EP0789713A1 - Antibodies - Google Patents

Antibodies

Info

Publication number
EP0789713A1
EP0789713A1 EP95936025A EP95936025A EP0789713A1 EP 0789713 A1 EP0789713 A1 EP 0789713A1 EP 95936025 A EP95936025 A EP 95936025A EP 95936025 A EP95936025 A EP 95936025A EP 0789713 A1 EP0789713 A1 EP 0789713A1
Authority
EP
European Patent Office
Prior art keywords
antibody
die
antibodies
conjugate according
antibody conjugate
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.)
Withdrawn
Application number
EP95936025A
Other languages
German (de)
French (fr)
Inventor
Frederick Charles Kull, Jr.
Mary Elizabeth Fling
Julie Beth Stimmel
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.)
Wellcome Foundation Ltd
Original Assignee
Wellcome Foundation Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Wellcome Foundation Ltd filed Critical Wellcome Foundation Ltd
Publication of EP0789713A1 publication Critical patent/EP0789713A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • 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
    • 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]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1093Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody conjugates with carriers being antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to antibodies which are capable of being conjugated at specific sites, to processes for the site-directed conjugation of such antibodies, to antibodies that have been conjugated at a specific site and to the use of such antibodies in therapy and diagnosis.
  • Antibodies are globular proteins which represent a vital component of the mammalian immune response to foreign disease inducing agents. Antibodies may be manufactured ex vivo by any of a number of methods, and such antibodies, particularly monoclonal antibodies and/or fragments thereof, have proved valuable as both diagnostic and therapeutic agents.
  • the utility of antibodies stems from their unique antigen specificity, i.e.. their ability to chemically recognise and remain bound to discrete chemical moieties such as pathogen antigens or tumour-associated antigens.
  • Another aspect of their utility is their diversity, i.e., the ability of mammals (and now other processes such as phage display) to create a very large variety of discrete, genetically defined antibodies (monoclonal antibodies).
  • a final aspect of their utility is their capacity to interact via their "constant" regions. This latter aspect determines other sets of properties, for example, those properties common to isotypes such as interaction with effector cells, complement or other binding moieties like protein A.
  • an antibody or fragment may be enhanced by chemically coupling one or more further molecular moieties (referred to herein as substances) either directly or indirectly that convey properties to the conjugate that are not naturally present in the constituents alone.
  • properties may include a reporter function such as a dye or a radionuclide, an enzymatic function, a second binding function (such as with biotin- avidin), a drug (such as adriamycin), a cytotoxic function (such as with ricin), a chelator, or a chemical linkage moiety that may, in turn render the antibody capable of subsequent reaction with any of a variety of molecular moieties.
  • an antibody conjugate comprises an antibody and an active substance (substance) which is either directly or indirectly conjugated to the antibody.
  • the active substance is a radionuclide
  • the radionuclide may be directly conjugated to the antibody or alternatively it may be indirectly conjugated to the antibody via a chelator such as for example TMT or even a chelator which is in turn linked to the antibody by a further protein reactive group (cross-linker) such as. for example, bromoacetyl.
  • the active substance could be a molecular chimaera for use in enzyme-prodrug therapy, such a chimaera comprises a transcriptional regulatory DNA sequence capable of being activated in a mammalian cell such as a cancer cell and a DNA sequence operatively linked to the transcriptional regulatory DNA sequence and encoding a heterologous enzyme capable of catalysing the conversion of a prodrug which is administered subsequently,into an agent toxic to the cancer cell.
  • This molecular chimera may be directly conjugated to the antibody or alternatively, it may be contained within a viral vector or liposome with the viral vector or liposome then being attached to the antibody (see European application No.90309430.8).
  • conjugation reactions exploit the chemical functional groups that occur naturally in the antibody.
  • a common approach is to target primary amines (mainly the ⁇ amino groups of lysine residues).
  • Another common example of this approach includes the iodination (such as l- ⁇ I) of tyrosine residues.
  • iodination such as l- ⁇ I
  • a disadvantage of this approach is that it is random. That is, lysine or tyrosine residues may occur throughout the structure and therefore the natural properties that these residues help convey ⁇ such as, for example, antigen recognition, complement reactivity or effector cell interaction — may be compromised.
  • the behaviour of the conjugate is the average of the behaviour of all the individual unique components, some of which may be entirely useless or detrimental.
  • the random approach is also known to affect antigen recognition.
  • Site-directed conjugation i.e.. conjugation to a specific amino acid residue within the antibody structure, would convey the advantage that the resulting conjugate is not a mixture of different products.
  • the properties conveyed by the antibody to the conjugate such as antigen reactivity or pharmacokinetic stability, can thus be ascribed to a defined chemical structure.
  • the site may be selected so as to be spatially removed from areas known to convey antigen-binding properties.
  • the variable region of an antibody is known to contain the antigen binding site and the CH2 domain of the heavy chain is known to contain effector (FcR) and complement (Clq) interactive residues.
  • FcR effector
  • Clq complement
  • glycosylation conjugation procedure presents the risk of altering carbohydrate that might be required for antigen or effector interaction (Lund, et al., Molec. Immunmol 27: 1 145, 1990; Isaacs, et al. J. Immunol. 148: 3062, 1992).
  • Another example of site-directed conjugation to antibodies involves the creation, by site- directed mutagenesis, of a free thiol on the antibody.
  • Antibodies naturally contain cvstine residues whose thiol groups are joined by disulfide bridges. The position of the naturally occurring cvstine residues is highly conserved among species indicating that these residues are essential for the structure and function of antibodies.
  • Antibodies do not naturally contain free sulfhydryi groups. It is hypothetically attractive to engineer an antibody to possess a cysteine the thiol group of which is neither oxidised nor compromises the fidelity of the natural sulfhydryi bridges.
  • Sulfhydryi groups of cysteine residues may of course be exploited for conjugation by numerous conjugation chemistries that are rather specific for sulfhydryls such as maleimides. alkyl and aryl halides, ⁇ -haloacyls and pyridyl disulfides. However, it has been determined that variant monoclonal antibodies that have been designed with unnatural cysteine residues do not de facto possess free thiol groups available for conjugation.
  • the present invention provides a monoclonal antibody comprising a cysteine residue exposed on the surface of the antibody such that the residue is capable of being conjugated to a substance and wherein the antibody is immunochemically functional, the term immunochemically functional primarily referring to the antibody ' s ability to bind but also encompassing effector functions if these are present.
  • variable region of the antibody contains the antigen binding site
  • preferred sites for conjugation are the surface residues on d e surface of the variable region which are not involved in antigen binding such as, for example, the sSv heavy chain - light chain linker peptide as well as the surface residues of the constant region of the antibody which encompass the constant region of the light chain, the CH1.CH2, and CH3 domains of the heavy chain and also includes the hinge region.
  • all residues on the surface of the antibody which are not involved in antigen binding in particular those which are not part of the CDRs, are suitable for conjugation to a substance.
  • the preferred sites for conjugation are the same but excluding the CH2 domain which is known to contain the effector functions.
  • the cysteine residue is in a substantially reduced form. More preferably, die reduced cysteine residue is in the CH3 domain of the heavy chain and more preferably at position 442 within the CH3 domain. Another preferred position for the reduced cysteine residue is the heavy chain - light chain linker peptide.
  • Novel cys442 antibodies are capable of being expressed by their producer cells in a manner indicating both monomeric IgG and aggregated forms. Although the presence of aggregate suggested that the CVS442 variants were surface variants as had been observed in the works by Bodmer. et al. and by Shopes. surprisingly, most of the antibody was not in an aggregated form.
  • Monomeric IgG was readily purified, for example, by gel filtration chromatography, and the monomeric form was stable upon long term storage.
  • the monomeric IgG was found to possess no free thiol (Table 1 ). Whilst not wishing to be bound by theory, we believe that the thiol may be initially blocked (i.e. protected) by naturally occcurring adducts such as for example, glutathione.
  • the engineered thiol is reduced under controlled conditions that do not reduce the natural disulfide bonds. For example, milder conditions such as lower concentrations of the reductant which are not capable of reducing the natural disulfide bonds are found to be suitable for reducing the engineered thiol .
  • the reduced antibody sustains a monomeric form even when stored for prolonged periods of time at pH 8. (Thiols are known to be reactive by judicious manipulation of pH and oxygen).
  • the engineered antibody is rendered capable of site-directed chemical addition specifically at the engineered thiol.
  • the present invention is also directed to a monoclonal antibody comprising a cysteine residue exposed on the surface of d e antibody wherein, by controlled reduction, the antibody is rendered capable of site-directed chemical conjugation to a substance, said cysteine residue being introduced at a site which does not interfere with the immunochemical function of the antibody.
  • the antibodies according to the present invention are preferably monoclonal antibodies, or fragments thereof, the term antibody encompassing both antibodies and antibody fragments.
  • Antibodies according to the present invention can be from any species.
  • the antibodies may be chimaeric antibodies that have variable regions from one antibody and constant regions from another, such as a human antibody.
  • chimaeric antibodies may be species/species chimaeras or class/class chimaeras.
  • Such chimaeric antibodies may have one or more further modifications to improve antigen binding ability or to alter effector functioning.
  • Another form of altered antibody is a humanised antibody including a composite antibody, wherein the constant regions and die hypervariable regions other than the CDRs are transferred to the human framework.
  • Antibodies of the present invention include different isotypes such, for example, as Gl. G2. G4. Examples of antibodies are the 40KD antibody (CO/ 17.1. A) as disclosed in J. Cell Biol 125(2) 437-446. April 1994 and in Proc.Natl. Acad. Sci. 87, 3542-3546. May 1990, preferably the humanised anti-40 D antibody and in pa ⁇ icular humanised anti-40KD of the G4 isotype. A specific example of an anti-40KD antibody is 323/A3. preferably humanised 323/A3 and in particular humanised 323/A3 IgG4.
  • an antibody is an anti-folate recepter antibody as disclosed in A.Tomasetti et al. Federation of European Biochemical Societies Vol 317. 143-146, Feb 1993. preferably humanised anti-folate and in particular humanised anti-folate of the Gl isotype.
  • a specific example of an anti-folate antibody is MOV 18, preferably humanised MOV 18 IgGl.
  • Further examples of antibodies include anti-CEA. anti mucin. anti-20/200KD, anti-ganglioside, anti-digoxin. anti-CD4, anti-CD23, anti- CDw52 and more specifically Campath-IH which is a humanised anti-CDw52 antibody.
  • the antibody chain DNA sequences including die CDRs of Campati -1HTM are set out in EPO328404, die disclosure of which is hereby incorporated by reference. (Page, M.J., and Sydenham, M.A., High level expression of the humanised monoclonal antibody Campath-IH in Chinese Hamster Ovary cells. Biotech. 9: 64-68, 1991.).
  • Antibody fragments of use in the present invention include Fab, F(ab)2, Fv and fragments comprising synthetic peptide sequences eg. as generated by recombinant DNA technology.
  • Monoclonal antibodies of use in the invention may be prepared by any metJ od well known in the art or more particularly as described in GB 9022547.5. Purification may be carried out as described in EP-A-91917891.
  • Fragments may be prepared by any of the means known in the literature, for example Antibodies, a laboratory manual, eds. E. Harlow and D. Lane, Cold Spring Harbor Laboratory, 1988 or by molecular genetic means.
  • the invention also provides an antibody wherein the cysteine is conjugated eiti er directly or indirectly to a substance.
  • the substance When the substance is conjugated indirectly to the antibody it may be connected to the antibody via one or more linkage moieties such as for example chelators.
  • linkage moieties such as for example chelators.
  • Such substances which are connected to the antibody via one or more linkage moieties are commonly known as "bifunctional substances".
  • linkage moieties may, for example, be a functional chemical moiety such as maleimide or bromoacetyl. that is capable of covalent attachment to thiol functional groups within proteins such as antibodies.
  • the linkage moiety may also utilise a chemical spacer (e.g. a p-benzyl group) tiiat functions as a bridge between the substance(s) and the antibody attachment moiety.
  • conjugated substance examples include dyes, radionuclides. enzymes, drugs, cytotoxins. and biotin/avidin. Specific examples of a drug and cytotoxin being adriamvcin and ricin respectively. Specific examples of chelators include d e following.
  • PA-DOT A ( ⁇ -[2-(/7-nitrophenyl)ethyl]- 1 ,4,7, 10- 1 -acetic-4.7.10-tris(methylacetic)acid);
  • TMT (6,6"-bis[NN",N'"-tetra(carboxymethyl)aminomethyl)-4'-(3-amino-4- methoxyphenyl)-2.2':6'.2"-terpyridine);
  • IB4M-DTPA N,N',N" ! N"-pentakis(carboxymethyl)-2-[(4-aminophenyl)methyl]-6- memyldied ylenetriamine
  • TRITA (l,4,7,10-tetraazacyclotridecane-N,N',N",N'"-tetraacetic acid);
  • the substance may be attached to d e free thiol of d e antibody by eitiier direct or indirect methods.
  • 99 m ⁇ c may be directly attached to the antibody by a modification of procedures similar to the Schwartz method (Schwartz. A., and Steinstrasser. A., J. Nucl. Med. 18:721 , 1987), wherein reduction of the natural disulfides would not be necessary.
  • Metallic radionuclides such as 9 ⁇ t 186R C 177L U> H I In and ⁇ Cu may be attached by eidier indirect prelabelling methods, wherein radionuclide is first added to bifunctional chelator then conjugated to antibody, or by indirect postlabelling methods, wherein radionuclide is added to preformed chelator- antibody conjugate.
  • Chelators of the present invention may be linked to antibodies by any hetero- or homo-bifunctional cross linker (i.e. chemical spacer) capable of linking a chelator to a thiol group in the antibody (M.McCall et al.. Bioconjugate Chem. 1.
  • chelators in combination with cross-linkers include:
  • bromoacetyl-DOTA (2-[p-(bromoacetamido)benzyl- 1.4,7, 10-tetraazacyclo-dodecane-
  • bromacetyl-TRITA (2-[p-(bromoacetamido)benzyl]-l .4,7.10-tetraazacyclotridecane-
  • bromacetyl-DOTA also called BAD
  • BAD bromacetyl-DOTA
  • the chelator of use in the present invention is eidier DOTA or TMT and die protein reactive group (cross-linker) is either bromoacetyl or a maleimide. most preferably bromoacetyl.
  • Radionuclides which may be used in accordance witii die present invention include those appropriate for obtaining in vivo radio immunotherapy and/or imaging of a target cell or tissue.
  • Radioimmunotherapy a high dose of energy must be delivered to die target site in order that cellular DNA is damaged; both ⁇ and ⁇ emitting radionuclides produce emissions in a suitable energy range.
  • ⁇ -emitters are eidier shorter lived or decay to hazardous daughter products.
  • die radionuclide of choice for radioimmunotherapy will usually be a ⁇ -emitter.
  • radionuclide For imaging die radiation must interact as little as possible witii the body tissue yet produce a strong signal for external detection. Hence a gamma emitting radionuclide is most suitable for imaging.
  • the radionuclide For botii imaging and radioimmunodierapy the radionuclide must possess a half-life suitable to permit activity or detection after the elapsed time between administration and binding to the target site.
  • the radiolabelled antibody must travel from the bloodstream to d e extracellular fluids of the target via the endothelial pores. Large antibodies or antibody/chelator complexes may diffuse slowly and a radionuclide half-life of between several hours and several days is desirable.
  • the radionuclide is selected from the group comprising of 195 ⁇ , 57 ⁇ 1, 5?Co. 105 Ag, 68 Cu, 52M . 52 Fe, ⁇ ⁇ In. u 3m In ⁇ 99m Tc? 67 Ga . 1 ⁇ 166 Tm? l67 Tm . 146 Gd, 157 D y 5 95 mNb , 103 Ru , 97 Ru, 99 Ru , 101 mRh , 201 T1 . 203 Hg , 197 Hg , 203p b . 99 ⁇ 48 Cr . 57 Co , 125 ⁇ , 131 ⁇ , 35 S, 153 Sm, 88 ⁇ 90 ⁇ 186 Re, 188 Re , 211 AL 212 B i. 212 Pb and 177 Lu .
  • radionuclide is selected fr , om th . e group composing I l l, In, 67 ⁇ C,u, 186 D Re. 188 ⁇ Re. 177- Lu, 99 m ⁇ Tc. 131, I. 88 v Y,
  • the radionuclide is l ⁇ Lu 153 s m 90 ⁇ and U lin.
  • the invention also provides a radiolabelled antibody comprising an antibody of the present invention conjugated eidier directly or indirectly to a radionuclide, in particular chelator-antibody conjugate that may be labelled witii ⁇ Y or 17?Lu v a DOTA. or TMT.
  • the invention also provides metiiods for producing antibodies capable of being conjugated at specific sites and for site-directed conjugation of antibodies according to the invention.
  • a conjugated antibody of die invention in therapy and diagnosis.
  • antibodies according to the invention for the diagnosis and/or tiierapy of conditions which are detectable or amenable to dierapy with dyes, radionuclides enzymes, drugs and cytotoxins.
  • These antibody complexes are useful in treating cancers such as lymphomas and leukaemias and in particular small cell and non small cell lung cancer, prostatic cancer as well as ovarian cancer.
  • an antibody complex according to the invention for use in the imaging and/or treatment of cancers and associated metastases.
  • T-cell mediated disorders including severe vasculitis.
  • rheumatoid ardiritis, systemic lupis also autoimmune disorders such as multiple sclerosis, graft vs host disease, psoriasis, juvenile onset diabetes, Sjogrens' disease, thyroid disease, myasthenia gravis, transplant rejection and astiima.
  • the invention also provides the use of a conjugated antibody described above in the manufacture of a medicament for the treatment or imaging of any of the aformentioned disorders.
  • a method of treatment of conditions amenable to therapy and diagnosis witii a conjugated antibody complex according to d e invention comprising administering a tiierapeutically efficacious amount of antibody complex to a mammal requiring such treatment.
  • metiiods of treatment of cancers such as lymphomas and leukaemias and in particular small cell and non-small cell lung cancer, prostatic cancer as well as ovarian cancer and most particularly metiiods of treatment of cancers and associated metastases.
  • T-cell mediated disorders including severe vasculitis, rheumatoid arthritis, systemic lupis and also autoimmune disorders such as multiple sclerosis, graft vs host disease, psoriasis, juvenile onset diabetes, Sjogren's disease, thyroid disease, myasd enia gravis, transplant rejection and astiima.
  • compositions containing conjugated antibodies according to the present invention which comprise a conjugated monoclonal antibody or fragment tiiereof and one or more pharmaceutically acceptable excipients.
  • compositions include, in addition to conjugated antibodies a physiologically acceptable diluent or carrier possibly in admixture with odier agents such as other antibodies or an antibiotic.
  • Suitable carriers include but are not limited to physiological saline, phosphate buffered saline, phosphate buffered saline glucose and buffered saline. Routes of administration are routinely parenteral including intravenous, intramuscular, subcutaneous and intraperitoneal injection or delivery.
  • radioimmunod erapy die dosages of compositions containing antibody conjugated to radionuclides according to die invention will vary witii the condition being treated and the recipient of the treatment, but will be in the range of to about 1- lOOmg for an adult patient, preferably 1-lOmg, most preferably 5mg, usually administered as an infusion.
  • a repeat dosing regime may be preferable wherein 10 mg are administered for 1 day followed after weeks or months by a second treatment.
  • the dosages of such compositions will vary witii the condition being imaged and the recipient of die treatment, but will be in the range 1 to about lOOmg, preferably 1-10 and most preferably 5mg for an adult patient.
  • Kits can also be supplied for use with the subject conjugated antibodies in d e protection against or detection of a cellular activity or for the presence of a selected antigen.
  • a monoclonal antibody conjugated of the present invention may be provided, usually in a lyophilized form in a container, either alone or in conjunction with additional antibodies specific for the desired cell type.
  • the conjugated antibodies which may be conjugated to a dye, radionuclide.
  • tiiis will usually be present in a separate vial.
  • the second antibody is typically conjugated to a label and formulated in an analogous manner with die antibody formulations described above.
  • the kit will also contain a set of instructions for use.
  • FIG. 1 Immunoreactivitv of variant antibodies.
  • the two antibodies were biosyndietically labelled in situ with 3 ⁇ to the same specific activity (0.3 mCi/nmole) and purified (35S-ligand) as described above. Equilibrium competition was carried out in parallel where each ligand was competed for by its respective unlabelled form. Binding was performed in a total volume of 0.2ml containing 5 x 10 ⁇ fixed Wein 133 cells, -0.1 nM of the indicated radiolabeled form of C1H and the indicated concentration of competitor.
  • the binding buffer contained phosphate-buffered saline, 2% bovine calf serum. 0.01% triton XI 00, and 0.02% sodium azide (binding buffer) overnight at 4°.
  • the cells were centrifuged, washed 3 times with cold binding buffer and radioactivity determined. Points depict die average of triplicate measurements ⁇ SEM (bars).
  • the two antibodies were biosyntiietically labelled in situ witii 35$ o the same specific activity (0.3 mCi/nmole) and purified (35s-ligand) as described above. Equilibrium competition was carried out in parallel where each ligand was competed for by its respective unlabelled form. The two profiles indicate that the antibodies have identical antigen binding potencies.
  • diiol groups were introduced onto nonreduced antibody using the procedure of McCall, et al, Bioconjugate Chem. 1: 222-226, 1990. (The latter procedure is a random conjugation process tiiat employs 2-iminod ⁇ iolane to introduce thiol groups onto the ⁇ amino groups of lysine residues.) Both tiiiol- containing antibody preparations were then conjugated as described in Example 2.
  • Equal amounts of d e labelled conjugates were subjected to reducing SDS PAGE, wherein the antibody heavy and li ht chain subunits were separated by virtue of their size.
  • the gel was stained for protein (left side) and for radioactivity by autoradiography (right side).
  • the figure shows that the addition of the conjugate was localised to the heavy chain for the specific labelling procedure, whereas for the random process, botii subunits were labelled.
  • Example 1 production of variant monoclonal antibodies with a cvs substitution.
  • Campath-IH and anti-digoxin variants were introduced into the heavy chain of various isotypes by conventional molecular genetic means. For example, genetic constructs of human IgG2. IgG4, Campath-IH and anti-digoxin (botii IgGl) were obtained from within Wellcome Laboratories. The terminal portion of the CH3 region was excised with the restriction enzymes Nsil(5') and EcoRl(3') and replaced with an annealed double stranded oligonucleotide ligated at die respective restriction sites. Antigen specificity was introduced onto constant regions by respective replacement of the variable and CHI regions.
  • a ser228 ⁇ P r0 was introduced into IgG4 in order to match die G4 sequence initially reported by Pink, et al, Biochem. J. 117: 33-47, 1970.
  • An IgG4 cys442 variant entitled G4mc was further modified by changing tiiree residues in the CH2 region: leu235 ⁇ ala. gly237 ⁇ *ala and glu3 ⁇ g— ⁇ ala. these latter changes were introduced based on rationale supplied by Winter, et al. tiiat such changes might reduce antibody interaction with Fc gamma receptors and complement Clq (Duncan, et al, Nature 332: 563-564, 1988: Duncan and Winter, Nature 332: 738- 740. 1988).
  • (b) 323/A3 variant The murine antibody 323/A3 reacts witii an epitope on human epithelial tissues that may be useful in the identification of treatment of adenocarcinoma (Edwards, et al. Cancer Res. 46: 1306-1317, 1986).
  • the complimentarity determining regions within the variable region of 323/A3 were first "humanized” and grafted onto a human IgGI isotype. To prepare a cys variant, the cD ⁇ A expression construct was ligated in frame with d e cD ⁇ A encoding die constant region of Campath-IH IgG4 cys442 variant.
  • the humanized 323/A3 IgG4 cys442 variant was expressed in ⁇ SO cells and purified by conventional means.
  • (c) 323/A3 sFv fragment A single chain sFv fragment of humanized 323/A3 was constructed by conventional PCR and cloning techniques. Cys variant constructs were produced by introducing a cys residue substitutions into the linker region. For example, the conventional linker region (gly4ser three repeat) was altered to contain gly4serglv2cys2sergly4ser by site-directed mutagenesis. The variants were expressed in E. coli and purified by affinity chromatography.
  • Mov- 18 variant (d) Mov- 18 variant.
  • Mov- 18 reacts with a folate binding protein that is prominently expressed on ovarian cancer tissue (Miotti. et al., Intl. J. Cancer 39: 297-303. 1987).
  • a human IgGl isotype cDNA was cloned from a public source mRNA library by using reverse transcriptase.
  • the variable region of Mov- 18 was humanized and ligated to die human Gl constant region.
  • Cys442 was introduced into the heavy chain cDNA by site- directed mutagenesis.
  • the humanized Mov- 18 IgGl cys442 variant was expressed in NSO cells and purified by conventional means.
  • Antibody solutions were prepared in degassed buffer, such as 1 OOmM sodium phosphate or trimethylammonium phosphate at pH> 8.0, preferably pH 8.0 - 8.5. at a convenient concentration, for example 100 ⁇ M (15 mg/ml).
  • An amount of reductant was mixed witii the antibody solution to achieve the desired extent of reduction.
  • fixed volumes of a 50% gel slurry of a solid phase reductant such as Reduce-I ⁇ u-J (Pierce) were stined into the antibody solution.
  • the reductant capacity of Reduce- ImmTM g e j as assumed from the manufacturer to be 30 ⁇ mole/ml packed gel.
  • a 250 ⁇ l volume of 50% slurry was added to 1 ml of 100 ⁇ M antibody solution to generate a mixture with a 30-fold excess reductant capacity (mole reductant/mole antibody).
  • the antibody-reductant mixture was shaken for 1 hr at room temperature, centrifuged and the solution subjected to additional procedures such as free tiiiol determination. pH reduction, purification or conjugation as described in Example 3.
  • antibody protein was reduced with soluble reductant such as mercaptoediylamine.
  • soluble reductant such as mercaptoediylamine.
  • protein was concentrated to 200-300 ⁇ M in 0.1 M sodium phosphate, pH 6.0, 5 mM DTPA.
  • Mercaptoediylamine was added to a final concentration 10-fold in excess of the protein concentration and mixed gently for 1 hour at room temperature.
  • the reduced protein was then separated from reductant and prepared for conjugation by conventional means such as gel filtration.
  • protein solution was gel filtered through Bio Spin 30 columns (Bio-Rad Laboratories) that had been pre-equilibrated in 0.1 M tetrametiiylammonium phosphate pH 8.2. 25 uM DTPA for 2 min at 150 x g.
  • Example 3 Site directed corrugation of reduced variant antibody.
  • a reduced ser442 ⁇ cys variant, Campath-IH G4mc was conjugated to 2-[p- (bromoacetamido)benzyl]- 1 ,4,7, 10-tetraazacyclododecane-N,N'N",N" -tetraacetic acid (bromoacetyi-DOTA).
  • Bromoacetyi-DOTA was a bifunctional moiety wherein one aspect was a chelator that had been prelabelled with 5?Co and the second aspect was a reactive group capable of covalent attachment to a free sulfhydryi.
  • reduced protein at a concentration of 150 - 200 uM in tetrametiiylammonium phosphate, pH 8.2. 25 ⁇ M DTPA, was added to a 10-fold excess of bromoacetylTMT in a metal- free reaction vial. The reaction was carried out for 24 hours at room termperature. and die conjugate was isolated by gel filtration as described above.
  • random conjugate was prepared by introducing thiol groups into nonreduced Campatii- 1 H g4mc as has been described previously for Lym- 1. (The latter process follows the procedure of McCall, et al, Bioconjugate Chem.
  • Iminothiolated Campath-IH G4mc was conjugated to bromoacetyi-DOTA to the same extent (chelators per antibody) as site directed conjugate.
  • the conjugates were rendered radioactive by mixing with 9 ⁇ YC 13 in plastic ware by addition of the following reagents: an 8X volume of monoclonal antibody conjugate (> 25 mg/mL. 0.1 M ammonium acetate pH 6.7), a 2X volume of 90N. and cold yttrium up to a final concentration of 10 ⁇ M. Cold yttrium was added first.
  • a reduced ser442" ⁇ cys variant Campath-IH was conjugated to TMT, tiiat is covalently attached via a thioether linkage to the cys442 residues in die heavy chains as illustrated in Figure 4.
  • the reaction may be regulated to produce conjugates that contain an average of 1 - 2 chelators per antibody.
  • Conjugate is purified free of unreacted bifunctional chelator by gel filtration in metal-free conditions and is stable in a buffered, metal-free environment.
  • Conjugate chelation was radiolabeled in metal-free plasticware using the best metal-free reagents available.
  • Carrier-free 90 YC 13 was purchased from Dupont/New England Nuclear, Amersham and odier sources. The specific activity was typically 5 mCi in 10-30 ⁇ L 0.05 N HC1, specific activity 5.6 X 10 5 Ci/g. Prior to use, the 90 ⁇ ci was buffered witii 0.1 volume of 6 M ammonium acetate to ⁇ pH 5.8.
  • Chelation was performed by adding the following reagents in sequence and incubating for up to 90 min at room temperature: a IX volume of cold yttrium (lOO ⁇ M yttrium in 0.1 M ammonium acetate, pH 6.8.), a 2X volume of 9 ⁇ acetate, and an 8X volume of monoclonal antibody conjugate (25 mg/ml 0.1 M ammonium acetate, pH 6.5).
  • Non- chelated radiometal was "scavenged" by d e addition of DTPA to a final concentration of 500 ⁇ M and a 10X volume of 0.1 M ammonium citrate, pH 6.5.
  • the mixture was incubated at room temperature for 30 minutes and fractionated by "spin column gel filtration," i.e., applied to a 1 mL Bio-Spin 30 (Bio-Rad Laboratories) tiiat had been pre-equilibrated in phosphate-buffered saline and centrifuged at 150 x g for 2 minutes. Spin column gel filtration was repeated for a total of two centrifugations. The efficiency of chelation (ability to chelate all the radiometal) and scavenging (ability to remove non-chelated radioactivity from radiolabeled conjugate) was monitored by thin layer chromatography as described by Meares et al, Anal. Biochem. 142, 68-78. 1984.
  • Example 7 Large scale site directed co ⁇ iugation of reduced variant antibody.
  • G4 ser442 ⁇ cys variant labelled "G4mc" and a natural G4 control were exposed to solid phase reductant as described in Example 2.
  • the thiol content was determined by Ellman's reagent and is expressed relative to moles antibody. (It is assumed tiiat 2 moles of tiiiol were reduced per antibody ti iol.)
  • the sem for triplicate measurements was ⁇ 0.1 SH/antibody.

Abstract

The present invention relates to antibodies which are capable of being conjugated at specific sites, to processes for the site-directed conjugation of such antibodies, to antibodies that have been conjugated at a specific site and to the use of such antibodies in therapy and diagnosis.

Description

ANTIBODIES
The present invention relates to antibodies which are capable of being conjugated at specific sites, to processes for the site-directed conjugation of such antibodies, to antibodies that have been conjugated at a specific site and to the use of such antibodies in therapy and diagnosis.
Antibodies are globular proteins which represent a vital component of the mammalian immune response to foreign disease inducing agents. Antibodies may be manufactured ex vivo by any of a number of methods, and such antibodies, particularly monoclonal antibodies and/or fragments thereof, have proved valuable as both diagnostic and therapeutic agents. The utility of antibodies stems from their unique antigen specificity, i.e.. their ability to chemically recognise and remain bound to discrete chemical moieties such as pathogen antigens or tumour-associated antigens. Another aspect of their utility is their diversity, i.e., the ability of mammals (and now other processes such as phage display) to create a very large variety of discrete, genetically defined antibodies (monoclonal antibodies). A final aspect of their utility is their capacity to interact via their "constant" regions. This latter aspect determines other sets of properties, for example, those properties common to isotypes such as interaction with effector cells, complement or other binding moieties like protein A.
The usefulness of an antibody or fragment may be enhanced by chemically coupling one or more further molecular moieties (referred to herein as substances) either directly or indirectly that convey properties to the conjugate that are not naturally present in the constituents alone. Such properties may include a reporter function such as a dye or a radionuclide, an enzymatic function, a second binding function (such as with biotin- avidin), a drug (such as adriamycin), a cytotoxic function (such as with ricin), a chelator, or a chemical linkage moiety that may, in turn render the antibody capable of subsequent reaction with any of a variety of molecular moieties. Thus an antibody conjugate comprises an antibody and an active substance (substance) which is either directly or indirectly conjugated to the antibody. For example, when the active substance is a radionuclide, the radionuclide may be directly conjugated to the antibody or alternatively it may be indirectly conjugated to the antibody via a chelator such as for example TMT or even a chelator which is in turn linked to the antibody by a further protein reactive group (cross-linker) such as. for example, bromoacetyl. Similarly, the active substance could be a molecular chimaera for use in enzyme-prodrug therapy, such a chimaera comprises a transcriptional regulatory DNA sequence capable of being activated in a mammalian cell such as a cancer cell and a DNA sequence operatively linked to the transcriptional regulatory DNA sequence and encoding a heterologous enzyme capable of catalysing the conversion of a prodrug which is administered subsequently,into an agent toxic to the cancer cell. This molecular chimera may be directly conjugated to the antibody or alternatively, it may be contained within a viral vector or liposome with the viral vector or liposome then being attached to the antibody (see European application No.90309430.8).
Many means have been described for the chemical addition or crosslinking of molecular entities to antibodies and their fragments, and these reactions are commonly referred to as conjugation reactions. Conjugation reactions exploit the chemical functional groups that occur naturally in the antibody. For example, a common approach is to target primary amines (mainly the ε amino groups of lysine residues). Another common example of this approach includes the iodination (such as l-^I) of tyrosine residues. A disadvantage of this approach is that it is random. That is, lysine or tyrosine residues may occur throughout the structure and therefore the natural properties that these residues help convey ~ such as, for example, antigen recognition, complement reactivity or effector cell interaction — may be compromised. The behaviour of the conjugate is the average of the behaviour of all the individual unique components, some of which may be entirely useless or detrimental. The random approach is also known to affect antigen recognition.
Site-directed conjugation, i.e.. conjugation to a specific amino acid residue within the antibody structure, would convey the advantage that the resulting conjugate is not a mixture of different products. The properties conveyed by the antibody to the conjugate, such as antigen reactivity or pharmacokinetic stability, can thus be ascribed to a defined chemical structure. Furthermore, the site may be selected so as to be spatially removed from areas known to convey antigen-binding properties. For example, the variable region of an antibody is known to contain the antigen binding site and the CH2 domain of the heavy chain is known to contain effector (FcR) and complement (Clq) interactive residues. Thus, additions to the CH3 domain of the heavy chain may avoid compromising these functions. - j -
Two examples of site-directed conjugation procedures to antibodies have been described. The conjugation of antibodies to the carbohydrate portion of antibodies has been detailed (O'Shannessy and Quarles. J. Immunol. Met 99: 153-161, 1987). Human IgG antibodies have one common glycosylation site at asn297 in the CH2 domain. (Note that all antibody amino acid residues described herein are numbered according to the EU index. Kabat, et al.. Sequences of proteins of immunological interest, 5th ed.. NIH publication No. 91-3242, 1991. The common isotypes are referred to as Gl, G2 G4 etc.). The glycosylation conjugation procedure presents the risk of altering carbohydrate that might be required for antigen or effector interaction (Lund, et al., Molec. Immunmol 27: 1 145, 1990; Isaacs, et al. J. Immunol. 148: 3062, 1992).
Another example of site-directed conjugation to antibodies involves the creation, by site- directed mutagenesis, of a free thiol on the antibody. Antibodies naturally contain cvstine residues whose thiol groups are joined by disulfide bridges. The position of the naturally occurring cvstine residues is highly conserved among species indicating that these residues are essential for the structure and function of antibodies. Antibodies do not naturally contain free sulfhydryi groups. It is hypothetically attractive to engineer an antibody to possess a cysteine the thiol group of which is neither oxidised nor compromises the fidelity of the natural sulfhydryi bridges. Sulfhydryi groups of cysteine residues may of course be exploited for conjugation by numerous conjugation chemistries that are rather specific for sulfhydryls such as maleimides. alkyl and aryl halides, α-haloacyls and pyridyl disulfides. However, it has been determined that variant monoclonal antibodies that have been designed with unnatural cysteine residues do not de facto possess free thiol groups available for conjugation. (Since antibodies consist of two identical heavy and two identical light chains and depending on how the gene has been engineered and expressed, genetic replacement of a residue on one chain can result in two new residues in the complete H2-L2 structure; furthermore, the two residues need not be chemically identical due to their spatial arrangement and neighbourhood.) In one known example of an engineered thiol (Bodmer. et al. US # 5,219,996 and Lyons, et al, Protein Eng. 3: 703, 1990), an available free thiol was observed only when a cysteine residue was introduced into a discreet "concave" molecular pocket within the antibody structure — a pocket only accessible to small molecules (0.13 - 0.5 mm diameter) and inaccessible for forming disulfide bridges within the same or other antibody chains. Cysteine residues introduced on "convex" or "flat" surfaces of the monoclonal antibodv were found not to contain free thiols. One s illed in the an would therefore conclude that surface cysteine residues would form disulfide bridges within the same and/or another antibody chain and thus distort the macromolecular structure and functions of the antibody.
The published literature indicates that engineered surface thiols alter antibody structure and function. For example, both monomers and dimers (IgG-IgG) were observed in the crude antibody-producing cell supernatant of a ser444→cys variant (Shopes, J. Immunol. 148: 2918. 1992). Relatedly, a procedure was described for the production of dimeric antibodies also using a ser444→cys variant (Caron, et al, J. Exp. Med. 176: 1 191-1 195, 1992). The dimeric IgGs from both of these examples were found to have enhanced effector functions in vitro. In these examples and in the earlier example of Bodmer, et al, no physicochemical evidence was provided to indicate whether dimers formed specifically at the engineered site or to what extent normal disulfide bridges may have been altered. Finally, a "tethered" antibody has been described that was produced by creating a se 19— »cys variant (Shopes, Mol. Immunol 30: 603, 1993). The variant allegedly generated dimers and an interchain structural variant or "tethered" antibody. Mixtures of structural aberrations compromise the goal of producing a defined chemical entity.
It was therefore with some surprise and contrary to expectation that we found that monoclonal antibodies containing a cysteine residue exposed on the surface of the antibody, are, in fact, amenable to site-directed conjugation, especially those containing a ser442→cys heavy chain variant.
Accordingly, the present invention provides a monoclonal antibody comprising a cysteine residue exposed on the surface of the antibody such that the residue is capable of being conjugated to a substance and wherein the antibody is immunochemically functional, the term immunochemically functional primarily referring to the antibody's ability to bind but also encompassing effector functions if these are present. Bearing in mind that the variable region of the antibody contains the antigen binding site the preferred sites for conjugation are the surface residues on d e surface of the variable region which are not involved in antigen binding such as, for example, the sSv heavy chain - light chain linker peptide as well as the surface residues of the constant region of the antibody which encompass the constant region of the light chain, the CH1.CH2, and CH3 domains of the heavy chain and also includes the hinge region. Thus all residues on the surface of the antibody which are not involved in antigen binding, in particular those which are not part of the CDRs, are suitable for conjugation to a substance. If the antibody has effector functions, the preferred sites for conjugation are the same but excluding the CH2 domain which is known to contain the effector functions. The cysteine residue is in a substantially reduced form. More preferably, die reduced cysteine residue is in the CH3 domain of the heavy chain and more preferably at position 442 within the CH3 domain. Another preferred position for the reduced cysteine residue is the heavy chain - light chain linker peptide. Novel cys442 antibodies are capable of being expressed by their producer cells in a manner indicating both monomeric IgG and aggregated forms. Although the presence of aggregate suggested that the CVS442 variants were surface variants as had been observed in the works by Bodmer. et al. and by Shopes. surprisingly, most of the antibody was not in an aggregated form. Monomeric IgG was readily purified, for example, by gel filtration chromatography, and the monomeric form was stable upon long term storage. The monomeric IgG was found to possess no free thiol (Table 1 ). Whilst not wishing to be bound by theory, we believe that the thiol may be initially blocked (i.e. protected) by naturally occcurring adducts such as for example, glutathione.
We have discovered that the engineered thiol is reduced under controlled conditions that do not reduce the natural disulfide bonds. For example, milder conditions such as lower concentrations of the reductant which are not capable of reducing the natural disulfide bonds are found to be suitable for reducing the engineered thiol . The reduced antibody sustains a monomeric form even when stored for prolonged periods of time at pH 8. (Thiols are known to be reactive by judicious manipulation of pH and oxygen). Thus, by controlled reduction, the engineered antibody is rendered capable of site-directed chemical addition specifically at the engineered thiol.
Thus the present invention is also directed to a monoclonal antibody comprising a cysteine residue exposed on the surface of d e antibody wherein, by controlled reduction, the antibody is rendered capable of site-directed chemical conjugation to a substance, said cysteine residue being introduced at a site which does not interfere with the immunochemical function of the antibody.
The antibodies according to the present invention are preferably monoclonal antibodies, or fragments thereof, the term antibody encompassing both antibodies and antibody fragments. Antibodies according to the present invention can be from any species. The antibodies may be chimaeric antibodies that have variable regions from one antibody and constant regions from another, such as a human antibody. Thus, chimaeric antibodies may be species/species chimaeras or class/class chimaeras. Such chimaeric antibodies may have one or more further modifications to improve antigen binding ability or to alter effector functioning. Another form of altered antibody is a humanised antibody including a composite antibody, wherein the constant regions and die hypervariable regions other than the CDRs are transferred to the human framework. Additional amino acids in the framework or constant regions of such antibodies may be altered if required to restore binding. Thus the antibodies of use in the present invention include any altered antibodies in which the amino acid sequence is not one which exists in nature. However, CDR-grafted antibodies are most preferred. Antibodies of the present invention include different isotypes such, for example, as Gl. G2. G4. Examples of antibodies are the 40KD antibody (CO/ 17.1. A) as disclosed in J. Cell Biol 125(2) 437-446. April 1994 and in Proc.Natl. Acad. Sci. 87, 3542-3546. May 1990, preferably the humanised anti-40 D antibody and in paπicular humanised anti-40KD of the G4 isotype. A specific example of an anti-40KD antibody is 323/A3. preferably humanised 323/A3 and in particular humanised 323/A3 IgG4.
Another example of an antibody is an anti-folate recepter antibody as disclosed in A.Tomasetti et al. Federation of European Biochemical Societies Vol 317. 143-146, Feb 1993. preferably humanised anti-folate and in particular humanised anti-folate of the Gl isotype. A specific example of an anti-folate antibody is MOV 18, preferably humanised MOV 18 IgGl. Further examples of antibodies include anti-CEA. anti mucin. anti-20/200KD, anti-ganglioside, anti-digoxin. anti-CD4, anti-CD23, anti- CDw52 and more specifically Campath-IH which is a humanised anti-CDw52 antibody. The antibody chain DNA sequences including die CDRs of Campati -1H™ are set out in EPO328404, die disclosure of which is hereby incorporated by reference. (Page, M.J., and Sydenham, M.A., High level expression of the humanised monoclonal antibody Campath-IH in Chinese Hamster Ovary cells. Biotech. 9: 64-68, 1991.).
Antibody fragments of use in the present invention include Fab, F(ab)2, Fv and fragments comprising synthetic peptide sequences eg. as generated by recombinant DNA technology. Monoclonal antibodies of use in the invention may be prepared by any metJ od well known in the art or more particularly as described in GB 9022547.5. Purification may be carried out as described in EP-A-91917891.
Fragments may be prepared by any of the means known in the literature, for example Antibodies, a laboratory manual, eds. E. Harlow and D. Lane, Cold Spring Harbor Laboratory, 1988 or by molecular genetic means.
The invention also provides an antibody wherein the cysteine is conjugated eiti er directly or indirectly to a substance. When the substance is conjugated indirectly to the antibody it may be connected to the antibody via one or more linkage moieties such as for example chelators. Such substances which are connected to the antibody via one or more linkage moieties are commonly known as "bifunctional substances". Such linkage moieties may, for example, be a functional chemical moiety such as maleimide or bromoacetyl. that is capable of covalent attachment to thiol functional groups within proteins such as antibodies. The linkage moiety may also utilise a chemical spacer (e.g. a p-benzyl group) tiiat functions as a bridge between the substance(s) and the antibody attachment moiety.
Examples of the conjugated substance are dyes, radionuclides. enzymes, drugs, cytotoxins. and biotin/avidin. Specific examples of a drug and cytotoxin being adriamvcin and ricin respectively. Specific examples of chelators include d e following.
DOTA ( 1.4,7, 10-tetraazacyclododecane-NN' N",N" -tetraacetic acid):
PA-DOT A(α-[2-(/7-nitrophenyl)ethyl]- 1 ,4,7, 10- 1 -acetic-4.7.10-tris(methylacetic)acid);
TMT (6,6"-bis[NN",N'"-tetra(carboxymethyl)aminomethyl)-4'-(3-amino-4- methoxyphenyl)-2.2':6'.2"-terpyridine);
IB4M-DTPA (NN,N',N"!N"-pentakis(carboxymethyl)-2-[(4-aminophenyl)methyl]-6- memyldied ylenetriamine);
CHX-A-DTPA (-^-[2----mino-3-( -aminobenzyl)propyl]-trαrø-cyclohexane-1.2-diamine-
NN',N',N",N"-pentaacetic acid);
TRITA (l,4,7,10-tetraazacyclotridecane-N,N',N",N'"-tetraacetic acid); and
TETA 1,4.8.1 l-tetraazacyclotetradecane-NN',N",N" -tetraacetic acid When the substance is a radionuclide, it may be attached to d e free thiol of d e antibody by eitiier direct or indirect methods. For example, 99mχc may be directly attached to the antibody by a modification of procedures similar to the Schwartz method (Schwartz. A., and Steinstrasser. A., J. Nucl. Med. 18:721 , 1987), wherein reduction of the natural disulfides would not be necessary. Metallic radionuclides such as 9 γt 186RC 177LU> H I In and ^^Cu may be attached by eidier indirect prelabelling methods, wherein radionuclide is first added to bifunctional chelator then conjugated to antibody, or by indirect postlabelling methods, wherein radionuclide is added to preformed chelator- antibody conjugate. Chelators of the present invention may be linked to antibodies by any hetero- or homo-bifunctional cross linker (i.e. chemical spacer) capable of linking a chelator to a thiol group in the antibody (M.McCall et al.. Bioconjugate Chem. 1. 222- 226, 1990) or by use of cross linkers described in Pierce "Immuno Technology Catalog and Handbook" 1992, pages E8 to E39 and in Parker. Chem.Soc.Rev. [1990], __, 271- 291 and the metiiods referred to therein.
Examples of chelators in combination with cross-linkers include:
bromoacetyl-DOTA (2-[p-(bromoacetamido)benzyl- 1.4,7, 10-tetraazacyclo-dodecane-
NN',N",N'"-tetraacetic acid); bromacetyl-TRITA (2-[p-(bromoacetamido)benzyl]-l .4,7.10-tetraazacyclotridecane-
N.N, N", N'"-tetraacetic acid); bromoacetyl-TMT (2-(^-(bromoacetamido)benzyl]-6.6''-bis[N.N"N''- tetra(carboxymethyl)aminomethyl)-4'-(3-amino-4'-methoxyphenyl)-2.2' : 6'.2 "- terpyridine);
The bifunctional chelator cross-linker combination. bromacetyl-DOTA, also called BAD, is described in M.J. McCall, H. Diril. and C.F. Meares, Bioconjugate Chem. 1: 222-226. 1990
Most particularly the chelator of use in the present invention is eidier DOTA or TMT and die protein reactive group (cross-linker) is either bromoacetyl or a maleimide. most preferably bromoacetyl.
Chelators of use in the present invention may be prepared by any known technique (for example M.McCall et al.. ibid) Radionuclides which may be used in accordance witii die present invention include those appropriate for obtaining in vivo radio immunotherapy and/or imaging of a target cell or tissue. For radioimmunotherapy, a high dose of energy must be delivered to die target site in order that cellular DNA is damaged; both α and β emitting radionuclides produce emissions in a suitable energy range. However α-emitters are eidier shorter lived or decay to hazardous daughter products. Hence die radionuclide of choice for radioimmunotherapy will usually be a β-emitter. For imaging die radiation must interact as little as possible witii the body tissue yet produce a strong signal for external detection. Hence a gamma emitting radionuclide is most suitable for imaging. For botii imaging and radioimmunodierapy the radionuclide must possess a half-life suitable to permit activity or detection after the elapsed time between administration and binding to the target site. The radiolabelled antibody must travel from the bloodstream to d e extracellular fluids of the target via the endothelial pores. Large antibodies or antibody/chelator complexes may diffuse slowly and a radionuclide half-life of between several hours and several days is desirable. In a particular aspect of die present invention the radionuclide is selected from the group comprising of 195^^, 57^1, 5?Co. 105Ag, 68Cu, 52M . 52Fe, ι πIn. u3mInι 99mTc? 67Ga. 1 ^ 166Tm? l67Tm. 146Gd, 157Dy5 95mNb, 103Ru, 97Ru, 99Ru, 101mRh, 201T1. 203Hg, 197Hg, 203pb. 99^ 48Cr. 57Co, 125τ, 131τ, 35S, 153Sm, 88γ 90γ 186Re, 188Re, 211AL 212Bi. 212Pb and 177Lu.
In a more particular embodiment of the present invention d e radionuclide is selected fr , om th . e group composing I l l, In, 67^ C,u, 186D Re. 188ϋ Re. 177- Lu, 99 m τTc. 131, I. 88v Y,
90γ. 21 1 At 212Bi 212Pb 57Co, 153Sm., 88γ 90γ md 177Lu.
In a more particular embodiment of the present invention the radionuclide is l^Lu 153 sm 90γ and U lin. The invention also provides a radiolabelled antibody comprising an antibody of the present invention conjugated eidier directly or indirectly to a radionuclide, in particular chelator-antibody conjugate that may be labelled witii υY or 17?Lu v a DOTA. or TMT. The invention also provides metiiods for producing antibodies capable of being conjugated at specific sites and for site-directed conjugation of antibodies according to the invention.
According to another aspect of the present invention there is provided the use of a conjugated antibody of die invention in therapy and diagnosis. In particular there is prύvided the use of antibodies according to the invention for the diagnosis and/or tiierapy of conditions which are detectable or amenable to dierapy with dyes, radionuclides enzymes, drugs and cytotoxins. These antibody complexes are useful in treating cancers such as lymphomas and leukaemias and in particular small cell and non small cell lung cancer, prostatic cancer as well as ovarian cancer.. In a most particular aspect of the present invention there is provided an antibody complex according to the invention for use in the imaging and/or treatment of cancers and associated metastases. They may also be used for example as immunosuppressives and more particularly for the treatment of T-cell mediated disorders including severe vasculitis. rheumatoid ardiritis, systemic lupis, also autoimmune disorders such as multiple sclerosis, graft vs host disease, psoriasis, juvenile onset diabetes, Sjogrens' disease, thyroid disease, myasthenia gravis, transplant rejection and astiima.
The invention also provides the use of a conjugated antibody described above in the manufacture of a medicament for the treatment or imaging of any of the aformentioned disorders.
According to anoti er aspect of the present invention there is provided a method of treatment of conditions amenable to therapy and diagnosis witii a conjugated antibody complex according to d e invention comprising administering a tiierapeutically efficacious amount of antibody complex to a mammal requiring such treatment. In particular there are provided metiiods of treatment of cancers such as lymphomas and leukaemias and in particular small cell and non-small cell lung cancer, prostatic cancer as well as ovarian cancer and most particularly metiiods of treatment of cancers and associated metastases. They may also be used in a metiiod of treatment of T-cell mediated disorders including severe vasculitis, rheumatoid arthritis, systemic lupis and also autoimmune disorders such as multiple sclerosis, graft vs host disease, psoriasis, juvenile onset diabetes, Sjogren's disease, thyroid disease, myasd enia gravis, transplant rejection and astiima.
There is also provided in the present invention a pharmaceutically acceptable composition containing conjugated antibodies according to the present invention which comprise a conjugated monoclonal antibody or fragment tiiereof and one or more pharmaceutically acceptable excipients.
Such compositions include, in addition to conjugated antibodies a physiologically acceptable diluent or carrier possibly in admixture with odier agents such as other antibodies or an antibiotic. Suitable carriers include but are not limited to physiological saline, phosphate buffered saline, phosphate buffered saline glucose and buffered saline. Routes of administration are routinely parenteral including intravenous, intramuscular, subcutaneous and intraperitoneal injection or delivery.
In respect of radioimmunod erapy die dosages of compositions containing antibody conjugated to radionuclides according to die invention will vary witii the condition being treated and the recipient of the treatment, but will be in the range of to about 1- lOOmg for an adult patient, preferably 1-lOmg, most preferably 5mg, usually administered as an infusion. A repeat dosing regime may be preferable wherein 10 mg are administered for 1 day followed after weeks or months by a second treatment.
In respect of imaging the dosages of such compositions will vary witii the condition being imaged and the recipient of die treatment, but will be in the range 1 to about lOOmg, preferably 1-10 and most preferably 5mg for an adult patient.
Kits can also be supplied for use with the subject conjugated antibodies in d e protection against or detection of a cellular activity or for the presence of a selected antigen. Thus, a monoclonal antibody conjugated of the present invention may be provided, usually in a lyophilized form in a container, either alone or in conjunction with additional antibodies specific for the desired cell type. The conjugated antibodies, which may be conjugated to a dye, radionuclide. enzyme, drug, cytotoxin, chelator or biotin/avidin, are included in die kits witii buffers, such as Tris, phosphate, carbonate, etc., stabilisers, biocides, inert proteins, e.g., serum albumin, or the like, and a set of instructions for use. Generally, these materials will be present in less tiian about 5% wt. based on the amount of active antibody, and usually present in total amount of at least about 0.001% wt. based again on the antibody concentration. Frequently, it will be desirable to include an inert extender or excipient to dilute die active ingredients, where the excipient may be present in from about 1 to 99% wt. of the total composition. Where a second antibody capable of binding to the chimeric antibody is employed in an assay, tiiis will usually be present in a separate vial. The second antibody is typically conjugated to a label and formulated in an analogous manner with die antibody formulations described above. Generally the kit will also contain a set of instructions for use.
Description of Figures
Figure 1 . Immunoreactivitv of variant antibodies.
a) Five distinct antibody preparations bearing the Campath-IH antigen specificity — 2 natural isotypes (Gl and G4) and 3 variants (G4m. G4c and G4mc), the G4c and G4mc containing ser 442 cys substitutions — were constructed and expressed in NSO cells as described in example 1. The antibodies were labelled in situ witii ^^S and purified in the following manner. Two x 10 ' washed producer cells were incubated for 48-72hrs at 37° in 5ml methionine cystine-free DMEM (ICN Biomedicals. Costa Mesa. CA) that contained 6μg cystine/ml. 3μg methionine/ml. and 8-10mCi of 3 s methionine eg. Tran ^S-label (ICN). The supernatant was harvested by centrifugation. dialized and concentrated (Centricon, Amicon. Beverly, MA). The antibody was purified by protein A chromatography (HPLC Dynamax Hydropore-protein A mini column [Rainin, Woburn. MA]), eluted witii 1 M acetic acid, concentrated by ultrafiltration eg. verus a PM 30 membrane (Amicon) and further fractionated by HPLC gel filtration eg. S-5 200A diol, YMC, Willmington. NC. The protein A eluants resolved on gel filtration commensurate with unlabelled antibodies and had specific activites ~ 2μCi/μg. Immunoreactivitv was determined on Fixed Wein 133 Cl cells using the method of Lindmo et al. (J. Immunol Meth. 72: 77-84, 1984). Note that the v intercept, die inverse of which is a measure of immunoreactivity, was equivalent for all the preparations. (The slopes are a function of the specific activity and not d e immunoreactivity.) b) Equilibrium specific binding competition for Campath-IH Gl and G4mc on fixed Wein 133 Cl cells. The two antibodies were biosyndietically labelled in situ with 3 § to the same specific activity (0.3 mCi/nmole) and purified (35S-ligand) as described above. Equilibrium competition was carried out in parallel where each ligand was competed for by its respective unlabelled form. Binding was performed in a total volume of 0.2ml containing 5 x 10^ fixed Wein 133 cells, -0.1 nM of the indicated radiolabeled form of C1H and the indicated concentration of competitor. The binding buffer contained phosphate-buffered saline, 2% bovine calf serum. 0.01% triton XI 00, and 0.02% sodium azide (binding buffer) overnight at 4°. The cells were centrifuged, washed 3 times with cold binding buffer and radioactivity determined. Points depict die average of triplicate measurements ± SEM (bars). The two antibodies were biosyntiietically labelled in situ witii 35$ o the same specific activity (0.3 mCi/nmole) and purified (35s-ligand) as described above. Equilibrium competition was carried out in parallel where each ligand was competed for by its respective unlabelled form. The two profiles indicate that the antibodies have identical antigen binding potencies.
More definitive evidence for site-directed conjugation was obtained by radiolabeling a monoclonal antibody chelator conjugate prepared as described in Example 3. Peptide mapping (enzymatic digestion, fractionation of peptides and peptide amino acid sequence analysis) determined that all radioactive peaks were composed of heavy chain. C-teπninal peptide fragments that contained cys442 -chelator adduct.
Figure 2. Site specific conjugation as indicated bv SDS PAGE.
A ser442→ cys variant, Campath-IH G4mc, was specifically reduced as described in Example 2. By way of comparison, diiol groups were introduced onto nonreduced antibody using the procedure of McCall, et al, Bioconjugate Chem. 1: 222-226, 1990. (The latter procedure is a random conjugation process tiiat employs 2-iminodιiolane to introduce thiol groups onto the ε amino groups of lysine residues.) Both tiiiol- containing antibody preparations were then conjugated as described in Example 2. Equal amounts of d e labelled conjugates were subjected to reducing SDS PAGE, wherein the antibody heavy and li ht chain subunits were separated by virtue of their size. The gel was stained for protein (left side) and for radioactivity by autoradiography (right side). Lanes, left to right: Random, protein stain; specific, protein stain; random, autorad; specific, autorad. The figure shows that the addition of the conjugate was localised to the heavy chain for the specific labelling procedure, whereas for the random process, botii subunits were labelled.
Figure 3.
Biodistribution of 90Y-TMT-Campath-1H G4mc conjugate. Reduced Campath-IH G4mc was conjugated to bromoacetyl-TMT, radiolabeled witii 90γcι-, and biodistribution carried out in tumor-bearing mice as described above for bromoacetyl- DOTA. bars show die average % injected dose/g tissue (% ID/g) conected for decay for 5 mice.
The following Examples are illustrative of the present invention and not intended to constitute any limitation thereof:
Example 1 production of variant monoclonal antibodies with a cvs substitution.
(a) Campath-IH and anti-digoxin variants. CVS442 was introduced into the heavy chain of various isotypes by conventional molecular genetic means. For example, genetic constructs of human IgG2. IgG4, Campath-IH and anti-digoxin (botii IgGl) were obtained from within Wellcome Laboratories. The terminal portion of the CH3 region was excised with the restriction enzymes Nsil(5') and EcoRl(3') and replaced with an annealed double stranded oligonucleotide ligated at die respective restriction sites. Antigen specificity was introduced onto constant regions by respective replacement of the variable and CHI regions. A ser228~ Pr0 was introduced into IgG4 in order to match die G4 sequence initially reported by Pink, et al, Biochem. J. 117: 33-47, 1970. An IgG4 cys442 variant entitled G4mc was further modified by changing tiiree residues in the CH2 region: leu235→ala. gly237~ *ala and glu3 \ g— ►ala. these latter changes were introduced based on rationale supplied by Winter, et al. tiiat such changes might reduce antibody interaction with Fc gamma receptors and complement Clq (Duncan, et al, Nature 332: 563-564, 1988: Duncan and Winter, Nature 332: 738- 740. 1988).
(b) 323/A3 variant. The murine antibody 323/A3 reacts witii an epitope on human epithelial tissues that may be useful in the identification of treatment of adenocarcinoma (Edwards, et al. Cancer Res. 46: 1306-1317, 1986). The complimentarity determining regions within the variable region of 323/A3 were first "humanized" and grafted onto a human IgGI isotype. To prepare a cys variant, the cDΝA expression construct was ligated in frame with d e cDΝA encoding die constant region of Campath-IH IgG4 cys442 variant. The humanized 323/A3 IgG4 cys442 variant was expressed in ΝSO cells and purified by conventional means.
(c) 323/A3 sFv fragment. A single chain sFv fragment of humanized 323/A3 was constructed by conventional PCR and cloning techniques. Cys variant constructs were produced by introducing a cys residue substitutions into the linker region. For example, the conventional linker region (gly4ser three repeat) was altered to contain gly4serglv2cys2sergly4ser by site-directed mutagenesis. The variants were expressed in E. coli and purified by affinity chromatography.
(d) Mov- 18 variant. Mov- 18 reacts with a folate binding protein that is prominently expressed on ovarian cancer tissue (Miotti. et al., Intl. J. Cancer 39: 297-303. 1987). A human IgGl isotype cDNA was cloned from a public source mRNA library by using reverse transcriptase. The variable region of Mov- 18 was humanized and ligated to die human Gl constant region. Cys442 was introduced into the heavy chain cDNA by site- directed mutagenesis. The humanized Mov- 18 IgGl cys442 variant was expressed in NSO cells and purified by conventional means.
Example 2. Reduction of variant monoclonal antibodies.
Antibody solutions were prepared in degassed buffer, such as 1 OOmM sodium phosphate or trimethylammonium phosphate at pH> 8.0, preferably pH 8.0 - 8.5. at a convenient concentration, for example 100 μM (15 mg/ml). An amount of reductant was mixed witii the antibody solution to achieve the desired extent of reduction. For example, fixed volumes of a 50% gel slurry of a solid phase reductant such as Reduce-Iπu-J (Pierce) were stined into the antibody solution. The reductant capacity of Reduce- ImmTM gej as assumed from the manufacturer to be 30 μmole/ml packed gel. A 250 μl volume of 50% slurry was added to 1 ml of 100 μM antibody solution to generate a mixture with a 30-fold excess reductant capacity (mole reductant/mole antibody). The antibody-reductant mixture was shaken for 1 hr at room temperature, centrifuged and the solution subjected to additional procedures such as free tiiiol determination. pH reduction, purification or conjugation as described in Example 3.
Alternatively, antibody protein was reduced with soluble reductant such as mercaptoediylamine. For example, protein was concentrated to 200-300 μM in 0.1 M sodium phosphate, pH 6.0, 5 mM DTPA. Mercaptoediylamine was added to a final concentration 10-fold in excess of the protein concentration and mixed gently for 1 hour at room temperature. The reduced protein was then separated from reductant and prepared for conjugation by conventional means such as gel filtration. Commonly, protein solution was gel filtered through Bio Spin 30 columns (Bio-Rad Laboratories) that had been pre-equilibrated in 0.1 M tetrametiiylammonium phosphate pH 8.2. 25 uM DTPA for 2 min at 150 x g.
Example 3. Site directed corrugation of reduced variant antibody. A reduced ser442→ cys variant, Campath-IH G4mc, was conjugated to 2-[p- (bromoacetamido)benzyl]- 1 ,4,7, 10-tetraazacyclododecane-N,N'N",N" -tetraacetic acid (bromoacetyi-DOTA). Bromoacetyi-DOTA was a bifunctional moiety wherein one aspect was a chelator that had been prelabelled with 5?Co and the second aspect was a reactive group capable of covalent attachment to a free sulfhydryi. The bifunctional chelator and a metiiod for labelling it with ^Co has been described previously (Mears, et al, Analytical Biochem. 142: 68-78, 1984: ). Briefly, a solution of bromoacetyl DOTA was trace labelled with ^Co. and then a 10-fold molar excess was added to antibody solution. The conjugation reaction was carried out for 2 hrs at 37° and stopped by separation of the reactants by gel filtration using a Bio Spin 30 column (Bio Rad) as recommended by the manufacturer. Bifunctional chelators, such as bromoacetylTMT, were also employed prior to radiolabeling. The termperature and duration of d e conjugation reaction was varied for maximal site-specific conjugation. For example, reduced protein at a concentration of 150 - 200 uM in tetrametiiylammonium phosphate, pH 8.2. 25 μM DTPA, was added to a 10-fold excess of bromoacetylTMT in a metal- free reaction vial. The reaction was carried out for 24 hours at room termperature. and die conjugate was isolated by gel filtration as described above.
Example 4, Muring biodis-i- -tion o conjugates,
A reduced ser442→ cys variant, Campatii-IH G4mc, was conjugated to bromoacetyl DOTA by the site directed procedure described above without prelabelling. By way of comparison, random conjugate was prepared by introducing thiol groups into nonreduced Campatii- 1 H g4mc as has been described previously for Lym- 1. (The latter process follows the procedure of McCall, et al, Bioconjugate Chem. I: 222-226, 1990 and employs 2-iminodιiolane to introduce thiol groups randomly onto die ε amino groups of lysine residues.) Iminothiolated Campath-IH G4mc was conjugated to bromoacetyi-DOTA to the same extent (chelators per antibody) as site directed conjugate. The conjugates were rendered radioactive by mixing with 9^YC 13 in plastic ware by addition of the following reagents: an 8X volume of monoclonal antibody conjugate (> 25 mg/mL. 0.1 M ammonium acetate pH 6.7), a 2X volume of 90N. and cold yttrium up to a final concentration of 10 μM. Cold yttrium was added first. followed by 90γ and conjugate. Chelation was allowed to occur at room temperature for 90 minutes. Ten μg (2 μCi) were injected intravenously per mouse into mice bearing a subcutaneous tumour (CHO-10/D4) that expressed die Campath-IH antigen. Mice were sacrificed at d e indicated times, tissues excised, weighed and radioactivity determined. The results are shown in Table II below and are expressed as the % injected dose per gram tissue (average of 5 mice per group +/- sem). Note the diminution of normal tissue deposition and increase in tumour deposition in die site- directed (direct) conjugate groups relative to the random groups. Figure 3 illustrates the biodistribution of 90 Y-TMT-Campath- 1 HG4mc.
Example 5
A reduced ser442"^cys variant. Campath-IH was conjugated to TMT, tiiat is covalently attached via a thioether linkage to the cys442 residues in die heavy chains as illustrated in Figure 4. The reaction may be regulated to produce conjugates that contain an average of 1 - 2 chelators per antibody. Conjugate is purified free of unreacted bifunctional chelator by gel filtration in metal-free conditions and is stable in a buffered, metal-free environment.
Immunoconjugate chelation. Conjugates were radiolabeled in metal-free plasticware using the best metal-free reagents available. Carrier-free 90 YC 13 was purchased from Dupont/New England Nuclear, Amersham and odier sources. The specific activity was typically 5 mCi in 10-30 μL 0.05 N HC1, specific activity 5.6 X 105 Ci/g. Prior to use, the 90γci was buffered witii 0.1 volume of 6 M ammonium acetate to ~ pH 5.8. Chelation was performed by adding the following reagents in sequence and incubating for up to 90 min at room temperature: a IX volume of cold yttrium (lOOμM yttrium in 0.1 M ammonium acetate, pH 6.8.), a 2X volume of 9 γ acetate, and an 8X volume of monoclonal antibody conjugate (25 mg/ml 0.1 M ammonium acetate, pH 6.5). Non- chelated radiometal was "scavenged" by d e addition of DTPA to a final concentration of 500μM and a 10X volume of 0.1 M ammonium citrate, pH 6.5. The mixture was incubated at room temperature for 30 minutes and fractionated by "spin column gel filtration," i.e., applied to a 1 mL Bio-Spin 30 (Bio-Rad Laboratories) tiiat had been pre-equilibrated in phosphate-buffered saline and centrifuged at 150 x g for 2 minutes. Spin column gel filtration was repeated for a total of two centrifugations. The efficiency of chelation (ability to chelate all the radiometal) and scavenging (ability to remove non-chelated radioactivity from radiolabeled conjugate) was monitored by thin layer chromatography as described by Meares et al, Anal. Biochem. 142, 68-78. 1984. More than 90% of the radiometal was routinely chelated. (The extent of chelation was dependent on die acid and metal content of die supplied radiometal.) After scavenging, die fraction of " γ that was tightly bound to conjugate was typically >98% when analyzed by HPLC gel filtration, thin layer chromatography or SDS PAGE.
90γ.postlabeled conjugates have been prepared with specific activities of up to 10 mCi/mg which is 30-fold below the theoretical capacity. Although every chelator is available to accept radiometal. higher specific activity causes radiolysis (a function of time and concentration) that can be reduced by inclusion of an anti-oxidant such as ascorbic acid. In principle, chelation can be optimized for complete efficiency given a consistent and high quality supply of radiometal and thus eliminate the need for scavenging.
Example 6. Large scale reduction of variant monclonal antibodies
Dilute ultrafiltered IgG product to 50mg/ml (+/- lmg/ml) and measure a volume V. Add V/10 of freshly prepared stock solution (13mg/ml ME A in 50mM phosphate and 5mM EDTA at pH7.0), mixing well during addition to ensure even distribution of reductant. Leave at ambient temperature for 60 minutes. The sample is applied to a gel filtration column such as, for example, Sephadex G25 or Superdex 75 or 30. The IgG peak is monitored at absorbance of 280nm and die peak collected and subjected to additional procedures such as free thiol determination, pH adjustment, purification or conjugation as described in Example 3.
Example 7. Large scale site directed coηiugation of reduced variant antibody.
Add 10 fold molar excess of bromoacetyl-TMT as a lOmM stock made up in 0.1 M Hepes + 25μM DTPA at pH8.4 over the IgG from example 6. Mix well and leave at ambient temperature for a minimum of 21 hours, maximum 37 hours. The conjugated antibody is then loaded onto a Superdex 200 column. The IgG peak is monitored at absorbance of 280nm and the monomer peak collected and subjected to standard analytical procedures such as estimation of binding, protein content etc. ________
Free thiol content following exposure to solid-phase reductant
A G4 ser442→ cys variant labelled "G4mc" and a natural G4 control were exposed to solid phase reductant as described in Example 2. The thiol content was determined by Ellman's reagent and is expressed relative to moles antibody. (It is assumed tiiat 2 moles of tiiiol were reduced per antibody ti iol.) The sem for triplicate measurements was ± 0.1 SH/antibody.
Molar Ex Gel Moles -SH/Antibody
______ __4
Expt.1
100 4.6 .6
30 3.0 10 1.2 .1
J .6 0 0 .1 0
Expt.2
50 J.J .J 30 2.3 20 1.7 10 1.0 - .5 0 .1
Iab JI
Murine biodistribution of ^--Y-coηjugates
% ID/g (avg -/- se ^
24 hour 72 hour 168 hour direct
BLOOD 13.47 1.75 8.80 1.67 5.93 0.80
SPLEEN 3.51 0.63 2.46 0.50 2.83 0.39
TUMOUR 17.01 3.16 24.78 8.41 31.36 4.96
LIVER 5.24 0.62 3.32 0.69 2.56 0.28
LUNG 5.60 0.49 5.00 0.70 3.99 1.32
KIDNEY 5.09 0.30 5.21 0.65 4.16 0.54
BONE 1.56 0.19 1.09 0.18 0.95 0.15
random
BLOOD 11.97 3.96 9.48 2.00 6.66 0.76
SPLEEN 5.79 1.42 4.61 0.90 3.55 2.07
TUMOUR 15.42 5.52 22.37 9.29 29.91 4.70
LIVER 10.07 2.99 6.75 1.31 4.34 0.82
LUNG 6.19 0.95 4.56 1.22 3.67 1.08
KIDNEY 5.38 1.41 6.84 1.41 5.18 0.50
BONE 1.76 0.46 1.39 0.35 1.25 0.14

Claims

_________
1. An immunochemically functional monoclonal antibody comprising a cysteine residue exposed on the surface of the antibody such that die residue is capable of being conjugated to a substance.
2. An antibody according to claim 1, wherein the cysteine residue is in the variable region of the antibody but not part of the CDRs.
3. An antibody according to claim 1, wherein the cysteine residue is in die constant region .
4. An antibody according to claim 3, wherein the cysteine residue is in the CH3 domain of the heavy chain.
5. An antibody according to claim 4. wherein the cysteine residue is at position 442 within the CH3 domain.
6. An antibody according to any of the preceding claims, wherein the antibody binds to a 40KD antigen or folate receptor antigen.
7. An antibody according to any of d e preceding claims, wherein the antibody is humanised.
8. An antibody according to any of the preceding claims, wherein the antibody is a Gl or G4 isotype.
9. An antibody conjugate comprising an antibody according to any of the preceding claims and a substance, which is directly or indirectly conjugated to the cysteine- residue of the antibody.
10. An antibody conjugate according to claim 9, wherein the substance is indirectly conjugated via a chelator.
1 1. An antibody conjugate according to claim 9, wherein die substance is indirectly conjugated via a linker and a chelator.
12. An antibody conjugate according to claim 10 or 1 1. wherein the chelator is TMT or DOTA.
13. An antibody conjugate according to claim 1 1 or 12. wherein the linker is bromoacetyl.
14. An antibody conjugate according to any of claims 10 to 13, wherein die substance is Y or Lu.
15. An antibody conjugate according to claim 9, wherein the substance is a molecular chimaera for use in enzyme-prodrug therapy.
16. An antibody conjugate according to claim 15. wherein die chimaera comprises a transcriptional regulatory DNA sequence capable of being activated in a mammalian cell and a DNA sequence operatively linked to die transcriptional regulatory DNA sequence and encoding a heterologous enzyme capable of catalysing the conversion of the prodrug into an agent toxic to die cancer cell.
17. An antibody conjugate according to claim 16. wherein the transcriptional regulatory DNA sequence is a tissue- or cancer- specific transcriptional regulatory DNA sequence.
18. An antibody conjugate according to claim 15 or 17, wherein the chimera is contained witiiin a viral vector or liposome.
19. Use of an antibody conjugate according to any of claims 9 to 18, for the treatment and diagnosis of cancers and associated metastasis.
20. Use of an antibody conjugate according to claims 9 to 14, for the treatment of small cell and non-small cell lung cancer, prostatic cancer and associated metastasis.
21. Use of an antibody conjugate according to claims 15 to 18, for the treatment of ovarian cancer.
22. Use of an antibody conjugate according to claims 10 to 14 for radioimmuno¬ therapy.
23. A pharmaceutically acceptable composition comprising conjugated antibodies according to any of claims 9 to 18 togetiier with a physiologically acceptable diluent or carrier.
24. A pharmaceutically acceptable composition according to claim 23 for radioimmunodierapy, wherein the dosage is 1 to 10 mg.
EP95936025A 1994-11-05 1995-11-03 Antibodies Withdrawn EP0789713A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
GB9422383 1994-11-05
GB9422383A GB9422383D0 (en) 1994-11-05 1994-11-05 Antibodies
PCT/GB1995/002585 WO1996014339A1 (en) 1994-11-05 1995-11-03 Antibodies

Publications (1)

Publication Number Publication Date
EP0789713A1 true EP0789713A1 (en) 1997-08-20

Family

ID=10763976

Family Applications (1)

Application Number Title Priority Date Filing Date
EP95936025A Withdrawn EP0789713A1 (en) 1994-11-05 1995-11-03 Antibodies

Country Status (6)

Country Link
EP (1) EP0789713A1 (en)
JP (1) JPH10508482A (en)
AU (1) AU3811495A (en)
GB (1) GB9422383D0 (en)
WO (1) WO1996014339A1 (en)
ZA (1) ZA959336B (en)

Families Citing this family (112)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6897044B1 (en) 1999-01-28 2005-05-24 Biogen Idec, Inc. Production of tetravalent antibodies
GB0030378D0 (en) 2000-12-13 2001-01-24 Cyclacel Ltd Compound
WO2004021996A2 (en) 2002-09-06 2004-03-18 The Government Of The United States Of America, Represented By The Secretary, Dept. Of Health And Human Services Backbone-substituted bifunctional dota ligands, complexes and compositions thereof, and methods of using same
TWI353991B (en) 2003-05-06 2011-12-11 Syntonix Pharmaceuticals Inc Immunoglobulin chimeric monomer-dimer hybrids
EP2385069A3 (en) 2003-11-12 2012-05-30 Biogen Idec MA Inc. Neonatal Fc rReceptor (FcRn)- binding polypeptide variants, dimeric Fc binding proteins and methods related thereto
US8008453B2 (en) 2005-08-12 2011-08-30 Amgen Inc. Modified Fc molecules
US20100297103A1 (en) * 2006-09-14 2010-11-25 Medical & Biological Laboratories Co., Ltd. Antibody having enhanced adcc activity and method for production thereof
KR20100107501A (en) * 2008-01-18 2010-10-05 메디뮨 엘엘씨 Cysteine engineered antibodies for site-specific conjugation
CN102341411A (en) 2008-12-31 2012-02-01 比奥根艾迪克Ma公司 Anti-lymphotoxin antibodies
CN102802661B (en) * 2009-06-22 2016-01-13 米迪缪尼有限公司 For the engineered Fc district of site-specific conjugation
JP5813641B2 (en) 2009-08-24 2015-11-17 アムニクス オペレーティング インコーポレイテッド Coagulation factor IX composition and methods of making and using the same
PL2496691T3 (en) 2009-11-02 2017-09-29 University Of Washington Therapeutic nuclease compositions and methods
NZ605400A (en) 2010-07-09 2015-05-29 Biogen Idec Hemophilia Inc Chimeric clotting factors
KR102006393B1 (en) 2011-04-29 2019-08-02 유니버시티 오브 워싱톤 스루 이츠 센터 포 커머셜리제이션 Therapeutic nuclease compositions and methods
SI2717898T1 (en) 2011-06-10 2019-07-31 Bioverativ Therapeutics Inc. Pro-coagulant compounds and methods of use thereof
WO2013012733A1 (en) 2011-07-15 2013-01-24 Biogen Idec Ma Inc. Heterodimeric fc regions, binding molecules comprising same, and methods relating thereto
UY34317A (en) 2011-09-12 2013-02-28 Genzyme Corp T cell antireceptor antibody (alpha) / ß
US20130108641A1 (en) 2011-09-14 2013-05-02 Sanofi Anti-gitr antibodies
JP6483442B2 (en) 2011-12-05 2019-03-13 エックス−ボディ インコーポレイテッド PDGF receptor beta-binding polypeptide
SG11201403764XA (en) 2012-01-12 2014-07-30 Biogen Idec Inc Chimeric factor viii polypeptides and uses thereof
PL3564260T3 (en) 2012-02-15 2023-03-06 Bioverativ Therapeutics Inc. Factor viii compositions and methods of making and using same
HUE043537T2 (en) 2012-02-15 2019-08-28 Bioverativ Therapeutics Inc Recombinant factor viii proteins
AR090352A1 (en) 2012-03-28 2014-11-05 Sanofi Sa ANTIBODIES AGAINST BRADICININE B1 RECEIVER LIGANDS
MX2014013637A (en) 2012-05-07 2015-02-05 Sanofi Sa Methods for preventing biofilm formation.
WO2013175276A1 (en) 2012-05-23 2013-11-28 Argen-X B.V Il-6 binding molecules
EP3693000B1 (en) 2012-06-08 2022-03-02 Bioverativ Therapeutics Inc. Procoagulant compounds
CA2875247A1 (en) 2012-06-08 2013-12-12 Biogen Idec Ma Inc. Chimeric clotting factors
WO2014008480A2 (en) 2012-07-06 2014-01-09 Biogen Idec Ma Inc. Cell line expressing single chain factor viii polypeptides and uses thereof
NZ703366A (en) 2012-07-11 2018-03-23 Amunix Operating Inc Factor viii complex with xten and von willebrand factor protein, and uses thereof
US9790268B2 (en) 2012-09-12 2017-10-17 Genzyme Corporation Fc containing polypeptides with altered glycosylation and reduced effector function
SI2895513T1 (en) 2012-09-12 2018-11-30 Genzyme Corporation Fc containing polypeptides with altered glycosylation and reduced effector function
KR102447350B1 (en) 2013-02-08 2022-09-23 노파르티스 아게 Specific sites for modifying antibodies to make immunoconjugates
PL2956477T3 (en) 2013-02-15 2021-06-14 Bioverativ Therapeutics Inc. Optimized factor viii gene
US9580511B2 (en) 2013-03-11 2017-02-28 Genzyme Corporation Site-specific antibody-drug conjugation through glycoengineering
SG10201913874TA (en) 2013-03-15 2020-03-30 Biogen Ma Inc Factor ix polypeptide formulations
EP3875106A1 (en) 2013-08-08 2021-09-08 Bioverativ Therapeutics Inc. Purification of chimeric fviii molecules
TW201722994A (en) 2013-08-13 2017-07-01 賽諾菲公司 Antibodies to Plasminogen Activator Inhibitor-1 (PAI-1) and uses thereof
UA118267C2 (en) 2013-08-13 2018-12-26 Санофі Antibodies to plasminogen activator inhibitor-1 (pai-1) and uses thereof
TW202003554A (en) 2013-08-14 2020-01-16 美商百歐維拉提夫治療公司 Factor VIII-XTEN fusions and uses thereof
ES2900425T3 (en) 2013-09-25 2022-03-16 Bioverativ Therapeutics Inc Column viral inactivation methods
WO2015066550A1 (en) 2013-10-31 2015-05-07 Resolve Therapeutics, Llc Therapeutic nuclease-albumin fusions and methods
EP3065769A4 (en) 2013-11-08 2017-05-31 Biogen MA Inc. Procoagulant fusion compound
CN117106095A (en) 2014-01-10 2023-11-24 比奥贝拉蒂治疗公司 Factor VIII chimeric proteins and uses thereof
WO2015123265A1 (en) * 2014-02-11 2015-08-20 Seattle Genetics, Inc. Selective reduction of proteins
PL3129067T3 (en) 2014-03-19 2023-05-08 Genzyme Corporation Site-specific glycoengineering of targeting moieties
WO2015143271A1 (en) 2014-03-21 2015-09-24 X-Body, Inc. Bi-specific antigen-binding polypeptides
US10160812B2 (en) 2014-04-11 2018-12-25 Medimmune, Llc Bispecific HER2 antibodies
EP3160478A4 (en) 2014-06-30 2018-05-16 Bioverativ Therapeutics Inc. Optimized factor ix gene
PE20170702A1 (en) 2014-09-26 2017-06-24 Bayer Pharma AG STABILIZED DERIVATIVES OF ADRENOMEDULIN AND THE USE OF THEM
CA2964123C (en) 2014-10-09 2023-09-05 Genzyme Corporation Glycoengineered antibody drug conjugates
BR112017007765B1 (en) 2014-10-14 2023-10-03 Halozyme, Inc COMPOSITIONS OF ADENOSINE DEAMINASE-2 (ADA2), VARIANTS THEREOF AND METHODS OF USING THE SAME
CA2988806A1 (en) * 2015-06-29 2017-01-05 Immunogen, Inc. Conjugates of cysteine engineered antibodies
BR112018002150A2 (en) 2015-08-03 2018-09-18 Bioverativ Therapeutics Inc factor ix fusion proteins and methods of manufacturing and using them
US20190022092A1 (en) 2015-09-15 2019-01-24 Acerta Pharma B.V. Therapeutic Combinations of a BTK Inhibitor and a GITR Binding Molecule, a 4-1BB Agonist, or an OX40 Agonist
US11753461B2 (en) 2016-02-01 2023-09-12 Bioverativ Therapeutics Inc. Optimized factor VIII genes
EP3478830B1 (en) 2016-07-01 2024-04-10 Resolve Therapeutics, LLC Optimized binuclease fusions and methods
TWI788307B (en) 2016-10-31 2023-01-01 美商艾歐凡斯生物治療公司 Engineered artificial antigen presenting cells for tumor infiltrating lymphocyte expansion
WO2018098258A2 (en) 2016-11-23 2018-05-31 Immunogen, Inc. Selective sulfonation of benzodiazepine derivatives
AU2017368328A1 (en) 2016-12-02 2019-07-18 Bioverativ Therapeutics Inc. Methods of inducing immune tolerance to clotting factors
CN110520150A (en) 2016-12-02 2019-11-29 比奥维拉迪维治疗股份有限公司 Use the method for chimeric coagulation factor therapies hemophilic arthosis
TW201837168A (en) 2017-01-06 2018-10-16 美商艾歐凡斯生物治療公司 Expansion of tumor infiltrating lymphocytes (TILS) with tumor necrosis factor receptor superfamily (TNFRSF) agonists and therapeutic combinations of TILS and TNFRSF agonists
US11357841B2 (en) 2017-01-06 2022-06-14 Iovance Biotherapeutics, Inc. Expansion of tumor infiltrating lymphocytes with potassium channel agonists and therapeutic uses thereof
CN110832070A (en) 2017-05-10 2020-02-21 艾欧凡斯生物治疗公司 Expansion of liquid tumor-derived tumor infiltrating lymphocytes and therapeutic uses thereof
CA3072334A1 (en) 2017-08-09 2019-02-14 Bioverativ Therapeutics Inc. Nucleic acid molecules and uses thereof
CA3073537A1 (en) 2017-08-22 2019-02-28 Sanabio, Llc Soluble interferon receptors and uses thereof
JP2021503885A (en) 2017-11-22 2021-02-15 アイオバンス バイオセラピューティクス,インコーポレイテッド Expanded culture of peripheral blood lymphocytes (PBL) from peripheral blood
US20210369775A1 (en) 2017-12-15 2021-12-02 Iovance Biotherapeutics, Inc. Systems and methods for determining the beneficial administration of tumor infiltrating lymphocytes, and methods of use thereof and beneficial administration of tumor infiltrating lymphocytes, and methods of use thereof
JP2021512126A (en) 2018-02-01 2021-05-13 バイオベラティブ セラピューティクス インコーポレイテッド Use of a lentiviral vector expressing factor VIII
CA3090795A1 (en) 2018-02-13 2019-08-22 Iovance Biotherapeutics, Inc. Expansion of tumor infiltrating lymphocytes (tils) with adenosine a2a receptor antagonists and therapeutic combinations of tils and adenosine a2a receptor antagonists
CN112154153A (en) 2018-03-28 2020-12-29 百时美施贵宝公司 Interleukin-2/interleukin-2 receptor alpha fusion proteins and methods of use
US20190375822A1 (en) 2018-05-18 2019-12-12 Bioverativ Therapeutics Inc. Methods of treating hemophilia a
WO2019236417A1 (en) 2018-06-04 2019-12-12 Biogen Ma Inc. Anti-vla-4 antibodies having reduced effector function
EP3817720A2 (en) 2018-07-03 2021-05-12 Bristol-Myers Squibb Company Fgf21 formulations
CN113227385A (en) 2018-08-09 2021-08-06 比奥维拉迪维治疗股份有限公司 Nucleic acid molecules and their use for non-viral gene therapy
TW202031273A (en) 2018-08-31 2020-09-01 美商艾歐凡斯生物治療公司 Treatment of nsclc patients refractory for anti-pd-1 antibody
WO2020096989A1 (en) 2018-11-05 2020-05-14 Iovance Biotherapeutics, Inc. Treatment of nsclc patients refractory for anti-pd-1 antibody
KR20210113261A (en) 2019-01-04 2021-09-15 리졸브 테라퓨틱스, 엘엘씨 Treatment of Sjogren's Disease Using Nuclease Fusion Proteins
MX2021010288A (en) 2019-03-01 2021-09-23 Iovance Biotherapeutics Inc Expansion of tumor infiltrating lymphocytes from liquid tumors and therapeutic uses thereof.
JP2022521850A (en) 2019-04-03 2022-04-12 ジェンザイム・コーポレーション Anti-alpha beta TCR-binding polypeptide with reduced fragmentation
CA3143584A1 (en) 2019-06-18 2020-12-24 Bayer Aktiengesellschaft Adrenomedullin-analogues for long-term stabilization and their use
US20210113634A1 (en) 2019-09-30 2021-04-22 Bioverativ Therapeutics Inc. Lentiviral vector formulations
WO2021158938A1 (en) 2020-02-06 2021-08-12 Bristol-Myers Squibb Company Il-10 and uses thereof
AU2021225962A1 (en) 2020-02-28 2022-10-20 Genzyme Corporation Modified binding polypeptides for optimized drug conjugation
CA3165342A1 (en) 2020-06-29 2022-01-06 James Arthur Posada Treatment of sjogren's syndrome with nuclease fusion proteins
WO2022076606A1 (en) 2020-10-06 2022-04-14 Iovance Biotherapeutics, Inc. Treatment of nsclc patients with tumor infiltrating lymphocyte therapies
JP2023546359A (en) 2020-10-06 2023-11-02 アイオバンス バイオセラピューティクス,インコーポレイテッド Treatment of NSCLC patients with tumor-infiltrating lymphocyte therapy
JP2024501452A (en) 2020-12-11 2024-01-12 アイオバンス バイオセラピューティクス,インコーポレイテッド Treatment of cancer patients with tumor-infiltrating lymphocyte therapy in combination with BRAF inhibitors and/or MEK inhibitors
WO2022133140A1 (en) 2020-12-17 2022-06-23 Iovance Biotherapeutics, Inc. Treatment with tumor infiltrating lymphocyte therapies in combination with ctla-4 and pd-1 inhibitors
JP2024500403A (en) 2020-12-17 2024-01-09 アイオバンス バイオセラピューティクス,インコーポレイテッド Treatment of cancer with tumor-infiltrating lymphocytes
EP4271791A2 (en) 2020-12-31 2023-11-08 Iovance Biotherapeutics, Inc. Devices and processes for automated production of tumor infiltrating lymphocytes
TW202241508A (en) 2021-01-29 2022-11-01 美商艾歐凡斯生物治療公司 Cytokine associated tumor infiltrating lymphocytes compositions and methods
TW202300014A (en) 2021-03-05 2023-01-01 美商艾歐凡斯生物治療公司 Tumor storage and cell culture compositions
EP4308691A1 (en) 2021-03-19 2024-01-24 Iovance Biotherapeutics, Inc. Methods for tumor infiltrating lymphocyte (til) expansion related to cd39/cd69 selection and gene knockout in tils
TW202305118A (en) 2021-03-23 2023-02-01 美商艾歐凡斯生物治療公司 Cish gene editing of tumor infiltrating lymphocytes and uses of same in immunotherapy
JP2024512029A (en) 2021-03-25 2024-03-18 アイオバンス バイオセラピューティクス,インコーポレイテッド Methods and compositions for T cell co-culture efficacy assays and use with cell therapy products
JP2024515189A (en) 2021-04-19 2024-04-05 アイオバンス バイオセラピューティクス,インコーポレイテッド Chimeric costimulatory receptors, chemokine receptors, and their uses in cellular immunotherapy - Patents.com
WO2022245754A1 (en) 2021-05-17 2022-11-24 Iovance Biotherapeutics, Inc. Pd-1 gene-edited tumor infiltrating lymphocytes and uses of same in immunotherapy
CA3226111A1 (en) 2021-07-22 2023-01-26 Iovance Biotherapeutics, Inc. Method for cryopreservation of solid tumor fragments
WO2023009716A1 (en) 2021-07-28 2023-02-02 Iovance Biotherapeutics, Inc. Treatment of cancer patients with tumor infiltrating lymphocyte therapies in combination with kras inhibitors
WO2023039488A1 (en) 2021-09-09 2023-03-16 Iovance Biotherapeutics, Inc. Processes for generating til products using pd-1 talen knockdown
CA3232700A1 (en) 2021-09-24 2023-03-30 Rafael CUBAS Expansion processes and agents for tumor infiltrating lymphocytes
EP4166569A1 (en) * 2021-10-18 2023-04-19 Fondazione IRCCS Istituto Nazionale dei Tumori Antibodies directed against alpha-folate receptor
AR127482A1 (en) 2021-10-27 2024-01-31 Iovance Biotherapeutics Inc SYSTEMS AND METHODS TO COORDINATE THE MANUFACTURE OF CELLS FOR PATIENT-SPECIFIC IMMUNOTHERAPY
WO2023086803A1 (en) 2021-11-10 2023-05-19 Iovance Biotherapeutics, Inc. Methods of expansion treatment utilizing cd8 tumor infiltrating lymphocytes
WO2023147488A1 (en) 2022-01-28 2023-08-03 Iovance Biotherapeutics, Inc. Cytokine associated tumor infiltrating lymphocytes compositions and methods
WO2023147486A1 (en) 2022-01-28 2023-08-03 Iovance Biotherapeutics, Inc. Tumor infiltrating lymphocytes engineered to express payloads
WO2023196877A1 (en) 2022-04-06 2023-10-12 Iovance Biotherapeutics, Inc. Treatment of nsclc patients with tumor infiltrating lymphocyte therapies
WO2023201369A1 (en) 2022-04-15 2023-10-19 Iovance Biotherapeutics, Inc. Til expansion processes using specific cytokine combinations and/or akti treatment
WO2023220608A1 (en) 2022-05-10 2023-11-16 Iovance Biotherapeutics, Inc. Treatment of cancer patients with tumor infiltrating lymphocyte therapies in combination with an il-15r agonist
WO2024011114A1 (en) 2022-07-06 2024-01-11 Iovance Biotherapeutics, Inc. Devices and processes for automated production of tumor infiltrating lymphocytes
WO2024030758A1 (en) 2022-08-01 2024-02-08 Iovance Biotherapeutics, Inc. Chimeric costimulatory receptors, chemokine receptors, and the use of same in cellular immunotherapies
WO2024062074A1 (en) 2022-09-21 2024-03-28 Sanofi Biotechnology Humanized anti-il-1r3 antibody and methods of use

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8720833D0 (en) * 1987-09-04 1987-10-14 Celltech Ltd Recombinant dna product
GB8903021D0 (en) * 1989-02-10 1989-03-30 Celltech Ltd Chemical compounds
GB8919607D0 (en) * 1989-08-30 1989-10-11 Wellcome Found Novel entities for cancer therapy
WO1991019515A1 (en) * 1990-06-21 1991-12-26 The Board Of Trustees Of The Leland Stanford Junior University Oligomeric immunoglobulin constant domain mutant with enhanced complement-mediated cytolytic activity

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9614339A1 *

Also Published As

Publication number Publication date
AU3811495A (en) 1996-05-31
GB9422383D0 (en) 1995-01-04
JPH10508482A (en) 1998-08-25
WO1996014339A1 (en) 1996-05-17
ZA959336B (en) 1997-05-05

Similar Documents

Publication Publication Date Title
WO1996014339A1 (en) Antibodies
JP3095415B2 (en) Preparation and use of immune complexes
US6511663B1 (en) Tri- and tetra-valent monospecific antigen-binding proteins
KR0135632B1 (en) Detection and treatment of infections and inflammatory
US9765155B2 (en) Covalent disulfide-linked diabodies and uses thereof
US6241961B1 (en) Radioimmuno conjugates for use in human therapy and method for their preparation
EP0385601A2 (en) Cross-linked antibodies and processes for their preparation
US20190298846A1 (en) Cell targeting conjugates
US6509451B1 (en) Cross-linked antibodies and processes for their preparation
EP4130046A1 (en) Platform for constructing multispecific antibody
EP0787138B1 (en) Radioactive phosphorous labeling of proteins for targeted radiotherapy
US5993813A (en) Family of high affinity, modified antibodies for cancer treatment
US6051225A (en) Family of high affinity, modified antibodies for cancer treatment
CA2026312A1 (en) Cross-linked antibodies and processes for their preparation
AU702975B2 (en) Recombinant proteins having multiple disulfide bonds and thiol-substituted conjugates thereof
Kerr et al. Application of monoclonal antibodies against cytosine deaminase for the in vivo clearance of a cytosine deaminase immunoconjugate
CA2066031C (en) Methods for reducing non-target retention of immunoconjugates and metabolites thereof
Leung et al. The effects of domain deletion, glycosylation, and long IgG3 hinge on the biodistribution and serum stability properties of a humanized IgG1 immunoglobulin, hLL2, and its fragments
US20230181772A1 (en) Engineered anti-prostate stem cell antigen fusion proteins and uses thereof
EP0569531B1 (en) Stabilized antibody fragments
CN116655793A (en) Preparation method and application of BCMA (bcmA-specific diagnosis and treatment) integrated molecular imaging probe
CN116925220A (en) IL20RB neutralizing antibody and medical application thereof
IL302111A (en) Radioactive complexes of anti-her2 antibody, and radiopharmaceutical
Kamigaki et al. Enhancement of tumor uptakes by stabilized Fab homo-oligomers of a chimeric monoclonal antibody against carcinoembryonic antigen.

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19970502

AK Designated contracting states

Kind code of ref document: A1

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

AX Request for extension of the european patent

Free format text: LT PAYMENT 970502;LV PAYMENT 970502;SI PAYMENT 970502

17Q First examination report despatched

Effective date: 19991027

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20020504