CCGGTCACCGTCTCTTCAGCGGATCCTCAGCTCTGCTATATCCTGGATG 3 GGGAGCTGCTCGAGTCTAAAGCTACTGTGGTGG 3' GCTGGATCCCAAACTCTGCTACC 3' CGGCTCGAGCTGTTAGCGAGGGGGC 3' CCGGTCACCGTCTCCTCAGGGTACCAAGTCTCTTTCTGC 3' CCCTCGAGTCATTTGTCTTGAGGGTC 3'
CCGGTCACCGTCTCTTCAGCGGATCCTACGATTCCGTGGCTCGGCCAC 3' GGAGATAGAAGCTTGCCAGG 3' CCTGGCAAGCTTCTATCTCC 3'
Figure imgf000026_0003
GGCTCGAGTCTACACCAAGTGAGTTGGG 3'
C Expression of the chimeric SCFVR-T/C genes.
Transfection of 20μg of pRSVscFvR7/f DNA into 20xl06 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. 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 xCr release (29). All determinations were performed in triplicate.
D. Imrπunoprecipitation and immunoblotting. Washed pellets containing 108 cells were lysed in 1 ml of 1% digitonin, 0.12% Triton X-100 in lOmM Tris.HCl - saline buffer pH 7.4 containing lOmM EDTA, ImM phenylmethyi sulfonyl fluoride (Sigma) , 10 μg/ml aprotinin and 10 μg/ml leupeptin (Boehringer Mannheim, GmbH) . After 20 min at 0°C and centrifugation at 12000Xg for 15 min, aliquots of the supernatants were incubated with antibodies and then precipitated with second antibodies and Protein G - Sepharose (Pharmacia) as described (32) .
Alternatively, 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. To avoid destruction of the Sp6 idiotope, 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) .
E„ Results. To produce a chimeric receptor with an antigen binding site of a given antibody and the signalling gamma or zeta chains, we have adopted the scFv design (16, 17) which allows combining both entities into one continuous molecule. In engineering the pRSVscFvR7/r expression vector (Figs. 1,2), harboring the VL and VH of the Sp6 anti-TNP mAb (37) , we introduced elements that enable its usage as a modular expression cassette to accommodate scFvs from different antibodies in combination with gamma, zeta or other chains. This was achieved by using oligonucleotide primers composed of sequences common to the majority of the 5' and 3' sequences of either VL or VH 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) . We have chosen to use the 5' -VL -linker-VE-3' design, which was found suitable for the expression of a variety of single-chain antibodies and their fragments in bacteria (17) , but the converse, 5' -VH -linker-VL -3' , alignment (16) can be used as well.
Introduction of the chimeric scFvR7 gene into the MD.45 murine CTL hybridoma (STA series of transfectants) or its MD45.27J TCRα.- mutant, which does not express surface TCR/CD3 complex (STB series) , resulted in the expression of the chimeric molecule on the cell surface of selected clones as revealed by staining with the anti-Sp6 idiotypic antibody (Fig. 3) . Similar staining was observed for STZ, which was derived by transfecting MD45.27J with the scFvRf chimeric gene. 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 scFvR7 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) . Immunoblo ting analysis of cell lysates prepared from representative STA and STB transfectants using either antiidiotypic mAb 20.5 or polyclonal anti-human gamma antibodies, revealed 4 distinct bands of apparent molecular weight of 36, 54-62, 74-80 and 85-90 kDa, which did not appear in the parental cells (Fig. 4) . Under reducing conditions one species, which corresponds to the predicted 36 kDa monomeric form of the scFvR7, was apparent, indicating the multimeric nature of the molecule. The band with apparent 75 kDa molecular weight corresponds to the homodimeric molecule, and the nature of the 90 kDa species is unknown. It might represent a novel gamma-associated polypeptide, analogous to the one recently reported (31) . This species can be detected only in immunoblots of cell lysates and is not apparent after surface iodination and immunoprecipitation (Fig. 5B) , suggesting the intracellular origin of the molecule. The appearance of bands in the range of 54-62 kDa was more pronounced in the STB transfectant. It might represent heterodimers between the chimeric scFvR7 chain and endogenous zeta and probably eta chains of the CD3 complex. We therefore electrophoresed anti-Sp6 immunoprecipita es made from STB lysates, blotted the gels, and developed it with anti-Sp6, anti-gamma or anti-mouse zeta/eta antibodies (Fig. 5A) . Both the anti-idiotypic and the anti-gamma antibodies revealed the four bands from the transfected cells; however, the anti-zeta (which cross-reacts with the mouse eta chain) differentially developed only the 60 kDa species. Immunoprecipitation of surface-iodinated proteins with either anti-Sp6 or anti-gamma antibodies (Fig. 5B) demonstrates a main species of 75 kDa under non-reducing conditions. This is the homodimer of the chimeric chain.
Example 2: Expression of scFvR-y/C as functional receptors
To test whether the chimeric scFvR7 or scFvRf can function as an active receptor molecule, we studied the ability of the transfected hybridomas to undergo antigen-specific stimulation. 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-2b target cells (29) , while its MD45.27J mutant cannot be stimulated through its TCR due to the absence of an a chain. Upon introduction of the chimeric Sp6-scFvR7, both of these cells could be specifically triggered to produce IL-2 following incubation with TNP-modified stimulator cells (Fig. 6A) or plastic- immobilized TNP-fowl gamma globulin (TNP-F7G) (Fig. 6B) . 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. While STA responded to plastic-bound TNP-F7G in consistent manner, STB and STZ (transfected with the scFvR7 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. Indeed, costimulation with TNP-F7G plus either phorbol 12-myristate 13-acetate (PMA) or Ca*'1" ionophore resulted in enhancement of IL-2 production (data unshown) . Incubation with soluble TNP- proteins even at high hapten/protein ratios did not result in activation but rather specifically inhibited triggering by immobilized antigen (Fig. 6B) or cell-bound hapten. The activation of GTAe.20, a transfectant expressing a two-chain chimeric TCR (9) , was also inhibited by soluble TNF-F7G.
Identical concentrations of antigen were needed to cause 50% inhibition (IC50) of STA and GTAe.20 (Fig. 6B) , indicating that the single-chain and the double-chain Fv display the same relative affinity to TNP. Finally, we tested the ability of the chimeric receptors to mediate specific target cell lysis by incubating them with 5 Cr labeled cells. As shown in Fig. 7, only the cells transfected with the Sp6-scFvR7 or -scFvRr could lyse TNP-modified target cells in a dose-related fashion. This cytolytic activity was specific to TNP as soluble TNP-F7G blocked it (not shown) and unmodified A.20 cells were not affected by the transfectants. It is demonstrated here for the first time that 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. 5 and 6), strongly suggests that the gamma and zeta chains are capable of autonomous activation of T cells. Yet, because of the low level of heterodimers between the scFvR7 and the endogenous zeta and eta chains (Figs. 3 and 4), the possibility of some contribution by the residual zeta (or eta) chain in the signalling process cannot be excluded. Nonetheless, the present results clearly indicate that the TCR chains do not take part in this process, thus confirming and complementing recent observations in which antibody cross-linking through the extracellular domains of CD4, CD8, IL-2 receptor, or CD16 joined to the cytoplasmic tail of either one of the gamma/zeta family members resulted in T cell activation (26-28) . Like scFvR7/f , chimeric CD4 or CD16-gamma/zeta molecules expressed in cytotoxic lymphocytes could direct specific cytolysis against appropriate target cells (26, 38) . Analysis of mutations within the intracellular 18-residue motif, which has been recently assigned to account for the activity of the gamma/zeta chain, revealed that the ability to mediate calcium responsiveness can be separated from the ability to support cytolysis (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. The finding that immobilization of antigen is needed for efficient stimulation through scFvR /f and that soluble multimeric ligand (such as TNP-protein) did not trigger, but rather inhibited, receptor-mediated activation through cell- cr plastic-bound TNP (Fig. 5B) , indicates that mere engagement or even cross-linking of adjacent gamma or zeta chains does not result in T cell activation (as manifested by IL-2 release) . The dependence on ligand immobilization for efficient T cell triggering has been reported also for cTCR-mediated signalling (8, 9) , and the mechanisms underlying this are as yet unclear. Using the hybridoma transfected cells, different variants were obtained which differ in their ability to respond to immobilized antigen or to TNP-modified stimulator cells of various origin. Because these variants express surface receptors and respond to stimuli which bypass the TCR (such as with PMA + Ca++ ionophore) , it was reasoned that they are deficient in one of the components along the pathway leading to the costimulatory signal, required for optimal cytokine release (39) . Indeed, the fact that the addition of either PMA or ionomycin to the immobilized antigen increased the response of most of these clones (not shown) , strongly support this assumption.
Example 3s Targeting of Cytolytic Lymphocytes to Neu/HER2 Expressjtier Cells Using Chimeric Single-Chaiπ Fv Receptors
Cell surface molecules essential for the transformed phenotype or growth of malignant cells are attractive targets for anti-cancer immunotherapy. Antibodies specific to
Neu/HER2, a human adenocarcinoma-associated growth factor receptor, were demonstrated to have tumor inhibitory capacity.
Yet, the inefficient accessibility of antibodies to solid tumor limits their clinical use. To redirect effector lymphocytes tc adenocarcinomas, we constructed and functionally expressed in T cells chimeric single-chain receptor genes incorporating both the antigen binding domain of anti-Neu/HER2 antibodies and the
7 or ζ signal transducing subunits of the T cell receptor/CD3 and the immunoglobulin Fc receptor complexes.
Surface expression of the anti-Neu/HER2 chimeric genes in cytotoxic T cell hybridomas endowed them with specific Neu/HER2 recognition enabling their activation for interleukin-2 production and lysis of cells overexpressing Neu/HER2. These chimeric genes can be used for the immunotherapy of cancer.
To establish the feasibility of the chimeric recept approach to retarget cytolytic lymphocytes to tumor cells, we have used anti-Neu/HER2 antibodies. 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. Out of a panel of monoclonal antibodies (mAbs) specific to the extracellular portion of the Neu/HER2 protein (41) , we selected 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) . In this example, we show that 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.
In this example, the following materials and method were used.
A„ Cells and Antibodies, 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. CNCM 1-1262. Anti-N29 idiotypic antiserum was prepared b immunizing rabbits with purified N29 protein and adsorption o the immune serum on a normal mouse Ig-agarose column. Rabbit anti-CD3_T and anti-FcεR y antibodies were kindly provided by Dr. J. -P. Kinet. B„ Construction and Trans ection of Chimeric Genes,
Chimeric scFvN29R7 or scFvN29R genes were constructed from single chain Fv DNA (in the VL-linker-VH alignment), amplified by PCR from cDNA prepared of hybridoma producing the N29 anti- HER2 mAb, and either γ 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 μg 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. Detection of Soluble Receptor, Cell lysates were prepared from the transfectants by adding 100 μl 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 μg/ml aprotinin and 10 μg/ml leupeptin (Boehringer Mannheim, GmbH) to a pellet of 5xl06 cells. -After 30 min. at 0°C and centrifugation, the nuclei-free supernatant was added to wells of a microtitre plate precoated with 5 μg/well of purified HER2X protein. 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 Iμg/ml of anti-human ζ 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 OD6 g 0. D, IL-2 Production and Cytotoxic Assays.
Stimulator cells (3xl04 /well) were cultured in 96-well microculture plates for at least 6 hours until adherent. For stimulation of transfectants with purified HER2X, 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 (105/200 μl/well) in DMEM supplemented with 10% fetal calf serum and 10" 5 M of 2-/9- mercaptoethanol. Following 20-24 hrs in culture, 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) . To measure the cytotoxic activity, the transfectants and their parental hybridomas were co- incubated with 51Cr labeled target cells at various effector to target ratios for 16 hrs. The 51Cr release assay was performed as described previously (16) .
E, Results„ Genes coding for single chain Fv of N29 fused to either human 7 or f chains were prepared in the pRSVscFvR vector, and used to transfect the murine MD45 allospecific CTL hybridoma or its TCR α-mutant MD45.27J which does not express the TCR/CD3 complex. Surface expression of the chimeric chains on the hybridoma cells was detected using anti-idiotypic antibodies specific to the N29 anti-Neu/HER2 mAb (Fig. 8) . The integrity of the 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 ζ 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. Three transfectants, N29Ύ1 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. Incubation of the scFvR-expressing cells together with human cancer cells, which express Neu/HER2 on their surface, resulted in a marked activation as measured by the production of IL-2 (Fig. 10a) . 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 51Cr release assay. When 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. Following incubation with an scFvR7-expressing T cell hybridoma (N297D (Fig. 12) , a substantial level of specific lysis was obtained. The scFvRf expressing hybridoma (N29_*18) gave only a marginal specific 51Cr release signal when compared with the untransfected hybridomas. The cytolytic effect was Neu/HER2- specific, since untransfected NIH-3T3 fibroblasts did not undergo killing. Likewise, the parental MD45.27J cells did not cause any significant 51Cr release. The high levels of spontaneous 51Cr 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 51Cr release from human tumors (such as SKBR-3 breast and N87 gastric carcinoma cell lines, Fig. 11) , than the parental hybridomas.
This study demonstrates that T cells expressing chimeric receptor genes utilizing single-chain Fv of anti-tumor 5 antibodies can be redirected to tumor cells. Binding of the scFvR to the tumor antigen either in its isolated, immobilized form or in a cellular context was sufficient to trigger T cell activation and mediate target cell lysis. These results extend the previous examples using the anti-TNP scFvR7/f . In all 0 these instances, activated T cells or T cell lines have been used.
Example 4- Frnir-i-ional Expression of scFvR with Anti-IgE Specificity ς Allergic diseases are characterized by elevated synthesis of IgE upon stimulation by environmental allergens. 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 Fcε receptors (FcεRI) on mast cells and basophils, thus sensitizing them. Following encountering allergen the FcεRI-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. In this example, we take advantage of the fact that both 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. The present example demonstrates the feasibility of this approach in an in vi tro system, utilizing anti-mouse IgE antibodies.
In this example, the following materials and methods 5 were used. A, Generation and transfection of anti-IgE specific scFvR genes. The antibody which has been chosen is 84.1c, a rat mAb specific to an epitope on the murine Cε3 (43) . The advantage of using 84.1c mAb is that it reacts with free IgE and not with mast-cell bound IgE, thus it was reasoned that it recognizes an epitope closely related to the FcεRI binding site on IgE. The basic strategy for construction of the chimeric genes encoding the 84.1c mAb VL and VH in a continuous single chain Fv linked to the constant region of the TCR a or β chains (Cα or Cβ) 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'Cκ and 3'CH heavy primers employing "M-MLV-reverse transcriptase (BRL) . We amplified the VH and Vκ by PCR using the mouse consensus oligonucleotide primers similar to the ones described above for the Sp6 anti- TNP scFv (44) . The Vκ-3' primer and the VH-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. Following digestion of the purified PCR product with Xba I and Sal I (Vκ) and Xba I and BstE II (VH) , the fragments were ligated and introduced into the Xba I and BstE II sites of PRSVLJ CQ! or Cβ expression cassettes. These 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 pRSV2neo the leader of the 38c.13 K-light chain 3' to the RSV LTR and downstream, either the Co. or Cβ of the human TCR. The Cα. and Cβ were PCR- amplified from human TCR clones using primers 9 and 12 from Table I for Co; and 11 and 13 for Cβ. Because we found previously (46) that the SRα promoter (47) drives transcription in T cells better than the RSV LTR, we adopted it here for the anti-IgE scFvR expression. For this purpose, we used the pBJlneo plasmid (47) . We cut out the complete scFv at the SnaBI sites from the pRSVscFvQ3/Cα vectors and introduced it into the EcoRV of the pBJlneo vector. Fig. 13 describes the construction of the SRα based vector (pBJ-sc84. β ) .
The 84.1c based scFvQS chimeric gene was introduced into either the murine MD45 hybridoma (29) or the human Jurkat T cell leukemia β 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.
B. Expression of the anti-IgE scFvR in T cells. To determine the integrity of the chimeric genes, their ability to encode for surface receptor and to study the molecular nature of the receptor, we first transfected the Cβ based chimeric gene into the human leukemic Jurkat cell mutant JRT3.5, lacking the TCR β chain. In the absence of anti-84.1c idiotype antibodies', we screened the transfectants obtained for the reappearance of surface CD3 by immunofluorescence, using anti- CD3 antibodies, expecting that the chimeric scFvCS chain will associate with the endogenous TCR a chain and bring out the TCR/CD3 complex. Although in parallel experiments we could bring out the CD3 to the surface of JRT3.5 cells following transfection with VHCo. or VLCα cTCR chains, we could not demonstrate any CD3-specific staining of transfectants receiving the scFvC3 gene (data unshown) . We therefore monitored surface expression of the chimeric scFv genes in the transfectants by rosette formation using trinitrophenylated sheep red blood cells (TNP-SRBC) coated with anti-TNP IgE SPE-7 (49) . Fig. 14 represents such experiments depicting the rosettes and showing that they are specific to IgE and could be inhibited by adding IgE (and not control IgG antibody, Fig. 14C) . That the chimeric receptors contain both the antigen- binding moiety and TCR determinants in the same complex, was 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. Figure 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.
Next we checked, using the cytotoxic MD45 T cell hybridoma whether the scFvCβ can arm and trigger cytotoxic cells to eliminate IgE producing cells (known to express IgE on their surface) . To mimic the in vivo situation, as target cells we used murine splenic lymphocytes which were induced to produce IgE by cul uring them in the presence of lipopolysaccharide (LPS) and IL-4. LPS÷IL-4 are known to induce Ig class switch in B cells and specifically trigger IgE and IgGi formation (50) . In the experiments described in Fig. 18, we coincubated MD45 transfectants expressing the anti-IgE scFvQ3 with murine lymphocytes, added LPS+IL-4 and monitored both IgE and IgG accumulation into the supernatants of these cultures. As shown in the figure, IgE secretion was completely abrogated in cultures containing the scFvQS T cells. The effect was very specific as no effect on IgG production could be observed. The suppression of IgE production was most likely due to elimination of IgE producing cells by the redirected scFvCø-bearing CTL hybridomas. The inability of 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.
Example 5: Endowing R-nt-fhody Specificity to the Low Affinity Fc-rR (Fc-τRIIX) Using Chimeric scFv Joined to the CDlβα Chain
One of the most attractive candidates for the chimeric receptor approach in natural killer (NK) cells is the low affinity receptor for IgG ( (FC7RIHA) which is composed of the ligand binding CD16α polypeptide associated with the 7 chain (51, 52) . Triggering of NK cells via FC7RIII (either by anti-CD16 or through immune complexes) 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 (CD16IM) in NK (54). The FC7RHI- associated 7 chain serves also as part of the FcεRI 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). Like r and η , 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, ζ or η have been recently reported in studies mapping the regions of these molecules which take part in the signaling process.
It has been shown in previous examples that the binding domain of a specific antibody in the form of an scFv can serve as the recognition unit of the CD3 f (see also 44) , TCR Cβ and Fc-εRi/Fc7RIH 7 (44) . In the present example we report successful experiments in which we constructed and functionally expressed chimeric receptors composed of scFv and anti-TNP and the CD16α polypeptide of the FCΎRIII.
In this example, the following materials and methods were used. A. Design ^τι r-onstruction of chimeric scFv-CDlβo.,
For the scFv-CD16α 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 CD16α (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. Bo Expression of the chimeric scFv-CDlβg, A. Expression in mast cells. Since the
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 FcεRI (56) . These cells produce excess of 7 chain as part of the FcεRI and provide us with convenient function as the receptor-triggered degranulation assay. Following electroporation of the chimeric scFv-CD16α as well as the scFvR? and scFvRf genes and selection in G418, 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. Upon the addition of TNP-protein conjugates to the SCFVRT and scFvRf expressing RBL transfectants, cross-linking of adjacent receptors by the multivalent antigen triggered degranulation as measured by specific release of β-hexoseaminidase to the supernatant (Table II) .
Figure imgf000043_0001
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 on the other hand, 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) .
B. Expression in BW5147 cells. BW5147 is a murine thymoma which does not express surface TCR/CD3 because it does not transcribe either 7 or r chains. As expected, transfection of the BW5147 cells with chimeric scFvCDlδ DNA did not yield any detectable surface receptor, yet intracellular receptor could be detected by immunoblotting of lysates (not shown) . When 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-F7G) (Fig. 23, 24) .
Finally, we checked whether the chimeric scFvIL2R gene (made of the scFv of Sp6 and the β chain of the IL-2 receptor) can be expressed following transfection on the surface of RBL cells. 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 β -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. These results clearly demonstrate that the chimeric scFvIL2R can be expressed as a surface protein.
Example 6; Expression of Chimeric Single-Chain Fv Receptors
IT. BWTi.4-7 Thymoma
-Ti.T-r-t-ir-T-al Expression in BW5147 Thymo-T-a Cells
BW5147 (BW) is a murine thymoma cell line which do not express the TCR or FC7R 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 scFvR7 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, γ 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. In parallel, 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. As can be seen, both BW.Sp6-7 and BW.Sp6-f transfectants (which received weither scFvR7 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. When studied for CD3 expression, using 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) . However, the co-transfection of scFvCD16 and the 7 chain DNA yielded transfectants, like the BW.Sp6-CD16 shown in Fig. 26, which express the chimeric receptors. These results clearly prove that the CD16 chimeric chain was not sufficient for itself and needed the 7 chain for surface expression.
To study whether the chimeric receptors function in the BW cells, we tested the ability of transfectants to undergo stimulation for IL-2 production following stimulation with TNP modified A.20 cells (Fig. 27) or immobilized T P-F7G (Fig. 28) . Although BW cells do not produce any IL-2 following incubation with TNP-labeled antigen, the single-chain receptor expressing transfectants produced IL-2 following stimulation with either cellular or solid-phase antigen.
Taken together these studies demonstrate the appropriate expression of the chimeric chains as functional receptors: they bind ligand with antibody-type specificity on one end and signal for T cell stimulation on the other end. Although we have demonstrated here expression of the chimeric single chain receptors in non-TCR expressing T cells, it is reasonable to expect that natural killer cells, which make use of 7 and CD16 in their signaling FC7 receptor will behave in a similar way. All references cited herein, including journal articles or abstracts, published or corresponding U.S. or foreign patent applications, issued U.S. or foreign patents, or any other references, are entirely incorporated by reference herein, including all data, tables, figures, and text presented in the cited references. Additionally, the entire contents of the references cited within the references cited herein are also entirely incorporated by reference.
Reference to known method steps, conventional methods steps, known methods or conventional methods is not in any way an admission that any aspect, description or embodiment of the present invention is disclosed, taught or suggested in the relevant art.
The foregoing description of the specific embodiments will so fully reveal the general nature of the invention that others can, by applying knowledge within the skill of the art (including the contents of the references cited herein) , readily modify and/or adapt for various applications such specific embodiments, without undue experimentation, without departing from the general concept of the present invention. Therefore, such adaptations and modifications are intended to be within the meaning and range of equivalents of the disclosed embodiments, based on the teaching and guidance presented herein. It is to be understood that the phraseology or terminology herein is for the purpose of description and not of limitation, such that the terminology or phraseology of the present specification is to be interpreted by the skilled artisan in light of the teachings and guidance presented herein, in combination with the knowledge of one of ordinary skill in the art.
REFERENCES l) Lowder, J.N. et al. Cancer Surv. 4:359-375 (1985)
2) Waldmann, T.A. Science 252:1657-1662 (1991)
3) Jain, R.K. J. Natl. Cancer Inst. 81:64-66 (1989)
4) Mule, J.J. et al. Science 225:1487-1489 (1984)
5) Rosenberg, S.A. et al. Science 233:1318-1321 (1986) 6) Rosenberg, S.A. J. Clin. Oncol. i_:180-199 (1992)
7) Kuwana, Y. et al. Biochem. Biophys. Res. Comm. 149.:960-968 (1987) Gross, G. et al. Transplant. Proc. 21:127-130 (1989) Gross, G. et al. Proc. Natl. Acad. Sci. USA .86.:1002410028
(1989)
Becker, M.L.B. et al. Cell 5_8;9H-921 (1989)
Goverman, J. et al. Cell 6j3:929-939 (1990)
Gross, G. Ph.D. thesis (Weizmann Institute of Science,
Rehovot, Israel)
Culver, K. et al. Proc. Natl. Acad. Sci. USA JL8:31553159
(1988) Kasid, A. et al. Proc. Natl. Acad. Sci. USA 87:473-477
(1990)
Adam, M.A. et al. J. Virol. 6_5.:4985-4990 (1991) Huston, J.S. et al. Proc. Natl. Acad. Sci. USA 85:58795884
(1988)
Bird, R.E. et al. Science 241:423-426 (1988) Weissman, A. et al. EMBO J. -3:3651-3656 (1989) Bauer, A. et al. Proc. Natl. Acad. Sci. USA 88:3842-3846
(1991) Unkeless, J.C. et al. Annu. Rev. Immunol. -6:251-281
(1988)
Ravetch, J.V. and Kinet, J.-P. Annu. Rev. Immunol. 9.:457-
492 (1991)
Wirth uller, U. et al. J. Exp. Med. 175:1381-1390 (1992)
Orloff, D.G. et al. Nature (London) 342:189-191 (1990)
Lanier, L.G. et al. J. Immunol. 146.:1571-1576 (1991)
Vivier, E. et al. J. Immunol. 147:4263-4270 (1991)
Romeo, C. and Seed, B. Cell 64:1037-1046 (1991)
Irving, B.A. and Weiss, A. Cell j54.:891-901 (1991)
Letourneur , F. and Klausner, R.D. Proc. Natl. Acad. Sci.
USA -38.: 8905-8909 (1991)
Kaufmann, Y. et al . Proc. Natl. Acad. Sci. USA 78:2502-
2506 (1981)
Rusconi, S. and Kohler, G. Nature (London) 3_14_:330-334
(1985)
Figure imgf000047_0001
Schoneich, J. et al . J. Immunol. 148:2181-2185 (1992) 32) Eshhar, Z. et al. in Practical Approach to Tumor Immunology , ed. G. Gallagher (IRL, Oxford) in press (1992)
33) Kabat, E.A. et al. Sequences of Proteins of Immunological Importance (Dept. of Health and Human Services,
Washington, D.C.) 4th ed. (1987)
34) Colcher, D. et al. J. Nat. Cancer Inst. 82:1191-1197 (1990)
35) Kuster, H. et al. J. Biol. Chem. 265.:6448-6452 (1990) 36) Mosmann, T. J. Immunol. Methods 65_:55-63 (1983)
37) Kohler, G. and Milstein, C. Eur. J. Immunol. 6:511-519 (1976)
38) Romeo, C. et al. Cell 68_:889-897 (1992)
39) Weaver, C.T. and Unanue, E.R. Immunol. Today 11:49-53 (1990)
40) Eshhar, Z. and Gross, G. Br. J. Cancer 62, ISuppl. X), 27- 29 (1990)
41) Stancovski, I. et al. Proc. Natl. Acad. Sci. U.S.A. 88_:8691-8695 (1991) 42) Bacus, S. et al. Cancer Res. .52:2580-2589 (1992)
43) Schwarzbaum, S. et al. Eur. J. Immunol. JL9_:1915 (1989)
44) Eshhar, Z et al. Proc. Natl. Acad. Sci. USA 9_0:720 (1993)
45) Gross, G. and Eshhar, Z. FASEB 6:3370 (1992)
46) Yablonski, D. "Transfection and functional expression of r. cell receptor genes". M.Sc. Thesis presented to the
Feinberg Graduate School, The Weizmann Institute of Science, Rehovot, Israel (1987)
47) Lin, A. et al. Science 249.:677 (1990)
48) Weiss, A. and Stobo, J. J. Exp. Med. 160:1284 (1984) 49) Eshhar, Z. et al. J. Immunol. 124:775 (1980)
50) Snapper, C. et al. J. Immunol. 141:489 (1988)
51) Lanier, L. et al. Nature 342:803 (1989)
52) Anderson, P.M. et al. Proc. Natl. Acad. Sci. 87:2274
(1990) 53) Cassatella, M.A. et al. J. Exp. Med. 169:548 (1989) 54) Ravetch, J.V. and Perussia, B. J. Exp. Med. 170:481 (1989) 55) Lustgarten, J. and Eshhar, Z., in preparation
56) Barsumian, E.L. et al. Eur. J. Immunol. .11:317 (1981)]
57) Wegener, A.M. et al. Cell 68_:83 (1992)

Claims

1. A chimeric gene comprising a first gene segment encoding a single-chain Fv domain (scFv) of a specific antibody and a second gene segment encoding at least the transmembrane an cytoplasmic domains of an immune cell-triggering molecule which, upon transfection to immune cells, expresses the antibody- recognition site and the immune cell-triggering moiety into one continuous chain.
2. A chimeric gene according to claim 1 wherein the second gene segment further comprises partially or entirely the extracellular domain of the immune cell-triggering molecule.
3. A chimeric gene according to claim 1 or 2 wherein the first gene segment encodes the scFv domain of an antibody against tumor cells.
4. A chimeric gene according to claim 1 or 2 wherein the first gene segment encodes the scFv domain of an antibody against virus infected cells.
5. A chimeric gene according to claim 4 wherein the virus is HIV.
6. A chimeric gene according to claim 1 to 5 wherein the second gene segment encodes a lymphocyte receptor chain.
7. A chimeric gene according to claim 6 wherein the gene encodes a chain of the T cell receptor.
8. A chimeric gene according to claim 7 encoding a subunit of the T cell receptor.
9. A chimeric gene according to claim 8 comprising a gene segment encoding the a, β , i or δ chain of the antigen- specific T cell receptor.
10. A chimeric gene according to claim 1 or 2 wherein the second gene segment encodes a polypeptide of the TCR/CD3 complex.
11. A chimeric gene according to claim 10 encoding th zeta or eta isoform chain.
12. A chimeric gene according to claim 1 or 2 wherein the second gene segment encodes a subunit of the Fc receptor or
IL-2 receptor.
13. A chimeric gene according to claim 12 wherein the second gene segment encodes a common subunit of IgE and IgG binding Fc receptors.
1 . A chimeric gene according to claim 13 wherein sai subunit is the gamma chain.
15. A chimeric gene according to claim 14 comprising gene segment coding for the CD16α chain of the FC7RIII or FC7RII.
16. A chimeric gene according to claim 12 comprising gene segment coding for the a or β subunit of the IL-2 receptor.
17. An expression vector comprising a chimeric gene according to any of claims 1 to 16.
18. An immune cell endowed with antibody specificity transformed with an expression vector according to claim 17.
19. An immune cell endowed with antibody specificity comprising a chimeric gene according to any of claims 1 to 16.
20. An immune cell according to claim 18 or 19 selected from the group consisting of a natural killer cell, a lymphokine activated cell, a cytotoxic T cell, a helper T cell and a subtype thereof.
21. A method of treatment of a tumor in a patient comprising transforming lymphocyte cells of the patient with an expression vector comprising a chimeric gene according to claim 1 in which the first gene segment encodes a scFv domain of an antibody directed against the tumor cells, and administering the transformed and thus activated cells to the patient, said cells being targeted to the tumor cells thus causing tumor regression.
22. A method according to claim 21 wherein peripheral blood cells of the patient are transformed.
PCT/US1993/002506 1992-03-18 1993-03-18 Chimeric receptor genes and cells transformed therewith WO1993019163A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
AU39243/93A AU668156B2 (en) 1992-03-18 1993-03-18 Chimeric receptor genes and cells transformed therewith
AT93908414T ATE242800T1 (en) 1992-03-18 1993-03-18 CHIMERIC RECEPTOR GENES AND CELLS TRANSFORMED THEREWITH
DE69333038T DE69333038T2 (en) 1992-03-18 1993-03-18 CHIMERIC RECEPTOR GENES AND, ACCORDING TO THAT, TRANSFORMED CELLS
JP51673193A JP3643590B2 (en) 1992-03-18 1993-03-18 Chimeric receptor gene and cells transformed with the same
CA002132349A CA2132349C (en) 1992-03-18 1993-03-18 Chimeric receptor genes and cells transformed therewith
EP93908414A EP0638119B1 (en) 1992-03-18 1993-03-18 Chimeric receptor genes and cells transformed therewith
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 (en) 1993-09-30

Family

ID=26322411

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1993/002506 WO1993019163A1 (en) 1992-03-18 1993-03-18 Chimeric receptor genes and cells transformed therewith

Country Status (1)

Country Link
WO (1) WO1993019163A1 (en)

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 (en) * 1994-05-02 1995-11-09 Ciba-Geigy Ag Bifunctional protein, preparation and use
WO1997015669A1 (en) * 1995-10-24 1997-05-01 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Chimeric receptor genes and cells transformed therewith
WO1997020048A2 (en) * 1995-11-30 1997-06-05 Bristol-Myers Squibb Company Modified sfv molecules which mediate adhesion between cells and uses thereof
WO1997023613A2 (en) * 1995-12-21 1997-07-03 Celltech Therapeutics Ltd. Cell activation process and reagents therefor
WO1997035004A1 (en) * 1996-03-15 1997-09-25 Medical Research Council Cell stimulation
EP0821730A1 (en) * 1995-02-02 1998-02-04 Cell Genesys, Inc. Chimeric receptors for regulating cellular proliferation and effector function
WO1998038324A2 (en) * 1997-02-27 1998-09-03 Boehringer Mannheim Corporation Chimeric cell-surface receptors that undergo antigen-induced association
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 (en) * 1998-01-15 1999-07-22 Center For Molecular Medicine And Immunology Antibody/receptor targeting moiety for enhanced delivery of armed ligand
WO1999057268A1 (en) * 1998-05-06 1999-11-11 Celltech Therapeutics Limited Chimeric receptors
US6004811A (en) * 1991-03-07 1999-12-21 The Massachussetts General Hospital Redirection of cellular immunity by protein tyrosine kinase chimeras
WO2000018932A2 (en) * 1998-09-25 2000-04-06 Regeneron Pharmaceuticals, Inc. Receptor based antagonists and methods of making and using
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 (en) * 2000-02-22 2001-08-30 Ahuva Nissim Chimeric and tcr phage display libraries, chimeric and tcr reagents and methods of use thereof
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 (en) * 2012-09-04 2014-03-13 Cellectis Multi-chain chimeric antigen receptor and uses thereof
EP2126054B1 (en) 2007-01-31 2016-07-06 Yeda Research And Development Company Limited Redirected, genetically-engineered t regulatory cells and their use in suppression of autoimmune and inflammatory disease
WO2017123808A1 (en) 2016-01-15 2017-07-20 The J. David Gladstone Institutes Methods of treating disease by metabolic control of t-cell differentiation
US10342829B2 (en) 2012-05-25 2019-07-09 Cellectis Multi-chain chimeric antigen receptor and uses thereof
EP3298033B1 (en) 2015-05-18 2020-08-05 TCR2 Therapeutics Inc. Compositions and medical uses 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
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 (en) 2021-08-20 2023-02-23 大塚製薬株式会社 Combination drug
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 (en) * 1987-05-21 1988-12-01 Creative Biomolecules, Inc. Targeted multifunctional proteins

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1988009344A1 (en) * 1987-05-21 1988-12-01 Creative Biomolecules, Inc. Targeted multifunctional proteins

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 (80)

* 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 (en) * 1994-05-02 1995-11-09 Ciba-Geigy Ag Bifunctional protein, preparation and use
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 (en) * 1995-02-02 2000-04-19 Cell Genesys Inc Chimeric receptors for regulating cellular proliferation and effector function
EP0821730A1 (en) * 1995-02-02 1998-02-04 Cell Genesys, Inc. Chimeric receptors for regulating cellular proliferation and effector function
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
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
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 (en) * 1995-10-24 1997-05-01 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Chimeric receptor genes and cells transformed therewith
WO1997020048A2 (en) * 1995-11-30 1997-06-05 Bristol-Myers Squibb Company Modified sfv molecules which mediate adhesion between cells and uses thereof
WO1997020048A3 (en) * 1995-11-30 1997-09-25 Bristol Myers Squibb Co Modified SFV molecules which mediate adhesion between cells and uses thereof
US6699715B1 (en) 1995-11-30 2004-03-02 Bristol-Myers Squibb Co. Modified sFv molecules which mediate adhesion between cells and uses thereof
WO1997023613A3 (en) * 1995-12-21 1997-08-21 Celltech Therapeutics Ltd Cell activation process and reagents therefor
WO1997023613A2 (en) * 1995-12-21 1997-07-03 Celltech Therapeutics Ltd. Cell activation process and reagents therefor
WO1997035004A1 (en) * 1996-03-15 1997-09-25 Medical Research Council Cell stimulation
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
WO1998038324A2 (en) * 1997-02-27 1998-09-03 Boehringer Mannheim Corporation Chimeric cell-surface receptors that undergo antigen-induced association
WO1998038324A3 (en) * 1997-02-27 1999-01-28 Boehringer Mannheim Corp Chimeric cell-surface receptors that undergo antigen-induced association
US6339070B1 (en) 1997-05-10 2002-01-15 Zeneca Limited Gene construct encoding a heterologous prodrug-activating enzyme and a cell targeting moiety
WO1999036437A1 (en) * 1998-01-15 1999-07-22 Center For Molecular Medicine And Immunology Antibody/receptor targeting moiety for enhanced delivery of armed ligand
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 (en) * 1998-05-06 1999-11-11 Celltech Therapeutics Limited Chimeric receptors
US7083949B2 (en) 1998-09-25 2006-08-01 Regeneron Pharmaceuticals, Inc. Receptor based antagonists and methods of making and using
US7927583B2 (en) 1998-09-25 2011-04-19 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 (en) * 1998-09-25 2002-08-07 Regeneron Pharmaceuticals, Inc. IL-1 Receptor fusion proteins used as antagonists and methods of making and using
WO2000018932A3 (en) * 1998-09-25 2000-11-02 Regeneron Pharma Receptor based antagonists and methods of making and using
US7417134B2 (en) 1998-09-25 2008-08-26 Regeneron Pharmaceuticals, Inc. IL-1 receptor based cytokine traps and method of using
WO2000018932A2 (en) * 1998-09-25 2000-04-06 Regeneron Pharmaceuticals, Inc. Receptor based antagonists and methods of making and using
EP1229047A3 (en) * 1998-09-25 2002-10-02 Regeneron Pharmaceuticals, Inc. IL-1 Receptor fusion proteins used as antagonists and methods of making and using
WO2001062908A3 (en) * 2000-02-22 2002-04-25 Ahuva Nissim Chimeric and tcr phage display libraries, chimeric and tcr reagents and methods of use thereof
WO2001062908A2 (en) * 2000-02-22 2001-08-30 Ahuva Nissim Chimeric and tcr phage display libraries, chimeric and tcr reagents and methods of use thereof
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 (en) 2007-01-31 2016-07-06 Yeda Research And Development Company Limited Redirected, genetically-engineered t regulatory cells and their use in suppression of autoimmune and inflammatory disease
EP3097923A1 (en) 2007-01-31 2016-11-30 Yeda Research And Development Co., Ltd. Redirected, genetically-engineered t regulatory cells and their use in suppression of autoimmune and inflammatory disease
EP3097923B1 (en) 2007-01-31 2022-07-27 Yeda Research And Development Co., Ltd. Redirected, genetically-engineered t regulatory cells and their use in suppression of autoimmune and inflammatory disease
US10342829B2 (en) 2012-05-25 2019-07-09 Cellectis Multi-chain chimeric antigen receptor and uses thereof
WO2014039523A1 (en) * 2012-09-04 2014-03-13 Cellectis Multi-chain chimeric antigen receptor and uses thereof
CN104769103A (en) * 2012-09-04 2015-07-08 塞勒克提斯公司 Multi-chain chimeric antigen receptor and uses thereof
CN104769103B (en) * 2012-09-04 2018-06-08 塞勒克提斯公司 Multichain Chimeric antigen receptor and its purposes
US11304976B2 (en) 2015-02-18 2022-04-19 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11497767B2 (en) 2015-02-18 2022-11-15 Enlivex Therapeutics R&D 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
US11717539B2 (en) 2015-02-18 2023-08-08 Enlivex Therapeutics RDO 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
US11596652B2 (en) 2015-02-18 2023-03-07 Enlivex Therapeutics R&D Ltd Early apoptotic cells for use in treating sepsis
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 (en) 2015-05-18 2020-08-05 TCR2 Therapeutics Inc. Compositions and medical uses for tcr reprogramming using fusion proteins
EP3298033B2 (en) 2015-05-18 2023-07-12 TCR2 Therapeutics Inc. Compositions and medical uses for tcr reprogramming using fusion proteins
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
WO2017123808A1 (en) 2016-01-15 2017-07-20 The J. David Gladstone Institutes Methods of treating disease by metabolic control of t-cell differentiation
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
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
WO2023022235A1 (en) 2021-08-20 2023-02-23 大塚製薬株式会社 Combination drug

Similar Documents

Publication Publication Date Title
EP0638119B1 (en) Chimeric receptor genes and cells transformed therewith
US8211422B2 (en) Chimeric receptor genes and cells transformed therewith
WO1993019163A1 (en) Chimeric receptor genes and cells transformed therewith
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 (en) Immune cells expressing reverse universal chimeric antigen receptors for the targeting of several multiple antigens and methods for their production and their use to treat cancer, infectious diseases and autoimmune diseases
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.
US7446190B2 (en) Nucleic acids encoding chimeric T cell receptors
EP0758394B1 (en) Bifunctional protein, preparation and use
CN106755107B (en) A kind of CAR recruit and its application in oncotherapy
KR20180002604A (en) Chimeric antigen receptor (CAR) targeting compositions of hematological malignancies, compositions and uses thereof
JP2004537952A (en) Multispecific binding molecules and their uses
JP2022522654A (en) Chimeric cytokine receptor carrying PD-1 external domain
Sadelain T-cell engineering for cancer 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 (en) Targeted cytolysis of cancer cells
US20220306719A1 (en) Ultramodular igg3-based spacer domain and multi-function site for implementation in chimeric antigen receptor design
Zumla et al. Co-expression of human T cell receptor chains with mouse CD3 on the cell surface of a mouse T cell hybridoma
Mamalaki et al. In vitro and in vivo antitumor activity of a mouse CTL hybridoma expressing chimeric receptors bearing the single chain Fv from HER-2/neu-specific antibody and the γ-chain from Fc (ε) RI

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