US20030147808A1 - Single Chain Fv constructs of anti-ganglioside GD2 antibodies - Google Patents

Single Chain Fv constructs of anti-ganglioside GD2 antibodies Download PDF

Info

Publication number
US20030147808A1
US20030147808A1 US10/075,947 US7594702A US2003147808A1 US 20030147808 A1 US20030147808 A1 US 20030147808A1 US 7594702 A US7594702 A US 7594702A US 2003147808 A1 US2003147808 A1 US 2003147808A1
Authority
US
United States
Prior art keywords
scfv
antibody
peptide
streptavidin
variable region
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US10/075,947
Inventor
Nai-Kong Cheung
Steven Larson
Hong-Fen Guo
Ken Rivlin
Michel Sadelain
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sloan Kettering Institute for Cancer Research
Original Assignee
Sloan Kettering Institute for Cancer Research
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 Sloan Kettering Institute for Cancer Research filed Critical Sloan Kettering Institute for Cancer Research
Priority to US10/075,947 priority Critical patent/US20030147808A1/en
Assigned to SLOAN-KETTERING INSTITUTE FOR CANCER RESEARCH reassignment SLOAN-KETTERING INSTITUTE FOR CANCER RESEARCH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SADELAIN, MICHEL, RIVLIN, KEN, CHEUNG, NAI-KONG V., GUO, HONG-FEN, LARSON, STEVEN M.
Publication of US20030147808A1 publication Critical patent/US20030147808A1/en
Assigned to UNITED STATES DEPARTMENT OF ENERGY reassignment UNITED STATES DEPARTMENT OF ENERGY CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: SLOAN-KETTERING INST CAN RESEARCH
Abandoned legal-status Critical Current

Links

Images

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
    • C07K16/3076Immunoglobulins [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 against structure-related tumour-associated moieties
    • C07K16/3084Immunoglobulins [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 against structure-related tumour-associated moieties against tumour-associated gangliosides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6891Pre-targeting systems involving an antibody for targeting specific cells
    • A61K47/6897Pre-targeting systems with two or three steps using antibody conjugates; Ligand-antiligand therapies
    • A61K47/6898Pre-targeting systems with two or three steps using antibody conjugates; Ligand-antiligand therapies using avidin- or biotin-conjugated antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6891Pre-targeting systems involving an antibody for targeting specific cells
    • A61K47/6899Antibody-Directed Enzyme Prodrug Therapy [ADEPT]
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/36Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Actinomyces; from Streptomyces (G)
    • 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
    • C07K14/70517CD8
    • 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

Definitions

  • This application relates to single chain antibody constructs which specifically bind to the disialoganglioside G D2 , and to the use of such constructs for targeted delivery of imaging agents or therapeutic agents to human neuroectodermal derived cancers.
  • Gangliosides are acidic glycosphingolipids found on the outer surface of most cell membranes. 1 Many tumors have abnormal glycolipid composition and structure. Disialoganglioside G D2 has been found in a wide spectrum of human tumors, including neuroblastoma, osteosarcomas and other soft tissue sarcomas, medulloblastomas, high grade astrocytomas, melanomas, and small cell lung cancer. 2-4 Among glioblastoma multiforme and anaplastic astrocytoma, anti-G D2 demonstrated the most restrictive pattern when compared with anti-GD3 and anti-GM2 antibodies. 5,6
  • Gangliosides are ideal targets for monoclonal antibodies (MAb) because of the high antigen density, lack of modulation, relative homogeneity in many tumors and the possibility of up-regulation by cytokines. 7
  • the only normal tissues with high ganglioside expression are neurons, and biodistribution studies have shown that MAb do not localize to the nontumorous brain or spinal cord because of the blood brain barrier. In contrast, in patients with primary or metastatic brain tumors, specific antibodies can localize preferentially to tumor tissues, but not to normal brain. 8
  • Murine monoclonal antibodies have been prepared to ganglioside G D2 Using somatic cell hybridization, murine MAbs were produced against the ganglioside GD 2 . 9 They were shown to react with disialoganglioside G D2 , but not with GD3, GT1b, GD1b, GD1a, GM1, GM3 and GM4.
  • base-treatment step was omitted from the standard neuroblastoma ganglioside extraction procedure
  • immuno-thin-layer-chromatography (ITLC) using 3F8, 3G6 and other anti-G D2 MAbs revealed a new ganglioside band with Rf of 0.342, besides G D2 (Rf 0.183). 4 Immunochemical analysis showed that this new neuroblastoma ganglioside contained alkali-sensitive O-acetylated sialic acid residues recognized by MAb D1.1.
  • G D2 G D2 expression was analyzed in 67 solid tumors and normal tissues from children by using the antibody 3A7. 11 G D2 expression was found in 28 of 28 neuroblastomas, and was most abundant in stroma-poor tumors. Differentiating stroma-rich neuroblastomas, neuroblastic clusters, neurofibrils, and most ganglion-like cells were found to be G D2 positive, whereas Schwann's-cell stroma did not express G D2 . In ganglioneuromas, only a few ganglion-like cells showed G D2 , whereas all other structures were negative.
  • G D2 Scattered foci of G D2 were also found in some non-neuronal tumors, such as rhabdomyosarcomas and osteosarcomas, but not in lymphomas, Askin tumors, or most Wilm's tumors. 3A7 was also found to react with retinoblastomas. 12
  • Embryonal rhabdomyo-sarcoma and synovial sarcoma demonstrated substantially weaker staining by either MAb.
  • Ganglioside extraction and immuno-thin layer chromatography (ITLC) confirmed the identities of these gangliosides as G D2 and GD3 respectively
  • the efficacy of tumor targeting was then tested by imaging neuroblastoma patients with 131I-MAb. Radiolocalization was demonstrated in primary tumors of the mediastinum and abdomen, as well as metastatic disease in the lymph nodes, bone marrow and bone. 16,17 The specificity was validated by tumor and marrow biopsies, as well as by CT/MRI and bone scans. A comparison with 131I-meta-iodobenzylguanidine (MIBG) suggested that 131I-MAb was twice as sensitive in detecting metastatic sites of disease. The tumor uptake in patients was 0.08% of the injected dose per gm (compared to 0.002% for MIBG).
  • This high tumor uptake in vivo was a result of (1) the high density (5 ⁇ 106/cell) and homogeneity of the target antigen G D2 , and (2) the lack of uptake in the reticuloendothelial system.
  • G D2 specific antibodies include small cell lung cancer, 18 brain tumors, 8 and both osteosarcomas 13 and soft tissue sarcomas. 19
  • a phase I study to test the biological toxicity of “cold” anti-G D2 was carried out in 1987 in 17 patients with metastatic neuroblastoma or melanoma.
  • Acute self-limited toxicities of MAb treatment were severe pain requiring analgesics, fever, urticaria, hypertension, hypotension, anaphylactoid reactions of the respiratory tract, as well as significant decreases in blood counts and serum complement levels. There were no treatment related deaths.
  • the 5 neuroblastoma patients who are still alive and well (19 mos, 3y, 5y, 5y, 6y respectively after MAb treatment)
  • there are no acute or delayed neurological complications attributable to MAb therapy are no acute or delayed neurological complications attributable to MAb therapy.
  • one patient had chemo-radiotherapy-resistant stage IVS neuroblastoma, and the other 4 had poor risk stage IV neuroblastoma diagnosed at more than one year of age (2 relapsed neuroblastoma and 2 with refractory neuroblastoma prior to antibody treatment).
  • Marrow was infused when blood radioactivity decreased to ⁇ 0.01 uCi/ml in the first 18 patients and to ⁇ 1 uCi/ml in the last 4 patients.
  • Acute toxicities of 131I-MAb treatment included pain (19/23) during the infusion, fever (19/23), hyperbilirubinemia (6/23), and diarrhea. All patients developed grade 4 myelosuppression with sepsis in 7/23 patients (5 fungal, 2 bacterial), disseminated zoster in 1, and pneumocystis in 1.
  • 3 patients developed hypothyroidism.
  • Subsequent 14 patients were treated with synthroid or Cytomel for thyroid protection.
  • hybrdoma 3F8 was found to be negative for adventitious agents by MAP, S+L ⁇ , and XC plaque assays, as well as negative for reverse transcriptase.
  • MAP testing included screening for murine leukovirus, LCM virus isolation by intracerebral inoculation, murine saliva gland virus, mouse thymic virus, EDIM and LDH virus isolations.
  • Purified antibody e.g. 3F8 had to pass MAP and sterility testing (bacteria, mycoplasma, and fungal cultures), rabbit pyrogen testing, as well as safety testing in mice and guinea pigs. Conjugation to 131I by the chloramine-T method was supervised by Dr. Ronald Finn and Dr.
  • Radiolabeled antibody 3F8 must have >50% binding by in vitro antigen binding assay, >95% TCA precipitable and ⁇ 3% free iodine by radio-thin layer chromatography. Periodic testing of radiolabeled antibodies was performed to ensure sterility as well as the absence of pyrogen.
  • G D2 human neuroectodermal-derived cancers (melanoma, small cell lung cancer, neuroblastoma, brain tumors, sarcoma, HTLV-1 leukemia, retinoblastoma and osteosarcoma) and the preliminary clinical studies of monoclonal antibodies to G D2 in radioimmuno-scintigraphy and radioimmunotherapy have been encouraging, further optimization of antibodies for binding to G D2 would be desirable. It is an object of the present invention to provide such optimized antibodies and the DNA sequences coding therefore.
  • the antibodies of the present invention are recombinant antibody constructs comprising the variable regions of the heavy and light chains of anti-G D2 antibodies. These antibody constructs may be coupled to a label such as a radiolabel or to a protein such as streptavidin or pro-drug converting enzymes for use in imaging or therapeutic applications. The antibody constructs may also be transduced into T cells to produce populations of T cells which target G D2 -producing tumor cells.
  • FIG. 1 shows two alternative strategies for the use of the antibody constructs of the present invention in therapy.
  • This invention relates to optimized antibodies to G D2 , to DNA encoding such antibodies, and to the use of the antibodies and DNA in diagnostic assays and therapy.
  • the antibodies of the invention are antibody constructs comprising the variable regions of the heavy and light chains of anti-G D2 antibodies as a single chain Fv fragment.
  • Single-chain Fv fragments offer some of the best opportunities to achieve these results.
  • ScFv technology utilizes molecular biology methods to reduce antibodies to the minimal-required-unit of heavy and light chain variable regions tethered by a peptide linker which can be designed with versatile side chains for radioconjugation.
  • the anti-G D2 scFvs shown in Tables 2 and 3 have been prepared using the methods described in the examples.
  • the orientation VH-VL is used for the scFv.
  • the orientation VL-VH is necessary. Since the cDNA sequences of 3G6 and 3F8 are similar, we expect the binding properties of both of these antibodies to be very similar if not identical. Since our cloning strategies have been most successful with 3G6, this has been the focus of our research instead of 3F8.
  • Histidine-tag (His5) is inserted for ease of purification by Ni-column and Myc-tag is inserted to facilitate detection. Myc-tag and E-tag can be genetically removed if necessary for future clinical studies.
  • scFv-strep fusion proteins are expected to form tetramers with both antigen- and biotin-binding activity They are expected to be stable over a wide range of pH and range of physiologic temperatures.
  • All these single chains have strings of 5 histidine residues inserted at the carboxyl end for purification on the Nickel-column using FPLC. Washing was done at 10 mM Imidazole and scFv peak elution at 500 mM Imidazole. Further purification of scFv can be accomplished by size-exclusion using Sephadex HR75 and superose 6 (hi-resolution). scFv can be fuirther affinity purified by myc-affinity chromatography. 9E10 is a hydridoma (ATCC) that secretes the antibody specific for the myc-tag. From ascites the 9E10 IgG1 antibody is purified by protein-G affinity chromatography and used for chemical coupling to sepharose 4B.
  • ATCC hydridoma
  • Recombinant antibody constructs according to the invention can be coupled to metal labels such as 99m Tc for use in diagnostic imaging of G D2 expressing cells.
  • technetium can be chelated to the antibody construct via a heterobifuinctional linker such as succinimidyl-6-hydrazinonicotinate hydrochloride (SHNH).
  • SHNH succinimidyl-6-hydrazinonicotinate hydrochloride
  • SHNH succinimidyl-6-hydrazinonicotinate hydrochloride
  • Tc(V) precursors coupled readily and conveniently to the SHNH-modified protein to yield the desired 99mTc-radiolabeled conjugate.
  • 99mTc-3F8 localized rapidly and successfully to G D2 -positive xenografts.
  • SHNH-modified scFvs can be synthesized for conjugation to 99mTc using the techniques described in Schwartz et al., Bioconjugate Chem 2:333-336 (1991), which is incorporated herein by reference.
  • Metal chelation to scFv can also be accomplished via the streptavidin protein.
  • the rationale of pretargeting using scFv-streptavidin fusion proteins in radioimmunotherapy are 5-fold: (a) Large amounts of scFv can be used to saturate G D2 sites in vivo, without the accompanying blood and tissue toxicity from radioisotope, (b) radiolabel is injected at the time when the tumor-nontumor ratio of scFv is maximal, (c) a radiolabeled ligand is chosen such that it binds with high affinity (e.g.
  • scFv-streptavidin is a homo-tetramer, as such the antigen binding avidity is greatly amplified especially for high-density antigens (e.g. G D2 on neuroblastoma).
  • scFv-strep fusion proteins for both 5F11 and 3G6 have been made and purified.
  • scFv coupled to technetium provides a safe, camera-ready isotope, with fast-decay and therefore easy disposal.
  • 99m -technetium is optimal for imaging studies.
  • Other isotopes can also be used, including a positron-emitting technetium for PET imaging.
  • rhenium a therapeutic beta-emitting radionuclide
  • the scFv and scFv-streptavidin of the invention are also useful in a number of therapeutic applications, which is turn form aspects of the present invention
  • these approaches involve administration of scFV coupled to a therapeutic or pre-therapeutic moiety.
  • ScFv-streptavidin streptavidin being the pre-therapeutic moiety
  • a therapeutic agent (X) bound to biotin is then introduced. Binding of the biotin the streptavidin results in localization of the chemotherapeutic agent X at the site of the G D2 producing cells.
  • Other pre-therapeutic moieties include pro-drug converting enzymes. Directly therapeutic moieties such as toxins can also be used.
  • a second approach also illustrated in FIG. 1, utilizes a vector encoding ScFv is transduced into primary human lymphocytes (preferably along with a suicide gene such a HSV-TK).
  • the transduced lymphocytes now recognize and target G D2 , resulting in an immune response to the G D2 -producing cells.
  • the scFv or scFv-streptavidin can be incorporated in a fusion protein with therapeutic agents such as toxins or pro-drug converting enzymes, can be incorporated in a fusion protein with CD8 to facilitate the formation of G D2 -targeted lymphocytes, or can be coupled to viral coat proteins superantigen (SEA) to facilitate targeting of G D2 producing cells,
  • therapeutic agents such as toxins or pro-drug converting enzymes
  • CD8 to facilitate the formation of G D2 -targeted lymphocytes
  • SEA viral coat proteins superantigen
  • scFv and scFv-streptavidin can also be usefully combined in a fusion protein with CD8.
  • scFv-CD8 constructs can be transfected through retroviral vector into human and mouse lymphocytes. Since these scFv are permanently integrated into the cellular genome, these lymphocytes express scFv on their cell surface and through the CD8 cytoplasmic domain become activated upon antigen binding. scFv facilitates the homing of these cells to tumor sites, thus being effective in promoting both the localization and killing of tumors. With a suicide gene, thymidine kinase, also transfected, these cells can now be turned on and off as needed.
  • scFv-enzyme and scFv-enzyme-streptavidin conjugates can be used to provide targeted drug therapy using a technique known as ADEPT (antibody directed enzyme prodrug-therapy).
  • ADEPT antibody directed enzyme prodrug-therapy
  • Suitable enzymes for this technique include carboxypeptidase G2, alkaline phosphatase, and ⁇ -Lactamase.
  • a prodrug derivative e.g. cephalosporin derivative of doxo20
  • the enzyme beta-lactamase
  • tumor cells are exposed to a high local concentration (up to 10-fold higher than blood/tissue levels) of specific chemotherapeutic agents.
  • scFv with or without streptavidin
  • integration of scFv (with or without streptavidin) into viral coat proteins can be used to retarget these viruses in vivo.
  • viruses include adenovirus, retrovirus and herpes virus.
  • SEA Superantigen
  • ScFv-SEA and scFv-streptavidin-SEA can target T cells to lyse antigen-positive MHC-class II-negative human tumor cells.
  • SEA has been cloned (Betley et al: J. Bacteriology 170: 34-41, 1988) and the cDNA is available for making fusion proteins.
  • 5F11 hybridoma cells were processed for mRNA using a commercially available kit (Quick Prep Micro MRNA Purification, pharmacia Biotech) following the procedures outlined by the manufacturer. Briefly, hybridoma cells were cultured in ROPMI-1640 medium supplemented with 10% calf serum, 2 mmol/L L-glutamine (Sigma), 100 U/L penicillin and 100 ug/ml streptomycin sulfate (Sigma). The cell cultures were maintained at 37° C. under a water-saturated atmosphere of 5% CO 2 .
  • the mRNA preparation was used in the construction of the 5F11 scFv gene using the Mouse ScFv Module/Recombinant Phage Antibody System (Pharmacia Biotech). 5 ul of the mRNA preparation was reverse transcribed in a total volume of 11 ul of reaction mixture and 1 ul DTT solution for 1 hour at 37° C.
  • light primer mix and the heavy primer set were added respectively to generate quantities of the light (325 bp(and heavy (340 bp) chains.
  • 5 U Ampli Taq DNA polymerase (Perkin Elmer) was added.
  • the PCR cycle consisted of a 1 min denaturation step at 94° C.
  • PCR derived fragments were purified using glassmilk beads (Bio 101 Co.) and evaluated by electrophoresis on 1/5% agarose gel in TAE buffer with ethidium bromide visiualization.
  • both purified heavy chain and light chain fragments were added to ab appropriate PCR mixture containing linker-primer, dNTPs, PCR buffer and Ampli Taq DNA polymerase. Denaturation was performed at 94° C. for 1 minute, followed by a 4 minute annealing reaction at 63° C.
  • the heavy and light DNA were joined into a single chain with linker DNA after 7 thermocycles.
  • this single chain DNA as a template and restriction site primers (RS primers) containing either SfiI or NotI restriction sites
  • secondary PCR amplification was carried out for 30 cycles to amplify the ScFv DNA and add the restriction sites. This introduced the SfiI restrictions site at the 5′-end of the heavy chain and the NotI restriction site at the 3′-end of the light chain.
  • Amplified ScFv DNA was then purified by glassmilk beads and digested with SfiI and NotI.
  • Antibody-producing recombinant phage were selected by panning using the method of Ditzel, PNAS USA 91: 3710-3714 (1994) with slight modifications. 20 ul of GD2 (1 ul/ml) dissolved in ethanol were directly coated on a 96-well polystyrene plate and dried at rom temperature. Then 100 ul of the supernatant containing the phage library was added to each well and incubated for 2 hours. The plate was then washed 10 times with PBS containing 0 05% BSA to remove nonspecifically bound phage.
  • Antibody-positive recombinant phage captured by the G D2 antigen was eluted with 0.1 M HCl (pH 2.2 with solid glycine and 0.1% BSA) and neutralized with 2M Tris solution. Selected phage was then re-panned for two additional cycles to further enrich the GD2-binding recombinant phages.
  • the selected phage was used to reinfect E coli XL1-Blue cells. Clones were grown in 2XYT medium containing ampicillin (100 ug/ml) and 1% glucose at 30° C. until an OD 600 of 0.5 was obtained. Expression of ScFv antibody was induced by changing to a medium containing 100 uM IPTG and incubating overnight at 30° C. The supernatant obtained from the medium by centrifugation was directly added to a plate coated with GD2. The pellet was resuspended in PBD containing 1 mM EDTA and incubated on ice for 10 minutes. The periplasmic soluble antibody was collected by centrifugation again and added to the plate. After incubating at 37° C. for 32 hours, anti-E Tag antibody (Pharmacia Biotech) was used to specifically screen the binding of the ScFv fragment.
  • plasmid DNA from the 5F11-scFV in pCantab 5E vector was purified and amplified by PCR using two specially designed primers S6 and 318s.
  • S6 contains a NotI restriction site and 318s contains a PvuII restriction site so that amplified DNA can be restriction digested and inserted in the pSTE vector (Dr. Dubel, German Cancer Center).
  • the resulting vector 5FpoStMCH is the 5F11-scFv-streptavidin construct.
  • the streptavidin was digested with BamHI, leaving the scFV 5FpoMCH.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Cell Biology (AREA)
  • Epidemiology (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nanotechnology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Public Health (AREA)
  • Biotechnology (AREA)
  • Medical Informatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Recombinant antibody constructs comprise the variable regions of the heavy and light chains of anti-GD2 antibodies. These antibody constructs may be coupled to a label such as a radiolabel or to a protein such as streptavidin or pro-drug converting enzymes for use in imaging or therapeutic applications. The antibody constructs may also be transduced into T cells to produce populations of T cells which target GD2-producing tumor cells.

Description

    BACKGROUND OF THE INVENTION
  • This application relates to single chain antibody constructs which specifically bind to the disialoganglioside G[0001] D2, and to the use of such constructs for targeted delivery of imaging agents or therapeutic agents to human neuroectodermal derived cancers.
  • Gangliosides are acidic glycosphingolipids found on the outer surface of most cell membranes.[0002] 1 Many tumors have abnormal glycolipid composition and structure. Disialoganglioside GD2 has been found in a wide spectrum of human tumors, including neuroblastoma, osteosarcomas and other soft tissue sarcomas, medulloblastomas, high grade astrocytomas, melanomas, and small cell lung cancer.2-4 Among glioblastoma multiforme and anaplastic astrocytoma, anti-GD2 demonstrated the most restrictive pattern when compared with anti-GD3 and anti-GM2 antibodies.5,6
  • Gangliosides are ideal targets for monoclonal antibodies (MAb) because of the high antigen density, lack of modulation, relative homogeneity in many tumors and the possibility of up-regulation by cytokines.[0003] 7 The only normal tissues with high ganglioside expression are neurons, and biodistribution studies have shown that MAb do not localize to the nontumorous brain or spinal cord because of the blood brain barrier. In contrast, in patients with primary or metastatic brain tumors, specific antibodies can localize preferentially to tumor tissues, but not to normal brain.8
  • Murine monoclonal antibodies have been prepared to ganglioside G[0004] D2 Using somatic cell hybridization, murine MAbs were produced against the ganglioside GD2.9 They were shown to react with disialoganglioside GD2, but not with GD3, GT1b, GD1b, GD1a, GM1, GM3 and GM4. When base-treatment step was omitted from the standard neuroblastoma ganglioside extraction procedure, immuno-thin-layer-chromatography (ITLC) using 3F8, 3G6 and other anti-GD2 MAbs revealed a new ganglioside band with Rf of 0.342, besides GD2 (Rf 0.183).4 Immunochemical analysis showed that this new neuroblastoma ganglioside contained alkali-sensitive O-acetylated sialic acid residues recognized by MAb D1.1.
  • Of 15 anti-G[0005] D2 MAbs studied, 13 reacted strongly with the novel ganglioside. 3F8 was chosen for our initial clinical studies because of its being an IgG3 and its strong binding in vitro to GD2. Based on the cDNA sequence and the anti-idiotype cross-reactivity, the antigen specificity and affinity of 3f8 and 3G6 were similar if not identical. We have chosen 3G6 for scFv development for ease of comparison with the other 14 MAbs which are IgM antibodies.
    TABLE 1
    SPECIFICITY OF ANTI-GD2 MAb.
    MAb GD2 GD3 “O-GD2 GD1b
    1A8 4+ +/− 1+
    1F9 4+ +/− 3+ 1+
    1H12 4+ +/− 3+ 1+
    2F7 3+ 1+
    3A7 3+
    3A10 4+ 1+ 2+ 1+
    3B4 4+ 3+ 1+
    3F8 4+ 4+
    3G6 4+ 2+
    4C11 4+ 3+ +/−
    5E11 4+ 4+
    5F4 4+ 1+ 2+ 1+
    5F11 2+ +/− 3+ 1+
    6E8 4+ 1+ 3+ 1+
    6H4 4+ +/− 3+ 1+
  • In order to determine the general applicability of the ganglioside G[0006] D2 as a target for immunotherapy, its expression in human cancers has been studied by imnmunostaining tumor specimens using these monoclonal antibodies. These anti-GD2 antibodies reacted with all the neuroblastoma surgical specimens tested to date in our laboratory. A recent update10 analyzed a series of 39 neuroblastomas. Staining of both primitive neuroblastic and differentiating ganglioneuromatous elements were seen, although tumor cell heterogeneity was noted in some. 23/39 tumors showed a more intense reactivity with MAb 3A7 than with 3F8, and this was particularly evident in the primitive neuroblastoma group. In a separate study, the expression of GD2 was analyzed in 67 solid tumors and normal tissues from children by using the antibody 3A7.11 GD2expression was found in 28 of 28 neuroblastomas, and was most abundant in stroma-poor tumors. Differentiating stroma-rich neuroblastomas, neuroblastic clusters, neurofibrils, and most ganglion-like cells were found to be GD2 positive, whereas Schwann's-cell stroma did not express GD2. In ganglioneuromas, only a few ganglion-like cells showed GD2, whereas all other structures were negative. Scattered foci of GD2 were also found in some non-neuronal tumors, such as rhabdomyosarcomas and osteosarcomas, but not in lymphomas, Askin tumors, or most Wilm's tumors. 3A7 was also found to react with retinoblastomas.12
  • Previous studies have shown that anti-G[0007] D2 antibodies reacted with the majority of osteosarcomas.13 Sixty freshly frozen human soft-tissue sarcomas were studied by avidin-biotin immunostaining using purified monoclonal antibodies 3F8 (anti-GD2) and R24 (anti-GD3).14 Ninety-three percent of the tumors tested by the immunohistochemical staining expressed GD2 and 88% expressed GD3. The intensity of expression varied among different histologic types. Liposarcoma, fibrosarcoma, malignant fibrous histiocytoma, leiomyosarcoma and spindle cell sarcoma reacted strongly with both antibodies. Embryonal rhabdomyo-sarcoma and synovial sarcoma demonstrated substantially weaker staining by either MAb. Ganglioside extraction and immuno-thin layer chromatography (ITLC) confirmed the identities of these gangliosides as GD2 and GD3 respectively
  • Among brain tumors, 3F8 and 3A7 have also shown excellent reactivities. Two separate studies were carried out: the first study in collaboration with Dr. Paul Zeltzer of Texas and the second with Dr. Ira Bergman (now Associate Professor of Neurology at the University of Pittsburgh) in our laboratory. In the first study, 12/15 medulloblastoma and 16/18 astrocytoma were positive, the majority staining homogeneously. In the second study, similar results were obtained. Medulloblastoma and a number of brain tumors reacted strongly with 3F8 and 3A7. The pattern of reactivity was generally homogeneous. For small cell lung cancer, all have reacted homogeneously in vitro using immunoperoxidase techniques. [0008]
  • Despite in vitro evidence for exquisite specificity of these antibodies for the ganglioside G[0009] D2 on neuroblastoma cells, a critical test of in vivo delivery is the actual amount of MAb uptake in the tumors. Biodistribution of 131I-anti-GD2 antibody was tested in preclinical experiments using athymic mice xenogratted with human neuroblastoma.
  • Between 8 to 50% injected dose of 131I-MAb/gm of tumor was found, with variability depending primarily on the size of the tumor.[0010] 15 There was no localization to GD2-negative tumors like Ewing's sarcoma. Pooled mouse IgG and an irrelevant MAb also did not localize to neuroblastoma xenografts. Both small tumors (50 mg) and large tumors (over 2 g) showed radiolocalization with this technique. Optimal tumor to normal tissue ratios were rapidly reached by 24 to 48 hours. There was no increased uptake in the reticuloendothelial system, and the MAb did not cross the intact blood-brain barrier. The efficacy of tumor targeting was then tested by imaging neuroblastoma patients with 131I-MAb. Radiolocalization was demonstrated in primary tumors of the mediastinum and abdomen, as well as metastatic disease in the lymph nodes, bone marrow and bone.16,17 The specificity was validated by tumor and marrow biopsies, as well as by CT/MRI and bone scans. A comparison with 131I-meta-iodobenzylguanidine (MIBG) suggested that 131I-MAb was twice as sensitive in detecting metastatic sites of disease. The tumor uptake in patients was 0.08% of the injected dose per gm (compared to 0.002% for MIBG). This high tumor uptake in vivo was a result of (1) the high density (5×106/cell) and homogeneity of the target antigen GD2, and (2) the lack of uptake in the reticuloendothelial system. A number of human cancers has been imaged using GD2 specific antibodies. These include small cell lung cancer,18 brain tumors,8 and both osteosarcomas13 and soft tissue sarcomas.19
  • A phase I study to test the biological toxicity of “cold” anti-G[0011] D2 was carried out in 1987 in 17 patients with metastatic neuroblastoma or melanoma. A subsequent phase II study was carried out in 16 patients with stage IV neuroblastoma. Acute self-limited toxicities of MAb treatment were severe pain requiring analgesics, fever, urticaria, hypertension, hypotension, anaphylactoid reactions of the respiratory tract, as well as significant decreases in blood counts and serum complement levels. There were no treatment related deaths. Among the 5 neuroblastoma patients who are still alive and well (19 mos, 3y, 5y, 5y, 6y respectively after MAb treatment), there are no acute or delayed neurological complications attributable to MAb therapy. Among the survivors, one patient had chemo-radiotherapy-resistant stage IVS neuroblastoma, and the other 4 had poor risk stage IV neuroblastoma diagnosed at more than one year of age (2 relapsed neuroblastoma and 2 with refractory neuroblastoma prior to antibody treatment).
  • More recently, a phase I study to determine the radiological toxicity was carried out. Twenty-three patients (11 M and 21 F, ranging from 0.3 to 24.2 years of age at diagnosis) with refractory neuroblastoma (22 stage IV, 1 stage IIIU), were treated with 131I-3F8 at 7 dose levels, namely 6, 8, 12, 16, 20, 24, and 28 mCi/kg. Radiation dose to the blood was calculated based on blood clearance; total body dose was based on total body clearance, and the tumor/organ dose on regions of interest calculations from serial gamma imagings. 21/23 patients were rescued with autologous bone marrow; one patient received GM-CSF alone, one died of progressive disease before marrow reinfusion. Marrow was infused when blood radioactivity decreased to <0.01 uCi/ml in the first 18 patients and to <1 uCi/ml in the last 4 patients. Acute toxicities of 131I-MAb treatment included pain (19/23) during the infusion, fever (19/23), hyperbilirubinemia (6/23), and diarrhea. All patients developed grade 4 myelosuppression with sepsis in 7/23 patients (5 fungal, 2 bacterial), disseminated zoster in 1, and pneumocystis in 1. Despite orally administered saturated solution of potassium iodide, 3 patients developed hypothyroidism. Subsequent 14 patients were treated with synthroid or Cytomel for thyroid protection. No other significant extramedullary toxicities have been encountered in patients followed past 20 months (50+, 40+, 30+, 26+, 23+, mos) from the time of 131I-MAb treatment. Fourteen patients have died 11 of disease and 3 from infections during the cytopenic period, and in 4 patients follow-up is still short. Responses were seen in both soft tissue masses and bone marrow. Average tumor dose was 150 rad/mCi/kg. We concluded that when 131I-MAb was administered intravenously (6-28 mCi/kg), significant toxicities were encountered, including myelosuppression and their infectious complications, pain, fever, as well as hypothyroidism. Autologous marrow rescue could reverse marrow aplasia and thyroid supplement was essential to prevent thyroid damage. Although severe extramedullary toxicities were not seen, improvement in the pharmacokinetics of the radioconjugates will reduce significantly the marrow toxicity. [0012]
  • To date, a total of >95 patients have been treated with antibody 3F8, and more than 120 imaging studies have been carried out on different ongoing protocols. Among pediatric patients, no neuropathy has been reported, either sensory or motor in nature. More than two thirds of these patients mounted HAMA response, mostly low titer and not persistent. There was no correlation of HAMA with toxicity. Nevertheless, in view of the neuropathy seen with other anti-G[0013] D2 antibodies 14.2a and 14.18 (similar in reactivity patterns to 3F8), we want to improve the specificity to reduce side effects. All of these clinical trials have been carried out using antibodies produced at Memorial Sloan-Kettering Cancer Center using guidelines of the Office of Biologics Research and Review Center for Drugs and Biologics, Food and Drug Administration. For quality assurance, hybrdoma 3F8 was found to be negative for adventitious agents by MAP, S+L−, and XC plaque assays, as well as negative for reverse transcriptase. MAP testing included screening for murine leukovirus, LCM virus isolation by intracerebral inoculation, murine saliva gland virus, mouse thymic virus, EDIM and LDH virus isolations. Purified antibody (e.g. 3F8) had to pass MAP and sterility testing (bacteria, mycoplasma, and fungal cultures), rabbit pyrogen testing, as well as safety testing in mice and guinea pigs. Conjugation to 131I by the chloramine-T method was supervised by Dr. Ronald Finn and Dr. Steven Larson in the Department of Nuclear Medicine. Specific activity of iodine-131 was >600 mCi/ug iodide. Radiolabeled antibody 3F8 must have >50% binding by in vitro antigen binding assay, >95% TCA precipitable and <3% free iodine by radio-thin layer chromatography. Periodic testing of radiolabeled antibodies was performed to ensure sterility as well as the absence of pyrogen.
  • Although the wide expression of G[0014] D2 in human neuroectodermal-derived cancers (melanoma, small cell lung cancer, neuroblastoma, brain tumors, sarcoma, HTLV-1 leukemia, retinoblastoma and osteosarcoma) and the preliminary clinical studies of monoclonal antibodies to GD2 in radioimmuno-scintigraphy and radioimmunotherapy have been encouraging, further optimization of antibodies for binding to GD2 would be desirable. It is an object of the present invention to provide such optimized antibodies and the DNA sequences coding therefore.
  • It is a further object of the invention to provide methods of using the optimized antibodies and DNA sequences in diagnostic assays and therapeutic techniques. [0015]
  • SUMMARY OF THE INVENTION
  • The antibodies of the present invention are recombinant antibody constructs comprising the variable regions of the heavy and light chains of anti-G[0016] D2 antibodies. These antibody constructs may be coupled to a label such as a radiolabel or to a protein such as streptavidin or pro-drug converting enzymes for use in imaging or therapeutic applications. The antibody constructs may also be transduced into T cells to produce populations of T cells which target GD2-producing tumor cells.
  • BRIEF DESCRIPTION OF THE DRAWING
  • FIG. 1 shows two alternative strategies for the use of the antibody constructs of the present invention in therapy.[0017]
  • DETAILED DESCRIPTION OF THE INVENTION
  • This invention relates to optimized antibodies to G[0018] D2, to DNA encoding such antibodies, and to the use of the antibodies and DNA in diagnostic assays and therapy. The antibodies of the invention are antibody constructs comprising the variable regions of the heavy and light chains of anti-GD2 antibodies as a single chain Fv fragment. Single-chain Fv fragments (scFv) offer some of the best opportunities to achieve these results. ScFv technology utilizes molecular biology methods to reduce antibodies to the minimal-required-unit of heavy and light chain variable regions tethered by a peptide linker which can be designed with versatile side chains for radioconjugation.
  • The anti-G[0019] D2 scFvs shown in Tables 2 and 3 have been prepared using the methods described in the examples. For 5F11 the orientation VH-VL is used for the scFv. For 3G6 the orientation VL-VH is necessary. Since the cDNA sequences of 3G6 and 3F8 are similar, we expect the binding properties of both of these antibodies to be very similar if not identical. Since our cloning strategies have been most successful with 3G6, this has been the focus of our research instead of 3F8. Histidine-tag (His5) is inserted for ease of purification by Ni-column and Myc-tag is inserted to facilitate detection. Myc-tag and E-tag can be genetically removed if necessary for future clinical studies. These svFv variants were constructed to test the effect of (1) the detection-tag (E-tag or Myc-tag), (2) the presence of His5-tag, and (3) the position of His5-tag (carboxyl-end-terminal versus internal) on (a) antibody specificity, (b) affinity, and (c) ease of purification.
  • In order to increase the avidity of the scFv, we have synthesized two scFv variants: (1) Cysteine residue at the carboxyl terminal of the scFv for dimerization (5FpoMCH of Table 2 and 3GpoMCH of Table 3): Free sulhydryl groups are blocked by acetylation and the monomer separated from the dimer by size-exclusion chromatography FPLC on Sephadex HR75 (PharTnacia). (2) Streptavidin at the carboxyl end for dimerization and tetramerization (5FpoStMCH of table 1 and 3GpoStMCH of table 2): Streptavidin is a homo-tetrameric protein that binds one biotin molecule per subunit with a very high affinity (Kd=4×10−14). scFv-strep fusion proteins are expected to form tetramers with both antigen- and biotin-binding activity They are expected to be stable over a wide range of pH and range of physiologic temperatures. [0020]
    TABLE 2
    scFv
    Expression 5FpcHE 5FphM 5FphHM 5FpoMCH 5FpoStMCH*
    Vector pCantab pHEN pHEN pOPE pOPE
    Tag
    E-tag +
    Myc-tag + + + +
    His-tag + + + +
    Bacteria Host
    phagemid XL1-blue XL1-blue XL1-blue
    phage hb2151 hb2151 hb2151 JM109 JM109
    Restriction sites:
    5′ Ncol + +
    5′ PvuII + +
    5′ Sfil +
    2′ NOT1 + + + + +
    Binding*
    GD2 elisa + + + + +
    ITLC + + + + +
    Western blot** + + + + +
    purification nd nd nd + +
  • [0021]
    TABLE 3
    scFv
    Expression 3 GphM 3 GphHM 3 GPoMCH 3 GpoStMCH*
    Vector pHEN pHEN pOPE pOPE
    Tag
    E-tag −/
    Myc-tag + + + +
    His-tag + + +
    Bacteria Host
    phagemid XL1-blue XL1-blue
    phage hb2151 hb2151 JM109 JM109
    Restriction sites:
    5′ Ncol + + + +
    5′ Sfil
    3′ NOTl + + + +
    Binding**
    GD2 elisa + + + +
    ITLC nd nd + nd
    anti-id nd nd + nd
    Western blot** + + + +
    Purification nd nd + nd
  • The 5F11-scFv, 3G6-scFv, 5F11-scFv-streptavidin, 3G6-scFv-streptavidin DNA sequences are shown below, with the linker sequences between the scFv and the streptavidin shown in lower case letters. [0022]
    SF11-scFv
    CAGGTGAAACTGCAGCAGTCAGGACCTGAACTGGTGNAGCCTGGGGCTTCAG
    TGAAGATATCCTGCAAGACTTCTGGANACAAATTCACTGAATACACCATGCAC
    TGGGTGAAGCAGAGCCATGGAAAGAGCCTTGAGTGGATTGGAGGTATTAAT
    CCTAACAATGGTGGTACTAACTACAAGCAGAAGTTCAAGGGCAAGGCCACAT
    TGACTGTAGACAAGTCCTCCAGCACAGCCTACATGGAGCTCCGCAGCCTGAC
    ATCTGAGGATTCTGCAGTCTATTACTGTGCAAGAGATACTACGGTCCCGTTTG
    CTTACTGGGTCCAAGGGACCACGGTCACCGTCTCCTCAGGTGGAGGCGGTTC
    AGGCGGAGGTGGCTCTGGCGGTGGCGGATCGGACATCGAGCTCACTCAGTCT
    CCAGCAATCATGTCTGCATCTCCAGGGGAGAAGGTCACCATGACCTGCAGTG
    GCAGCTCAAGTATAAGTTACATGCACTGGTACCAGCAGAAGCCTGTCACCTCC
    CCCAAAAGATGGATTTATGACACATCCAAACTGGCTTCTGGAGTCCCTGCTCG
    CTTCAGTGGCAGTGGGTCTGGGACCTCTTATTCTCTCACAATCAGCAGCATGG
    AGGCTGTAGATGCTGCCACTTATTACTGCCATCAGCGGAGTAGTTACCCGCTC
    ACGTTCGGTGCTGGGACACAGTTGGAAATAAAACGG
    3G6-scFv
    AGTATTGTGATGACCCAGACTCCCAAATTCCTGCTTGTATCAGCAGGAGACAG
    GGTTACCATAACCTGCAAGGCCAGTCAGAGTGTGAGTAATGATGTGGCTTGG
    TACCAACAGAAGCCAGGGCAGTCTCCGAAACTGCTGATATACTCTGCATCCAA
    TCGCTACACTGGAGTCCCTGATCGCTTCACTGGCAGTGGATATGGGACGGAT
    TTCACTTTCACCATCAGCACTGTGCAGGCTGAAGACCTGGCAGTTTATTTCTG
    TCAGCAGGATTATAGCTCGCTCGGAGGGGGGACCAAGCTGGAAATAAAAGG
    TGGAGGCGGTTCAGGCGGAGGTGGCTCTGGCGGTGGCGGATCGCAGGTGCA
    GGTGAAGGAGTCAGGACCTGGCCTGGTGGCGCCCTCACAGAGCCTGTCCATC
    ACTTGCACTGTCTCTGGGTTTTCATTAACCAATTATGGTGTACACTGGGTTCG
    CCAGCCTCCAGGAAAGGGTCTGGAGTGGCTGGGAGTAATATGGGCTGGTGG
    AAGCACAAATTATAATTCGGCTCTTATGTCCAGACTGAGCATCAGCAAGGACA
    ACTCCAAGAGCCAAGTTTTCTTAAAAATGAACAGTCTGCAAACTGATGACACA
    GCCATGTACTACTGTGCCAGTCGGGGGGGTAACTACGGCTATGCTTTGGACT
    ACTGGGGTCAAGGAACCTCAGTCACCGTCTCCTCA
    5F11-scFv-Streptavidin
    CAGGTGAAACTGCAGCAGTCAGGACCTGAACTGGTGNAGCCTGGGGCTTCAG
    TGAAGATATCCTGCAAGACTTCTGGANACAAATTCACTGAATACACCATGCAC
    TGGGTGAAGCAGAGCCATGGAAAGAGCCTTGAGTGGATTGGAGGTATTAAT
    CCTAACAATGGTGGTACTAACTACAAGCAGAAGTTCAAGGGCAAGGCCACAT
    TGACTGTAGACAAGTCCTCCAGCACAGCCTACATGGAGCTCCGCAGCCTGAC
    ATCTGAGGATTCTGCAGTCTATTACTGTGCAAGAGATACTACGGTCCCGTTTG
    CTTACTGGGTCCAAGGGACCACGGTCACCGTCTCCTCAGGTGGAGGCGGTTC
    AGGCGGAGGTGGCTCTGGCGGTGGCGGATCGGACATCGAGCTCACTCAGTCT
    CCAGCAATCATGTCTGCATCTCCAGGGGAGAAGGTCACCATGACCTGCAGTG
    GCAGCTCAAGTATAAGTTACATGCACTGGTACCAGCAGAAGCCTGTCACCTCC
    CCCAAAAGATGGATTTATGACACATCCAAACTGGCTTCTGGAGTCCCTGCTCG
    CTTCAGTGGCAGTGGGTCTGGGACCTCTTATTCTCTCACAATCAGCAGCATGG
    AGGCTGTAGATGCTGCCACTTATTACTGCCATCAGCGGAGTAGTTACCCGCTC
    ACGTTCGGTGCTGGGACACAGTTGGAAATAAAACGGgcggccgctggatccggtgctgct
    GAAGCAGGTATCACCGGCACCTGGTACAACCAGCTCGGCTCGACCTTCATCGT
    GACCGCGGGCGCCGACGGCGCCCTGACCGGAACCTACGAGTCGGCCGTCGG
    CAACGCCGAGAGCCGCTACGTCCTGACCGGTCGTTACGACAGCGCCCCGGCC
    ACCGACGGCAGCGGCACCGCCCTCGGTTGGACGGTGGCCTGGAAGAATAACT
    ACCGCAACGCCCACTCCGCGACCACGTGGAGCGGCCAGTACGTCGGCGGCGC
    CGAGGCGAGGATCAACACCCAGTGGCTGCTGACCTCCGGCACAACCGAGGCC
    AACGCCTGGAAGTCCACGCTGGTCGGCCACGACACCTTCACCAAGGTGAAGC
    CATACATCACCATCATCAT
    3G6-scFv-streptavidin
    AGTATTGTGATGACCCAGACTCCCAAATTCCTGCTTGTATCAGCAGGAGACAG
    GGTTACCATAACCTGCAAGGCCAGTCAGAGTGTGAGTAATGATGTGGCTTGG
    TACCAACAGAAGCCAGGGCAGTCTCCGAAACTGCTGATATACTCTGCATCCAA
    TCGCTACACTGGAGTCCCTGATCGCTTCACTGGCAGTGGATATGGGACGGAT
    TTCACTTTCACCATCAGCACTGTGCAGGCTGAAGACCTGGCAGTTTATTTCTG
    TCAGCAGGATTATAGCTCGCTCGGAGGGGGGACCAAGCTGGAAATAAAAGG
    TGGAGGCGGTTCAGGCGGAGGTGGCTCTGGCGGTGGCGGATCGCAGGTGCA
    GGTGAAGGAGTCAGGACCTGGCCTGGTGGCGCCCTCACAGAGCCTGTCCATC
    ACTTGCACTGTCTCTGGGTTTTCATTAACCAATTATGGTGTACACTGGGTTCG
    CCAGCCTCCAGGAAAGGGTCTGGAGTGGCTGGGAGTAATATGGGCTGGTGG
    AAGCACAAATTATAATTCGGCTCTTATGTCCAGACTGAGCATCAGCAAGGACA
    ACTCCAAGAGCCAAGTTTTCTTAAAAATGAACAGTCTGCAAACTGATGACACA
    GCCATGTACTACTGTGCCAGTCGGGGGGGTAACTACGGCTATGCTTTGGACT
    ACTGGGGTCAAGGAACCTCAGTCACCGTCTCCTCAgcggccgctggatccggtgctgctGAA
    GCAGGTATCACCGGCACCTGGTACAACCAGCTCGGCTCGACCTTCATCGTGAC
    CGCGGGCGCCGACGGCGCCCTGACCGGAACCTACGAGTCGGCCGTCGGCAAC
    GCCGAGAGCCGCTACGTCCTGACCGGTCGTTACGACAGCGCCCCGGCCACCG
    ACGGCAGCGGCACCGCCCTCGGTTGGACGGTGGCCTGGAAGAATAACTACCG
    CAACGCCCACTCCGCGACCACGTGGAGCGGCCAGTACGTCGGCGGCGCCGAG
    GCGAGGATCAACACCCAGTGGCTGCTGACCTCCGGCACAACCGAGGCCAACG
    CCTGGAAGTCCACGCTGGTCGGCCACGACACCTTCACCAAGGTGAAGCCGTC
    CGCCGCCTCCGGATCCGAACAAAAGCTGATCTCAGAAGAAGATCTATGCATA
    CATCACCATATCAT
  • All these single chains have strings of 5 histidine residues inserted at the carboxyl end for purification on the Nickel-column using FPLC. Washing was done at 10 mM Imidazole and scFv peak elution at 500 mM Imidazole. Further purification of scFv can be accomplished by size-exclusion using Sephadex HR75 and superose 6 (hi-resolution). scFv can be fuirther affinity purified by myc-affinity chromatography. 9E10 is a hydridoma (ATCC) that secretes the antibody specific for the myc-tag. From ascites the 9E10 IgG1 antibody is purified by protein-G affinity chromatography and used for chemical coupling to sepharose 4B. [0023]
  • Recombinant antibody constructs according to the invention can be coupled to metal labels such as [0024] 99mTc for use in diagnostic imaging of GD2 expressing cells. For example, technetium can be chelated to the antibody construct via a heterobifuinctional linker such as succinimidyl-6-hydrazinonicotinate hydrochloride (SHNH). SHNH is used to synthesize hydrazino-modified antibody. At a conjugation ratio of 1.8:1 of SHNH to antibody, immunoreactivity was preserved. Tc(V) precursors coupled readily and conveniently to the SHNH-modified protein to yield the desired 99mTc-radiolabeled conjugate. 99mTc-3F8 localized rapidly and successfully to GD2-positive xenografts. SHNH-modified scFvs can be synthesized for conjugation to 99mTc using the techniques described in Schwartz et al., Bioconjugate Chem 2:333-336 (1991), which is incorporated herein by reference.
  • Metal chelation to scFv can also be accomplished via the streptavidin protein. The rationale of pretargeting using scFv-streptavidin fusion proteins in radioimmunotherapy are 5-fold: (a) Large amounts of scFv can be used to saturate G[0025] D2 sites in vivo, without the accompanying blood and tissue toxicity from radioisotope, (b) radiolabel is injected at the time when the tumor-nontumor ratio of scFv is maximal, (c) a radiolabeled ligand is chosen such that it binds with high affinity (e.g. 111I-biotin binding to streptavidin) with fast blood-clearance, (d) a ligand construction where the isotope can be modified to optimize microdosimetry (e.g. SHNH-biotin) (e) the scFv-streptavidin is a homo-tetramer, as such the antigen binding avidity is greatly amplified especially for high-density antigens (e.g. GD2 on neuroblastoma). scFv-strep fusion proteins for both 5F11 and 3G6 have been made and purified. Both in vitro and in vivo studies are being carried out to test the concept of pretargeting, where scFv-strep is first allowed to bind (or target) to GD2-positive tumors through the scFv. After the excess or nonbinding scFv-strep is washed off (or cleared from the body), a radiolabeled-biotin ligand is allowed to bind to the streptavidin moiety. Different radiolabels can be coupled to biotin using SHNH (99mTc) or DTPA (111In or yttrium).
  • scFv coupled to technetium provides a safe, camera-ready isotope, with fast-decay and therefore easy disposal. [0026] 99m-technetium is optimal for imaging studies. Other isotopes can also be used, including a positron-emitting technetium for PET imaging. Through the same side chain SHNH, rhenium (a therapeutic beta-emitting radionuclide) may also be attached
  • The scFv and scFv-streptavidin of the invention are also useful in a number of therapeutic applications, which is turn form aspects of the present invention In general, these approaches involve administration of scFV coupled to a therapeutic or pre-therapeutic moiety. For example, as shown in FIG. 1, ScFv-streptavidin (streptavidin being the pre-therapeutic moiety) is introduced into an organism suspected of harboring G[0027] D2 expressing cells, where it binds to any such cells present. A therapeutic agent (X) bound to biotin is then introduced. Binding of the biotin the streptavidin results in localization of the chemotherapeutic agent X at the site of the GD2 producing cells. Other pre-therapeutic moieties include pro-drug converting enzymes. Directly therapeutic moieties such as toxins can also be used.
  • A second approach, also illustrated in FIG. 1, utilizes a vector encoding ScFv is transduced into primary human lymphocytes (preferably along with a suicide gene such a HSV-TK). The transduced lymphocytes now recognize and target G[0028] D2, resulting in an immune response to the GD2-producing cells.
  • The scFv or scFv-streptavidin can be incorporated in a fusion protein with therapeutic agents such as toxins or pro-drug converting enzymes, can be incorporated in a fusion protein with CD8 to facilitate the formation of G[0029] D2-targeted lymphocytes, or can be coupled to viral coat proteins superantigen (SEA) to facilitate targeting of GD2 producing cells,
  • Direct conjugation of scfv or scFv-streptavidin to toxin replaces the cell-binding domain of natural toxins with the scFv, which serves as a tumor binding domain specific to G[0030] D2 expressing cells. ScFv-ricin-A-chain and scFv-pseudomonas toxin have been successfully constructed for other scFv. This coupling is advantageously performed at the DNA level, not at the protein level. For example, when the fusion protein of the heavy chain, the light chain and the linker is created by overlap PCR extension, a DNA coding for the toxin can also included, and then expressed along with the scFv.
  • scFv and scFv-streptavidin can also be usefully combined in a fusion protein with CD8. scFv-CD8 constructs can be transfected through retroviral vector into human and mouse lymphocytes. Since these scFv are permanently integrated into the cellular genome, these lymphocytes express scFv on their cell surface and through the CD8 cytoplasmic domain become activated upon antigen binding. scFv facilitates the homing of these cells to tumor sites, thus being effective in promoting both the localization and killing of tumors. With a suicide gene, thymidine kinase, also transfected, these cells can now be turned on and off as needed. [0031]
  • scFv-enzyme and scFv-enzyme-streptavidin conjugates can be used to provide targeted drug therapy using a technique known as ADEPT (antibody directed enzyme prodrug-therapy). Suitable enzymes for this technique include carboxypeptidase G2, alkaline phosphatase, and β-Lactamase. A prodrug derivative (e.g. cephalosporin derivative of doxo20) becomes activated to the active agent by the enzyme (beta-lactamase) targeted to the tumor by the scFv. Thus tumor cells are exposed to a high local concentration (up to 10-fold higher than blood/tissue levels) of specific chemotherapeutic agents. [0032]
  • Integration of scFv (with or without streptavidin) into viral coat proteins can be used to retarget these viruses in vivo. These viruses include adenovirus, retrovirus and herpes virus. [0033]
  • Superantigen (SEA) can stimulate T cells without the requirement of MHC.21 ScFv-SEA and scFv-streptavidin-SEA can target T cells to lyse antigen-positive MHC-class II-negative human tumor cells. SEA has been cloned (Betley et al: J. Bacteriology 170: 34-41, 1988) and the cDNA is available for making fusion proteins. [0034]
  • EXAMPLE 1
  • 5F11 hybridoma cells were processed for mRNA using a commercially available kit (Quick Prep Micro MRNA Purification, pharmacia Biotech) following the procedures outlined by the manufacturer. Briefly, hybridoma cells were cultured in ROPMI-1640 medium supplemented with 10% calf serum, 2 mmol/L L-glutamine (Sigma), 100 U/L penicillin and 100 ug/ml streptomycin sulfate (Sigma). The cell cultures were maintained at 37° C. under a water-saturated atmosphere of 5% CO[0035] 2. 5×106 cells were pelleted by centrifugation at 800 g and washed once with RNase-free phosphate buffered saline (pH 7.4), The recentrifuged cells were ysed directly in the extraction buffer. Poly(A)+RNA was purified by a single fractionation of oligo(dT)-cellulose and then eluted with elution buffer. The mRNA sample was precipitated for 1 hour at 100 ug glycogen, 40 ul of 2M potassium acetate and 1 ml absolute ethanol at −20° C. The nucleic acids were recovered by centrifugation at 10,000 g for 30 minutes, The sample was evaporated until dry and dissolved in 20 ul RNase-free water.
  • The mRNA preparation was used in the construction of the 5F11 scFv gene using the Mouse ScFv Module/Recombinant Phage Antibody System (Pharmacia Biotech). 5 ul of the mRNA preparation was reverse transcribed in a total volume of 11 ul of reaction mixture and 1 ul DTT solution for 1 hour at 37° C. For PCR amplification of immunoglobulin variable region, light primer mix and the heavy primer set were added respectively to generate quantities of the light (325 bp(and heavy (340 bp) chains. Following an initial 5 minute dwell at 95° C. , 5 U Ampli Taq DNA polymerase (Perkin Elmer) was added. The PCR cycle consisted of a 1 min denaturation step at 94° C. , a 2 min. annealing step at 55° C. and a 2 min extension step at 72° C. After 30 cycles of amplification, PCR derived fragments were purified using glassmilk beads (Bio 101 Co.) and evaluated by electrophoresis on 1/5% agarose gel in TAE buffer with ethidium bromide visiualization. For the assembly and fill-in reaction, both purified heavy chain and light chain fragments were added to ab appropriate PCR mixture containing linker-primer, dNTPs, PCR buffer and Ampli Taq DNA polymerase. Denaturation was performed at 94° C. for 1 minute, followed by a 4 minute annealing reaction at 63° C. The heavy and light DNA were joined into a single chain with linker DNA after 7 thermocycles. Using this single chain DNA as a template and restriction site primers (RS primers) containing either SfiI or NotI restriction sites, secondary PCR amplification was carried out for 30 cycles to amplify the ScFv DNA and add the restriction sites. This introduced the SfiI restrictions site at the 5′-end of the heavy chain and the NotI restriction site at the 3′-end of the light chain. Amplified ScFv DNA was then purified by glassmilk beads and digested with SfiI and NotI. [0036]
  • Purified scFv DNA was inserted into the pCantab 5e vector (Pharmacia Biotech) by ligation as SfiI/NotI sites in the vector. Competent [0037] E. coli XL 1-Blue cells were transformed with pCantab 5E phagemid containing the ScFv DNA following the method outlined in Stratagene protocols. For rescue of a recombinant phage antibody library, the helper phage M13 K07 was added.
  • Antibody-producing recombinant phage were selected by panning using the method of Ditzel, PNAS USA 91: 3710-3714 (1994) with slight modifications. 20 ul of GD2 (1 ul/ml) dissolved in ethanol were directly coated on a 96-well polystyrene plate and dried at rom temperature. Then 100 ul of the supernatant containing the phage library was added to each well and incubated for 2 hours. The plate was then washed 10 times with PBS containing 0 05% BSA to remove nonspecifically bound phage. Antibody-positive recombinant phage captured by the G[0038] D2 antigen was eluted with 0.1 M HCl (pH 2.2 with solid glycine and 0.1% BSA) and neutralized with 2M Tris solution. Selected phage was then re-panned for two additional cycles to further enrich the GD2-binding recombinant phages.
  • The selected phage was used to reinfect [0039] E coli XL1-Blue cells. Clones were grown in 2XYT medium containing ampicillin (100 ug/ml) and 1% glucose at 30° C. until an OD600 of 0.5 was obtained. Expression of ScFv antibody was induced by changing to a medium containing 100 uM IPTG and incubating overnight at 30° C. The supernatant obtained from the medium by centrifugation was directly added to a plate coated with GD2. The pellet was resuspended in PBD containing 1 mM EDTA and incubated on ice for 10 minutes. The periplasmic soluble antibody was collected by centrifugation again and added to the plate. After incubating at 37° C. for 32 hours, anti-E Tag antibody (Pharmacia Biotech) was used to specifically screen the binding of the ScFv fragment.
  • For construction of the 5FpoStMCH vector which contains the 5F11-scFV-streptavidin plasmid DNA, plasmid DNA from the 5F11-scFv in pCantab 5E vector (Pharmacia Biotech) was purified and amplified by PCR using two specially designed primers S6 and 318s. S6 contains a NotI restriction site and 318s contains a PvuII restriction site so that amplified DNA can be restriction digested and inserted in the pSTE vector (Dr. Dubel, German Cancer Center). The resulting vector 5FpoStMCH is the 5F11-scFv-streptavidin construct. The streptavidin was digested with BamHI, leaving the scFV 5FpoMCH. [0040]
  • EXAMPLE 2
  • Supernatant, periplasmic extract and cell extract from positive clones were fractionated on unreduced SDS-PAGE 12% SDS-polyacrylamide slab gels and buffer prepared according to Laemmli (1970). Electrophoresis was performed at 100V for 45 min. After completion of the run, western blot analysis was carried out as described by Towbin (1979). The nitrocellulose membranes were blocked by 1% nonfat milk in TBS solution for 1 hour and incubated with anti-E Tag antibody for 1 hour at room temperature. After incubating with HRP-conjugated goat anti-mouse Ig (Fisher Co.), the membrane was detected by ECL System (Amersham). The results showed a protein band with an apparent molecular mass of 31KD using anti-E Tag antibody which recognizes the sequence GAPVPVPDPLEPR. The same protein was not detected in control cells nor in cells without IPTG treatment to induce expression of the scFV. [0041]
  • EXAMPLE 3
  • Immunostaining thin layer chromatography was performed under conditions similar to those described by Tai et al (1987) GD2, GD3, GD1a, GD1b, GM2, GT1b and GL1000 were dissolved in ethanol and spotted on an HPTLC plate. The supernatant from ScFv 5F11 clone and 5F11 hybridoma cells were incubated with the spread plate. Immunostaining was visualized with the use of o-phenylenediamine dihydrochloride (Sigma). GD2 antigen was detected by both the ScFv supernatant and the 5F11 Mab, which appeared similar in specificity. Cross-reactions of the ScFv antibody with other glycolipids was not detected. [0042]
  • EXAMPLE 4
  • In constructing 3G6-scFv, the orientation VH-VL did not produce a functional scFV. Therefore the orientation VL-VH was used. cDNA of VH and VL of 3G6 hybridoma were linked through a custom built linker and inserted into the pHEN vector (DR. Greg Winter). NcoI and NotI restriction sites were built into the VH and VL linkers so that the scFV can be digested with these enzymes for insertion in the pSTE vector. Clone 7 was chosen and called 3GpoStMCH. Digestion of the streptavidin position of the gene left behind 3G6-scFv, now called 3GpoMCH. [0043]
  • The following references are cited above, and are incorporated herein by reference. [0044]
  • 1. Rodden F A, Wiegandt H, Bauer B L: Gangliosides: the relevance of current research to neurosurgery. J Neurosurg 74:606-619, 1991 [0045]
  • 2. Berra B, Gaini S M, Riboni L: Correlation between ganglioside distribution and histological grading of human astrocytoma. Int J Cancer 36:363-366, 1985 [0046]
  • 3. Traylor T D, Hogan E L: Gangliosides of human cerebral astrocytomas. J Neurochem 34:126-131, 1980 [0047]
  • 4. Ye J N, Cheung N K V: A novel O-acetylated ganglioside detected by anti-G[0048] D2 monoclonal antibodies. Int J Cancer 50-197-201, 1992
  • 5. Wikstrand C J, Fredman P, Svennerholm L, et al: Expression of gangliosides GM2, G[0049] D2, GD3, 3′-sioLM1, and 3′,6′ isoLD1 in CNS malignancies as defined by epitope-characterized monoclonal antibodies (Mabs). 9th International Conference on Brain Tumors Research and Therapy 1991 (abstract)
  • 6. Longee D C, Wikstrand C J, Mansson J E, et al: Disialoganglioside G[0050] D2 in human neuroectodermal tumor cell lines and gliomas. Acta-Neuropathology (Berl) 82:45-54, 1991
  • 7. Hoon D S, Banez M, Okun E, et al: Modulation of human melanoma cells by interleukin-4 and in combination with gamma-interferon or alpha-tumor necrosis factor. Cancer Res 51:2002-2008, 1991 [0051]
  • 8. Arbit E, Yeh S J, Cheung N K, Larson S M: Quantitative Immunoimaging of gliomas in humans with anti-ganglioside monoclonal antibodies. J Neurosurg 76:399a, 1991 [0052]
  • 9. Saito M, Yu R K, Cheung N K V: Ganglioside G[0053] D2 specificity of monoclonal antibodies to human neuroblastoma cell. Biochem Biophys Res Comm 127:1-4, 1985
  • 10. Lammie G A, Cheung N K V, Gerald W, et al: Ganglioside G[0054] D2 expression in the human nervous system and in neuroblastomas—an immunohistochemical study. Int J Oncol 3:909-915, 1993
  • 11. Sariola H, Terava H, Rapola J, Saarinen U M: Cell-Surface Ganglioside G[0055] D2 in the Immunohistochemical Detection and Differential Diagnosis of Neuroblastoma. AJCP 96:248-252, 1991
  • 12. Saarinen U M, Sariola H, Hovi L: Recurrent Disseminated Retinoblastoma Treated by High-dose Chemotherapy, Total Body Irradiation, and Autologous Bone Marrow Rescue. Am J Pediatr Hematol/Oncol 13:315-319, 1991 [0056]
  • 13. Heiner J, Miraldi F D, Kallick S, et al: In vivo targeting of G[0057] D2 specific monoclonal antibody in human osteogenic sarcoma xenografts. Cancer Res 47:5377-5381, 1987
  • 14. Chang H R, Cordon-Cardo C, Houghton A N, et al: Expression of disialogangliosides G[0058] D2 and GD3 by human soft tissue sarcomas. Cancer 70:633-638, 1992
  • 15. Cheung N K, Neely J E, Landmeier B, et al: Targeting of ganglioside G[0059] D2 monoclonal antibody to neuroblastoma. J Nuci Med 28:1577-1583, 1987
  • 16. Yeh S D, Larson S M, Burch L, et at: Radioimmunodetection of neuroblastoma with iodine-131-3F8: Correlation with biopsy, iodine-131-Metaiodobenzylguanidine (MIBG) and standard diagnostic modalities. J Nucl Med 32:769-776, 1991 [0060]
  • 17. Miraldi F D, Nelson A D, Kraly C, et al: Diagnostic imaging of human neuroblastoma with radiolabeled antibody. Radiology 161:413-418, 1986 [0061]
  • 18 Grant S C, Kostakoglu L, Kris M G, et al: Imaging of small cell lung carcinoma with the monoclonal antibody 3F8. Proc Am Soc Clin Oncol 10:265, 1991 (abstract) [0062]
  • 19. Yeh S D J, Casper E S, Cheung N K V, et al: Radioimmunoimaging of soft-tissue sarcoma with an anti-ganglioside monoclonal antibody 3F8. 5th Asia & Oceania Cong of Nucl Med & Biol Proceedings: 104, 1992 [0063]
  • 20. Svenson H P, Vrudhula V M, Ernswiler J E, et al: In Vitro and In Vivo Activities of a Doxorubicin Prodrug in Combination with Monoclonal Antibody β-Lactamase Conjugates. Cancer Res 55:2357-65, 1995 [0064]
  • 21. Dohlsten M, Abrahmsen L, Bjork P, et al: Monoclonal antibody-superantigen fission proteins: Tumor-specific agents for T-cell-based tumor therapy. Proc Natl Acad Sci (USA) 91:8945-8949, 1994 [0065]
  • 22. Dhingra K, Fritsch H. Murray J L, et at: Phase I Clinical and Pharmacological Study of Suppression of Human Antimouse Antibody Response to Monoclonal antibody L6 by Deosxysspergualin. Cancer Res 55:3060-67, 1995 [0066]
  • 23. Wnag C-Y, Huang L: p-H-sensitive immunoliposomes mediate target-cell-specific delivery and controlled expression of a foreign gene in mouse. Proc Natl Acad Sci (USA) 84:7851-55, 1987 [0067]
  • 24. Vieweg J, Boczkowski D, Roberson K M, et at: Efficient Gene Transfer with Adeno-associated Virus-based Plasmids Complexed to Cationic Liposomes for Gene Therapy of Human Prostate Cancer. Cancer Res 55:2366-2372, 1995 [0068]
  • 25. Lorimer I A J, Wikstrand C J, Batra S K, et al: Immunotixins That Target an Oncogenic Mutant Epidermal Growth Factor Receptor Expressed in Human Tumors. Clin Can Res 1:859-64, 1995 [0069]

Claims (16)

1. A recombinant single chain polynucleotide comprising a region encoding the variable region of the light chain of an anti-GD2 antibody linked to a region encoding the variable region of the heavy chain of an anti-GD2 antibody.
2. The recombinant polynucleotide of claim 1, further comprising a region encoding an additional protein.
3. The recombinant polynucleotide of claim 1, wherein the additional protein is streptavidin.
4. The recombinant polynucleotide of claim 1, wherein the additional protein is a drug-converting enzyme.
5. A recombinant single chain peptide comprising the variable region of the light chain of an anti-GD2 antibody linked to the variable region of the heavy chain of an anti-GD2 antibody.
6. The peptide according to claim 5, wherein the peptide is labeled with a radiolabel.
7. The peptide according to claim 6, wherein the radiolabel is 99mTc.
8. The peptide according to claim 5, wherein the peptide further comprises a drug-converting enzyme.
9. The peptide according to claim 5, wherein the peptide further comprises streptavidin.
10. The peptide according to claim 5, wherein the peptide further comprises CD8.
11. T cells expressing a recombinant single chain peptide comprising the variable region of the light chain of an anti-GD2 antibody linked to the variable region of the heavy chain of an anti-GD2 antibody.
12. A method for assaying for the presence of cells expressing GD2 in tissue comprising combining the tissue with a recombinant single chain peptide comprising the variable region of the light chain of an anti-GD2 antibody linked to the variable region of the heavy chain of an anti-GD2 antibody and a detectable label.
13. A method for targeted delivery of a therapeutic agent to cells expressing GD2 in tissue comprising combining the tissue with a recombinant single chain peptide comprising the variable region of the light chain of an anti-GD2 antibody linked to the variable region of the heavy chain of an anti-GD2 antibody and a therapeutic or pre-therapeutic moiety.
14. The method according to claim 13, wherein the pre-therapeutic moiety is a pro-drug converting enzyme.
15. The method according to claim 13, wherein the pre-therapeutic moiety is streptavidin.
16. The method according to claim 13 wherein the therapeutic moiety is a toxin.
US10/075,947 1996-03-20 2002-02-13 Single Chain Fv constructs of anti-ganglioside GD2 antibodies Abandoned US20030147808A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/075,947 US20030147808A1 (en) 1996-03-20 2002-02-13 Single Chain Fv constructs of anti-ganglioside GD2 antibodies

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US1370396P 1996-03-20 1996-03-20
US09/142,974 US6451995B1 (en) 1996-03-20 1997-03-20 Single chain FV polynucleotide or peptide constructs of anti-ganglioside GD2 antibodies, cells expressing same and related methods
US10/075,947 US20030147808A1 (en) 1996-03-20 2002-02-13 Single Chain Fv constructs of anti-ganglioside GD2 antibodies

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US1997/004427 Continuation WO1997034634A1 (en) 1996-03-20 1997-03-20 Single chain fv constructs of anti-ganglioside gd2 antibodies
US09/142,974 Continuation US6451995B1 (en) 1996-03-20 1997-03-20 Single chain FV polynucleotide or peptide constructs of anti-ganglioside GD2 antibodies, cells expressing same and related methods

Publications (1)

Publication Number Publication Date
US20030147808A1 true US20030147808A1 (en) 2003-08-07

Family

ID=21761285

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/142,974 Expired - Lifetime US6451995B1 (en) 1996-03-20 1997-03-20 Single chain FV polynucleotide or peptide constructs of anti-ganglioside GD2 antibodies, cells expressing same and related methods
US10/075,947 Abandoned US20030147808A1 (en) 1996-03-20 2002-02-13 Single Chain Fv constructs of anti-ganglioside GD2 antibodies

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/142,974 Expired - Lifetime US6451995B1 (en) 1996-03-20 1997-03-20 Single chain FV polynucleotide or peptide constructs of anti-ganglioside GD2 antibodies, cells expressing same and related methods

Country Status (2)

Country Link
US (2) US6451995B1 (en)
WO (1) WO1997034634A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014144763A3 (en) * 2013-03-15 2014-12-31 Memorial Sloan Kettering Cancer Center High affinity anti-gd2 antibodies
US9688772B2 (en) 2010-06-19 2017-06-27 Memorial Sloan Kettering Cancer Center Anti-GD2 antibodies

Families Citing this family (190)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997040140A1 (en) * 1996-04-22 1997-10-30 The Wistar Institute Of Anatomy And Biology Gd2 anti-idiotypic antibodies and uses thereof
DE19744531A1 (en) * 1997-10-09 1999-05-27 Klaus Dr Rer Nat Bosslet Binding molecules against receptor-ligand complexes
CA2318576C (en) * 1997-12-01 2009-04-14 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Antibodies, including fv molecules, and immunoconjugates having high binding affinity for mesothelin and methods for their use
US6809184B1 (en) 1997-12-01 2004-10-26 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Antibodies, including FV molecules, and immunoconjugates having high binding affinity for mesothelin and methods for their use
CA2374398C (en) 1999-05-27 2011-03-15 Ira Pastan Immunoconjugates having high binding affinity
US7144991B2 (en) 1999-06-07 2006-12-05 Aletheon Pharmaceuticals, Inc. Streptavidin expressed gene fusions and methods of use thereof
JP2003501096A (en) * 1999-06-07 2003-01-14 ネオルクス コーポレイション Streptavidin expressing gene fusion and method of using the same
WO2001023573A1 (en) * 1999-09-30 2001-04-05 Kyowa Hakko Kogyo Co., Ltd. Human type complementation-determining domain transplanted antibody against ganglioside gd2 and derivative of this antibody
WO2001097854A2 (en) * 2000-06-21 2001-12-27 Universite Catholique De Louvain Method for the selective survival or selective growth of a target cell by the use of a conjugate, its use in therapeutics and/or diagnostics
CA2490542C (en) 2002-05-23 2013-07-16 Mark I. Greene Fas peptide mimetics and uses thereof
US7446190B2 (en) 2002-05-28 2008-11-04 Sloan-Kettering Institute For Cancer Research Nucleic acids encoding chimeric T cell receptors
WO2005081898A2 (en) 2004-02-20 2005-09-09 The Trustees Of The University Of Pennsylvania Binding peptidomimetics and uses of the same
GB0410627D0 (en) * 2004-05-12 2004-06-16 Scancell Ltd Specific binding members
TWI422594B (en) 2007-02-02 2014-01-11 Baylor Res Inst Agents that engage antigen-presenting cells through dendritic cell asialoglycoprotein receptor (dc-asgpr)
US8057798B2 (en) 2007-02-02 2011-11-15 Baylor Research Institute Vaccines based on targeting antigen to DCIR expressed on antigen-presenting cells
CA2729499A1 (en) 2008-06-30 2010-01-07 Morphotek, Inc. Anti-gd2 antibodies and methods and uses related thereto
IL292038A (en) 2014-04-23 2022-06-01 Juno Therapeutics Inc Methods for enriching or producing immune cell populations for adoptive therapy
JP6836500B2 (en) 2014-05-16 2021-03-03 ベイラー リサーチ インスティテュートBaylor Research Institute Methods and compositions for treating autoimmune and inflammatory conditions
EP3169773B1 (en) 2014-07-15 2023-07-12 Juno Therapeutics, Inc. Engineered cells for adoptive cell therapy
TWI805109B (en) 2014-08-28 2023-06-11 美商奇諾治療有限公司 Antibodies and chimeric antigen receptors specific for cd19
WO2016064929A1 (en) 2014-10-20 2016-04-28 Juno Therapeutics, Inc. Methods and compositions for dosing in adoptive cell therapy
CA2966538A1 (en) 2014-11-05 2016-05-12 Juno Therapeutics, Inc. Methods for transduction and cell processing
CA2969456A1 (en) 2014-12-03 2016-06-09 Juno Therapeutics, Inc. Methods and compositions for adoptive cell therapy
MA41346A (en) 2015-01-12 2017-11-21 Juno Therapeutics Inc POST-TRANSCRIPTIONAL REGULATORY ELEMENTS OF MODIFIED HEPATITIS
ES2818103T3 (en) 2015-01-16 2021-04-09 Juno Therapeutics Inc ROR1-specific chimeric antigen receptors and antibodies
WO2016166568A1 (en) 2015-04-16 2016-10-20 Juno Therapeutics Gmbh Methods, kits and apparatus for expanding a population of cells
US20180161368A1 (en) 2015-05-29 2018-06-14 Juno Therapeutics, Inc. Composition and methods for regulating inhibitory interactions in genetically engineered cells
MA42895A (en) 2015-07-15 2018-05-23 Juno Therapeutics Inc MODIFIED CELLS FOR ADOPTIVE CELL THERAPY
US11142565B2 (en) 2015-09-24 2021-10-12 Abvitro Llc Broadly neutralizing anti-HIV-1 antibodies that bind to an N-glycan epitope on the envelope
WO2017053902A1 (en) 2015-09-25 2017-03-30 Abvitro Llc High throughput process for t cell receptor target identification of natively-paired t cell receptor sequences
MA45489A (en) 2015-10-22 2018-08-29 Juno Therapeutics Gmbh CELL CULTURE PROCESSES, ASSOCIATED KITS AND APPARATUS
MA45488A (en) 2015-10-22 2018-08-29 Juno Therapeutics Gmbh CELL CULTURE PROCESSES, KITS AND APPARATUS
WO2017068419A2 (en) 2015-10-22 2017-04-27 Juno Therapeutics Gmbh Methods, kits, agents and apparatuses for transduction
MA44314A (en) 2015-11-05 2018-09-12 Juno Therapeutics Inc CHEMERICAL RECEPTORS CONTAINING TRAF-INDUCING DOMAINS, AND ASSOCIATED COMPOSITIONS AND METHODS
WO2017079703A1 (en) 2015-11-05 2017-05-11 Juno Therapeutics, Inc. Vectors and genetically engineered immune cells expressing metabolic pathway modulators and uses in adoptive cell therapy
MA43380A (en) 2015-12-03 2018-10-10 Juno Therapeutics Inc MODIFIED CHEMERICAL RECEPTORS AND ASSOCIATED COMPOSITIONS AND PROCESSES
MA43378A (en) 2015-12-03 2018-10-10 Juno Therapeutics Inc COMPOSITIONS AND METHODS FOR REDUCTION OF IMMUNE RESPONSE AGAINST CHEMERIC ANTIGEN RECEPTORS
WO2017096331A1 (en) 2015-12-04 2017-06-08 Juno Therapeutics, Inc. Methods and compositions related to toxicity associated with cell therapy
MA43759A (en) 2016-03-16 2018-11-28 Jason Connor PROCESSES FOR THE ADAPTIVE DESIGN OF A TREATMENT REGIME AND ASSOCIATED TREATMENTS
EP3430548A1 (en) 2016-03-16 2019-01-23 Juno Therapeutics, Inc. Methods for determining dosing of a therapeutic agent and related treatments
US11518814B2 (en) 2016-03-22 2022-12-06 Seattle Children's Hospital Early intervention methods to prevent or ameliorate toxicity
WO2017193107A2 (en) 2016-05-06 2017-11-09 Juno Therapeutics, Inc. Genetically engineered cells and methods of making the same
MX2018014599A (en) 2016-05-27 2019-02-28 Aadigen Llc Peptides and nanoparticles for intracellular delivery of genome-editing molecules.
JP2019517518A (en) 2016-06-03 2019-06-24 メモリアル スローン ケタリング キャンサー センター Adoptive cell therapy as an early treatment option
MA45341A (en) 2016-06-06 2019-04-10 Hutchinson Fred Cancer Res METHODS FOR TREATING B-LYMPHOCYTE MALIGNITIES USING ADOPTIVE CELL THERAPY
MA45491A (en) 2016-06-27 2019-05-01 Juno Therapeutics Inc CMH-E RESTRICTED EPITOPES, BINDING MOLECULES AND RELATED METHODS AND USES
WO2018005559A1 (en) 2016-06-27 2018-01-04 Juno Therapeutics, Inc. Method of identifying peptide epitopes, molecules that bind such epitopes and related uses
EP3490585B1 (en) 2016-07-29 2023-05-17 Juno Therapeutics, Inc. Immunomodulatory polypeptides and related compositions and methods
CN109804089A (en) 2016-07-29 2019-05-24 朱诺治疗学股份有限公司 For assessing the present or absent method of duplicating virus
MA45784A (en) 2016-07-29 2019-06-05 Juno Therapeutics Inc ANTI-BODY ANTI-IDIOTYPES DIRECTED AGAINST ANTI-CD19 ANTIBODY
BR112019004662A2 (en) 2016-09-12 2019-05-28 Juno Therapeutics Inc infusion bioreactor bag sets
JP2019536437A (en) 2016-10-03 2019-12-19 ジュノー セラピューティクス インコーポレイテッド HPV-specific binding molecules
EP3525803B1 (en) 2016-10-13 2022-12-07 Juno Therapeutics, Inc. Immunotherapy methods and compositions involving tryptophan metabolic pathway modulators
EP3534940A1 (en) 2016-11-03 2019-09-11 Juno Therapeutics, Inc. Combination therapy of a cell based therapy and a microglia inhibitor
EP3534938A2 (en) 2016-11-03 2019-09-11 Juno Therapeutics, Inc. Combination therapy of a t cell therapy and a btk inhibitor
WO2018089423A1 (en) 2016-11-09 2018-05-17 Musc Foundation For Research Development Cd38-nad+ regulated metabolic axis in anti-tumor immunotherapy
BR112019011065A2 (en) 2016-12-03 2019-10-01 Juno Therapeutics Inc methods for determining car cell dosage
MA46995A (en) 2016-12-03 2019-10-09 Acerta Pharma Bv METHODS AND COMPOSITIONS FOR THE USE OF THERAPEUTIC T-LYMPHOCYTES IN COMBINATION WITH KINASE INHIBITORS
KR20190104529A (en) 2016-12-03 2019-09-10 주노 쎄러퓨티크스 인코퍼레이티드 Modulation of CAR-T Cells
MA46998A (en) 2016-12-05 2019-10-09 Juno Therapeutics Inc PRODUCTION OF MODIFIED CELLS FOR ADOPTIVE CELL THERAPY
US11821027B2 (en) 2017-01-10 2023-11-21 Juno Therapeutics, Inc. Epigenetic analysis of cell therapy and related methods
MX2019008538A (en) 2017-01-20 2019-11-05 Juno Therapeutics Gmbh Cell surface conjugates and related cell compositions and methods.
CN117357638A (en) 2017-02-17 2024-01-09 弗雷德哈钦森癌症中心 Combination therapy for the treatment of BCMA-related cancers and autoimmune disorders
BR112019017767A2 (en) 2017-02-27 2020-04-07 Juno Therapeutics Inc compositions, articles of manufacture and methods related to dosing in cell therapy
CN110582288A (en) 2017-02-28 2019-12-17 恩多塞特公司 Compositions and methods for CAR T cell therapy
AU2018234640B2 (en) 2017-03-14 2024-03-14 Juno Therapeutics, Inc. Methods for cryogenic storage
AU2018250336A1 (en) 2017-04-07 2019-09-26 Juno Therapeutics, Inc. Engineered cells expressing prostate-specific membrane antigen (PSMA) or a modified form thereof and related methods
CN110709700A (en) 2017-04-14 2020-01-17 朱诺治疗学股份有限公司 Method for evaluating cell surface glycosylation
EP4083063A3 (en) 2017-04-18 2023-01-04 FUJIFILM Cellular Dynamics, Inc. Antigen-specific immune effector cells
CA3060526A1 (en) 2017-04-27 2018-11-01 Juno Therapeutics Gmbh Oligomeric particle reagents and methods of use thereof
IL270250B1 (en) 2017-05-01 2024-02-01 Juno Therapeutics Inc Combination of a cell therapy and an immunomodulatory compound
EP3652537A4 (en) * 2017-05-05 2021-10-20 Memorial Sloan Kettering Cancer Center Modular self assembly disassembly (sada) technologies
CN111225675B (en) 2017-06-02 2024-05-03 朱诺治疗学股份有限公司 Articles and methods of treatment using adoptive cell therapy
EP3631468A1 (en) 2017-06-02 2020-04-08 Juno Therapeutics, Inc. Articles of manufacture and methods related to toxicity associated with cell therapy
EP3538645B1 (en) 2017-06-20 2021-01-20 Institut Curie Immune cells defective for suv39h1
AU2018288863A1 (en) 2017-06-22 2020-01-30 Board Of Regents, The University Of Texas System Methods for producing regulatory immune cells and uses thereof
AU2018291032A1 (en) 2017-06-29 2020-01-16 Juno Therapeutics, Inc. Mouse model for assessing toxicities associated with immunotherapies
CA3070573A1 (en) 2017-07-29 2019-02-07 Juno Therapeutics, Inc. Reagents for expanding cells expressing recombinant receptors
BR112020001605A2 (en) 2017-08-09 2020-08-11 Juno Therapeutics Inc methods for producing genetically modified cell compositions and related compositions
CA3070575A1 (en) 2017-08-09 2019-02-14 Juno Therapeutics, Inc. Methods and compositions for preparing genetically engineered cells
US20210071258A1 (en) 2017-09-01 2021-03-11 Juno Therapeutics, Inc. Gene expression and assessment of risk of developing toxicity following cell therapy
EP3679370A1 (en) 2017-09-07 2020-07-15 Juno Therapeutics, Inc. Methods of identifying cellular attributes related to outcomes associated with cell therapy
CN109517820B (en) 2017-09-20 2021-09-24 北京宇繁生物科技有限公司 gRNA of target HPK1 and HPK1 gene editing method
CN111954679A (en) 2017-10-03 2020-11-17 朱诺治疗学股份有限公司 HPV specific binding molecules
US11851679B2 (en) 2017-11-01 2023-12-26 Juno Therapeutics, Inc. Method of assessing activity of recombinant antigen receptors
US11564946B2 (en) 2017-11-01 2023-01-31 Juno Therapeutics, Inc. Methods associated with tumor burden for assessing response to a cell therapy
BR112020008478A2 (en) 2017-11-01 2020-10-20 Editas Medicine, Inc. methods, compositions and components for editing crispr-cas9 of tgfbr2 in t cells for immunota-rapy
US20210132042A1 (en) 2017-11-01 2021-05-06 Juno Therapeutics, Inc. Methods of assessing or monitoring a response to a cell therapy
EP3704230A1 (en) 2017-11-01 2020-09-09 Juno Therapeutics, Inc. Process for generating therapeutic compositions of engineered cells
JP7447006B2 (en) 2017-11-01 2024-03-11 ジュノー セラピューティクス インコーポレイテッド Chimeric antigen receptor specific for B cell maturation antigen (BCMA)
CA3080509A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Process for producing a t cell composition
MX2020004243A (en) 2017-11-01 2020-09-25 Juno Therapeutics Inc Antibodies and chimeric antigen receptors specific for b-cell maturation antigen.
WO2019090202A1 (en) 2017-11-06 2019-05-09 Editas Medicine, Inc. Methods, compositions and components for crispr-cas9 editing of cblb in t cells for immunotherapy
AU2018359907A1 (en) 2017-11-06 2020-05-07 Fred Hutchinson Cancer Center Combination of a cell therapy and a gamma secretase inhibitor
CN111556789A (en) 2017-11-10 2020-08-18 朱诺治疗学股份有限公司 Closed system cryogenic vessel
WO2019109053A1 (en) 2017-12-01 2019-06-06 Juno Therapeutics, Inc. Methods for dosing and for modulation of genetically engineered cells
WO2019113557A1 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Process for producing a composition of engineered t cells
CN112041430A (en) 2017-12-08 2020-12-04 朱诺治疗学股份有限公司 Serum-free medium formulations for culturing cells and methods of use thereof
EP3720480A2 (en) 2017-12-08 2020-10-14 Juno Therapeutics, Inc. Phenotypic markers for cell therapy and related methods
CN112204048A (en) 2017-12-15 2021-01-08 朱诺治疗学股份有限公司 anti-CCT5 binding molecules and methods of use thereof
US11919937B2 (en) 2018-01-09 2024-03-05 Board Of Regents, The University Of Texas System T cell receptors for immunotherapy
US11779602B2 (en) 2018-01-22 2023-10-10 Endocyte, Inc. Methods of use for CAR T cells
EP3746569A1 (en) 2018-01-31 2020-12-09 Juno Therapeutics, Inc. Methods and reagents for assessing the presence or absence of replication competent virus
KR20200128014A (en) 2018-01-31 2020-11-11 셀진 코포레이션 Adoptive cell therapy and combination therapy with checkpoint inhibitors
US20210046159A1 (en) 2018-03-09 2021-02-18 Ospedale San Raffaele S.R.L. Il-1 antagonist and toxicity induced by cell therapy
PE20201345A1 (en) 2018-04-05 2020-11-25 Juno Therapeutics Inc RECEIVERS OF T-CELLS, AND DESIGNED CELLS THAT EXPRESS THEM
JP2021520202A (en) 2018-04-05 2021-08-19 ジュノー セラピューティクス インコーポレイテッド Methods for Producing Cells Expressing Recombinant Receptors and Related Compositions
SG11202010642TA (en) 2018-05-03 2020-11-27 Juno Therapeutics Inc Combination therapy of a chimeric antigen receptor (car) t cell therapy and a kinase inhibitor
AU2019318560A1 (en) 2018-08-09 2021-02-25 Juno Therapeutics, Inc. Processes for generating engineered cells and compositions thereof
EP3833759A1 (en) 2018-08-09 2021-06-16 Juno Therapeutics, Inc. Methods for assessing integrated nucleic acids
CA3110089A1 (en) 2018-08-28 2020-03-05 Fred Hutchinson Cancer Research Center Methods and compositions for adoptive t cell therapy incorporating induced notch signaling
AU2019339464A1 (en) 2018-09-11 2021-03-25 Juno Therapeutics, Inc. Methods for mass spectrometry analysis of engineered cell compositions
KR20210098450A (en) 2018-10-31 2021-08-10 주노 테라퓨틱스 게엠베하 Method for cell selection and stimulation and device therefor
BR112021008289A2 (en) 2018-11-01 2021-10-26 Juno Therapeutics, Inc. METHODS FOR TREATMENT USING CHIMERIC ANTIGEN RECEPTORS SPECIFIC FOR B CELL MATURATION ANTIGEN
US20210393691A1 (en) 2018-11-06 2021-12-23 Juno Therapeutics, Inc. Process for producing genetically engineered t cells
MX2021005366A (en) 2018-11-08 2021-09-10 Juno Therapeutics Inc Methods and combinations for treatment and t cell modulation.
CN113271963A (en) 2018-11-16 2021-08-17 朱诺治疗学股份有限公司 Methods of administering engineered T cells for treatment of B cell malignancies
WO2020106621A1 (en) 2018-11-19 2020-05-28 Board Of Regents, The University Of Texas System A modular, polycistronic vector for car and tcr transduction
US20220031749A1 (en) 2018-11-28 2022-02-03 Board Of Regents, The University Of Texas System Multiplex genome editing of immune cells to enhance functionality and resistance to suppressive environment
BR112021010248A2 (en) 2018-11-29 2021-08-17 Board Of Regents, The University Of Texas System methods for ex vivo expansion of natural killer cells and their use
WO2020113188A2 (en) 2018-11-30 2020-06-04 Juno Therapeutics, Inc. Methods for dosing and treatment of b cell malignancies in adoptive cell therapy
SG11202105502RA (en) 2018-11-30 2021-06-29 Juno Therapeutics Inc Methods for treatment using adoptive cell therapy
PE20212198A1 (en) 2019-01-29 2021-11-16 Juno Therapeutics Inc ANTIBODIES AND CHIMERIC RECEPTORS OF SPECIFIC ANTIGENS TO ORPHAN RECEPTOR 1, RECEPTOR TYROSINE KINASE TYPE (ROR1)
AU2020265741A1 (en) 2019-05-01 2021-11-25 Editas Medicine, Inc. Cells expressing a recombinant receptor from a modified TGFBR2 Locus, related polynucleotides and methods
MX2021013223A (en) 2019-05-01 2022-02-17 Juno Therapeutics Inc Cells expressing a chimeric receptor from a modified cd247 locus, related polynucleotides and methods.
CA3139162A1 (en) 2019-05-17 2020-11-26 Cancer Prevention Pharmaceuticals, Inc. Methods for treating familial adenomatous polyposis
US20220228101A1 (en) 2019-06-07 2022-07-21 Juno Therapeutics, Inc. Automated t cell culture
WO2020252218A1 (en) 2019-06-12 2020-12-17 Juno Therapeutics, Inc. Combination therapy of a cell-mediated cytotoxic therapy and an inhibitor of a prosurvival bcl2 family protein
WO2021013950A1 (en) 2019-07-23 2021-01-28 Mnemo Therapeutics Immune cells defective for suv39h1
WO2021035194A1 (en) 2019-08-22 2021-02-25 Juno Therapeutics, Inc. Combination therapy of a t cell therapy and an enhancer of zeste homolog 2 (ezh2) inhibitor and related methods
WO2021041994A2 (en) 2019-08-30 2021-03-04 Juno Therapeutics, Inc. Machine learning methods for classifying cells
AU2020343407A1 (en) 2019-09-02 2022-03-24 INSERM (Institut National de la Santé et de la Recherche Médicale) Immunotherapy targeting tumor neoantigenic peptides
JP2022547168A (en) 2019-09-09 2022-11-10 スクライブ・セラピューティクス・インコーポレイテッド Compositions and methods for use in immunotherapy
WO2021078910A1 (en) 2019-10-22 2021-04-29 Institut Curie Immunotherapy targeting tumor neoantigenic peptides
MX2022005145A (en) 2019-10-30 2022-06-29 Juno Therapeutics Gmbh Cell selection and/or stimulation devices and methods of use.
JP2022554353A (en) 2019-11-07 2022-12-28 ジュノー セラピューティクス インコーポレイテッド T cell therapy and (S)-3-[4-(4-morpholin-4-ylmethyl-benzyloxy)-1-oxo-1,3-dihydro-isoindol-2-yl]-piperidine-2,6- Combination with Dione
MX2022006715A (en) 2019-12-06 2022-09-23 Juno Therapeutics Inc Methods related to toxicity and response associated with cell therapy for treating b cell malignancies.
JP2023504736A (en) 2019-12-06 2023-02-06 ジュノー セラピューティクス インコーポレイテッド Anti-idiotypic antibodies against GPRC5D target binding domains and related compositions and methods
CN115916817A (en) 2019-12-06 2023-04-04 朱诺治疗学股份有限公司 Anti-idiotypic antibodies directed against BCMA-targeting binding domains and related compositions and methods
BR112022014501A2 (en) 2020-01-24 2022-09-20 Juno Therapeutics Inc METHODS FOR DOSING AND TREATMENT OF FOLLICULAR LYMPHOMA AND MARGINAL ZONE LYMPHOMA IN ADOPTIVE CELL THERAPY
EP4097218A1 (en) 2020-01-28 2022-12-07 Juno Therapeutics, Inc. Methods for t cell transduction
BR112022015968A2 (en) 2020-02-12 2022-10-11 Juno Therapeutics Inc BCMA-Targeted ANTIGEN RECEPTOR T-CELL COMPOSITIONS AND METHODS AND USES THEREOF
MX2022009832A (en) 2020-02-12 2023-02-14 Juno Therapeutics Inc Cd19-directed chimeric antigen receptor t cell compositions and methods and uses thereof.
EP4107173A1 (en) 2020-02-17 2022-12-28 Board of Regents, The University of Texas System Methods for expansion of tumor infiltrating lymphocytes and use thereof
US20230149462A1 (en) 2020-04-10 2023-05-18 Juno Therapeutics, Inc. Methods and uses related to cell therapy engineered with a chimeric antigen receptor targeting b-cell maturation antigen
WO2021228999A1 (en) 2020-05-12 2021-11-18 Institut Curie Neoantigenic epitopes associated with sf3b1 mutations
US20230178239A1 (en) 2020-05-13 2023-06-08 Juno Therapeutics, Inc. Methods of identifying features associated with clinical response and uses thereof
CN115835873A (en) 2020-05-13 2023-03-21 朱诺治疗学股份有限公司 Method for generating donor batch cells expressing recombinant receptor
AU2021275239A1 (en) 2020-05-21 2022-12-15 Board Of Regents, The University Of Texas System T cell receptors with VGLL1 specificity and uses thereof
WO2021260186A1 (en) 2020-06-26 2021-12-30 Juno Therapeutics Gmbh Engineered t cells conditionally expressing a recombinant receptor, related polynucleotides and methods
AU2021316727A1 (en) 2020-07-30 2023-03-02 INSERM (Institut National de la Santé et de la Recherche Médicale) Immune cells defective for SOCS1
KR20230095918A (en) 2020-08-05 2023-06-29 주노 쎄러퓨티크스 인코퍼레이티드 Anti-idiotype antibodies to the ROR1-target binding domain and related compositions and methods
AU2021377699A1 (en) 2020-11-13 2023-06-15 Catamaran Bio, Inc. Genetically modified natural killer cells and methods of use thereof
WO2022133030A1 (en) 2020-12-16 2022-06-23 Juno Therapeutics, Inc. Combination therapy of a cell therapy and a bcl2 inhibitor
WO2022150731A1 (en) 2021-01-11 2022-07-14 Sana Biotechnology, Inc. Use of cd8-targeted viral vectors
US20240108654A1 (en) 2021-03-03 2024-04-04 Juno Therapeutics, Inc. Combination of a t cell therapy and a dgk inhibitor
EP4304635A1 (en) 2021-03-11 2024-01-17 Mnemo Therapeutics Tumor neoantigenic peptides and uses thereof
KR20240006721A (en) 2021-03-11 2024-01-15 엥스띠뛰 퀴리 Membrane-transforming neoantigen peptide
EP4304632A2 (en) 2021-03-11 2024-01-17 Mnemo Therapeutics Tumor neoantigenic peptides
KR20230159851A (en) 2021-03-22 2023-11-22 주노 쎄러퓨티크스 인코퍼레이티드 How to Determine the Potency of a Therapeutic Cell Composition
CN117321200A (en) 2021-03-22 2023-12-29 朱诺治疗学股份有限公司 Method for assessing efficacy of viral vector particles
JP2024513054A (en) 2021-03-29 2024-03-21 ジュノー セラピューティクス インコーポレイテッド Combination of CAR T cell therapy and immunomodulatory compounds for the treatment of lymphoma
BR112023019847A2 (en) 2021-03-29 2023-11-07 Juno Therapeutics Inc METHODS FOR DOSING AND TREATMENT WITH A COMBINATION OF A CHECKPOINT INHIBITOR THERAPY AND A CAR T CELL THERAPY
WO2022234009A2 (en) 2021-05-06 2022-11-10 Juno Therapeutics Gmbh Methods for stimulating and transducing t cells
TW202321457A (en) 2021-08-04 2023-06-01 美商薩那生物科技公司 Use of cd4-targeted viral vectors
WO2023014922A1 (en) 2021-08-04 2023-02-09 The Regents Of The University Of Colorado, A Body Corporate Lat activating chimeric antigen receptor t cells and methods of use thereof
WO2023105000A1 (en) 2021-12-09 2023-06-15 Zygosity Limited Vector
TW202342757A (en) 2021-12-17 2023-11-01 美商薩那生物科技公司 Modified paramyxoviridae attachment glycoproteins
WO2023115039A2 (en) 2021-12-17 2023-06-22 Sana Biotechnology, Inc. Modified paramyxoviridae fusion glycoproteins
WO2023126458A1 (en) 2021-12-28 2023-07-06 Mnemo Therapeutics Immune cells with inactivated suv39h1 and modified tcr
WO2023139269A1 (en) 2022-01-21 2023-07-27 Mnemo Therapeutics Modulation of suv39h1 expression by rnas
WO2023147515A1 (en) 2022-01-28 2023-08-03 Juno Therapeutics, Inc. Methods of manufacturing cellular compositions
WO2023150518A1 (en) 2022-02-01 2023-08-10 Sana Biotechnology, Inc. Cd3-targeted lentiviral vectors and uses thereof
WO2023178348A1 (en) 2022-03-18 2023-09-21 The Regents Of The University Of Colorado, A Body Corporate Genetically engineered t-cell co-receptors and methods of use thereof
WO2023180552A1 (en) 2022-03-24 2023-09-28 Institut Curie Immunotherapy targeting tumor transposable element derived neoantigenic peptides in glioblastoma
WO2023193015A1 (en) 2022-04-01 2023-10-05 Sana Biotechnology, Inc. Cytokine receptor agonist and viral vector combination therapies
WO2023196921A1 (en) 2022-04-06 2023-10-12 The Regents Of The University Of Colorado, A Body Corporate Granzyme expressing t cells and methods of use
WO2023196933A1 (en) 2022-04-06 2023-10-12 The Regents Of The University Of Colorado, A Body Corporate Chimeric antigen receptor t cells and methods of use thereof
WO2023211972A1 (en) 2022-04-28 2023-11-02 Medical University Of South Carolina Chimeric antigen receptor modified regulatory t cells for treating cancer
WO2023213969A1 (en) 2022-05-05 2023-11-09 Juno Therapeutics Gmbh Viral-binding protein and related reagents, articles, and methods of use
WO2023220655A1 (en) 2022-05-11 2023-11-16 Celgene Corporation Methods to overcome drug resistance by re-sensitizing cancer cells to treatment with a prior therapy via treatment with a t cell therapy
EP4279085A1 (en) 2022-05-20 2023-11-22 Mnemo Therapeutics Compositions and methods for treating a refractory or relapsed cancer or a chronic infectious disease
WO2023230581A1 (en) 2022-05-25 2023-11-30 Celgene Corporation Methods of manufacturing t cell therapies
WO2023250400A1 (en) 2022-06-22 2023-12-28 Juno Therapeutics, Inc. Treatment methods for second line therapy of cd19-targeted car t cells
WO2024006960A1 (en) 2022-06-29 2024-01-04 Juno Therapeutics, Inc. Lipid nanoparticles for delivery of nucleic acids
WO2024044779A2 (en) 2022-08-26 2024-02-29 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for delta-like ligand 3 (dll3)
WO2024054944A1 (en) 2022-09-08 2024-03-14 Juno Therapeutics, Inc. Combination of a t cell therapy and continuous or intermittent dgk inhibitor dosing
WO2024062138A1 (en) 2022-09-23 2024-03-28 Mnemo Therapeutics Immune cells comprising a modified suv39h1 gene
WO2024081820A1 (en) 2022-10-13 2024-04-18 Sana Biotechnology, Inc. Viral particles targeting hematopoietic stem cells
WO2024100604A1 (en) 2022-11-09 2024-05-16 Juno Therapeutics Gmbh Methods for manufacturing engineered immune cells

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4675287A (en) * 1984-07-26 1987-06-23 Scripps Clinic And Research Foundation Monoclonal antibody directed to human ganglioside GD2
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5302370A (en) * 1989-09-11 1994-04-12 Institut Fur Diagnostikforschung Gmbh Chelating agents for forming complexes with radioactive isotopes, metal complexes thereof and use thereof in diagnosis and therapy
US5359046A (en) * 1990-12-14 1994-10-25 Cell Genesys, Inc. Chimeric chains for receptor-associated signal transduction pathways
US5405990A (en) * 1992-07-23 1995-04-11 Zeneca Limited Chemical compounds
US5851527A (en) * 1988-04-18 1998-12-22 Immunomedics, Inc. Method for antibody targeting of therapeutic agents
US5912172A (en) * 1988-05-04 1999-06-15 Yeda Research And Development Co. Ltd. Endowing lymphocytes with antibody specificity
US5973116A (en) * 1993-01-15 1999-10-26 Imperial Cancer Research Technology Limited Compounds for targeting

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992018629A1 (en) * 1991-04-19 1992-10-29 Genetics Institute, Inc. Recombinant 3f8-type antibodies
WO1993019163A1 (en) * 1992-03-18 1993-09-30 Yeda Research And Development Co, Ltd. Chimeric receptor genes and cells transformed therewith
DE69334095T2 (en) * 1992-07-17 2007-04-12 Dana-Farber Cancer Institute, Boston Method for intracellular binding of targeted molecules
US5540926A (en) * 1992-09-04 1996-07-30 Bristol-Myers Squibb Company Soluble and its use in B cell stimulation

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4675287A (en) * 1984-07-26 1987-06-23 Scripps Clinic And Research Foundation Monoclonal antibody directed to human ganglioside GD2
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5851527A (en) * 1988-04-18 1998-12-22 Immunomedics, Inc. Method for antibody targeting of therapeutic agents
US5912172A (en) * 1988-05-04 1999-06-15 Yeda Research And Development Co. Ltd. Endowing lymphocytes with antibody specificity
US5302370A (en) * 1989-09-11 1994-04-12 Institut Fur Diagnostikforschung Gmbh Chelating agents for forming complexes with radioactive isotopes, metal complexes thereof and use thereof in diagnosis and therapy
US5359046A (en) * 1990-12-14 1994-10-25 Cell Genesys, Inc. Chimeric chains for receptor-associated signal transduction pathways
US5405990A (en) * 1992-07-23 1995-04-11 Zeneca Limited Chemical compounds
US5973116A (en) * 1993-01-15 1999-10-26 Imperial Cancer Research Technology Limited Compounds for targeting

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9688772B2 (en) 2010-06-19 2017-06-27 Memorial Sloan Kettering Cancer Center Anti-GD2 antibodies
US10287365B2 (en) 2010-06-19 2019-05-14 Memorial Sloan Kettering Cancer Center Anti-GD2 antibodies
WO2014144763A3 (en) * 2013-03-15 2014-12-31 Memorial Sloan Kettering Cancer Center High affinity anti-gd2 antibodies
US10167341B2 (en) 2013-03-15 2019-01-01 Memorial Sloan Kettering Cancer Center High affinity anti-GD2 antibodies
RU2680267C2 (en) * 2013-03-15 2019-02-19 Мемориал Слоан Кеттеринг Кэнсер Сентер High-affinity anti-gd2 antibodies

Also Published As

Publication number Publication date
US6451995B1 (en) 2002-09-17
WO1997034634A1 (en) 1997-09-25

Similar Documents

Publication Publication Date Title
US6451995B1 (en) Single chain FV polynucleotide or peptide constructs of anti-ganglioside GD2 antibodies, cells expressing same and related methods
Whitlow et al. An improved linker for single-chain Fv with reduced aggregation and enhanced proteolytic stability
EP1912674B1 (en) Bispecific single chain fv antibody molecules and methods of use thereof
US6071515A (en) Dimer and multimer forms of single chain polypeptides
CA2195556C (en) Polyspecific immunoconjugates and antibody composites for targeting the multidrug resistant phenotype
Melani et al. Targeting of interleukin 2 to human ovarian carcinoma by fusion with a single-chain Fv of antifolate receptor antibody
US7183388B2 (en) Anti-MUC-1 single chain antibodies for tumor targeting
US6174691B1 (en) Minimum recognition unit of a PEM mucin tandem repeat specific monoclonal antibody
US20180155449A1 (en) Covalent disulfide-linked diabodies and uses thereof
Lin et al. A genetically engineered anti-CD45 single-chain antibody-streptavidin fusion protein for pretargeted radioimmunotherapy of hematologic malignancies
US20040071696A1 (en) Bispecific single chain Fv antibody molecules and methods of use thereof
WO1999062526A2 (en) Use of genetically engineered antibodies to cd38 to treat multiple myeloma
JPH05502236A (en) Chimeric antibodies for the detection and treatment of infectious and inflammatory lesions
JP2002510968A (en) Recombinant antibody-enzyme fusion protein
US20070148088A1 (en) Cytotoxic therapy
JPH07507440A (en) Antibody constructs with increased binding affinity
ES2293642T3 (en) RECOMBINANT PEPTIDES DERIVED FROM ANTIBODY MC3 ANTI-BA46, METHODS OF USE OF THE SAME AND METHODS OF HUMANIZATION OF ANTIBODY PEPTIDES.
US20030077676A1 (en) Anti-MUC-1 single chain antibodies for tumor targeting
WO2023025304A1 (en) Polypeptide fusion molecule close to natural molecule
JP2004507205A (en) Polypeptide for detection and removal of CA19-9 antigen positive cells
US6440386B1 (en) Stabilized radiophosphate-labeled proteins
US20030232010A2 (en) Improved cytotoxic therapy
Kobayashi et al. Methods to avoid adverse effect of circulating antigen on biodistribution of 125I-labeled antiTac dsFv: Preinjection of intact antibody versus clearance of antigen with avidin-biotin system
Molimi et al. Targeting of Interleukin 2 to Human Ovarian Carcinoma by Fusion with a Single-Chain Fv of Antifolate Receptor Antibody1
Cheng 99m Tc-Hydrazinonicotinamide-anti-TAG-72 CC49 tetravalent single-chain Fv monoclonal antibody 99m Tc-HYNIC-CC49 [sc (Fv) 2] 2 MAb

Legal Events

Date Code Title Description
AS Assignment

Owner name: SLOAN-KETTERING INSTITUTE FOR CANCER RESEARCH, NEW

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CHEUNG, NAI-KONG V.;LARSON, STEVEN M.;GUO, HONG-FEN;AND OTHERS;REEL/FRAME:012951/0477;SIGNING DATES FROM 19970527 TO 19970612

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: UNITED STATES DEPARTMENT OF ENERGY, DISTRICT OF COLUMBIA

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:SLOAN-KETTERING INST CAN RESEARCH;REEL/FRAME:052893/0724

Effective date: 20151002