WO2011159847A2 - Receptor tyrosine kinase-like orphan receptor 1 (ror1) single chain fv antibody fragment conjugates and methods of use thereof - Google Patents

Receptor tyrosine kinase-like orphan receptor 1 (ror1) single chain fv antibody fragment conjugates and methods of use thereof Download PDF

Info

Publication number
WO2011159847A2
WO2011159847A2 PCT/US2011/040595 US2011040595W WO2011159847A2 WO 2011159847 A2 WO2011159847 A2 WO 2011159847A2 US 2011040595 W US2011040595 W US 2011040595W WO 2011159847 A2 WO2011159847 A2 WO 2011159847A2
Authority
WO
WIPO (PCT)
Prior art keywords
conjugate
antibody
scfv
peptide
seq
Prior art date
Application number
PCT/US2011/040595
Other languages
French (fr)
Other versions
WO2011159847A3 (en
Inventor
Thomas J. Kipps
Jianqiang Yu
Original Assignee
The Regents Of The University Of California
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Regents Of The University Of California filed Critical The Regents Of The University Of California
Priority to US13/703,864 priority Critical patent/US9242014B2/en
Publication of WO2011159847A2 publication Critical patent/WO2011159847A2/en
Publication of WO2011159847A3 publication Critical patent/WO2011159847A3/en
Priority to US14/973,486 priority patent/US9938350B2/en
Priority to US15/908,659 priority patent/US20180362657A1/en
Priority to US17/093,474 priority patent/US11548953B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C07K14/70521CD28, CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/468Immunoglobulins having two or more different antigen binding sites, e.g. multifunctional antibodies
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57492Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds localized on the membrane of tumor or cancer cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants

Definitions

  • the present invention relates generally to single chain Fv fragments of tumor- associated antigen binding antibodies.
  • the invention further relates to single chain Fv conjugate molecules and their use in the deliver ⁇ ' of biologically active molecules to cancer cells as well as in directing T cells to respond to tumor-associated antigen expressing cancer cells.
  • CLL cells that are not expressed in other lymphoid tissues. Coupled with the observation that CLL cells also express a highly restricted immunoglobulin repertoire, cells likely express distinctive antigens that also could be targeted for immunotherapy. Conceivably, chronic lymphocytic leukemia (CLL.) cells in particular also might have such leukemia-associated antigens (LAA).
  • CLL. chronic lymphocytic leukemia
  • Implicated in the abnormal immune function are immime-suppressive factors and an acquired functional deficiency of CD 154. Furthermore, CLL cells are particularly poor at antigen presentation, which appears in part secondary to inadequate leukemia-cell expression of immune co-stimulatory/adhesion molecules.
  • TAA binding antibodies have been used to develop potential cancer vaccines with mixed degrees of success. Many such TAAs represent developmental or differentiation antigens that have restricted expression. Fc containing antibodies to TAAs are often cleared relatively quickly from circulation and may be recognized only by one type of MHC molecule, limiting their usefulness in therapy.
  • Activation of CLL cells via CD40-ligation can reverse its immune-suppressive phenotype.
  • CLL cells transduced with an adenovirus encoding the ligand for CD40 can function as more effective antigen-presenting cells (APCs).
  • APCs effective antigen-presenting cells
  • they can effect ligation of CD40 on bystander leukemia B cells and stimulate autologous leukemia-reactive T cells both in vitro and in vivo.
  • targeting of CD 154 modified T cells to CLL. cells remains to be achieved.
  • Tyrosine kinases are important mediators of the signaling cascade, determining key roles in diverse biological processes like growth, differentiation, metabolism and apoptosis in response to external and internal stimuli. Studies ha ve implicated the role of tyrosine kinases in the pathophysiology of cancer. Schlessinger J. (2000) Cell, 103:211-225; and Robinson et al. (2000) Oncogene, 19:5548-5557.
  • RNAi to RORl was found as one of the most potent in inducing apoptosis among the set of RNAi targeting each of 73 different kinase-encoding genes. MacKeigan et al. (2005) Nat Cell Biol, 7:591-600, However, these investigators did not examine the expression or function of ROR l protein in these cells.
  • RORl is a membrane-receptor with an intracellular kinase-like domain and extracellular Frizzled-like cysteine-rich domain, which is common to receptors of members of the Wnt-family, RORl is member of the ROR family that is evoiutionarily conserved among Caenorhavditis elegans, Drosophi!a, mice and humans.
  • Wilson C Goberdhan D C,
  • Dror a potential neurotrophic receptor gene, encodes a Drosophila homolog of the vertebrate Ror family of Trk-related receptor tyrosine kinases. Proc Natl Acad Sci USA. 1993; 90:7109- 71 13; Oishi et al.
  • RORl is expressed primarily in developing cephalic neural crest in the dorsal part of the diencephalons and mid-hind brain boundary during embryogenesis. In most species examined, expression of RORl apparently attenuates during embryonic development, becoming negligible at term. RORl mRNA was reported to express in infant brain, renal cancer and colon cancer.
  • RORl at both mRNA and protein level, was highly expressed in CLL B cells but not normal B cells. Moreover, it was found that ROR l is a receptor for Wni5a, which could induce activation of NF-KB when co-expressed with RORl in HEK293 cells and enhance survival of C LL cells in vitro, Another study found that RORl was expressed in acute lymphocytic leukemia (ALL) as well.
  • ALL acute lymphocytic leukemia
  • NP__005003.2 and M_005012 cancer cells which express the WntSa protein, such as CLL ceils, not only bind RORl but have a survival advantage conferred as a consequence.
  • RORl is an embryonic protein that is expressed uniquely on certain cancer cells, including in CLL, small lymphocytic lymphoma, marginal cell B-Cell lymphoma, Burkett's Lymphoma, and other cancers (e.g., breast cancers), but not on normal adult tissues and cells. It is therefore a potential TAA target for modulation of an immune response to RORl expressing cancer cells ("RORl cancers").
  • the present invention is based on the seminal generation of an anti-RORl single- chain variable fragment (scFv) antibody, as well as conjugates thereof which include a biologically active molecule.
  • conjugates may comprise a chimeric receptor to direct T ceils to respond to ROR1 cancer cells.
  • the scFv may be used to target ceils expressing RQR 1 for therapeutic and diagnostic purposes,
  • the present invention provides an antibody single-chain variable fragment (scFv) conjugate.
  • the conjugate includes an scFv having an amino acid sequence at least 90% identical to SEQ ID NO: 1 or SEQ ID NO: 2, and a biologically active molecule.
  • the biologically active molecule is selected from the group consisting of a polypeptide, a peptidomimetic, a nucleic acid molecule, a lipid, a drug compound, chemotherapeutic agent, a iiposome, an antagonist and an agonist.
  • the present invention provides an isolated peptide including an amino acid sequence at least 90% identical to SEQ ID NO: 1 or SEQ ID NO: 2.
  • the conjugate and peptide include an amino acid sequence at least 95% identical to SEQ ID NO: 1 or SEQ ID NO: 2, In exemplary embodiments, the conjugate and peptide has a binding specificity of an antibody as produced by a hybridoma having Accession No. PTA 8634 by the American Type Culture Collection.
  • the present invention provides an isolated nucleic acid encoding the conjugate or peptide of the in vention.
  • the present invention provides a pharmaceutical composition including the conjugate or peptide of the invention an optionally a pharmaceutically acceptable carrier,
  • the present invention provides a method of producing a scFv antibody conjugate.
  • the method includes transforming a host cell with an expression construct including a nucleic acid molecule encoding a scFv antibody and a polypeptide, the scFv peptide including an amino acid sequence at least 90% identical to SEQ ID NO: 1 or SEQ ID NO: 2; and culturhig the host cell under conditions suitable for producing the conjugate, thereby producing the protein conjugate.
  • the present invention provides a method for detecting RO 1 protein in a sample, utilizing a detectably labeled scFv of the in vention.
  • the present invention provides a method of targeting a biologically active molecule to a cell having an ROR1 receptor.
  • the method includes contacting the cell with an scFv conjugate of the invention.
  • the present invention provides a kit to detect the presence of ROR1 protein in a sample from a subject that is known or suspected to contain cancer cells.
  • the kit includes the an scFv antibody conjugate or peptide of the invention and instructions for its use in an assay environment.
  • the present invention provides a method for treating a R.OR1 cancer in a human subject using an scFv antibody conjugate or peptide of the invention.
  • the present invention provides a method for treating or preventing CLL in which ROR1 binding of WntSa on CLL ceils confers a survival advantage thereon by administering an scFv antibody conjugate or peptide of the invention.
  • the present invention provides a method of monitoring a therapeutic regimen for treating a subject having or at risk of having an RORl cancer using using an scFv antibody conjugate or peptide of the invention.
  • Figure 1 shows the amino acid sequence (SEQ ID NO: 1) of an anti-RGRl mAb scFv.
  • the amino acid sequence includes a leader sequence (amino acid residues 1 to 25) and VL (amino acid residues 25 to 132) and VH (amino acid residues 151 to 269) chains joined by a linker sequence (amino acid residues 133 to 150).
  • Figure 2 shows the amino acid sequence (SEQ ID NO: 2) of an anti-RORl mAb scFv after cleavage of the leader during processing of the polypeptide
  • Figure 3 shows the nucleotide sequence encoding the anti-RORl mAb scFv (SEQ ID NO: 3) of SEQ ID NO: 1.
  • Figure 4 shows the nucleotide sequence encoding the anti-RORl mAb scFv (SEQ ID NO: 4) of SEQ ID NO: 2.
  • Figure 5 shows plots of fluorescence intensity. The left plot shows intensity in a Jurkat T cell line and the right shows a Jurkat T cell line transduced to express an ROR.1 conjugate including anti-RORl niAb scFv, an IgG4 CH domain, a transmembrane domain of CD28, and a cytoplasmic domain of the zeta chain for a T-cell receptor (CD247) to generate a chimeric antigen receptor (ROR1-CAR).
  • ROR.1 conjugate including anti-RORl niAb scFv, an IgG4 CH domain, a transmembrane domain of CD28, and a cytoplasmic domain of the zeta chain for a T-cell receptor (CD247) to generate a chimeric antigen receptor (ROR
  • the shaded histogram indicates the autofluorescence of the unstained ceils.
  • the open histogram demonstrates the fluorescence of cells inc ubated with a soluble ROR1 protein (RORl-Ig) and then stained with anti-Ig phycoerythrin (PE).
  • Figure 6 shows the results of F ACS analysis for PC D A 3.1 CH 2 CH 3 stable scFV clones targeting of Endemic African Burkitt's lymphoma B cells (EW36). Results for non- ROR1 expressing CHO cells are provided for comparison.
  • the present invention is based on the seminal generation of as anti-RORl scFv antibody and conjugates thereof including a biologically-active molecule.
  • Such conjugates may comprise a chimeric receptor to direct T cells to respond to ROR1 cancer cells.
  • the scFv may be used to target cells expressing ROR1 for therapeutic and diagnostic purposes.
  • RORl encodes a type I membrane receptor tyrosine kinase that initially was identified using oligonucleotide primers targeting sequences encoding amino acid sequences common to tyrosine kinase domains of different proteins. This protein appears highly conserved throughout evolution. RORl is evolutionary conserved among Caenorhabditis elegans (C. elegans), Aplysia, Drosophd ' a melanogaster, Xenopus, mice, and humans. In rodents, RORl is expressed primarily in developing cephalic neural crest in die dorsal part of the diencephalons and mid-hind brain boundary during embryogenesis. Work in
  • Caenorhabditis elegans (C. elegans) indicated that the RORI-type kinases might be involved in the regulation of cell motility and in asymmetric cell division during embryogenesis.
  • the ROR protein in C. elegans apparently has both kinase-dependent and kinase independent ROR-family receptor tyrosine kinases are characterized by the intracellular tyrosine kinase domains, highly related to those of the Trk-family receptor tyrosine kinases, and by the extracellular Frizzled-like cysteine-rich domains and kringle domains, which are common to receptors of the Wnt-family members.
  • An ortholog to RORl, namely ROR2 has been found interact physically with WntSa to activate non-canonical Wnt- signaling.
  • ROR l interacts physically with WntSa.
  • the interaction of RORl with WntSa was implicated in studies demonstrating that co-transfection of expression vectors encoding NF- ⁇ reporter constructs, RORl , and WntSa, but not other Wnt factors, could induce activation of NF-NB in a mutually dose-dependent fashion.
  • This activity was determined to be independent of expression of LPR5/6, which ordinarily serves as a co- receptor for Wnt receptors.
  • Physical interaction of RORl with Wnt5a was observed using recombinant proteins, demonstrating that RORl has binding activity for Wnt5a independent of LPR5/6. Nonetheless, Wnt a apparently could not activate the canonical Wnt-signaling pathway when co-expressed with RORl in 293 cells.
  • ⁇ -catenin apparently can physically complex with NF-KB, resulting in reduction of NF- ⁇ D A binding, transactivation activity, and target gene expression.
  • NB kinase-alpha (Ii a) involved in the phosphorylation ofNF- f B2/pl00, can phosphorylate and stabilize ⁇ -catenin
  • the kinase involved in activation of the canonical p50/p65 NF- ⁇ pathway namely ⁇ , can enhance ⁇ -catenin degradation.
  • the inability of RORl to activate the LEF/TCF-signaling may be secondary in part to its capacity to activate ⁇ , leading to reduced stability of p-catenin and enhanced activity of NF- ⁇ , which in itself could potentially play a role in cancer development.
  • CLL Crohn's disease
  • neoplastic cells induced by interaction with tissue stromal cells that might elaborate Wnt5a and other factors that activate NF- ⁇ .
  • CLL cells derive a survival benefit from interactions with marrow stromal cells, nurse-like cells, or dendritic cells, which are found in the leukemia-infiltrated marro or lymphoid tissues of patients with this disease. It is noteworthy in this regard that dendritic cells have been found to express high- levels of WntSa.
  • RORl appears to be at least in part responsible for the survival-signal triggered by co-culture with Wnt5a-expressing CHO cells. This is indicated by findings that serum obtained after treatment with autologous Ad-CD 154- CLL cells could neutralize the capacity of CHO-Wnt5a cells to enhance the survival of CLL cells over that of CLL cells co-cultured with CHO cells or CLL cells cultured alone.
  • antisenim with CHO-R.OR1 cells abrogated the capacity of the post- treatment serum to neutralize the activity of CHO-Wnt5a cells.
  • anti-RQRl antibodies could be responsible for some of the size-reductions observed in the lymph nodes of patients who had received infusions of autologous Ad-CD 154-CLL cells.
  • antibody variants or fragments that maintain specific binding with the native ROR1 protein are necessary as they provide increased flexibility in designing a wide variety of antibody conjugates that may be used for therapeutic and diagnostic purposes.
  • the present invention provides an antibody single-chain variable fragment (scFv) conjugate.
  • the conjugate includes an scFv having an amino acid sequence at least 90%, 95% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 2 as shown in Figures 1 and 2, and a biologically active molecule.
  • the conjugate and peptide has a binding specificity of an antibody as produced by a hybridoma having Accession No. PTA 8634 by the American Type Culture Collection.
  • the present invention provides an isolated peptide including an amino acid sequence at least 90%, 95% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 2.
  • a “conjugate” as used herein generally refers to a molecule which contains a peptide including an amino acid sequence at least 90%, 95% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 2, conjugated with a biologically active molecule. Typically, the conjugate is capable of being targeted to a cell expressing R.OR.1.
  • biologically active molecule refers to a molecule that has a biological effect in a cell.
  • the active molecule may be an inorganic molecule, an organic molecule, a small organic molecule, a drug compound, a peptide, a polypeptide, a peptidomimetic, a nucleic acid molecule, a lipid, a drug compound, chemotherapeutic agent, a liposome, an antagonist or an agonist.
  • the active molecule may be doxorubicin.
  • specific binding refers to antibody binding to a predetermined antigen.
  • the antibody binds with an affinity corresponding to a KD of about 10 "8 M or less, and binds to the predetermined antigen with an affinity (as expressed by K D ) that is at least 1 0 fold less, and preferably at least 100 fold less than its affinity for binding to a nonspecific antigen (e.g. , BSA, casein) other than the predetermined antigen or a closely-related antigen.
  • the antibody can bind with an affinity corresponding to a KA of about 10 6 or 10 9 M "1 or higher, and binds to the
  • predetermined antigen with an affinity (as expressed by KA) tha is at least 10 fold higher, and preferably at least 100 fold higher than its affinity for binding to a non-specific antigen (e.g. , BSA, casein) other than the predetermined antigen or a closely-related antigen.
  • a non-specific antigen e.g. , BSA, casein
  • the scFv antibody conjugate or peptide of the in vention wil l have the same A or KD as an antibody produced by the hybridoma having ATCC accession number PTA 8634.
  • the term 3 ⁇ 4" (sec " '), as used herein, is intended to refer to the dissociation rate constant of a particular antibody-antigen interaction. This value is also referred to as the off value.
  • the term "KD" (3VT 1 ), as used herein, is intended to refer to the dissociation equilibrium constant of a particular antibody-antigen interaction.
  • k a (M 'l sec _1 ), as used herein, is intended to refer to the association rate constant of a particular antibody-antigen interaction.
  • KA (M), as used herein, is intended to refer to the association equilibrium constant of a particular antibody-antigen interaction.
  • Naturally occurring antibodies are generally tetramers containing two light chains and two heavy chains.
  • antibodies can be cleaved with the proteolytic enzyme papain, which causes each of the heavy chains to break, producing three separate sub units.
  • the two units that consist of a light chain and a fragment of the heavy chain approximately equal in mass to the light chain are called the Fab fragments (i.e., the "antigen binding" fragments).
  • the third unit, consisting of two equal segments of the heavy chain, is called the Fc fragment.
  • the Fc fragment is typically not involved in antigen-antibody binding, but is important in later processes involved in ridding the body of the antigen.
  • Fab and F(ab')2 fragments are smaller tha intact antibody molecules, more antigen-binding domains are available than when whole antibody molecules are used.
  • Proteolytic cleavage of a typical IgG molecule with papain is known to produce two separate antigen binding fragments called Fab fragments which contain an intact light chain linked to an amino terminal portion of the contiguous heavy chain via by disulfide linkage.
  • the remaining portion of the papain-digested immunoglobin molecule is known as the Fc fragment and consists of the carboxy terminal portions of the antibody left intact and linked together via disulfide bonds.
  • an antibody is digested with pepsin, a fragment known as an F(ab')2 fragment is produced which lacks the Fc region but contains both antigen-binding domains held together by disulfide bonds between contiguous light and heavy chains (as Fab fragments) and also disulfide linkages between the remaining portions of the contiguous heavy chains (Handbook of Experimental immunology. Vol 1 : immunocheniistry, Weir, D. M, Editor, Biackwell Scientific Publications, Oxford (1986)).
  • altered antibodies e.g., chimeric, humanized, CDR-grafted, bifunctional, antibody polypeptide dimers (i.e., an association of two polypeptide chain components of an antibody, e.g. , one arm of an antibody including a heavy chain and a light chain, or an Fab fragment including VL, VH, CL and CH antibody domains, or an F fragment comprising a VL domain and a VH domain
  • single chain antibodies e.g. , an scFv (i.e. , single chain Fv) fragment including a VL domain linked to a VH domain by a linker, and the like
  • an scFv i.e. , single chain Fv
  • scFvs single chain Fv fragments
  • scFvs can be dimerized to produce a diabody.
  • scFvs bind a specific epitope of interest and can be produced using any of a variety of recombinant bacterial phage-based methods, for example as described in Lowman et al. (1 991) Biochemistry, 30, 10832-10838; Clackson et ai. (1991 ) Nature 352, 624-628; and Cwirla et al. (1990) Proc. Natl. Acad. Sci. USA 87, 6378-6382. These methods are usually based on producing genetically altered filamentous phage, such as recombinant M 13 or fd phages, which display on the surface of the phage particle a
  • recombinant fusion protein containing the antigen-binding scFv antibody as the amino terminal region of the fusion protein and the minor phage coat protein g3p as the carboxy terminal region of the fusion protein.
  • Such recombinant phages can be readily grown and isolated using well-known phage methods.
  • the intact phage particles can usually be screened directly for the presence (display) of an antigen-binding scFv on their surface without the necessity of isolating the scFv away from the phage particl e.
  • scFv standard reverse transcriptase protocols are used to first produce cDNA from mR A isolated from a hybridoma that produces an mAb for ROR-1 antigen.
  • the cDNA molecules encoding the variable regions of the heavy and light chains of the mAb can then be amplified by standard polymerase chain reaction (PGR) methodology using a set of primers for mouse immunoglobulin heavy and light variable regions (Clackson (1991 ) Nature, 352, 624-628),
  • PGR polymerase chain reaction
  • the amplified cDNAs encoding mAb heavy and light chain variable regions are then linked together with a linker oligonucleotide in order to generate a recombinant scFv DNA molecule.
  • the scFv DNA is ligated into a filamentous phage plasmid designed to fuse the amplified cDNA sequences into the 5' region of the phage gene encoding the minor coat protein called g3p, Escherichia coli bacterial cells are than transformed with the recombinant phage plasmids, and filamentous phage grown and harvested.
  • the desired recombinant phages display antigen-binding domains fused to the amino terminal region of the minor coat protein.
  • Such "display phages” can then be passed over immobilized antigen, for example, using the method known as "panning", see Parmley and Smith (1989) Adv. Exp, Med. Biol.
  • Selection for increased antigen-binding ability may be made by adjusting the conditions under which binding takes place to require a tighter binding activity.
  • Another method to select for enhanced antigen-binding activity is to alter nucleotide sequences within the cDN A encoding the binding domain of the scFv and subject recombinant phage
  • an scFv can be produced in a free form using an appropriate vector in conjunction with E. coli strain HB2151. These bacteria actually secrete scFv in a soluble form, free of phage components (Hoogenboom et al. (1991) Nucl, Acids Res. 19, 4133- 4137).
  • the purification of soluble scFv from the HB2151 bacteria culture medium can be accomplished by affinity chromatography using antigen molecules immobilized on a solid support such as AFFIGELTM (BioRad, Hercules, Calif.).
  • scFvs are even smaller molecules than Fab or F(ab') 2 fragments, they can be used to attain even higher densities of antigen binding sites per unit of surface area when immobilized on a solid support material than possible using whole antibodies, F(ab f ) 2 , or Fab fragments. Furthermore, recombinant antibody technology offers a more stable genetic source of antibodies, as compared with hybridomas. Recombinant antibodies can also be produced more quickly and economically using standard bacterial phage production methods.
  • nucleic acids encoding one is inserted into expression vectors.
  • the light and heavy chains can be cloned in the same or different expression vectors.
  • Expression vectors are typically replicable in the host organisms either as episomes or as an integral part of the host chromosome.
  • E. coli is one procaryotic host particularly useful for expressing antibodies of the present invention.
  • Other microbial hosts suitable for use include bacilli, such as Bacillus s btilus, and other enterobacteriaceae, such as Salmonella, Serratia, and various Pseudomonas species.
  • expression vectors typically contain expression control sequences compatible with the host ceil (e.g., an origin of replication) and regulator ⁇ ' sequences such as a lactose promoter system, a tryptopha (trp) promoter system, a beta-lactamase promoter system, or a promoter system from phage lambda.
  • regulator ⁇ ' sequences such as a lactose promoter system, a tryptopha (trp) promoter system, a beta-lactamase promoter system, or a promoter system from phage lambda.
  • Other microbes, such as yeast may also be used for expression
  • Saccharomyces is a preferred host, with suitable vectors having expression control sequences, such as promoters, including 3-phosphogiycerate kinase or other glycolytic enzymes, and an origin of replication, termination sequences and the like as desired.
  • Mammalian tissue ceil culture can also be used to express and produce the antibodies of the present invention (see, e.g., Winnacker, From Genes to Clones VCH Publishers, N.Y., 1987).
  • Eukaryotic cells are preferred, because a number of suitable host cell lines capable of secreting intact antibodies have been developed.
  • Preferred suitable host cells for expressing nucleic acids encoding the immunoglobulins of the invention include: monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line; baby hamster kidney cells (BHK, ATCC CCL 10); Chinese hamster ovary-cells (CHO); mouse Sertoli cells; monkey kidney cells (CV1 ATCC CCL 70); african green monkey kidney cells (VERO-76, ATCC CRL, 1587); human cervical carcinoma cells (HELA, ATCC CCL, 2); canine kidney cells (MDC , ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL, 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL51); and TRI cells.
  • monkey kidney CV1 line transformed by SV40 COS-7, ATCC CRL 1651
  • human embryonic kidney line BHK, ATCC C
  • the vectors containing the polynucleotide sequences of interest can be transferred into the host cell.
  • Calcium chloride transfection is commonly utilized for prokaryotic cells, whereas calcium phosphate treatment or eiectroporation can be used for other cellular hosts (see, e.g., Sambrook et al, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, 2nd ed., 1989).
  • cel l lines expressing immunoglobulin products are cell selected. Cell lines capable of stable expression are preferred (i.e., undiminished le vels of expression after fifty passages of the cell line),
  • the scFv or other immunoglobulin Forms of the present invention can be purified according to standard procedures of the art, including ammonium sulfate precipitation, affinity columns, column chromatography, gel electrophoresis and the like (see, e.g., Scopes, Protein Purification, Springer- Verlag, N.Y., 1982). Substantially pure
  • immunoglobulins of at least about 90 to 95% homogeneity are preferred, and 98 to 99% or more homogeneity most preferred.
  • a labeled antibody or a detectably labeled antibody is generally an antibody (or antibody fragment which retains binding specificity), having an attached detectable label.
  • the detectable label is normally attached by chemical conjugation, but where the label is a polypeptide, it could alternatively be attached by genetic engineering techniques. Methods for production of detectably labeled proteins are well known in the art.
  • Detectable labels known in the art include radioisotopes, fiuorophores, paramagnetic labels, enzymes (e.g., horseradish peroxidase), or other moieties or compounds which either emit a detectable signal (e.g., radioactivity, fluorescence, color) or emit a detectable signal after exposure of the label to its substrate.
  • Various detectable label/substrate pairs e.g., horseradish
  • Antibodies may be humanized by replacing sequences of the Fv variable region which are not directly involved in antigen binding with equivalent sequences from human Fv variable regions.
  • General reviews of humanized chimeric antibodies are provided by Morrison et al, (Science 229: 12024207 (1985)) and by Oi et al. (BioTechniques 4:214 (1986)).
  • Those methods include isolating, manipulating, and expressing the nucleic acid sequences that encode all or part of immunoglobulin Fv variable regions from at least one of a heavy or light chain. Sources of such nucleic acid are wel l known to those skil led in the art and, for example, may be obtained from for example, an antibody producing hybridoma.
  • the recombinant DNA encoding the humanized or chimeric antibody, or fragment thereof, can then be cloned into an appropriate expression vector.
  • Humanized antibodies can alternatively be produced by CDR substitution (U.S. Pat. No. 5,225,539; Jones, Nature 321:552-525 (1986); Verhoeyan et al, Science 239:1534 (1988); and Beidler, J. Immunol. 141 :4053-4060 (1988)).
  • the antibody used in the conjugate is a humanized or CDR-grafted form of an antibody produced by the hybridoma having ATCC accession number PTA 2439.
  • the antibody is a humanized or CDR-grafted form of antibody mAb 3E10.
  • the CDR regions can include amino acid substitutions such as 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid differences from those shown in the figures. In some instances, there are anywhere from 1-5 amino acid differences.
  • an scFv antibody having the binding specificity of an antibody as produced by the hybridoma having ATCC accession number PTA 8634 includes antibody fragments having at least 90% or 95% sequence identity to SEQ ID NO:l or SEQ ID NO: 2 and retain the same binding specificity as that produced by the hybridoma having ATCC accession number PTA 8634, such as variants modified by mutation to improve the utility thereof (e.g., improved ability to target specific cell types and the like). Such variants include those wherein one or more conservative substitutions are introduced into the heavy chain and/or the light chain of the scFv.
  • variants include those wherein one or more substitutions are introduced into the heavy chain nucleotide sequence and/or the light chain nucleotide sequence of the scFv.
  • the variant has a light chain and/or heavy chain having a nucleotide sequence at least 80% or at least 90% or at least 95% identical to the nucleotide sequence set forth in SEQ ID NO: 3 or SEQ ID NO: 4.
  • the conjugates include polypeptides (i.e., protein conjugates), they may be designed as fusion protein conjugate such that the conjugate can be expressed in a host ceil as a fusion protein.
  • portions of the conjugate can be chemically linked by peptide bonds or by a chemical or peptide linker molecule of various types well known in the ail as discussed further herein.
  • linkers may be used to link portions of the conjugates described herein.
  • linker is any bond, small molecule, or other vehicle which allows the substrate and the active molecule to be targeted to the same area, tissue, or cell, for example by physically linking the individual portions of the conjugate.
  • a cleavable or degradable linker may be used.
  • the linker is a chemical bond between one or more substrates and one or more therapeutic moieties.
  • the bond may be covalent or ionic.
  • An example of a therapeutic complex where the linker is a chemical bond would be a fusion protein.
  • the chemical bond is acid sensitive and the pH sensitive bond is cleaved upon going from the blood stream (pH 7.5) to the transcytotic vesicle or the interior of the cell (pH about 6.0).
  • the bond may not be acid sensitive, but may be cleavable by a specific enzyme or chemical which is subsequently added or naturally found in the microenvironment of the targeted site.
  • the bond may be a bond that is cleaved under reducing conditions, for example a disulfide bond.
  • the bond may not be cleavab!e.
  • Any kind of acid cleavable or acid sensitive linker may be used.
  • acid cleavable bonds include, but are not limited to: a class of organic acids known as cipolycarbox lic alkenes. This class of molecule contains at least three carboxylic acid groups (COOH) attached to a carbon chain that contains at least one double bond. These molecules as well as how they are made and used is disclosed in Shen, et al. U.S. Pat. No. 4,631 ,190.
  • molecules such asamino-sulfhydryl cross-linking reagents which are cleavable under mildly acidic conditions may be used. These molecules are disclosed in U.S. Pat. No. 4,569,789.
  • the acid cleavable linker may be a time-release bond, such as a biodegradable, hydrolyzable bond.
  • Typical biodegradable carrier bonds include esters, amides or urethane bonds, so thai typical carriers are polyesters, polyamides, polyurethanes and other condensation polymers having a molecular weight between about 5,000 and 1,000,000.
  • linkers are cleaved by natural acidic conditions, or alternatively, acid conditions can be induced at a target site as explained in U.S. Pat. No. 4,171,563.
  • linking reagents which contain cleavable disulfide bonds (reducable bonds) include, but are not limited to "DPDPB", l,4-di-[3'-(2'-pyridyldithio) propionamido] butane; "SADP", (N-succinimidyl (4-azidophenyl) l,3'-dithiopropionate); "Sulfo-SADP” (Sulfosuccinimidyl (4-azidophenyldithio) propionate; "DSP"-Dithio bis
  • linking reagents cleavable by oxidation are “DST”-disucciiiimidyi tartarate; and “Sulfo-DST”-disuccinimidyS tartarate.
  • non-cleavable linkers are "Si fQ-LC-SMPT"-(sulfosuccinimidyl 6- [alphamethyl-alpha-(2-pyridylthio)toluamido ⁇ hexanoate; "SMPT”; "ABH”-Azidobenzoyl hydrazide; ''NHS-ASA' ⁇ N-Hydroxysuccinimidyl-'-l-azidosalicyclic acid; "SASD”- Sulfos ' uccinimidyl 2-(pazidosalicylarmdo) ethyl- 1 ,3-dithiopropionate; " APDP"-N-
  • APG "p-Azidophenyl glyoxal monohydrate; "SANPAH”-N-Succiminidyl-6 (4'-azido-2 - mitrophenyl-amimo) hexanoate; "Sulfo-SANPAH”-Sulfosuccimmidyl6-(4'-azido-2'- mtrophenylamino) hexanoate; "A B-NOS" N-5-A.zido-2-nitrobenzyoyioxysuccinimide;
  • SA D Sulfosuccinimidyl-2-(m-azido-o- ⁇ opionate; , TNP-DTP"-p-nitrophenyl-2-diazo-3,3,3-trifluoropropionate;
  • SCC Suecinimidyl-4-(N- maleimidomethyl)cyclohexane- 1 -carboxylate;
  • Sulfo-SMCC Sulfosucciiiimidyl-4-(N- maleimidomethyl)cyclohexane-l-carboxy- late;
  • MS m- aleimidobenzoyl-N- hydroxysuccinimide ester;
  • smfo-MBS -m-Maleimidobenzoyl-N-hydroxysulfosuccinimide ester;
  • SIAB N-Succinimidyl (4-iodoaceiyl)armnobenz ownership;
  • SuliSIA Suli
  • the linker is a small molecule such as a peptide linker.
  • the peptide linker is not cleavable.
  • die peptide linker is cleavable by base, under reducing conditions, or by a specific enzyme.
  • the enzyme is indigenous.
  • the small peptide may be cleavable by an non-indigenous enzyme which is administered after or in addition to the therapeutic complex.
  • the small peptide may be cleaved under reducing conditions, for example, when the peptide contains a disulfide bond.
  • the small peptide may be pH sensitive.
  • the peptide linker may also be useful as a peptide tag (e.g. , myc or His6 (SEQ ID NO: 9)) or may be one or more repeats of the known linker sequence GGGG S ( SEQ ID NO: 10), The skilled artisan will recognize that the linker sequence may be varied depending on the polypeptide portions to be linked to form the conjugate. Additional peptide linkers and tags are known in the art, such as epitope tags, affinity tags, solubility enhancing tags, and the like.
  • tags and linkers examples include, haemagglutinin (HA) epitope, myc epitope, chitin binding protein (CBP), maltose binding protein (MBP), giutathione-S-transferase (GST), calmodulin binding peptide, biotin carboxyl carrier protein (BCCP), FLAG octapeptide, nus, green fluorescent protein (GFP), thioredoxin (TRX), poly(NANP), V5, S-protein, streptavidin, SBP, poly(Arg), DsbA, c-myc- tag, HAT, cellulose binding domain, softag 1, softag.
  • HA haemagglutinin
  • CBP chitin binding protein
  • MBP maltose binding protein
  • GST giutathione-S-transferase
  • calmodulin binding peptide biotin carboxyl carrier protein
  • BCCP biotin carboxyl carrier protein
  • the peptide linker has the formula (amino acid) n, where n is an integer between 2 and 100, preferably wherein the peptide comprises a polymer of one or more amino acids.
  • the linker is a cleavable linker including, poly (ethylene glycol) (PEG) and a dipeptide,L,-alany! ⁇ L ⁇ vaime (A!a-Va!), cleavable by the enzyme diermoiysin.
  • PEG poly (ethylene glycol)
  • A!a-Va! dipeptide,L,-alany! ⁇ L ⁇ vaime
  • the chemical and peptide linkers can be bonded between the scFv and the active molecule by techniques known in the art for conjugate synthesis, i.e. using genetic engineering, or chemically.
  • the conjugate synthesis can be accomplished chemically via the appropriate antibody by classical coupling reactions of proteins to other moieties at appropriate functional groups.
  • Examples of the functional groups present in proteins and utilized normally for chemical coupling reactions are outlined as follows.
  • the thiol group (cysteines in proteins) may be reacted with a compound containing a thiol-reactive group to the formation of a thioether group or disulfide group.
  • the free amino group (at the amino terminus of a protein or on a lysine) in amino acid residues may be reacted with a compound containing an electropliilic group, such as an activated carboxy group, to the formation of an amide group.
  • a compound containing an electropliilic group such as an activated carboxy group
  • Free carboxy groups in amino acid residues may be transformed to a reactive carboxy group and then reacted with a compound containing an amino group to the formation of an amide group.
  • the linker or biologically active molecule may alternatively be a liposome or nanoparticle containing a biologically active molecule.
  • Many methods for the preparation of liposomes are well known in the art. For example, the reverse phase evaporation method, freezethaw methods, extrusion methods, and dehydration-rehydration methods (see, Storm et al, PSTT 1 : 19-31 (1998)).
  • PEGylated or "stealth' ' ' liposomes are especially useful. For example, such liposomes al lo for deliver ⁇ ' of highly charged therapeutic molecules such as siRNA into ceils.
  • the liposomes may be produced in a solution containing the active molecule, such as doxorubicin, so that the substance is encapsulated during polymerization.
  • the liposomes can be polymerized first, and the biologically active substance can be added later by resuspending the polymerized liposomes in a solution of a biologically active substance and treating with sonication to affect encapsulation of the active molecule.
  • the liposomes can be polymerized in the presence of the substrate such that the substrate becomes a part of the phospholipid bilayer.
  • the liposome contains the active molecule on the inside and the substrate on the outside.
  • the liposomes contemplated in the present invention can comprise a variety of structures,
  • the liposomes can be multilamellar large vesicles (MLV), oligolamellar vesicles (OLV), unilamellar vesicles (UV), small unilamellar vesicles (SUV), medium sized unilamellar vesicles(MUV), large unilamellar vesicles (LUV), giant unilamellar vesicles (GUV), or multivesicul ar vesicl es (MVV).
  • MMV multilamellar large vesicles
  • OCV unilamellar vesicles
  • UV unilamellar vesicles
  • SUV small unilamellar vesicles
  • MUV medium sized unilamellar vesicles
  • LUV large unilamellar vesicles
  • GMV giant unilamellar vesic
  • the liposome is a "micromachine" that evulses pharmaceuticals for example by the application of specific frequency radio waves.
  • the liposomes can be degraded such that they will release the active molecule in the targeted cell, for example, the liposomes may be acid or alkaline, sensitive, or degraded in the presence of a low or high pH, such that the active molecule is released within the cell.
  • the liposomes may be uncharged so that they will be taken up by the targeted cell.
  • the liposomes may also be pH sensitive or sensitive to reducing conditions.
  • liposome which may be advantageously used in the present invention is that identified in U.S. Pat. No. 6,004,534.
  • a method of producing modified liposomes which are prepared by polymerization of double and triple bond-containing monomelic phospholipids is disclosed.
  • These liposomes have surprisingly enhanced stability against the harsh en vironment of the gastrointestinal tract. Thus, they have utility for oral and/or mucosal deliver of the active molecule. It has also been shown that the liposomes may be absorbed into the systemic circulation and lymphatic circulation.
  • the liposomes are generally prepared by polymerization (i.e., radical initiation or radiation) of double and triple bond-containing monomeric phospholipids.
  • the linker can also be a liposome having a long blood circulation time.
  • liposomes are well known in the art (see U.S. Pat. Nos., 5,013,556; 5,225,212; 5,213,804; 5,356,633; and 5,843,473). Liposomes having long blood circulation time are characterized by having a portion of their
  • phosphoslipids derivatized with polyethylene glycol (PEG) or other similar polymer in some embodiments, the end of the PEG molecule distal to the phospholipid may be activated so a to be chemically reactive.
  • PEG polyethylene glycol
  • Such a reactive PEG molecule can be used to link a substrate to the liposome.
  • a reactive PEG molecule is the maleimide derivative of PEG described in U.S. Pat. No. 5,527,528).
  • the linker may be a microcapsule, a nanoparticle, a magnetic particle, and the like (see Kumar, J. Pharm. Sci, 2:234-258 (2000); and Gill et al., Trends Biotechnol. 18(1 l):469-79 (2000)), with the lipophilic active molecule on or in the container, and the container functioning as the linker in the therapeutic complex.
  • the linker may be a photocleavable linker.
  • a 1-2- (nitrophenyl)ethyl moiety can be cleaved using 300 to 360 run light.
  • the photocleavable linker would allow activation and action of the drug in an even more specific area, for example a particular part of the organ.
  • the light could be localized using a catheter into the vessel.
  • light may be used to localize treatment to a specific part of the digestive tract and the light may be manipulated through a natural orifice to the area.
  • the light can be surgically manipulated to the area.
  • the linker may not be cleavabie, but the active molecule or substrate is.
  • the active molecule is a prodrug and the enzyme which cleaves the prodrug is administered with the therapeutic complex.
  • the enzyme is part of the therapeutic complex or indigenous and the prodrug is administered separately,
  • the enzyme or prodrug which is administered separately is administered within about 48 hours of the first administration.
  • the prodrug or enzyme which is administered separately may be administered between about 1 min and 24 hours, alternatively between about 2 min and 8 hours.
  • the prodrug or enzyme which is administered separately may be readministered at a later date and may continue to be administered until the effect of the drag is not longer needed.
  • the subject conjugates can be delivered via an expression construct to cells, including a nucleic acid that provides a coding sequence for a therapeutic protein.
  • the expression construct can encode a therapeutic protein that is secreted by the transduced cell.
  • nucleic acid refers to polynucleotides such as
  • DNA deoxyribonucleic acid
  • RNA ribonucleic acid
  • the term should also be understood to include, as appropriate to the context or as applicable to the embodiment being described, both single-stranded polynucleotides (such as antisense) and double-stranded polynucleotides (such as siRNAs).
  • a "protein coding sequence” or a sequence that "encodes” a particular polypeptide or peptide is a nucleic acid sequence that is transcribed (in the case of DN A) and is translated (in the case of mRNA) into a polypeptide in vitro or in vivo when placed under the control of appropriate regulatory sequences.
  • the boundaries of the coding sequence are determined by a start codon at the 5' (amino) terminus and a translation stop codon at the 3' (carboxyl) terminus.
  • a coding sequence can include, but is not limited to, cDNA from prokaryotic or eukaryotic mRNA, genomic DNA sequences from prokaryotic or eukaryotic DNA, and even synthetic DNA sequences.
  • a transcription termination sequence will usually be located 3' to the coding sequence.
  • vector refers to a nucleic acid molecule capable of transporting another nucl eic acid to which it has been l inked.
  • a genomic integrated vector or "integrated vector” which can become integrated into the chromosomal DNA of the host cell.
  • an episonial vector e.g., a nucleic acid capable of extra-chromosomal replication.
  • vectors capable of directing the expression of genes to which they are operative! ⁇ ' linked are referred to herein as "expression vectors.”
  • expression vectors In the present specification, "plasmid” and “vector” are used interchangeably unless otherwise clear from the context.
  • regulatory elements controlling transcription can be generally derived from mammalian, microbial, viral or insect genes.
  • the ability to replicate in a host, usually conferred by an origin of replication, and a selection gene to facilitate recognition of transfomiants may additionally be incorporated.
  • Vectors derived from viruses, such as retroviruses, adenoviruses, and the like, may be employed.
  • the present invention provides a method of producing a protein.
  • the method includes transforming a host cell with an expression construct, and culturing the host ceil under conditions suitable for producing the conjugate.
  • the expression construct includes a nucleic acid molecule encoding a protein conjugate including an scFv and a peptide, wherein the scFv includes an amino acid sequence at least 90%, at least 95% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 2.
  • Vectors suitable for use in preparation of proteins and/or protein conjugates include those selected from bacuiovirus, phage, plasmid, phagemid, cosmid, fosmid, bacterial artificial chromosome, viral DNA, Pi-based artificial chromosome, yeast plasmid, and yeast artificial chromosome.
  • the viral DNA vector can be selected from vaccinia, adenovirus, foul pox virus, pseudorabies and a derivative of SV40.
  • Suitable bacterial vectors for use in practice of the invention methods include pQE70 L ', pQE60 1M , pQE-9 1M , pBLUESCRIPT 1M SK, pBLUESCRIPTTM KS, pTRC99aTM, p 223-3TM, pDR540TM, PACTM and pRJT2TTM.
  • Suitable eukaryotic vectors for use in practice of the invention methods include p W LNEO ,M , pXTITM, pSG5TM, pSV 3TM, pBPVTM, pMSGTM, and p8VLSV40TM, Suitable eukaryotic vectors for use in practice of the invention methods include p WL EO j M , pXTI jM , pSG5 tM , pSVK3TM, pBPVTM, pMSGTM, and pSVLSV40TM.
  • a suitable regulatory region for example from lad, lacZ, T3, T7, apt, lambda PR, PL, trp, CM V immediate early, HSV thymidine kinase, early and late SV40, retroviral LTR, and mouse metallothionein-I regulatory regions.
  • Host cells in which the vectors containing the polynucleotides encoding the protein conjugates can be expressed include, for example, a bacterial cell, a eukaryotic cell, a yeast ceil, an insect cell, or a plant cell.
  • a bacterial cell for example, E, coli, Bacillus, Streptomyces, Pichia pastoris.
  • Salmonella typhimurium, Drosophila S2, Spodoptera SJ9, CHO, COS (e.g. COS-7), or Bowes melanoma cells are all suitable host cells for use in practice of the invention methods.
  • Conjugates in which the biologically active molecule is an antibody or sacFv molecule may be generated using methods known in the art,
  • methods for attaching a drug or other small molecule pharmaceutical to protein include bifunctionai chemical linkers such as N-succinimidyl (4-iodoacetyl)-aminobenzoate; sulfosuccinimidyl(4- iodoacetyl)-aminobenzoate; 4-succinimidyl-oxycarbonyl- -(2-pyridyldithio) toluene;
  • a conjugate of the present invention may further contain a detectable label.
  • labels are known in the art. and include radio-isotopes and fluorescent labels.
  • the reporter can be a label such as a fluorophore, a ehromophore, a radioisotope.
  • Confocal imagining can also be used to detect internalization of a label as it provides sufficient spatial resolution to distinguish between fluorescence on a cell surface and fluorescence within a cell; alternatively, confocal imaging can be used to track the movement of compounds over time.
  • die conjugates of the present invention may include a biologically active molecule that is a nucleic acid molecule or analog thereof.
  • conjugates may be utilized to deliver nucleic acids, or analogs thereof to a targeted tissue or cell type
  • protein expression can be specifically down-regulated using oligonucleotides such as, for example, antisense molecules, locked nucleic acids (LNA), peptide nucleic acids (PNA), morpholino nucleic acids and small interfering RNAs (siRNA) of various chemistries
  • oligonucleotides such as, for example, antisense molecules, locked nucleic acids (LNA), peptide nucleic acids (PNA), morpholino nucleic acids and small interfering RNAs (siRNA) of various chemistries
  • expression constructs may be delivered to cells, to induce expression of a desired gene product.
  • the expression construct that is delivered typically is part of a vector in which a regulator ⁇ ' element such as a promoter is operabiy linked to the nucleic acid of interest.
  • the promoter can be constitutive or inducible.
  • constitutive promoters include cytomegalovirus (CM V) promoter and the Rous sarcoma vims promoter.
  • CM V cytomegalovirus
  • Rous sarcoma vims promoter Rous sarcoma vims promoter.
  • inducible refers to both up-regulation and down regulation.
  • An inducible promoter is a promoter that is capable of directly or indirectly activating transcription of one or more DNA sequences or genes in response to an inducer. In the absence of an inducer, the DNA sequences or genes will not be transcribed.
  • the inducer can be a chemical agent such as a protein, metabolite, growth regulator, phenolic compound, or a physiological stress imposed directly by, for example heat, or indirectly through the action of a pathogen or disease agent such as a virus.
  • the inducer also can be an illumination agent such as light and light's various aspects, which include wavelength, intensity, fluorescence, direction, and duration.
  • an inducible promoter is the tetracycline (tet)-on promoter system, which can be used to regulate transcription of the nucleic acid
  • tet tetracycline
  • a mutated Tet repressor (TetR) is fused to the activation domain of herpes simplex VP 16 (transactivator protein) to create a tetracycline-controlled transcriptional activator (tTA), which is regulated by tet or doxycycline (dox).
  • tetR mutated Tet repressor
  • tTA tetracycline-controlled transcriptional activator
  • dox tetracycline-controlled transcriptional activator
  • Alterna tive inducible systems include the ecdysone or rapamycin systems.
  • Ecdysone is an insect molting hormone whose production is controlled by a lieterodimer of the ecdysone receptor and the product of the ultraspiracle gene (USP). Expression is induced by treatment with ecdysone or an analog of ecdysone such as muristerone A.
  • Additional regulator ⁇ ' elements that may be useful in vectors, include, but are not limited to, polyadenylation sequences, translation control sequences (e.g., an internal ribosome entry segment, IRES), enhancers, or iiitrons, Such elements may not be necessary, although they may increase expression by affecting transcription, stability of the mRNA, translational efficiency, or the like. Such elements can be included in a nucleic acid construct as desired to obtain optimal expression of the nucleic acids in the celi(s). Sufficient expression, however, may sometimes be obtained without such additional elements.
  • Vectors also can include other elements.
  • a vector can include a nucleic acid that encodes a signal peptide such that the encoded polypeptide is directed to a particular cellular location (e.g., a signal secretion sequence to cause the protein to be secreted by the cell) or a nucleic acid that encodes a selectable marker.
  • Non-limiting examples of selectable markers include puroraycin, adenosine deaminase (A DA), aminoglycoside phosphotransferase (neo, G418, APH), dihydrofolate reductase (DHFR), hygroniycin-B-phosphtransferase, thymidine kinase (T ), and xanthin-guanine phosphoribosyl transferase (XGPRT).
  • a DA adenosine deaminase
  • DHFR dihydrofolate reductase
  • DHFR dihydrofolate reductase
  • hygroniycin-B-phosphtransferase hygroniycin-B-phosphtransferase
  • T thymidine kinase
  • XGPRT xanthin-guanine phosphoribosyl transfera
  • Viral vectors can be used to form the conjugates, and include adenovirus, adeno- associated virus (AAV), retroviruses, lentiviruses, vaccinia virus, measles viruses, herpes viruses, and bovine papilloma virus vectors (see, Kay et al., Proc. Natl. Acad. Sci. USA 94: 12744-12746 (1997) for a re view of viral and non-viral vectors). Viral vectors are modified so the nativ e tropism and pathogenicity of t he v irus has been altered or removed. The genome of a virus also can be modified to increase its infectivity and to accommodate packaging of the nucleic acid encoding the polypeptide of interest.
  • AAV adeno- associated virus
  • retroviruses retroviruses
  • lentiviruses lentiviruses
  • vaccinia virus measles viruses
  • herpes viruses herpes viruses
  • Non-viral vectors can also be used in the subject conjugates.
  • the mammalian serum protein that is encoded by the vector is selected from the group consisting of a tissue-type plasminogen activator, a receptor of a tissue-type plasminogen activator, a streptokinase, a staphylokinase, a urokinase, and coagulation factors.
  • the invention also provides a method for treating associated with the formation of clots in its circulation, including the step of administering to the mammal a conjugate that causes the recombinant expression and secretion into the blood, such as from transduced muscle cells, of a therapeutically effective amount of such a mammalian serum protein.
  • An aptamer is a DNA or RNA molecule that has been selected from a random or biased pool of oligonucleic acids, based on its ability to bind to a target molecule.
  • Aptamers can be selected which bind nucleic acids, proteins, small organic compounds and specific cell surfaces, and several have been developed which bind to proteins which are associated with disease states.
  • Aptamers are in general more easily manufactured and are more amenable to chemical modification than are antibodies, and they can be "evolved" for tighter binding to the target by an iterative process of random modification and affinity-based selection.
  • the evolved aptamers often have antibody-like specificities, and are therefore expected to have utility in those applications, such as therapeutics and in vitro and in vivo diagnostics.
  • the method includes administering to a patient a scFv conjugate, coding sequence, vector or aptamer of the present invention,
  • the method includes contacting the cell or tissue with an scFv conjugate , coding sequence, vector or aptamer of the present invention.
  • the scFv antibody conjugates, coding sequences, vectors or aptamers of the present invention may be employed as therapeutic or prophylactic pharmacological agents in any subject in which it is desirable to administer, in vitro, ex vivo, or in vivo the subject antagonists that bind RORl; e.g., to treat CLL.
  • Typical subjects for treatment or management according to the methods herein are subjects presenting with a ROR l cancer; e.g., CLL,.
  • the antagonists described herein specifically recognize RORl protein, found in lymphoma samples but not expressed in cells of normal adults, and therefore can be used for detecting and/or neutralizing these biomolecules, and/or blocking their interactions with other biomolecules, in vitro or in vivo.
  • the methods herein may be used to target therapeutic antibodies, or nucleic acids encoding them, to particular target ceils.
  • the antibodies may be, for example, monoclonal antibodies, polyclonal antibodies, single-chain antibodies, or bi-specific antibodies.
  • Therapeutic antibodies are known to include, but are not limited to, Anti EGFr antibodies (e.g., panitumamab, Erbitux (cetuximab), matuzumab, IMC-IIF 8, TheraCIM liR3), denosumab, Avastin (bevacizumab), Anti-HGF antibodies, Elumira (adalimumab), Anti-Ang-2 antibodies, Herceptin (trastuzuniab), Remicade (infliximab), Anti-CD20 antibodies, rituximab, Synagis (palivizumab), Mylotarg (gemtuzumab oxogamicin), Raptiva (efalizumab), Tysabri (nataiizumab), Zenapax (dacliximab), NeutroSpec (Technetium (,sup.99mTc) fanolesomab), tocilizumab, ProstaScint (Indium-Ill labele
  • the selected scFv antibody conjugate, coding sequence, vector or aptamer may be administered alone, or in combination with, or conjugated to, one or more combinatorial therapeutic agents.
  • the scFV molecules described herein may exert a beneficial effect in the subject by a variety of mechanisms.
  • scFv antibody conjugates that specifically bind ROR1 are purified and administered to a patient to neutralize one or more forms of ROR1 , to block one or more activities of RORl, or to block or inhibit an interaction of one or more forms of RORl with another biomolec le; e.g., to treat CLL or other ROR l cancers.
  • the scFV molecules of the invention may include humanized antibodies, and can be combined for therapeutic use with additional active or inert, ingredients, e.g., in conventional pharmaceutically acceptable carriers or diluents, e.g., immunogenic adjuvants, and optionally with adjunctive or combinatorial ly active molecules such as antiinflammatory and anti-fibrinolytic drugs.
  • additional active or inert, ingredients e.g., in conventional pharmaceutically acceptable carriers or diluents, e.g., immunogenic adjuvants, and optionally with adjunctive or combinatorial ly active molecules such as antiinflammatory and anti-fibrinolytic drugs.
  • the scFv conjugates, coding sequences, vectors or aptamers described herein are coordinately administered with, co-formulated with, or coupled to (e.g., covalently bonded) a combinatorial therapeutic agent, for example a radionuclide, a differentiation inducer, a drug, or a toxin.
  • a combinatorial therapeutic agent for example a radionuclide, a differentiation inducer, a drug, or a toxin.
  • a combinatorial therapeutic agent for example a radionuclide, a differentiation inducer, a drug, or a toxin.
  • a combinatorial therapeutic agent for example a radionuclide, a differentiation inducer, a drug, or a toxin.
  • a combinatorial therapeutic agent for example a radionuclide, a differentiation inducer, a drug, or a toxin.
  • Suitable drugs for use in such combinatorial treatment formulations and methods include methotrexate, and
  • Suitable toxins include ricin, abrin, diptheria toxin, cholera toxin, gelonin, Pseudotnonas exotoxin, Shigella toxin, and pokeweed antiviral protein.
  • These combinatorial therapeutic agents can be coupled to an anti-RORl antibody either directly or indirectly (e.g., via a linker group). A direct reaction between an agent and an antibody is possible when each possesses a substituent capable of reacting with the other.
  • a nucleophilic group such as an amino or sulfhydryi group
  • a carbonyl-containing group such as an anhydride or an acid halide, or with an alky] group containing a good leaving group (e.g., a halide) on the other.
  • a linker group can also serve to increase the chemical reactivity of a substituent on an agent or an antibody, and thus increase the coupling efficiency.
  • polyfunctional reagents both homo- and hetero-functional (such as those described in the catalog of the Pierce Chemical Co., Rockford, 111.), may be employed as a linker group.
  • Coupling may be affected, for example, through amino groups, carboxyl groups, sulfhydryi groups or oxidized carbohydrate residues.
  • kits of the invention may contain an scFv antibody that specifically binds ROR1 optionally linked to an appropriate carrier, a freeze-dried preparation or a solution of an enzyme- labeled monoclonal antibody which can bind to the same antigen together with the monoclonal antibody or of a polyclonal antibody labeled with the enzyme in the same manner, a standard solution of purified ROR.1, a buffer solution, a washing solution, pipettes, a reaction container and the like.
  • kits optionally include labeling and/or instructional materials providing directions (i.e., protocols) for the practice of the methods described herein in an assay environment.
  • instructional materials typically comprise written or printed materials, they are not limited to such. Any medium capable of storing such instructions and communicating them to an end user is contemplated. Such media include, but are not limited to electronic storage media (e.g., magnetic discs, tapes, cartridges, chips), optical media (e.g., CD ROM), and the like. Such media may include addresses to internet sites that provide such instructional materials,
  • the scFV having SEQ ID NO: 3 was inserted upstream of the sequence encoding an IgG4 CH domain, the transmembrane domain of CD28 and cytoplasmic domain of the zeta chain for the T-cell receptor (CD247) to generate a chimeric antigen receptor (CAR).
  • the construct was inserted into a lenti-virus vector for transduction of T cells (Jurkat).
  • the T cells were examined for their capacity to bind a soluble ROR1 protein (ROR1- Ig).
  • ROR1- Ig soluble ROR1 protein
  • Cell-bound RGRl-ig could be detected by staining the cells with an anti-ig antibody that was labeled with a fluorescence dye, phycoerythrin (PE), This allowed monitoring of changes in fluorescence of the T ceils caused by binding the RORl protein using flow cytometry.
  • PE phycoerythrin

Abstract

Compositions including an antibody single-chain variable fragment (scFv) conjugate that specifically binds to ROR1 tumor-associated antigen are provided. The anti-ROR1 scFv antibody and conjugates may include a biologically-active molecule. Such conjugates may comprise a chimeric receptor to direct T cells to respond to ROR1 cancer cells, Methods to use the scFV conjugates to target cells expressing ROR1 for therapeutic and diagnostic purposes are also provided.

Description

BACKGROUND OF THE INVENTION
FIELD OF THE INVENTION
[0001 ] The present invention relates generally to single chain Fv fragments of tumor- associated antigen binding antibodies. The invention further relates to single chain Fv conjugate molecules and their use in the deliver}' of biologically active molecules to cancer cells as well as in directing T cells to respond to tumor-associated antigen expressing cancer cells.
BACKGROUND INFORMATION
[0002] Microarray analyses have revealed that there are genes expressed by CLL cells that are not expressed in other lymphoid tissues. Coupled with the observation that CLL cells also express a highly restricted immunoglobulin repertoire, cells likely express distinctive antigens that also could be targeted for immunotherapy. Conceivably, chronic lymphocytic leukemia (CLL.) cells in particular also might have such leukemia-associated antigens (LAA).
[0003] However, CLL, patients typically develop hypogammmaglobulinemia and worsening immune deficiency, which impairs their immune response to LAA-targeted vaccmes.
Implicated in the abnormal immune function are immime-suppressive factors and an acquired functional deficiency of CD 154. Furthermore, CLL cells are particularly poor at antigen presentation, which appears in part secondary to inadequate leukemia-cell expression of immune co-stimulatory/adhesion molecules.
[0004] Indeed, despite the presence of tumor-associated antigens (TAAs) like LAAs expressed in different cancers, cancer patients typically are unable to mount an effective immune response against existing or developing tumors. TAA binding antibodies have been used to develop potential cancer vaccines with mixed degrees of success. Many such TAAs represent developmental or differentiation antigens that have restricted expression. Fc containing antibodies to TAAs are often cleared relatively quickly from circulation and may be recognized only by one type of MHC molecule, limiting their usefulness in therapy. [0005] Activation of CLL cells via CD40-ligation can reverse its immune-suppressive phenotype. Furthermore, CLL cells transduced with an adenovirus encoding the ligand for CD40 (Ad-CD 154) can function as more effective antigen-presenting cells (APCs). In addition, they can effect ligation of CD40 on bystander leukemia B cells and stimulate autologous leukemia-reactive T cells both in vitro and in vivo. However, targeting of CD 154 modified T cells to CLL. cells remains to be achieved.
[0006] Tyrosine kinases are important mediators of the signaling cascade, determining key roles in diverse biological processes like growth, differentiation, metabolism and apoptosis in response to external and internal stimuli. Studies ha ve implicated the role of tyrosine kinases in the pathophysiology of cancer. Schlessinger J. (2000) Cell, 103:211-225; and Robinson et al. (2000) Oncogene, 19:5548-5557. MacKeigan and colleagues used a large-scale RNAi approach to identify kinases that might regulate survival and apoptosis of a human tumor cell line (HeLa), RNAi to RORl was found as one of the most potent in inducing apoptosis among the set of RNAi targeting each of 73 different kinase-encoding genes. MacKeigan et al. (2005) Nat Cell Biol, 7:591-600, However, these investigators did not examine the expression or function of ROR l protein in these cells.
[0007] RORl is a membrane-receptor with an intracellular kinase-like domain and extracellular Frizzled-like cysteine-rich domain, which is common to receptors of members of the Wnt-family, RORl is member of the ROR family that is evoiutionarily conserved among Caenorhavditis elegans, Drosophi!a, mice and humans. Wilson C, Goberdhan D C, Steller H. Dror, a potential neurotrophic receptor gene, encodes a Drosophila homolog of the vertebrate Ror family of Trk-related receptor tyrosine kinases. Proc Natl Acad Sci USA. 1993; 90:7109- 71 13; Oishi et al. (1997) J Biol. Chem., 272: 1 1916-1 1923; Masiakowski et al. (1992) J Biol. Chem., 267:261 81-26190; Forrester et al. (2002) Cell Moi Life Sci,, 59:83-96; and Oishi et al. (1999) Genes Cells, 4:41-56. In rodents, RORl is expressed primarily in developing cephalic neural crest in the dorsal part of the diencephalons and mid-hind brain boundary during embryogenesis. In most species examined, expression of RORl apparently attenuates during embryonic development, becoming negligible at term. RORl mRNA was reported to express in infant brain, renal cancer and colon cancer. In a recent study, it was found that RORl , at both mRNA and protein level, was highly expressed in CLL B cells but not normal B cells. Moreover, it was found that ROR l is a receptor for Wni5a, which could induce activation of NF-KB when co-expressed with RORl in HEK293 cells and enhance survival of C LL cells in vitro, Another study found that RORl was expressed in acute lymphocytic leukemia (ALL) as well. Shabani et al. (2007) Tumour Biol., 28:318-326; and Baskar et a!. (2008) Clin Cancer Res., 14:396-404.
[0008] Expression of full-length RORl in numerous cancer cell lines and samples, but not other tissues, including blood or splenic lymphocytes of non- leukemic patients or normal adult donors, and also generated mouse anti-sera against full-length human RORl . Fukuda et al., Blood: ASH Annua] Meeting Abstracts 2004 104, Abstract 772 (2004) (incorporated herein by reference in its entirety). The polypeptide and coding sequences for RORl have been reported elsewhere and are also incorporated herein by this reference (see, e.g., Accession Nos.
NP__005003.2 and M_005012). Surprisingly, it has also been discovered that cancer cells which express the WntSa protein, such as CLL ceils, not only bind RORl but have a survival advantage conferred as a consequence.
[0009] Thus, RORl is an embryonic protein that is expressed uniquely on certain cancer cells, including in CLL, small lymphocytic lymphoma, marginal cell B-Cell lymphoma, Burkett's Lymphoma, and other cancers (e.g., breast cancers), but not on normal adult tissues and cells. It is therefore a potential TAA target for modulation of an immune response to RORl expressing cancer cells ("RORl cancers").
[0010] Yet, as noted, patients with CLL typically develop disease-related
hypogammaglobulinemia and respond poorly to vaccines. The progressive acquired immune deficiency associated with CLL accounts for much of the morbidity related to this disease. However, as shown previously, following treatment with autologous Ad-CD 154-transduced CLL cells, most patients had increased serum IgM and IgG and developed a specific antibody response against adenovirus and some developed anti-CLL autoantibodies. Although virus infections occasionally can induce autoantibodies, autoantibodies were not detected against other blood cells or human CD 154. Likewise, there were no increases in the titer of antibodies to a recall antigen, tentanus toxoid, except in one patient who was immunized with tetanus toxoid following the second infusion of autologous Ad-CD 154-CLL ceils. Therefore, CLL patients could respond well against other vaccines administered during the course of such treatment, potentially allowing for generation of protective immunity against infectious agents that commonly afflict patients with this disease. The invention provides such a vaccine, as well as targeting means for drug delivery to RORl cancer cells. SUMMARY OF THE INVENTION
[0011] The present invention is based on the seminal generation of an anti-RORl single- chain variable fragment (scFv) antibody, as well as conjugates thereof which include a biologically active molecule. Such conjugates may comprise a chimeric receptor to direct T ceils to respond to ROR1 cancer cells. Thus, the scFv may be used to target ceils expressing RQR 1 for therapeutic and diagnostic purposes,
[0012] In one aspect, the present invention provides an antibody single-chain variable fragment (scFv) conjugate. The conjugate includes an scFv having an amino acid sequence at least 90% identical to SEQ ID NO: 1 or SEQ ID NO: 2, and a biologically active molecule. In various embodiments, the biologically active molecule is selected from the group consisting of a polypeptide, a peptidomimetic, a nucleic acid molecule, a lipid, a drug compound, chemotherapeutic agent, a iiposome, an antagonist and an agonist.
[0013] In another aspect, the present invention provides an isolated peptide including an amino acid sequence at least 90% identical to SEQ ID NO: 1 or SEQ ID NO: 2.
[0014] In various embodiments, the conjugate and peptide include an amino acid sequence at least 95% identical to SEQ ID NO: 1 or SEQ ID NO: 2, In exemplary embodiments, the conjugate and peptide has a binding specificity of an antibody as produced by a hybridoma having Accession No. PTA 8634 by the American Type Culture Collection.
[0015] In another aspect, the present invention provides an isolated nucleic acid encoding the conjugate or peptide of the in vention.
[0016] In another aspect, the present invention provides a pharmaceutical composition including the conjugate or peptide of the invention an optionally a pharmaceutically acceptable carrier,
[0017] In another aspect, the present invention provides a method of producing a scFv antibody conjugate. The method includes transforming a host cell with an expression construct including a nucleic acid molecule encoding a scFv antibody and a polypeptide, the scFv peptide including an amino acid sequence at least 90% identical to SEQ ID NO: 1 or SEQ ID NO: 2; and culturhig the host cell under conditions suitable for producing the conjugate, thereby producing the protein conjugate. [0018] In another aspect, the present invention provides a method for detecting RO 1 protein in a sample, utilizing a detectably labeled scFv of the in vention.
[0019] in another aspect, the present invention provides a method of targeting a biologically active molecule to a cell having an ROR1 receptor. The method includes contacting the cell with an scFv conjugate of the invention.
[0020] In another aspect, the present invention provides a kit to detect the presence of ROR1 protein in a sample from a subject that is known or suspected to contain cancer cells. The kit includes the an scFv antibody conjugate or peptide of the invention and instructions for its use in an assay environment.
[0021] in another aspect, the present invention provides a method for treating a R.OR1 cancer in a human subject using an scFv antibody conjugate or peptide of the invention.
[0022] In another aspect, the present invention provides a method for treating or preventing CLL in which ROR1 binding of WntSa on CLL ceils confers a survival advantage thereon by administering an scFv antibody conjugate or peptide of the invention.
[0023] In another aspect, the present invention provides a method of monitoring a therapeutic regimen for treating a subject having or at risk of having an RORl cancer using using an scFv antibody conjugate or peptide of the invention.
BRIEF DESCRI PTION OF THE DRAWINGS
[0024] Figure 1 shows the amino acid sequence (SEQ ID NO: 1) of an anti-RGRl mAb scFv. The amino acid sequence includes a leader sequence (amino acid residues 1 to 25) and VL (amino acid residues 25 to 132) and VH (amino acid residues 151 to 269) chains joined by a linker sequence (amino acid residues 133 to 150).
[0025] Figure 2 shows the amino acid sequence (SEQ ID NO: 2) of an anti-RORl mAb scFv after cleavage of the leader during processing of the polypeptide,
[0026] Figure 3 shows the nucleotide sequence encoding the anti-RORl mAb scFv (SEQ ID NO: 3) of SEQ ID NO: 1.
[0027] Figure 4 shows the nucleotide sequence encoding the anti-RORl mAb scFv (SEQ ID NO: 4) of SEQ ID NO: 2. [0028] Figure 5 shows plots of fluorescence intensity. The left plot shows intensity in a Jurkat T cell line and the right shows a Jurkat T cell line transduced to express an ROR.1 conjugate including anti-RORl niAb scFv, an IgG4 CH domain, a transmembrane domain of CD28, and a cytoplasmic domain of the zeta chain for a T-cell receptor (CD247) to generate a chimeric antigen receptor (ROR1-CAR). The shaded histogram indicates the autofluorescence of the unstained ceils. The open histogram demonstrates the fluorescence of cells inc ubated with a soluble ROR1 protein (RORl-Ig) and then stained with anti-Ig phycoerythrin (PE).
[0029] Figure 6 shows the results of F ACS analysis for PC D A 3.1 CH2CH3 stable scFV clones targeting of Endemic African Burkitt's lymphoma B cells (EW36). Results for non- ROR1 expressing CHO cells are provided for comparison.
DETAILED DESCRIPTION OF THE INVENTION
[0030] The present invention is based on the seminal generation of as anti-RORl scFv antibody and conjugates thereof including a biologically-active molecule. Such conjugates may comprise a chimeric receptor to direct T cells to respond to ROR1 cancer cells. Thus, the scFv may be used to target cells expressing ROR1 for therapeutic and diagnostic purposes.
[0031] Before the present methods are described, it is to be understood that this invention is not limited to particular compositions, methods, and experimental conditions described, as such compositions, methods, and conditions may vary. It is also to be understood that the terminology used herein is for purposes of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only in the appended claims.
[0032] As used in this specification and the appended claims, the singular forms "a", "an", and "the" include plural references unless the context clearly dictates otherwise. Thus, for example, references to "the method" includes one or more methods, and/or steps of the type described herein which will become apparent to those persons skilled in the art upon reading this disclosure and so forth.
[0033] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. [0034] As shown previously, some patients developed IgG anti-CLL autoantibodies, which reacted with RORl . These post-treatment antisera reacted with a protein of about.125 kD found in lysates of CLL cells or CHO-ROR1 cells, but not in lysates prepared from human blood l ymphocytes of normal donors or non-transfected CHO cells. Th a th ese antisera reacted with RORl was corroborated via ELISA using a recombinant RORl fusion-protein. Because th ese antisera appeared specific for CLL cells, the expression of RORl on other adult tissues was studied. These studies revealed that expression of the RORl protein was restricted to CLL B-cells and was not found on the non-leukemic blood or marrow mononuclear cells of patients with CLL, potential ly allowing for detection of CLL cells in the blood or marrow of patients with early-stage disease or minimal residual disease after therapy. Furthermore,, RORl was not found on normal adult tissues or lymphoid cells, including CD5-positive B cells. As such, RORl appears to represent a specific leukemia-associated antigen (LAA).
[0035] RORl encodes a type I membrane receptor tyrosine kinase that initially was identified using oligonucleotide primers targeting sequences encoding amino acid sequences common to tyrosine kinase domains of different proteins. This protein appears highly conserved throughout evolution. RORl is evolutionary conserved among Caenorhabditis elegans (C. elegans), Aplysia, Drosophd' a melanogaster, Xenopus, mice, and humans. In rodents, RORl is expressed primarily in developing cephalic neural crest in die dorsal part of the diencephalons and mid-hind brain boundary during embryogenesis. Work in
Caenorhabditis elegans (C. elegans) indicated that the RORI-type kinases might be involved in the regulation of cell motility and in asymmetric cell division during embryogenesis.
[0036] Furthermore, the ROR protein in C. elegans, apparently has both kinase-dependent and kinase independent ROR-family receptor tyrosine kinases are characterized by the intracellular tyrosine kinase domains, highly related to those of the Trk-family receptor tyrosine kinases, and by the extracellular Frizzled-like cysteine-rich domains and kringle domains, which are common to receptors of the Wnt-family members. An ortholog to RORl, namely ROR2, has been found interact physically with WntSa to activate non-canonical Wnt- signaling.
[0037] As previously described, ROR l interacts physically with WntSa. The interaction of RORl with WntSa was implicated in studies demonstrating that co-transfection of expression vectors encoding NF-κΒ reporter constructs, RORl , and WntSa, but not other Wnt factors, could induce activation of NF-NB in a mutually dose-dependent fashion. This activity was determined to be independent of expression of LPR5/6, which ordinarily serves as a co- receptor for Wnt receptors. Physical interaction of RORl with Wnt5a was observed using recombinant proteins, demonstrating that RORl has binding activity for Wnt5a independent of LPR5/6. Nonetheless, Wnt a apparently could not activate the canonical Wnt-signaling pathway when co-expressed with RORl in 293 cells.
[0038] Prior studies suggested that there is crosstalk between the Wnt-signaiing pathway and the NF-KB-signaling pathway, β-catenin apparently can physically complex with NF-KB, resulting in reduction of NF-κΒ D A binding, transactivation activity, and target gene expression. Whereas NB kinase-alpha (Ii a), involved in the phosphorylation ofNF- f B2/pl00, can phosphorylate and stabilize β-catenin, the kinase involved in activation of the canonical p50/p65 NF-κΒ pathway, namely ΙκΚΒ, can enhance β-catenin degradation.
Conceivably, the inability of RORl to activate the LEF/TCF-signaling may be secondary in part to its capacity to activate ΙκΚΡ, leading to reduced stability of p-catenin and enhanced activity of NF-κΒ, which in itself could potentially play a role in cancer development.
Furthermore, expression of RORl in CLL could contribute to the growth and/or survival of neoplastic cells induced by interaction with tissue stromal cells that might elaborate Wnt5a and other factors that activate NF-κΒ. Even though found circulating in the blood, CLL cells derive a survival benefit from interactions with marrow stromal cells, nurse-like cells, or dendritic cells, which are found in the leukemia-infiltrated marro or lymphoid tissues of patients with this disease. It is noteworthy in this regard that dendritic cells have been found to express high- levels of WntSa.
[0039] it has also previously been determined that co-culture of CLL, cells with CHO- WntSa cells maintained significantly higher viability over time in vitro than the same CLL cells co-cultured with CHO cells, which serves to control for other factors that might influence CLL-cell survival. These studies provides the first e vidence that the survival of CLL cells can be enhanced in vitro by such Wnt factors, which presumably also might function to enhance the survival of CLL, cells in iymphoid-tissue microenvironments containing cells that express Wnt5 a.
[0040] Although there are other receptors for WntSa, RORl appears to be at least in part responsible for the survival-signal triggered by co-culture with Wnt5a-expressing CHO cells. This is indicated by findings that serum obtained after treatment with autologous Ad-CD 154- CLL cells could neutralize the capacity of CHO-Wnt5a cells to enhance the survival of CLL cells over that of CLL cells co-cultured with CHO cells or CLL cells cultured alone.
Absorption of such antisenim with CHO-R.OR1 cells abrogated the capacity of the post- treatment serum to neutralize the activity of CHO-Wnt5a cells. Conceivably, such anti-RQRl antibodies could be responsible for some of the size-reductions observed in the lymph nodes of patients who had received infusions of autologous Ad-CD 154-CLL cells.
[0041] Whole antibodies that can react with native ROR1 protein have also been described in U.S. Patent Application No. 12/545731 which is incorporated herein by reference.
However, antibody variants or fragments that maintain specific binding with the native ROR1 protein are necessary as they provide increased flexibility in designing a wide variety of antibody conjugates that may be used for therapeutic and diagnostic purposes.
[0042] In one aspect, therefore, the present invention provides an antibody single-chain variable fragment (scFv) conjugate. The conjugate includes an scFv having an amino acid sequence at least 90%, 95% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 2 as shown in Figures 1 and 2, and a biologically active molecule. In exemplary embodiments, the conjugate and peptide has a binding specificity of an antibody as produced by a hybridoma having Accession No. PTA 8634 by the American Type Culture Collection.
[0043] in another aspect, the present invention provides an isolated peptide including an amino acid sequence at least 90%, 95% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 2.
[0044] A "conjugate" as used herein generally refers to a molecule which contains a peptide including an amino acid sequence at least 90%, 95% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 2, conjugated with a biologically active molecule. Typically, the conjugate is capable of being targeted to a cell expressing R.OR.1.
[0045] As used herein, the phrase "biologically active molecule" refers to a molecule that has a biological effect in a cell. In certain embodiments the active molecule may be an inorganic molecule, an organic molecule, a small organic molecule, a drug compound, a peptide, a polypeptide, a peptidomimetic, a nucleic acid molecule, a lipid, a drug compound, chemotherapeutic agent, a liposome, an antagonist or an agonist.
[0046] For example, in various embodiments, the active molecule may be doxorubicin. [0047] As used herein, "specific binding" refers to antibody binding to a predetermined antigen. Typically, the antibody binds with an affinity corresponding to a KD of about 10"8 M or less, and binds to the predetermined antigen with an affinity (as expressed by KD) that is at least 1 0 fold less, and preferably at least 100 fold less than its affinity for binding to a nonspecific antigen (e.g. , BSA, casein) other than the predetermined antigen or a closely-related antigen. Alternatively, the antibody can bind with an affinity corresponding to a KA of about 106
Figure imgf000011_0001
or 109 M"1 or higher, and binds to the
predetermined antigen with an affinity (as expressed by KA) tha is at least 10 fold higher, and preferably at least 100 fold higher than its affinity for binding to a non-specific antigen (e.g. , BSA, casein) other than the predetermined antigen or a closely-related antigen. In some embodiments the scFv antibody conjugate or peptide of the in vention wil l have the same A or KD as an antibody produced by the hybridoma having ATCC accession number PTA 8634.
[0048] The term ¾" (sec"'), as used herein, is intended to refer to the dissociation rate constant of a particular antibody-antigen interaction. This value is also referred to as the off value. The term "KD" (3VT1), as used herein, is intended to refer to the dissociation equilibrium constant of a particular antibody-antigen interaction.
[0049] The term "ka" (M'lsec_1), as used herein, is intended to refer to the association rate constant of a particular antibody-antigen interaction. The term "KA" (M), as used herein, is intended to refer to the association equilibrium constant of a particular antibody-antigen interaction.
[0050] Naturally occurring antibodies are generally tetramers containing two light chains and two heavy chains. Experimentally, antibodies can be cleaved with the proteolytic enzyme papain, which causes each of the heavy chains to break, producing three separate sub units. The two units that consist of a light chain and a fragment of the heavy chain approximately equal in mass to the light chain are called the Fab fragments (i.e., the "antigen binding" fragments). The third unit, consisting of two equal segments of the heavy chain, is called the Fc fragment. The Fc fragment is typically not involved in antigen-antibody binding, but is important in later processes involved in ridding the body of the antigen.
[0051] Because Fab and F(ab')2 fragments are smaller tha intact antibody molecules, more antigen-binding domains are available than when whole antibody molecules are used.
Proteolytic cleavage of a typical IgG molecule with papain is known to produce two separate antigen binding fragments called Fab fragments which contain an intact light chain linked to an amino terminal portion of the contiguous heavy chain via by disulfide linkage. The remaining portion of the papain-digested immunoglobin molecule is known as the Fc fragment and consists of the carboxy terminal portions of the antibody left intact and linked together via disulfide bonds. If an antibody is digested with pepsin, a fragment known as an F(ab')2 fragment is produced which lacks the Fc region but contains both antigen-binding domains held together by disulfide bonds between contiguous light and heavy chains (as Fab fragments) and also disulfide linkages between the remaining portions of the contiguous heavy chains (Handbook of Experimental immunology. Vol 1 : immunocheniistry, Weir, D. M, Editor, Biackwell Scientific Publications, Oxford (1986)).
[0052] As readily recognized by those of skill in the art, altered antibodies (e.g., chimeric, humanized, CDR-grafted, bifunctional, antibody polypeptide dimers (i.e., an association of two polypeptide chain components of an antibody, e.g. , one arm of an antibody including a heavy chain and a light chain, or an Fab fragment including VL, VH, CL and CH antibody domains, or an F fragment comprising a VL domain and a VH domain), single chain antibodies (e.g. , an scFv (i.e. , single chain Fv) fragment including a VL domain linked to a VH domain by a linker, and the like) can also be produced by methods well known in the art.
[0053] Recombinant DNA methods have been developed which permit the production and selection of recombinant single chain Fv fragments (scFvs or scFv antibodies). Further, scFvs can be dimerized to produce a diabody. scFvs bind a specific epitope of interest and can be produced using any of a variety of recombinant bacterial phage-based methods, for example as described in Lowman et al. (1 991) Biochemistry, 30, 10832-10838; Clackson et ai. (1991 ) Nature 352, 624-628; and Cwirla et al. (1990) Proc. Natl. Acad. Sci. USA 87, 6378-6382. These methods are usually based on producing genetically altered filamentous phage, such as recombinant M 13 or fd phages, which display on the surface of the phage particle a
recombinant fusion protein containing the antigen-binding scFv antibody as the amino terminal region of the fusion protein and the minor phage coat protein g3p as the carboxy terminal region of the fusion protein. Such recombinant phages can be readily grown and isolated using well-known phage methods. Furthermore, the intact phage particles can usually be screened directly for the presence (display) of an antigen-binding scFv on their surface without the necessity of isolating the scFv away from the phage particl e. [0054] To produce an scFv, standard reverse transcriptase protocols are used to first produce cDNA from mR A isolated from a hybridoma that produces an mAb for ROR-1 antigen, The cDNA molecules encoding the variable regions of the heavy and light chains of the mAb can then be amplified by standard polymerase chain reaction (PGR) methodology using a set of primers for mouse immunoglobulin heavy and light variable regions (Clackson (1991 ) Nature, 352, 624-628), The amplified cDNAs encoding mAb heavy and light chain variable regions are then linked together with a linker oligonucleotide in order to generate a recombinant scFv DNA molecule. The scFv DNA is ligated into a filamentous phage plasmid designed to fuse the amplified cDNA sequences into the 5' region of the phage gene encoding the minor coat protein called g3p, Escherichia coli bacterial cells are than transformed with the recombinant phage plasmids, and filamentous phage grown and harvested. The desired recombinant phages display antigen-binding domains fused to the amino terminal region of the minor coat protein. Such "display phages" can then be passed over immobilized antigen, for example, using the method known as "panning", see Parmley and Smith (1989) Adv. Exp, Med. Biol. 251, 215-21 8; Cwiria et ai. (1990) Proc. Natl. Acad. Sci. USA 87, 6378-6382, to adsorb those phage particles containing scFv antibody proteins that are capable of binding antigen. The antigen-binding phage particles can then be amplified by standard phage infection methods, and the amplified recombinant phage population again selected for antigen- binding ability. Such successive rounds of selection for antigen-binding ability, followed by amplification, select for enhanced antigen -bin ding ability in the scFvs displayed on
recombinant phages. Selection for increased antigen-binding ability may be made by adjusting the conditions under which binding takes place to require a tighter binding activity. Another method to select for enhanced antigen-binding activity is to alter nucleotide sequences within the cDN A encoding the binding domain of the scFv and subject recombinant phage
populations to successi ve rounds of selection for antigen-binding activity and amplification (see Lowman et al. (1991) Biochemistry 30, 10832-10838; and Cwiria et ai. (1990) Proc. Natl. Acad. Sci. USA 87, 6378-6382).
[0055] Once an scFv is selected, it can be produced in a free form using an appropriate vector in conjunction with E. coli strain HB2151. These bacteria actually secrete scFv in a soluble form, free of phage components (Hoogenboom et al. (1991) Nucl, Acids Res. 19, 4133- 4137). The purification of soluble scFv from the HB2151 bacteria culture medium can be accomplished by affinity chromatography using antigen molecules immobilized on a solid support such as AFFIGEL™ (BioRad, Hercules, Calif.). [0056] Other developments in the recombinant antibody technology demonstrate possibilities for further improvements such as increased avidity of binding by polymerization of scFvs into dimers and tetramers (see Holliger et al, (1993) Proc, Natl. Acad, Sci. USA 90, 64446448),
[0057] Because scFvs are even smaller molecules than Fab or F(ab')2 fragments, they can be used to attain even higher densities of antigen binding sites per unit of surface area when immobilized on a solid support material than possible using whole antibodies, F(abf)2, or Fab fragments. Furthermore, recombinant antibody technology offers a more stable genetic source of antibodies, as compared with hybridomas. Recombinant antibodies can also be produced more quickly and economically using standard bacterial phage production methods.
[0058] To produce scFvs described herein recombinant!}', nucleic acids encoding one is inserted into expression vectors. The light and heavy chains can be cloned in the same or different expression vectors. The teachings of U.S. Patent No, 6,287,569 to Kipps et al., incorporated herein by reference in its entirety, and the methods provided herein can readily be adapted by those of skill in the art to create the scFvs of the present invention.
[0059] Expression vectors are typically replicable in the host organisms either as episomes or as an integral part of the host chromosome. E. coli is one procaryotic host particularly useful for expressing antibodies of the present invention. Other microbial hosts suitable for use include bacilli, such as Bacillus s btilus, and other enterobacteriaceae, such as Salmonella, Serratia, and various Pseudomonas species. In these prokaryotic hosts, one can also make expression vectors, which typically contain expression control sequences compatible with the host ceil (e.g., an origin of replication) and regulator}' sequences such as a lactose promoter system, a tryptopha (trp) promoter system, a beta-lactamase promoter system, or a promoter system from phage lambda. Other microbes, such as yeast, may also be used for expression, Saccharomyces is a preferred host, with suitable vectors having expression control sequences, such as promoters, including 3-phosphogiycerate kinase or other glycolytic enzymes, and an origin of replication, termination sequences and the like as desired. Mammalian tissue ceil culture can also be used to express and produce the antibodies of the present invention (see, e.g., Winnacker, From Genes to Clones VCH Publishers, N.Y., 1987). Eukaryotic cells are preferred, because a number of suitable host cell lines capable of secreting intact antibodies have been developed. Preferred suitable host cells for expressing nucleic acids encoding the immunoglobulins of the invention include: monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line; baby hamster kidney cells (BHK, ATCC CCL 10); Chinese hamster ovary-cells (CHO); mouse Sertoli cells; monkey kidney cells (CV1 ATCC CCL 70); african green monkey kidney cells (VERO-76, ATCC CRL, 1587); human cervical carcinoma cells (HELA, ATCC CCL, 2); canine kidney cells (MDC , ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL, 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL51); and TRI cells.
[0060] The vectors containing the polynucleotide sequences of interest can be transferred into the host cell. Calcium chloride transfection is commonly utilized for prokaryotic cells, whereas calcium phosphate treatment or eiectroporation can be used for other cellular hosts (see, e.g., Sambrook et al, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, 2nd ed., 1989). After introduction of recombinant DNA, cel l lines expressing immunoglobulin products are cell selected. Cell lines capable of stable expression are preferred (i.e., undiminished le vels of expression after fifty passages of the cell line),
[0061] Once expressed, the scFv or other immunoglobulin Forms of the present invention can be purified according to standard procedures of the art, including ammonium sulfate precipitation, affinity columns, column chromatography, gel electrophoresis and the like (see, e.g., Scopes, Protein Purification, Springer- Verlag, N.Y., 1982). Substantially pure
immunoglobulins of at least about 90 to 95% homogeneity are preferred, and 98 to 99% or more homogeneity most preferred.
[0062] A labeled antibody or a detectably labeled antibody is generally an antibody (or antibody fragment which retains binding specificity), having an attached detectable label. The detectable label is normally attached by chemical conjugation, but where the label is a polypeptide, it could alternatively be attached by genetic engineering techniques. Methods for production of detectably labeled proteins are well known in the art. Detectable labels known in the art include radioisotopes, fiuorophores, paramagnetic labels, enzymes (e.g., horseradish peroxidase), or other moieties or compounds which either emit a detectable signal (e.g., radioactivity, fluorescence, color) or emit a detectable signal after exposure of the label to its substrate. Various detectable label/substrate pairs (e.g., horseradish
peroxidase/diarninohenzidme, avidin/streptavidin, lucii "erase/3 uciferin), methods for labeling antibodies, and methods for using labeled antibodies are well known in the art (see, for example, Harlow and Lane, eds., 1988, Antibodies; A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.) Another technique which may also result in greater sensitivity consists of coupling the antibodies to low molecular weight haptens. These haptens can then be specifically detected by means of a second reaction. For example, it is common to use such haptens as hiotin, which reacts with avidin, or dinitrophenyl, pyridoxal, and fluorescein, which can react with specific antihapten antibodies.
[0063] Antibodies may be humanized by replacing sequences of the Fv variable region which are not directly involved in antigen binding with equivalent sequences from human Fv variable regions. General reviews of humanized chimeric antibodies are provided by Morrison et al, (Science 229: 12024207 (1985)) and by Oi et al. (BioTechniques 4:214 (1986)). Those methods include isolating, manipulating, and expressing the nucleic acid sequences that encode all or part of immunoglobulin Fv variable regions from at least one of a heavy or light chain. Sources of such nucleic acid are wel l known to those skil led in the art and, for example, may be obtained from for example, an antibody producing hybridoma. The recombinant DNA encoding the humanized or chimeric antibody, or fragment thereof, can then be cloned into an appropriate expression vector.
[0064] Humanized antibodies can alternatively be produced by CDR substitution (U.S. Pat. No. 5,225,539; Jones, Nature 321:552-525 (1986); Verhoeyan et al, Science 239:1534 (1988); and Beidler, J. Immunol. 141 :4053-4060 (1988)). Thus, in certain embodiments, the antibody used in the conjugate is a humanized or CDR-grafted form of an antibody produced by the hybridoma having ATCC accession number PTA 2439. In other embodiments the antibody is a humanized or CDR-grafted form of antibody mAb 3E10. For example, the CDR regions can include amino acid substitutions such as 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid differences from those shown in the figures. In some instances, there are anywhere from 1-5 amino acid differences.
[0065] As used herein, reference to "an scFv antibody having the binding specificity of an antibody as produced by the hybridoma having ATCC accession number PTA 8634" includes antibody fragments having at least 90% or 95% sequence identity to SEQ ID NO:l or SEQ ID NO: 2 and retain the same binding specificity as that produced by the hybridoma having ATCC accession number PTA 8634, such as variants modified by mutation to improve the utility thereof (e.g., improved ability to target specific cell types and the like). Such variants include those wherein one or more conservative substitutions are introduced into the heavy chain and/or the light chain of the scFv. [0066] Such variants include those wherein one or more substitutions are introduced into the heavy chain nucleotide sequence and/or the light chain nucleotide sequence of the scFv. in some embodiments the variant has a light chain and/or heavy chain having a nucleotide sequence at least 80% or at least 90% or at least 95% identical to the nucleotide sequence set forth in SEQ ID NO: 3 or SEQ ID NO: 4.
[0067] In embodiments where the conjugates include polypeptides (i.e., protein conjugates), they may be designed as fusion protein conjugate such that the conjugate can be expressed in a host ceil as a fusion protein. Alternatively, portions of the conjugate can be chemically linked by peptide bonds or by a chemical or peptide linker molecule of various types well known in the ail as discussed further herein.
[0068] A variety of linkers may be used to link portions of the conjugates described herein. The term "degradable linker" as used herein, refers to linker moieties that are capable of cleavage under various conditions. Conditions suitable for cleavage can include but are not limited to H, UV irradiation, enzymatic activity, temperature, hydrolysis, elimination, and substitution reactions, and thermodynamic properties of the linkage. The term "photolabile linker" as used herein, refers to linker moieties as are known in the art that are selectively cleaved under particular UV wavelengths. Compounds of the invention containing photolabile linkers can be used to deliver compounds to a target cell or tissue of interest, and can be subsequently released in the presence of a UV source.
[0069] The term "linker" as used herein is any bond, small molecule, or other vehicle which allows the substrate and the active molecule to be targeted to the same area, tissue, or cell, for example by physically linking the individual portions of the conjugate.
[0070] In certain embodiments, a cleavable or degradable linker may be used. In one embodiment the linker is a chemical bond between one or more substrates and one or more therapeutic moieties. Thus, the bond may be covalent or ionic. An example of a therapeutic complex where the linker is a chemical bond would be a fusion protein. In one embodiment, the chemical bond is acid sensitive and the pH sensitive bond is cleaved upon going from the blood stream (pH 7.5) to the transcytotic vesicle or the interior of the cell (pH about 6.0).
Alternatively, the bond may not be acid sensitive, but may be cleavable by a specific enzyme or chemical which is subsequently added or naturally found in the microenvironment of the targeted site. Alternatively, the bond may be a bond that is cleaved under reducing conditions, for example a disulfide bond.
[0071] Alternati vely, the bond may not be cleavab!e. Any kind of acid cleavable or acid sensitive linker may be used. Examples of acid cleavable bonds include, but are not limited to: a class of organic acids known as cipolycarbox lic alkenes. This class of molecule contains at least three carboxylic acid groups (COOH) attached to a carbon chain that contains at least one double bond. These molecules as well as how they are made and used is disclosed in Shen, et al. U.S. Pat. No. 4,631 ,190.
[0072] Alternatively, molecules such asamino-sulfhydryl cross-linking reagents which are cleavable under mildly acidic conditions may be used. These molecules are disclosed in U.S. Pat. No. 4,569,789.
[0073] Alternatively, the acid cleavable linker may be a time-release bond, such as a biodegradable, hydrolyzable bond. Typical biodegradable carrier bonds include esters, amides or urethane bonds, so thai typical carriers are polyesters, polyamides, polyurethanes and other condensation polymers having a molecular weight between about 5,000 and 1,000,000.
Examples of these carriers bonds are shown in U.S. Pat. No. 4,356, 166, Other acid cleavable linkers may be found in U.S. Pat. Nos. 4,569,789 and 4,631,190 or Blattner et al.
(Biochemistry 24: 1517-1524 (1984)). The linkers are cleaved by natural acidic conditions, or alternatively, acid conditions can be induced at a target site as explained in U.S. Pat. No. 4,171,563.
[0074] Examples of linking reagents which contain cleavable disulfide bonds (reducable bonds) include, but are not limited to "DPDPB", l,4-di-[3'-(2'-pyridyldithio) propionamido] butane; "SADP", (N-succinimidyl (4-azidophenyl) l,3'-dithiopropionate); "Sulfo-SADP" (Sulfosuccinimidyl (4-azidophenyldithio) propionate; "DSP"-Dithio bis
(succinimidylproprionate); "DTSSP"-3,3'-Dithio bis(sulfosuccinimidylpropionate); "DTBP"- dimethyl 3,3dithiobispropionimidate-2HCI.
[0075] Examples of linking reagents cleavable by oxidation are "DST"-disucciiiimidyi tartarate; and "Sulfo-DST"-disuccinimidyS tartarate. [0076] Examples of non-cleavable linkers are "Si fQ-LC-SMPT"-(sulfosuccinimidyl 6- [alphamethyl-alpha-(2-pyridylthio)toluamido}hexanoate; "SMPT"; "ABH"-Azidobenzoyl hydrazide; ''NHS-ASA'^N-Hydroxysuccinimidyl-'-l-azidosalicyclic acid; "SASD"- Sulfos'uccinimidyl 2-(pazidosalicylarmdo) ethyl- 1 ,3-dithiopropionate; " APDP"-N- |4-(p~ azidosalicylamido) buthy}-3' (2'-pyidyldithio) propionamide; "BASED"-Bis-[P-(4- azidosalicylamido) ethyl] disulfide; "HSAB"-N-hydroxys ccmimidyl-4 azidobenzoate;
"APG"-p-Azidophenyl glyoxal monohydrate; "SANPAH"-N-Succiminidyl-6 (4'-azido-2 - mitrophenyl-amimo) hexanoate; "Sulfo-SANPAH"-Sulfosuccimmidyl6-(4'-azido-2'- mtrophenylamino) hexanoate; "A B-NOS" N-5-A.zido-2-nitrobenzyoyioxysuccinimide;
"SA D"-Sulfosuccinimidyl-2-(m-azido-o-^ opionate; ,TNP-DTP"-p-nitrophenyl-2-diazo-3,3,3-trifluoropropionate; "SMCC"-Suecinimidyl-4-(N- maleimidomethyl)cyclohexane- 1 -carboxylate; "Sulfo-SMCC"-Sulfosucciiiimidyl-4-(N- maleimidomethyl)cyclohexane-l-carboxy- late; "MBS" m- aleimidobenzoyl-N- hydroxysuccinimide ester; "smfo-MBS"-m-Maleimidobenzoyl-N-hydroxysulfosuccinimide ester; "SIAB"-N-Succinimidyl (4-iodoaceiyl)armnobenzoaie; "SuliSIAB"-N-Sulfosueeinirnidyi (4-iodoacetyl)aminobenzoate; "SMPB"-Succinimidy[ 4-(pmalenimidophenyl) butyrate; "Sulfo- SMPB"-Sulfosuccinimidyl 4-(p-malenimidopheiiyl) butyrate; "DSS"-Disuccinimidyl suberate; "BSSS"-bis(sulfosuccinimidyl) suberate; "BMH"-Bis ma!eimidohexane; "DFDNB"~1,5~ difluoro-2,4-dinitrobenzene; "DMA" -dimethyl adipimidate 2HCI; "DMP"-Dimethyl pimelimidate-2HCI; "DMS"-dimethyl suberimidate-2-HCI; "SPDPN-succinimidyl-3-(2- pyridylthio) propionate; "Sulfo-HSAB"-SulfosuccinimidyI 4-(pazidophenyl) butyrate; "Suifo- SAPB"~Sulfosuecinirmdyl 4-(|^~azidophenylbutyrate); " ASIB "- 1 -9p-azidosalicylamido)-4- (iodoacetamido) butane; "ASBA"-4-(p-Azidosalicylamido) butylamine,
[0077] In another embodiment the linker is a small molecule such as a peptide linker. In one embodiment the peptide linker is not cleavable. In a further embodiment die peptide linker is cleavable by base, under reducing conditions, or by a specific enzyme. In one embodiment, the enzyme is indigenous. Alternatively, the small peptide may be cleavable by an non-indigenous enzyme which is administered after or in addition to the therapeutic complex. Alternatively, the small peptide may be cleaved under reducing conditions, for example, when the peptide contains a disulfide bond. Alternatively, the small peptide may be pH sensitive. [0078] The peptide linker may also be useful as a peptide tag (e.g. , myc or His6 (SEQ ID NO: 9)) or may be one or more repeats of the known linker sequence GGGG S ( SEQ ID NO: 10), The skilled artisan will recognize that the linker sequence may be varied depending on the polypeptide portions to be linked to form the conjugate. Additional peptide linkers and tags are known in the art, such as epitope tags, affinity tags, solubility enhancing tags, and the like. Examples of various additional tags and linkers that may be used with the present invention include, haemagglutinin (HA) epitope, myc epitope, chitin binding protein (CBP), maltose binding protein (MBP), giutathione-S-transferase (GST), calmodulin binding peptide, biotin carboxyl carrier protein (BCCP), FLAG octapeptide, nus, green fluorescent protein (GFP), thioredoxin (TRX), poly(NANP), V5, S-protein, streptavidin, SBP, poly(Arg), DsbA, c-myc- tag, HAT, cellulose binding domain, softag 1, softag. , small ubi qui tin-like modifier (SUMO), and ubiquitiii (Lib). Further examples include: poly(L-Giy), (Poly L-Glycine linkers): poly(L- Glu), (PolyL-Glutamine linkers); poly (L-Lys), (Poly L-Lysine linkers), In one embodiment, the peptide linker has the formula (amino acid) n, where n is an integer between 2 and 100, preferably wherein the peptide comprises a polymer of one or more amino acids.
[0079] In a further embodiment the linker is a cleavable linker including, poly (ethylene glycol) (PEG) and a dipeptide,L,-alany!~L~vaime (A!a-Va!), cleavable by the enzyme diermoiysin. This linker is advantageous because thermolysin-like enzyme has been reported to be expressed at the site of many tumors. Alternatively, an amino acid residue spacer may be used which contains the recognition site for die protease furin.
[0080] The chemical and peptide linkers can be bonded between the scFv and the active molecule by techniques known in the art for conjugate synthesis, i.e. using genetic engineering, or chemically. The conjugate synthesis can be accomplished chemically via the appropriate antibody by classical coupling reactions of proteins to other moieties at appropriate functional groups.
[0081 ] Examples of the functional groups present in proteins and utilized normally for chemical coupling reactions are outlined as follows. The carbohydrate structures may be oxidized to aldehyde groups that in turn are reacted with a compound containing the groupH2NNH-R (wherein R is the compound) to the formation of a C=NH-NH-R group. The thiol group (cysteines in proteins) may be reacted with a compound containing a thiol-reactive group to the formation of a thioether group or disulfide group. The free amino group (at the amino terminus of a protein or on a lysine) in amino acid residues may be reacted with a compound containing an electropliilic group, such as an activated carboxy group, to the formation of an amide group. Free carboxy groups in amino acid residues may be transformed to a reactive carboxy group and then reacted with a compound containing an amino group to the formation of an amide group.
[0082] The linker or biologically active molecule may alternatively be a liposome or nanoparticle containing a biologically active molecule. Many methods for the preparation of liposomes are well known in the art. For example, the reverse phase evaporation method, freezethaw methods, extrusion methods, and dehydration-rehydration methods (see, Storm et al, PSTT 1 : 19-31 (1998)). For use in cancer immunotherapy, PEGylated or "stealth''' liposomes are especially useful. For example, such liposomes al lo for deliver}' of highly charged therapeutic molecules such as siRNA into ceils.
[0083] The liposomes may be produced in a solution containing the active molecule, such as doxorubicin, so that the substance is encapsulated during polymerization. Alternatively, the liposomes can be polymerized first, and the biologically active substance can be added later by resuspending the polymerized liposomes in a solution of a biologically active substance and treating with sonication to affect encapsulation of the active molecule. The liposomes can be polymerized in the presence of the substrate such that the substrate becomes a part of the phospholipid bilayer. In one embodiment, the liposome contains the active molecule on the inside and the substrate on the outside.
[0084] The liposomes contemplated in the present invention can comprise a variety of structures, For example, the liposomes can be multilamellar large vesicles (MLV), oligolamellar vesicles (OLV), unilamellar vesicles (UV), small unilamellar vesicles (SUV), medium sized unilamellar vesicles(MUV), large unilamellar vesicles (LUV), giant unilamellar vesicles (GUV), or multivesicul ar vesicl es (MVV). Each of these liposome structures are well known in the art (see, Storm et al, PSTT 1 : 19-31 (1998)).
[0085] In one embodiment, the liposome is a "micromachine" that evulses pharmaceuticals for example by the application of specific frequency radio waves. In another embodiment, the liposomes can be degraded such that they will release the active molecule in the targeted cell, for example, the liposomes may be acid or alkaline, sensitive, or degraded in the presence of a low or high pH, such that the active molecule is released within the cell. Alternatively, the liposomes may be uncharged so that they will be taken up by the targeted cell. The liposomes may also be pH sensitive or sensitive to reducing conditions.
[0086] One type of liposome which may be advantageously used in the present invention is that identified in U.S. Pat. No. 6,004,534. In this application a method of producing modified liposomes which are prepared by polymerization of double and triple bond-containing monomelic phospholipids is disclosed. These liposomes have surprisingly enhanced stability against the harsh en vironment of the gastrointestinal tract. Thus, they have utility for oral and/or mucosal deliver of the active molecule. It has also been shown that the liposomes may be absorbed into the systemic circulation and lymphatic circulation. The liposomes are generally prepared by polymerization (i.e., radical initiation or radiation) of double and triple bond-containing monomeric phospholipids.
[0087] Advantageously, in other embodiments of the present invention, the linker can also be a liposome having a long blood circulation time. Such liposomes are well known in the art (see U.S. Pat. Nos., 5,013,556; 5,225,212; 5,213,804; 5,356,633; and 5,843,473). Liposomes having long blood circulation time are characterized by having a portion of their
phosphoslipids derivatized with polyethylene glycol (PEG) or other similar polymer, in some embodiments, the end of the PEG molecule distal to the phospholipid may be activated so a to be chemically reactive. Such a reactive PEG molecule can be used to link a substrate to the liposome. One example of a reactive PEG molecule is the maleimide derivative of PEG described in U.S. Pat. No. 5,527,528).
[0088] Alternatively, the linker may be a microcapsule, a nanoparticle, a magnetic particle, and the like (see Kumar, J. Pharm. Sci, 2:234-258 (2000); and Gill et al., Trends Biotechnol. 18(1 l):469-79 (2000)), with the lipophilic active molecule on or in the container, and the container functioning as the linker in the therapeutic complex.
[0089] Alternatively, the linker may be a photocleavable linker. For example, a 1-2- (nitrophenyl)ethyl moiety can be cleaved using 300 to 360 run light. It can be envisioned that the photocleavable linker would allow activation and action of the drug in an even more specific area, for example a particular part of the organ. The light could be localized using a catheter into the vessel. Alternatively, light may be used to localize treatment to a specific part of the digestive tract and the light may be manipulated through a natural orifice to the area. Alternatively, the light can be surgically manipulated to the area. [0090] Alternatively, the linker may not be cleavabie, but the active molecule or substrate is. An example of this is when the active molecule is a prodrug and the enzyme which cleaves the prodrug is administered with the therapeutic complex. Alternatively, the enzyme is part of the therapeutic complex or indigenous and the prodrug is administered separately, Preferably, the enzyme or prodrug which is administered separately is administered within about 48 hours of the first administration. Alternatively, the prodrug or enzyme which is administered separately may be administered between about 1 min and 24 hours, alternatively between about 2 min and 8 hours. The prodrug or enzyme which is administered separately, may be readministered at a later date and may continue to be administered until the effect of the drag is not longer needed.
[0091] In certain embodiments, the subject conjugates can be delivered via an expression construct to cells, including a nucleic acid that provides a coding sequence for a therapeutic protein. For instance, the expression construct can encode a therapeutic protein that is secreted by the transduced cell.
[0092] As used herein, the term "nucleic acid" refers to polynucleotides such as
deoxyribonucleic acid (DNA), and, where appropriate, ribonucleic acid (RNA). The term should also be understood to include, as appropriate to the context or as applicable to the embodiment being described, both single-stranded polynucleotides (such as antisense) and double-stranded polynucleotides (such as siRNAs).
[0093] A "protein coding sequence" or a sequence that "encodes" a particular polypeptide or peptide, is a nucleic acid sequence that is transcribed (in the case of DN A) and is translated (in the case of mRNA) into a polypeptide in vitro or in vivo when placed under the control of appropriate regulatory sequences. The boundaries of the coding sequence are determined by a start codon at the 5' (amino) terminus and a translation stop codon at the 3' (carboxyl) terminus. A coding sequence can include, but is not limited to, cDNA from prokaryotic or eukaryotic mRNA, genomic DNA sequences from prokaryotic or eukaryotic DNA, and even synthetic DNA sequences. A transcription termination sequence will usually be located 3' to the coding sequence.
[0094] As used herein, the term "vector" refers to a nucleic acid molecule capable of transporting another nucl eic acid to which it has been l inked. One type of vector is a genomic integrated vector, or "integrated vector," which can become integrated into the chromosomal DNA of the host cell. Another type of vector is an episonial vector, e.g., a nucleic acid capable of extra-chromosomal replication. Vectors capable of directing the expression of genes to which they are operative!}' linked are referred to herein as "expression vectors." In the present specification, "plasmid" and "vector" are used interchangeably unless otherwise clear from the context. In the expression vectors, regulatory elements controlling transcription can be generally derived from mammalian, microbial, viral or insect genes. The ability to replicate in a host, usually conferred by an origin of replication, and a selection gene to facilitate recognition of transfomiants may additionally be incorporated. Vectors derived from viruses, such as retroviruses, adenoviruses, and the like, may be employed.
[0095] in one embodiment, the present invention provides a method of producing a protein. The method includes transforming a host cell with an expression construct, and culturing the host ceil under conditions suitable for producing the conjugate. In various embodiments, the expression construct includes a nucleic acid molecule encoding a protein conjugate including an scFv and a peptide, wherein the scFv includes an amino acid sequence at least 90%, at least 95% or 100% identical to SEQ ID NO: 1 or SEQ ID NO: 2.
Vectors suitable for use in preparation of proteins and/or protein conjugates include those selected from bacuiovirus, phage, plasmid, phagemid, cosmid, fosmid, bacterial artificial chromosome, viral DNA, Pi-based artificial chromosome, yeast plasmid, and yeast artificial chromosome. For example, the viral DNA vector can be selected from vaccinia, adenovirus, foul pox virus, pseudorabies and a derivative of SV40. Suitable bacterial vectors for use in practice of the invention methods include pQE70L ', pQE601M, pQE-91M, pBLUESCRIPT1M SK, pBLUESCRIPT™ KS, pTRC99a™, p 223-3™, pDR540™, PAC™ and pRJT2T™. Suitable eukaryotic vectors for use in practice of the invention methods include p W LNEO ,M, pXTI™, pSG5™, pSV 3™, pBPV™, pMSG™, and p8VLSV40™, Suitable eukaryotic vectors for use in practice of the invention methods include p WL EOj M, pXTI jM, pSG5 tM, pSVK3™, pBPV™, pMSG™, and pSVLSV40™.
[0097] Those of skill in the art can select a suitable regulatory region to be included in such a vector, for example from lad, lacZ, T3, T7, apt, lambda PR, PL, trp, CM V immediate early, HSV thymidine kinase, early and late SV40, retroviral LTR, and mouse metallothionein-I regulatory regions. 2.4
[0098] Host cells in which the vectors containing the polynucleotides encoding the protein conjugates can be expressed include, for example, a bacterial cell, a eukaryotic cell, a yeast ceil, an insect cell, or a plant cell. For example, E, coli, Bacillus, Streptomyces, Pichia pastoris. Salmonella typhimurium, Drosophila S2, Spodoptera SJ9, CHO, COS (e.g. COS-7), or Bowes melanoma cells are all suitable host cells for use in practice of the invention methods.
[0099] Conjugates in which the biologically active molecule is an antibody or sacFv molecule may be generated using methods known in the art, For example, methods for attaching a drug or other small molecule pharmaceutical to protein include bifunctionai chemical linkers such as N-succinimidyl (4-iodoacetyl)-aminobenzoate; sulfosuccinimidyl(4- iodoacetyl)-aminobenzoate; 4-succinimidyl-oxycarbonyl- -(2-pyridyldithio) toluene;
sulfosuccinimidyi-6-[ -methyka-(pyridyldithioi)-tokmmido]he- xanoate; N-succinimidyl-3-(2- pyridyldithio)-proprionate; succinimidyl-6-[3(-(-2-pyridyidithio)-proprionamido]hexanoate; sulfosuccinimidy[-6-[3(-(-2-pyridyldithio)-propionaniido]hexanoate; 3-(2-pyridyldithio)- propionyl hydrazide, EUman's reagent, dichlorotriazinic acid, S~(2-tbiopyridyl)-L-cysteme, and the like. Further bifunctionai linking molecules are disclosed in U.S. Pat. Nos, 5,349,066;
5,618,528; 4,569,789; 4,952,394; and 5,137,877, each of which is incorporated herein by reference in its entirety.
[0100] In certain embodiments, a conjugate of the present invention may further contain a detectable label. Such labels are known in the art. and include radio-isotopes and fluorescent labels. As such, internalization of a compound e videncing passage through transporters can be detected by detecting a signal from within a cell from any of a variety of reporters. The reporter can be a label such as a fluorophore, a ehromophore, a radioisotope. Confocal imagining can also be used to detect internalization of a label as it provides sufficient spatial resolution to distinguish between fluorescence on a cell surface and fluorescence within a cell; alternatively, confocal imaging can be used to track the movement of compounds over time. In another approach, internalization of a compound is detected using a reporter that is a substrate for an enzyme expressed within a cell. Once the complex is internalized, the substrate is metabolized by the enzyme and generates an optical signal or radioactive decay that is indicative of uptake. Light emission can be monitored by commercial PMT-based instruments or by CCD-based imaging systems. In addition, assay methods utilizing LCMS detection of the transported compounds or electrophysiological signals indicative of transport activity are also employed, [0101] In some embodiments, die conjugates of the present invention may include a biologically active molecule that is a nucleic acid molecule or analog thereof. As such the conjugates may be utilized to deliver nucleic acids, or analogs thereof to a targeted tissue or cell type, For example, protein expression can be specifically down-regulated using oligonucleotides such as, for example, antisense molecules, locked nucleic acids (LNA), peptide nucleic acids (PNA), morpholino nucleic acids and small interfering RNAs (siRNA) of various chemistries, Alternatively, expression constructs may be delivered to cells, to induce expression of a desired gene product.
[0102] The expression construct that is delivered typically is part of a vector in which a regulator}' element such as a promoter is operabiy linked to the nucleic acid of interest. The promoter can be constitutive or inducible. Non-limiting examples of constitutive promoters include cytomegalovirus (CM V) promoter and the Rous sarcoma vims promoter. As used herein, "inducible" refers to both up-regulation and down regulation. An inducible promoter is a promoter that is capable of directly or indirectly activating transcription of one or more DNA sequences or genes in response to an inducer. In the absence of an inducer, the DNA sequences or genes will not be transcribed. The inducer can be a chemical agent such as a protein, metabolite, growth regulator, phenolic compound, or a physiological stress imposed directly by, for example heat, or indirectly through the action of a pathogen or disease agent such as a virus. The inducer also can be an illumination agent such as light and light's various aspects, which include wavelength, intensity, fluorescence, direction, and duration.
[0103] An example of an inducible promoter is the tetracycline (tet)-on promoter system, which can be used to regulate transcription of the nucleic acid, In this system, a mutated Tet repressor (TetR) is fused to the activation domain of herpes simplex VP 16 (transactivator protein) to create a tetracycline-controlled transcriptional activator (tTA), which is regulated by tet or doxycycline (dox). In the absence of antibiotic, transcription is minimal, while in the presence of tet or dox, transcription is induced, Alterna tive inducible systems include the ecdysone or rapamycin systems. Ecdysone is an insect molting hormone whose production is controlled by a lieterodimer of the ecdysone receptor and the product of the ultraspiracle gene (USP). Expression is induced by treatment with ecdysone or an analog of ecdysone such as muristerone A. [0104] Additional regulator}' elements that may be useful in vectors, include, but are not limited to, polyadenylation sequences, translation control sequences (e.g., an internal ribosome entry segment, IRES), enhancers, or iiitrons, Such elements may not be necessary, although they may increase expression by affecting transcription, stability of the mRNA, translational efficiency, or the like. Such elements can be included in a nucleic acid construct as desired to obtain optimal expression of the nucleic acids in the celi(s). Sufficient expression, however, may sometimes be obtained without such additional elements.
[0105] Vectors also can include other elements. For example, a vector can include a nucleic acid that encodes a signal peptide such that the encoded polypeptide is directed to a particular cellular location (e.g., a signal secretion sequence to cause the protein to be secreted by the cell) or a nucleic acid that encodes a selectable marker. Non-limiting examples of selectable markers include puroraycin, adenosine deaminase (A DA), aminoglycoside phosphotransferase (neo, G418, APH), dihydrofolate reductase (DHFR), hygroniycin-B-phosphtransferase, thymidine kinase (T ), and xanthin-guanine phosphoribosyl transferase (XGPRT). Such markers are useful for selecting stable transformants in culture.
[0106] Viral vectors can be used to form the conjugates, and include adenovirus, adeno- associated virus (AAV), retroviruses, lentiviruses, vaccinia virus, measles viruses, herpes viruses, and bovine papilloma virus vectors (see, Kay et al., Proc. Natl. Acad. Sci. USA 94: 12744-12746 (1997) for a re view of viral and non-viral vectors). Viral vectors are modified so the nativ e tropism and pathogenicity of t he v irus has been altered or removed. The genome of a virus also can be modified to increase its infectivity and to accommodate packaging of the nucleic acid encoding the polypeptide of interest.
[0107] Non-viral vectors can also be used in the subject conjugates. To further illustrate, in one embodiment, the mammalian serum protein that is encoded by the vector is selected from the group consisting of a tissue-type plasminogen activator, a receptor of a tissue-type plasminogen activator, a streptokinase, a staphylokinase, a urokinase, and coagulation factors. The invention also provides a method for treating associated with the formation of clots in its circulation, including the step of administering to the mammal a conjugate that causes the recombinant expression and secretion into the blood, such as from transduced muscle cells, of a therapeutically effective amount of such a mammalian serum protein. 97
[0108] An aptamer is a DNA or RNA molecule that has been selected from a random or biased pool of oligonucleic acids, based on its ability to bind to a target molecule. Aptamers can be selected which bind nucleic acids, proteins, small organic compounds and specific cell surfaces, and several have been developed which bind to proteins which are associated with disease states. Aptamers are in general more easily manufactured and are more amenable to chemical modification than are antibodies, and they can be "evolved" for tighter binding to the target by an iterative process of random modification and affinity-based selection. The evolved aptamers often have antibody-like specificities, and are therefore expected to have utility in those applications, such as therapeutics and in vitro and in vivo diagnostics.
[0109] According to another embodiment of the invention, there are pro vided methods for treating a disease or disorder in a cell or tissue expressing RORl, especially CLL. The method includes administering to a patient a scFv conjugate, coding sequence, vector or aptamer of the present invention,
[0110] In a related embodiment of the invention, there are provided methods for targeting a cell or tissue expressing RORl, especially CLL cells. The method includes contacting the cell or tissue with an scFv conjugate , coding sequence, vector or aptamer of the present invention.
[0111 ] The scFv antibody conjugates, coding sequences, vectors or aptamers of the present invention may be employed as therapeutic or prophylactic pharmacological agents in any subject in which it is desirable to administer, in vitro, ex vivo, or in vivo the subject antagonists that bind RORl; e.g., to treat CLL. Typical subjects for treatment or management according to the methods herein are subjects presenting with a ROR l cancer; e.g., CLL,. The antagonists described herein specifically recognize RORl protein, found in lymphoma samples but not expressed in cells of normal adults, and therefore can be used for detecting and/or neutralizing these biomolecules, and/or blocking their interactions with other biomolecules, in vitro or in vivo.
[0112] In certain embodiments, the methods herein may be used to target therapeutic antibodies, or nucleic acids encoding them, to particular target ceils. The antibodies may be, for example, monoclonal antibodies, polyclonal antibodies, single-chain antibodies, or bi-specific antibodies. Therapeutic antibodies are known to include, but are not limited to, Anti EGFr antibodies (e.g., panitumamab, Erbitux (cetuximab), matuzumab, IMC-IIF 8, TheraCIM liR3), denosumab, Avastin (bevacizumab), Anti-HGF antibodies, Elumira (adalimumab), Anti-Ang-2 antibodies, Herceptin (trastuzuniab), Remicade (infliximab), Anti-CD20 antibodies, rituximab, Synagis (palivizumab), Mylotarg (gemtuzumab oxogamicin), Raptiva (efalizumab), Tysabri (nataiizumab), Zenapax (dacliximab), NeutroSpec (Technetium (,sup.99mTc) fanolesomab), tocilizumab, ProstaScint (Indium-Ill labeled Capromab Pendetide), Bexxar (tositumomab), Zevaiin (ibrituniomab tiuxetan (IDEC-Y2B8) conjugated to yttrium 90), Xolair (omalizumab), MabThera (Rituximab), ReoPro (abciximab), MabCampath (alemtuzumab), Simulect
(basiliximab), LeukoScan (sulesomab), CEA-Scan (arcitumomab), Verluma (nofetumomab), Panorex (Edrecolomab), alemtuzumab, CDP 870, and nataiizumab.
[0113] In certain therapeutic embodiments, the selected scFv antibody conjugate, coding sequence, vector or aptamer may be administered alone, or in combination with, or conjugated to, one or more combinatorial therapeutic agents. When the scFV molecules described herein are administered alone as therapeutic agents, they may exert a beneficial effect in the subject by a variety of mechanisms. For example, in certain embodiments, scFv antibody conjugates that specifically bind ROR1 are purified and administered to a patient to neutralize one or more forms of ROR1 , to block one or more activities of RORl, or to block or inhibit an interaction of one or more forms of RORl with another biomolec le; e.g., to treat CLL or other ROR l cancers.
[0114] As discussed herein, the scFV molecules of the invention may include humanized antibodies, and can be combined for therapeutic use with additional active or inert, ingredients, e.g., in conventional pharmaceutically acceptable carriers or diluents, e.g., immunogenic adjuvants, and optionally with adjunctive or combinatorial ly active molecules such as antiinflammatory and anti-fibrinolytic drugs.
[0115] In other embodiments, the scFv conjugates, coding sequences, vectors or aptamers described herein are coordinately administered with, co-formulated with, or coupled to (e.g., covalently bonded) a combinatorial therapeutic agent, for example a radionuclide, a differentiation inducer, a drug, or a toxin. Various known radionuclides can be employed, that are well known in the art. Useful drugs for use in such combinatorial treatment formulations and methods include methotrexate, and pyrimidine and purine analogs. Suitable differentiation inducers include phorbol esters and butyric acid. Suitable toxins include ricin, abrin, diptheria toxin, cholera toxin, gelonin, Pseudotnonas exotoxin, Shigella toxin, and pokeweed antiviral protein. These combinatorial therapeutic agents can be coupled to an anti-RORl antibody either directly or indirectly (e.g., via a linker group). A direct reaction between an agent and an antibody is possible when each possesses a substituent capable of reacting with the other. For example, a nucleophilic group, such as an amino or sulfhydryi group, on one may be capable of reacting with a carbonyl-containing group, such as an anhydride or an acid halide, or with an alky] group containing a good leaving group (e.g., a halide) on the other. Alternatively, it may be desirable to couple a combinatorial therapeutic agent and an antibody via a linker group as a spacer to distance an antibody from the combinatorial therapeutic agent in order to avoid interference with binding capabilities, A linker group can also serve to increase the chemical reactivity of a substituent on an agent or an antibody, and thus increase the coupling efficiency. It will be further evident to those skilled in the art that a variety of bifunctional or
polyfunctional reagents, both homo- and hetero-functional (such as those described in the catalog of the Pierce Chemical Co., Rockford, 111.), may be employed as a linker group.
Coupling may be affected, for example, through amino groups, carboxyl groups, sulfhydryi groups or oxidized carbohydrate residues.
[0116] In carrying out various assay, diagnostic, and therapeutic methods of the invention, it is desirable to prepare in advance kits comprises a combination of scFv conjugate or peptide as described herein with other materials. For example, in the case of sandwich enzyme immunoassays, kits of the invention may contain an scFv antibody that specifically binds ROR1 optionally linked to an appropriate carrier, a freeze-dried preparation or a solution of an enzyme- labeled monoclonal antibody which can bind to the same antigen together with the monoclonal antibody or of a polyclonal antibody labeled with the enzyme in the same manner, a standard solution of purified ROR.1, a buffer solution, a washing solution, pipettes, a reaction container and the like. In addition, the kits optionally include labeling and/or instructional materials providing directions (i.e., protocols) for the practice of the methods described herein in an assay environment. While the instructional materials typically comprise written or printed materials, they are not limited to such. Any medium capable of storing such instructions and communicating them to an end user is contemplated. Such media include, but are not limited to electronic storage media (e.g., magnetic discs, tapes, cartridges, chips), optical media (e.g., CD ROM), and the like. Such media may include addresses to internet sites that provide such instructional materials,
[0117] Having described the invention in detail, it will be apparent that modifications, variations, and equivalent embodiments are possible without departing the scope of the invention defined in the appended claims, Furthermore, it should be appreciated that all examples in the present disclosure are provided as non-limiting examples.
EXAMPLE 1
GENERATION OF AN SCFv CONSTRUCT
[0118] This example illustrates generation of an scFv antibody conjugate of the present invention.
[0119] The scFV having SEQ ID NO: 3 was inserted upstream of the sequence encoding an IgG4 CH domain, the transmembrane domain of CD28 and cytoplasmic domain of the zeta chain for the T-cell receptor (CD247) to generate a chimeric antigen receptor (CAR).
[0120] The construct was inserted into a lenti-virus vector for transduction of T cells (Jurkat). The T cells were examined for their capacity to bind a soluble ROR1 protein (ROR1- Ig). Cell-bound RGRl-ig could be detected by staining the cells with an anti-ig antibody that was labeled with a fluorescence dye, phycoerythrin (PE), This allowed monitoring of changes in fluorescence of the T ceils caused by binding the RORl protein using flow cytometry.
[0121 The results are shown in Figure
j l
Figure imgf000032_0001
[0122] Dual parameter flow cytometry was done in a FACSCalibur instrument (BD
Biosciences; Immunocytometry Systems) using an anti-human IgG that reacted with cell bound scFV of the invention (as shown in SEQ.ID.No: 3), but not the cells used in these experiments. CHO cells (which do not express ROR1) and EW36, a human B-cell lymphoma cell line (National Cancer Institute) were stained with culture supernatants of cells transfected to express pCDNA 3.1 (Invitrogen) CH2 CH3 stable scFV (Figure 6).
[0123] In particular, approximately 5 x 105 cells were incubated for 20 minutes with the scFV, the cells were then washed and then incubated with an Alexa-488-conjugated goat anti- human-IgG antibody that does not react with EW36 cel ls in the absence of the cel l-bound scFV. Propidium iodide was added to stain dead cells, which could then be excluded from the analysis. Following incubation with the culture supernatants, the cells were washed prior to analysis using a FACSCalibur1M (Becton Dickinson, Mountain View, CA). The data analyzed with FlowJolM software (Tree Star, San Carlos, CA). A total of 20,000-gated events were collected for each sample in a list mode file, and data analysis was done using CellQuest j;Vl software (BD Biosciences). The relative log florescence of the cells is depicted on the x-axis. Cells staining with the Alexa-488 conjugated goat antibody are shifted to the right, A histogram of control unstained cells or cells stained with the Alexa-488 conjugated goat antibody in the absence of the scFV are provided for comparison.
[0124] The binding activity for ROR1 expressed on the cell surface membrane was assessed via flow cytometry. The scFV failed to react with CHO cells that do not express ROR1 (Figure 6A). In contrast, the scFV produced by clones 1, 4-5, 8-14 and 16 reacted with EW36 cells, allowing for detection of the cell-bound scFV using the Alexa-488-conjugated goat anti- human antibody (Figures 6B, E-F, 1-0 and Q-R). These data indicate that the single chain scFV of the invention specifically reacts with ROR1 expressed on the surface of viable cells.
[0125] Although the invention has been described with reference to the above example, it will be understood that modifications and variations are encompassed within the spirit and scope of the invention. Accordingly, the invention is limited only by the following claims.

Claims

What is claimed is:
1. An antibody single-chain variable fragment (scFv) conjugate comprising,
a scFv having an amino acid sequence at least 90% identical to SEQ ID NO: 1 or SEQ ID NO: 2; and
a biologically active molecule,
2. The conjugate of claim 1, wherein the amino acid sequence of the scFv is at least 95% identical to SEQ ID NO: 1 or SEQ ID NO: 2.
3. The conjugate of claim 1 , wherein the amino acid sequence of the scFv is 100% identical to SEQ ID NO: 1 or SEQ ID NO: 2.
4. The conjugate of claim 1, wherein the scFv has a binding specificity of an antibody as produced by a hybridoma having Accession No. PTA 8634 by the American Type Culture Collection.
5. The conjugate of claim 1, wherein the biologically active molecule is selected from the group consisting of a polypeptide, a peptidomimetic, a nucleic acid molecule, a lipid, a drug compound, chemotherapeutic agent, an antagonist and an agonist.
6. The conjugate of claim 5, wherein the biologically active molecule is a polypeptide.
7. The conjugate of claim 6, wherein the polypeptide is an antibody or an antibody fragment.
8. The conjugate of claim 7, wherein the antibody fragment is selected from the group consisting of a Fab fragment, a F(ab)2 fragment, an FV fragment, a single chain FV (scFV) fragment, a dsFV fragment, a CH fragment and a dimeric scFV.
9. The conjugate of claim 8, wherein the CH fragment is an IgG CH fragment.
10. The conjugate of claim 9, wherein the IgG CH fragment is of immunoglobulin class IgGl, IgG2, IgG3, IgG4 or IgGM.
1 1. The conjugate of claim 7, wherein the antibody is selected from the group consisting of a chimeric antibody, a humanized antibody, a CDR-grafted antibody, a bifunctional antibody, a single chain antibody, and an antibody polypeptide dimer.
12. The conjugate of claim 5, wherein the polypeptide is an enzyme, transcription factor, receptor, hormone, steroid or functional fragments thereof.
13. The conjugate of claim 12, wherein the polypeptide comprises CD! 54.
14. The conjugate of claim 5, wherein the nucleic acid molecule is a DN A molecule or an RNA molecule,
15. The conjugate of claim 1, wherein the scFv is conjugated to the biologically active molecule by a linker molecule.
16. The conjugate of claim 1, wherein the conjugate is a genetic fusion.
17. The conjugate of claim 1, further comprising a detectable label.
18. An isolated peptide comprising an amino acid sequence at least 90% identical to SEQ ID NO: 1 or SEQ ID NO: 2.
19. The peptide of claim 18, wherein the amino acid sequence is at least 95% identical to SEQ ID NO: 1 or SEQ ID NO: 2.
20. The peptide of claim 18, wherein the amino acid sequence is 100%» identical to SEQ
ID NO: 1 or SEQ ID NO: 2.
21. The peptide of claim 18, wherein the peptide has a binding specificity of an antibody as produced by a hvbridoma having Accession No. PTA 8634 by the American Type Culture Collection.
22, An isolated nucleic acid encoding the peptide of claim 18.
23. A pharmaceutical composition comprising the conjugate of claim 1 and a pharmaceutically acceptable carrier.
24. A method of producing a protein conjugate comprising,
i) transforming a host cell with an expression construct comprising,
a nucleic acid molecule encoding a protein conjugate comprising an scFv and a polypeptide, wherein the scFv peptide comprises an amino acid sequence at least 90% identical to SEQ ID NO: 1 or SEQ ID NO: 2; and
ii) culturing the host cell under conditions suitable for producing the conjugate, thereby producing the protein conj ugate.
25. A method for detectmg ROR l protein in a sample, the method comprising:
(a) contacting the sample with a detectably labeled peptide of claim 17; and
(b) detecting immunoreactiviiy between the detectably labeled peptide and RORl in the sample.
26. The method of claim 25, further comprising (c) determining whether an increase or decrease in an amount of RORl protein has occurred in the subject in comparison to a control level of RORl .
27. The method according to claim 25, wherein the sample is from a subject that is suspected or known to contain RORl cancer cells.
28. A method of targeting a biologically active molecule to a cell having an RORl receptor, comprising contacting the cell with the peptide conjugate of claim 1.
29. The method of claim 28, wherein the cell is in vivo or in vitro.
30. A kit to detect the presence of RORl protein in a sample from a subject that is known or suspected to contain cancer cells, comprising the antibody conjugate of claim 1 or the peptide of claim 18 and instructions for its use in an assay environment.
31. A method for treating a RORl cancer in a human subject, the method comprising administering to the subject in need thereof a therapeutically eftective amount of the antibody conjugate of claim 1 or the peptide of claim 18.
32. The method according to claim 31, wherein the RORl cancer is a lymphoma or adenocarcmoma.
33. The method according to claim 32, wherein the lymphoma is selected from the group consisting of CLL, small lymphocytic lymphoma, marginal cell B-Ceil lymphoma, and Burkett's Lymphoma, colon adenocarcinoma, and breast adenocarcinoma.
34. The method according to claim 33, wherein the lymphoma is CLL.
35. A method according to claim 31, wherein the antibody conjugate or peptide is administered in an amount of (i) about 0,05 mg to about 2.5 mg; (ii) about 0.1 mg to about 1 mg; or (iii) about 0.3 mg to about 0.5 mg.
36. A method for treating or preventing CLL in which RORl binding of Wnt5a on CLL cells confers a survival advantage thereon, comprising administering the antibody conjugate of claim 1 or the peptide of claim 18.
37. A method of monitoring a therapeutic regimen for treatmg a subject having or at risk of having an RORl cancer, comprising determining a change in activity or expression of RORl protein as a result of the treating of claim 36, thereby monitoring the therapeutic regimen in the subject.
PCT/US2011/040595 2010-06-15 2011-06-15 Receptor tyrosine kinase-like orphan receptor 1 (ror1) single chain fv antibody fragment conjugates and methods of use thereof WO2011159847A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US13/703,864 US9242014B2 (en) 2010-06-15 2011-06-15 Receptor tyrosine kinase-like orphan receptor 1 (ROR1) single chain Fv antibody fragment conjugates and methods of use thereof
US14/973,486 US9938350B2 (en) 2010-06-15 2015-12-17 Receptor tyrosine kinase-like orphan receptor 1 (ROR1) single chain Fv antibody fragment conjugates and methods of use thereof
US15/908,659 US20180362657A1 (en) 2010-06-15 2018-02-28 Receptor tyrosine kinase-like orphan receptor 1 (ror1) single chain fv antibody fragment conjugates and methods of use thereof
US17/093,474 US11548953B2 (en) 2010-06-15 2020-11-09 Receptor tyrosine kinase-like orphan receptor 1 (ROR1) single chain Fv antibody fragment conjugates and methods of use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US35509210P 2010-06-15 2010-06-15
US61/355,092 2010-06-15

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/703,864 A-371-Of-International US9242014B2 (en) 2010-06-15 2011-06-15 Receptor tyrosine kinase-like orphan receptor 1 (ROR1) single chain Fv antibody fragment conjugates and methods of use thereof
US14/973,486 Continuation US9938350B2 (en) 2010-06-15 2015-12-17 Receptor tyrosine kinase-like orphan receptor 1 (ROR1) single chain Fv antibody fragment conjugates and methods of use thereof

Publications (2)

Publication Number Publication Date
WO2011159847A2 true WO2011159847A2 (en) 2011-12-22
WO2011159847A3 WO2011159847A3 (en) 2012-05-18

Family

ID=45348854

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/040595 WO2011159847A2 (en) 2010-06-15 2011-06-15 Receptor tyrosine kinase-like orphan receptor 1 (ror1) single chain fv antibody fragment conjugates and methods of use thereof

Country Status (2)

Country Link
US (4) US9242014B2 (en)
WO (1) WO2011159847A2 (en)

Cited By (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012075158A1 (en) * 2010-12-01 2012-06-07 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric rabbit/human ror1 antibodies
EP2789630A1 (en) 2013-04-09 2014-10-15 EngMab AG Bispecific antibodies against CD3e and ROR1
WO2015142675A2 (en) 2014-03-15 2015-09-24 Novartis Ag Treatment of cancer using chimeric antigen receptor
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
WO2016055592A1 (en) 2014-10-09 2016-04-14 Engmab Ag Bispecific antibodies against cd3epsilon and ror1
WO2016057705A1 (en) 2014-10-08 2016-04-14 Novartis Ag Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
WO2016090034A2 (en) 2014-12-03 2016-06-09 Novartis Ag Methods for b cell preconditioning in car therapy
WO2016115559A1 (en) * 2015-01-16 2016-07-21 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for ror1
WO2016126608A1 (en) 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
WO2016172583A1 (en) 2015-04-23 2016-10-27 Novartis Ag Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
US9523695B2 (en) 2011-01-14 2016-12-20 The Regents Of The University Of California Therapeutic antibodies against ROR-1 protein and methods for use of same
WO2017027392A1 (en) 2015-08-07 2017-02-16 Novartis Ag Treatment of cancer using chimeric cd3 receptor proteins
WO2017091786A1 (en) 2015-11-23 2017-06-01 Novartis Ag Optimized lentiviral transfer vectors and uses thereof
WO2017114497A1 (en) 2015-12-30 2017-07-06 Novartis Ag Immune effector cell therapies with enhanced efficacy
WO2017127664A1 (en) 2016-01-20 2017-07-27 The Scripps Research Institute Ror1 antibody compositions and related methods
WO2017149515A1 (en) 2016-03-04 2017-09-08 Novartis Ag Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore
WO2017165683A1 (en) 2016-03-23 2017-09-28 Novartis Ag Cell secreted minibodies and uses thereof
WO2017181119A2 (en) 2016-04-15 2017-10-19 Novartis Ag Compositions and methods for selective protein expression
WO2018014001A1 (en) 2016-07-14 2018-01-18 Fred Hutchinson Cancer Research Center Multiple bi-specific binding domain constructs with different epitope binding to treat cancer
WO2018026819A2 (en) 2016-08-01 2018-02-08 Novartis Ag Treatment of cancer using a chimeric antigen receptor in combination with an inhibitor of a pro-m2 macrophage molecule
US9938350B2 (en) 2010-06-15 2018-04-10 The Regents Of The University Of California Receptor tyrosine kinase-like orphan receptor 1 (ROR1) single chain Fv antibody fragment conjugates and methods of use thereof
WO2018111340A1 (en) 2016-12-16 2018-06-21 Novartis Ag Methods for determining potency and proliferative function of chimeric antigen receptor (car)-t cells
WO2018140725A1 (en) 2017-01-26 2018-08-02 Novartis Ag Cd28 compositions and methods for chimeric antigen receptor therapy
WO2018144535A1 (en) 2017-01-31 2018-08-09 Novartis Ag Treatment of cancer using chimeric t cell receptor proteins having multiple specificities
WO2018160731A1 (en) 2017-02-28 2018-09-07 Novartis Ag Shp inhibitor compositions and uses for chimeric antigen receptor therapy
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
WO2018237335A1 (en) * 2017-06-23 2018-12-27 VelosBio Inc. Ror1 antibody immunoconjugates
WO2019084288A1 (en) 2017-10-25 2019-05-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2019081983A1 (en) 2017-10-25 2019-05-02 Novartis Ag Antibodies targeting cd32b and methods of use thereof
WO2019089798A1 (en) 2017-10-31 2019-05-09 Novartis Ag Anti-car compositions and methods
WO2019210153A1 (en) 2018-04-27 2019-10-31 Novartis Ag Car t cell therapies with enhanced efficacy
WO2019213282A1 (en) 2018-05-01 2019-11-07 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
WO2019237035A1 (en) 2018-06-08 2019-12-12 Intellia Therapeutics, Inc. Compositions and methods for immunooncology
WO2020012337A1 (en) 2018-07-10 2020-01-16 Novartis Ag 3-(5-amino-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and their use in the treatment of i karos family zinc finger 2 (ikzf2)-dependent diseases
WO2020128972A1 (en) 2018-12-20 2020-06-25 Novartis Ag Dosing regimen and pharmaceutical combination comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2020165833A1 (en) 2019-02-15 2020-08-20 Novartis Ag 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020165834A1 (en) 2019-02-15 2020-08-20 Novartis Ag Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
US10758556B2 (en) 2017-08-07 2020-09-01 Nbe-Therapeutics Ag Anthracycline-based antibody drug conjugates having high in vivo tolerability
US10968275B2 (en) 2016-02-02 2021-04-06 Fred Hutchinson Cancer Research Center Anti-ROR1 antibodies and uses thereof
WO2021123996A1 (en) 2019-12-20 2021-06-24 Novartis Ag Uses of anti-tgf-beta antibodies and checkpoint inhibitors for the treatment of proliferative diseases
WO2021252920A1 (en) 2020-06-11 2021-12-16 Novartis Ag Zbtb32 inhibitors and uses thereof
WO2021260528A1 (en) 2020-06-23 2021-12-30 Novartis Ag Dosing regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2022029573A1 (en) 2020-08-03 2022-02-10 Novartis Ag Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2022104061A1 (en) 2020-11-13 2022-05-19 Novartis Ag Combination therapies with chimeric antigen receptor (car)-expressing cells
WO2022215011A1 (en) 2021-04-07 2022-10-13 Novartis Ag USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
WO2022229853A1 (en) 2021-04-27 2022-11-03 Novartis Ag Viral vector production system
WO2023143315A1 (en) * 2022-01-29 2023-08-03 杭州中美华东制药有限公司 Ror1-targeted antibody or antigen-binding fragment thereof and use thereof
WO2023214325A1 (en) 2022-05-05 2023-11-09 Novartis Ag Pyrazolopyrimidine derivatives and uses thereof as tet2 inhibitors
US11932691B2 (en) 2021-01-25 2024-03-19 Fred Hutchinson Cancer Center Anti-ROR1 antibodies and uses thereof

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012012695A2 (en) * 2010-07-23 2012-01-26 Fred Hutchinson Cancer Research Center A method for the treatment of obesity
EP2997134B1 (en) 2013-05-14 2020-06-24 Board of Regents, The University of Texas System Human application of engineered chimeric antigen receptor (car) t-cells
KR102375998B1 (en) 2014-02-14 2022-03-21 더 보드 오브 리젠츠 오브 더 유니버시티 오브 텍사스 시스템 Chimeric antigen receptors and methods of making
PL3280729T3 (en) 2015-04-08 2022-08-22 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
CA2983293C (en) * 2015-04-24 2023-06-20 The Regents Of The University Of California Modulators of ror1-ror2 binding
CA2992551A1 (en) 2015-07-21 2017-01-26 Novartis Ag Methods for improving the efficacy and expansion of immune cells
KR20180069067A (en) 2015-10-30 2018-06-22 엔비이-테라퓨틱스 아게 Anti-ROR1 antibody
CN110225927B (en) 2016-10-07 2024-01-12 诺华股份有限公司 Chimeric antigen receptor for the treatment of cancer
CN110612119A (en) 2017-02-07 2019-12-24 西雅图儿童医院(Dba西雅图儿童研究所) Phospholipid ether (PLE) CAR T cell tumor targeting (CTCT) agents
EP3589295A4 (en) 2017-02-28 2020-11-04 Endocyte, Inc. Compositions and methods for car t cell therapy
BR112019024654A2 (en) * 2017-05-23 2020-06-09 Dragonfly Therapeutics Inc binding protein nkg2d, cd16 and ror1 or ror2
US10905750B2 (en) 2017-11-10 2021-02-02 Donald J. Davidson GRP78 antagonist that block binding of receptor tyrosine kinase orphan receptors as immunotherapy anticancer agents
EP3743082A4 (en) 2018-01-22 2021-11-03 Seattle Children's Hospital (DBA Seattle Children's Research Institute) Methods of use for car t cells
EP3759130A1 (en) 2018-02-26 2021-01-06 Fred Hutchinson Cancer Research Center Compositions and methods for cellular immunotherapy
WO2020018964A1 (en) 2018-07-20 2020-01-23 Fred Hutchinson Cancer Research Center Compositions and methods for controlled expression of antigen-specific receptors
CN110590951A (en) * 2019-09-27 2019-12-20 安萌得医药科技(上海)有限公司 ROR1 monoclonal antibody, preparation method and application thereof
JP2023538945A (en) * 2020-08-24 2023-09-12 エピムアブ、バイオセラピューティクス、(ホンコン)、リミテッド Anti-ROR1 antibodies and related bispecific binding proteins
WO2022133169A1 (en) 2020-12-18 2022-06-23 Century Therapeutics, Inc. Chimeric antigen receptor systems with adaptable receptor specificity
WO2023077026A1 (en) 2021-10-28 2023-05-04 Lyell Immunopharma, Inc. Methods for culturing cells expressing ror1-binding protein

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008076868A2 (en) * 2006-12-18 2008-06-26 Abbott Laboratories Methods and compositions related to modulation of receptor tyrosine kinase orphan receptor-1 (ror-1)
WO2008103849A2 (en) * 2007-02-21 2008-08-28 The Regents Of The University Of California Methods and compounds for lymphoma cell detection and isolation

Family Cites Families (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5843749A (en) 1991-07-26 1998-12-01 Regeneron Pharmaceuticals, Inc. Ehk and Ror tyrosine kinases
US20050144655A1 (en) 2000-10-31 2005-06-30 Economides Aris N. Methods of modifying eukaryotic cells
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US7105348B2 (en) 2000-10-31 2006-09-12 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US6586251B2 (en) 2000-10-31 2003-07-01 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US20140072979A1 (en) 2001-01-16 2014-03-13 Regeneron Pharmaceuticals, Inc. Isolating cells expressing secreted proteins
US20090137416A1 (en) 2001-01-16 2009-05-28 Regeneron Pharmaceuticals, Inc. Isolating Cells Expressing Secreted Proteins
US20140072980A1 (en) 2001-01-16 2014-03-13 Regeneron Pharmaceuticals, Inc. Isolating cells expressing secreted proteins
EP1392859B1 (en) 2001-01-16 2006-05-10 Regeneron Pharmaceuticals, Inc. Isolating cells expressing secreted proteins
WO2005070966A2 (en) 2004-01-16 2005-08-04 Regeneron Pharmaceuticals, Inc. Fusion polypeptides capable of activating receptors
US8298532B2 (en) 2004-01-16 2012-10-30 Regeneron Pharmaceuticals, Inc. Fusion polypeptides capable of activating receptors
WO2007146957A2 (en) 2006-06-13 2007-12-21 Irm Llc Ror1 as a therapeutic target for lung cancer
ATE540313T1 (en) 2007-05-03 2012-01-15 Regeneron Pharma IDENTIFICATION OF GERMINAL EMBRYONIC STEM CELLS
WO2010008069A1 (en) 2008-07-18 2010-01-21 国立大学法人名古屋大学 Cell proliferation inhibitor
US9316646B2 (en) 2009-04-23 2016-04-19 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anti-human ROR1 antibodies
US20120282177A1 (en) 2009-11-02 2012-11-08 Christian Rohlff ROR1 as Therapeutic and Diagnostic Target
EP2513146B1 (en) 2009-12-18 2017-05-03 Kancera AB Antibodies against ror1 capable of inducing cell death of cll
WO2011123859A1 (en) 2010-04-02 2011-10-06 Skyfire Labs, Inc. Assisted hybrid mobile browser
WO2011159847A2 (en) 2010-06-15 2011-12-22 The Regents Of The University Of California Receptor tyrosine kinase-like orphan receptor 1 (ror1) single chain fv antibody fragment conjugates and methods of use thereof
WO2012012695A2 (en) 2010-07-23 2012-01-26 Fred Hutchinson Cancer Research Center A method for the treatment of obesity
EP3828205A1 (en) 2010-10-01 2021-06-02 Oxford BioTherapeutics Ltd Anti-ror1 antibodies
GB201020995D0 (en) 2010-12-10 2011-01-26 Bioinvent Int Ab Biological materials and uses thereof
EP3392274A1 (en) 2011-08-12 2018-10-24 Omeros Corporation Anti-fzd10 monoclonal antibodies and methods for their use
EP2617319A1 (en) 2012-01-17 2013-07-24 Braun GmbH Head portion and handle portion of an oral care device
CN103792364B (en) 2012-10-31 2016-06-08 张宝弘 For detecting reagent and the application thereof of circulating tumor cell ROR1 albumen in peripheral blood
TW202206465A (en) 2012-11-14 2022-02-16 美商再生元醫藥公司 Recombinant cell surface capture proteins
EP2789630A1 (en) 2013-04-09 2014-10-15 EngMab AG Bispecific antibodies against CD3e and ROR1
RU2725520C2 (en) 2013-12-11 2020-07-02 Регенерон Фармасьютикалс, Инк. Methods and compositions for genome targeted modification

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008076868A2 (en) * 2006-12-18 2008-06-26 Abbott Laboratories Methods and compositions related to modulation of receptor tyrosine kinase orphan receptor-1 (ror-1)
WO2008103849A2 (en) * 2007-02-21 2008-08-28 The Regents Of The University Of California Methods and compounds for lymphoma cell detection and isolation

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BASKAR, S. ET AL.: 'Unique cell surface expression of receptor tyrosine kinase ROR1 in human B-cell chronic lymphocytic leukemia' CLIN. CANCER RES. vol. 14, no. 2, 15 January 2008, pages 396 - 404 *
DANESHMANESH, A. H. ET AL.: 'Rorl, a cell surface receptor tyrosine kinase is expressed in chronic lymphocytic leukemia and may serve as a putative target for therapy' INT. J. CANCER vol. 123, no. 5, 01 September 2008, pages 1190 - 1195 *

Cited By (65)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11548953B2 (en) 2010-06-15 2023-01-10 The Regents Of The University Of California Receptor tyrosine kinase-like orphan receptor 1 (ROR1) single chain Fv antibody fragment conjugates and methods of use thereof
US9938350B2 (en) 2010-06-15 2018-04-10 The Regents Of The University Of California Receptor tyrosine kinase-like orphan receptor 1 (ROR1) single chain Fv antibody fragment conjugates and methods of use thereof
WO2012075158A1 (en) * 2010-12-01 2012-06-07 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric rabbit/human ror1 antibodies
US9758586B2 (en) 2010-12-01 2017-09-12 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric rabbit/human ROR1 antibodies
US9933434B2 (en) 2011-01-14 2018-04-03 The Regents Of The University Of California Therapeutic antibodies against ROR-1 protein and methods for use of same
US11536727B2 (en) 2011-01-14 2022-12-27 The Regents Of The University Of California Therapeutic antibodies against ROR-1 protein and methods for use of same
US10627409B2 (en) 2011-01-14 2020-04-21 The Regents Of The University Of California Therapeutic antibodies against ROR-1 protein and methods for use of same
US10900973B2 (en) 2011-01-14 2021-01-26 The Regents Of The University Of California Therapeutic antibodies against ROR-1 protein and methods for use of same
US9523695B2 (en) 2011-01-14 2016-12-20 The Regents Of The University Of California Therapeutic antibodies against ROR-1 protein and methods for use of same
EP2789630A1 (en) 2013-04-09 2014-10-15 EngMab AG Bispecific antibodies against CD3e and ROR1
WO2015142675A2 (en) 2014-03-15 2015-09-24 Novartis Ag Treatment of cancer using chimeric antigen receptor
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
WO2016057705A1 (en) 2014-10-08 2016-04-14 Novartis Ag Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
WO2016055592A1 (en) 2014-10-09 2016-04-14 Engmab Ag Bispecific antibodies against cd3epsilon and ror1
WO2016090034A2 (en) 2014-12-03 2016-06-09 Novartis Ag Methods for b cell preconditioning in car therapy
WO2016115559A1 (en) * 2015-01-16 2016-07-21 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for ror1
US11919970B2 (en) 2015-01-16 2024-03-05 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for ROR1
EP3760644A1 (en) * 2015-01-16 2021-01-06 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for ror1
US10889652B2 (en) 2015-01-16 2021-01-12 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for ROR1
JP2018503380A (en) * 2015-01-16 2018-02-08 ジュノー セラピューティクス インコーポレイテッド Antibodies and chimeric antigen receptors specific for ROR1
JP2021061857A (en) * 2015-01-16 2021-04-22 ジュノー セラピューティクス インコーポレイテッド Antibodies and chimeric antigen receptors specific for ror1
WO2016126608A1 (en) 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
WO2016172583A1 (en) 2015-04-23 2016-10-27 Novartis Ag Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
WO2017027392A1 (en) 2015-08-07 2017-02-16 Novartis Ag Treatment of cancer using chimeric cd3 receptor proteins
WO2017091786A1 (en) 2015-11-23 2017-06-01 Novartis Ag Optimized lentiviral transfer vectors and uses thereof
WO2017114497A1 (en) 2015-12-30 2017-07-06 Novartis Ag Immune effector cell therapies with enhanced efficacy
EP4219689A2 (en) 2015-12-30 2023-08-02 Novartis AG Immune effector cell therapies with enhanced efficacy
WO2017127664A1 (en) 2016-01-20 2017-07-27 The Scripps Research Institute Ror1 antibody compositions and related methods
US10968275B2 (en) 2016-02-02 2021-04-06 Fred Hutchinson Cancer Research Center Anti-ROR1 antibodies and uses thereof
WO2017149515A1 (en) 2016-03-04 2017-09-08 Novartis Ag Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore
WO2017165683A1 (en) 2016-03-23 2017-09-28 Novartis Ag Cell secreted minibodies and uses thereof
WO2017181119A2 (en) 2016-04-15 2017-10-19 Novartis Ag Compositions and methods for selective protein expression
EP4219721A2 (en) 2016-04-15 2023-08-02 Novartis AG Compositions and methods for selective protein expression
WO2018014001A1 (en) 2016-07-14 2018-01-18 Fred Hutchinson Cancer Research Center Multiple bi-specific binding domain constructs with different epitope binding to treat cancer
WO2018026819A2 (en) 2016-08-01 2018-02-08 Novartis Ag Treatment of cancer using a chimeric antigen receptor in combination with an inhibitor of a pro-m2 macrophage molecule
WO2018111340A1 (en) 2016-12-16 2018-06-21 Novartis Ag Methods for determining potency and proliferative function of chimeric antigen receptor (car)-t cells
WO2018140725A1 (en) 2017-01-26 2018-08-02 Novartis Ag Cd28 compositions and methods for chimeric antigen receptor therapy
EP4043485A1 (en) 2017-01-26 2022-08-17 Novartis AG Cd28 compositions and methods for chimeric antigen receptor therapy
WO2018144535A1 (en) 2017-01-31 2018-08-09 Novartis Ag Treatment of cancer using chimeric t cell receptor proteins having multiple specificities
WO2018160731A1 (en) 2017-02-28 2018-09-07 Novartis Ag Shp inhibitor compositions and uses for chimeric antigen receptor therapy
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
WO2018237335A1 (en) * 2017-06-23 2018-12-27 VelosBio Inc. Ror1 antibody immunoconjugates
US10335496B2 (en) 2017-06-23 2019-07-02 VelosBio Inc. ROR1 antibody immunoconjugates
US10758556B2 (en) 2017-08-07 2020-09-01 Nbe-Therapeutics Ag Anthracycline-based antibody drug conjugates having high in vivo tolerability
WO2019081983A1 (en) 2017-10-25 2019-05-02 Novartis Ag Antibodies targeting cd32b and methods of use thereof
WO2019084288A1 (en) 2017-10-25 2019-05-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2019089798A1 (en) 2017-10-31 2019-05-09 Novartis Ag Anti-car compositions and methods
WO2019210153A1 (en) 2018-04-27 2019-10-31 Novartis Ag Car t cell therapies with enhanced efficacy
WO2019213282A1 (en) 2018-05-01 2019-11-07 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
WO2019237035A1 (en) 2018-06-08 2019-12-12 Intellia Therapeutics, Inc. Compositions and methods for immunooncology
WO2020012337A1 (en) 2018-07-10 2020-01-16 Novartis Ag 3-(5-amino-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and their use in the treatment of i karos family zinc finger 2 (ikzf2)-dependent diseases
WO2020128972A1 (en) 2018-12-20 2020-06-25 Novartis Ag Dosing regimen and pharmaceutical combination comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2020165834A1 (en) 2019-02-15 2020-08-20 Novartis Ag Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020165833A1 (en) 2019-02-15 2020-08-20 Novartis Ag 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2021123996A1 (en) 2019-12-20 2021-06-24 Novartis Ag Uses of anti-tgf-beta antibodies and checkpoint inhibitors for the treatment of proliferative diseases
WO2021252920A1 (en) 2020-06-11 2021-12-16 Novartis Ag Zbtb32 inhibitors and uses thereof
WO2021260528A1 (en) 2020-06-23 2021-12-30 Novartis Ag Dosing regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2022029573A1 (en) 2020-08-03 2022-02-10 Novartis Ag Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2022104061A1 (en) 2020-11-13 2022-05-19 Novartis Ag Combination therapies with chimeric antigen receptor (car)-expressing cells
US11932691B2 (en) 2021-01-25 2024-03-19 Fred Hutchinson Cancer Center Anti-ROR1 antibodies and uses thereof
WO2022215011A1 (en) 2021-04-07 2022-10-13 Novartis Ag USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
WO2022229853A1 (en) 2021-04-27 2022-11-03 Novartis Ag Viral vector production system
WO2023143315A1 (en) * 2022-01-29 2023-08-03 杭州中美华东制药有限公司 Ror1-targeted antibody or antigen-binding fragment thereof and use thereof
WO2023214325A1 (en) 2022-05-05 2023-11-09 Novartis Ag Pyrazolopyrimidine derivatives and uses thereof as tet2 inhibitors

Also Published As

Publication number Publication date
US20180362657A1 (en) 2018-12-20
US20130101607A1 (en) 2013-04-25
US11548953B2 (en) 2023-01-10
US20160304619A1 (en) 2016-10-20
US20210171655A1 (en) 2021-06-10
WO2011159847A3 (en) 2012-05-18
US9242014B2 (en) 2016-01-26
US9938350B2 (en) 2018-04-10

Similar Documents

Publication Publication Date Title
US11548953B2 (en) Receptor tyrosine kinase-like orphan receptor 1 (ROR1) single chain Fv antibody fragment conjugates and methods of use thereof
US10875913B2 (en) Methods of treatment using activatable anti-EGFR antibodies
RU2477728C1 (en) ScFv-BINDING SPARC
US7335744B2 (en) Prostate cancer specific internalizing human antibodies
US8313745B2 (en) Compositions against cancer antigen LIV-1 and uses thereof
US20070207510A1 (en) Methods and compounds for lymphoma cell detection and isolation
US8906342B2 (en) Compositions against cancer antigen LIV-1 and uses thereof
JP2002524024A (en) ErbB2 antibody to internalize
US20030215453A1 (en) Methods of therapy and diagnosis using immunotargeting of cells expressing VpreB1 protein
Marshall et al. Engineering and characterization of a novel fusion protein incorporating B7. 2 and an anti–ErbB-2 single-chain antibody fragment for the activation of Jurkat T cells
US20040048817A1 (en) Methods of immunotherapy and diagnosis
KR102647825B1 (en) Anti-HLA-DP monoclonal antibody and use thereof
WO2007101676A2 (en) Treatment, diagnosis and imaging of mantle cell lymphoma
WO2022232612A1 (en) Lassa virus-specific nanobodies and methods of their use

Legal Events

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

Ref document number: 11796403

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 13703864

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11796403

Country of ref document: EP

Kind code of ref document: A2