CA1232838A - Immunotoxin conjugates - Google Patents

Immunotoxin conjugates

Info

Publication number
CA1232838A
CA1232838A CA000456816A CA456816A CA1232838A CA 1232838 A CA1232838 A CA 1232838A CA 000456816 A CA000456816 A CA 000456816A CA 456816 A CA456816 A CA 456816A CA 1232838 A CA1232838 A CA 1232838A
Authority
CA
Canada
Prior art keywords
chain
toxin
conjugate
cell surface
antibody
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.)
Expired
Application number
CA000456816A
Other languages
French (fr)
Inventor
Ellen S. Vitetta
Jonathan W. Uhr
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.)
University of Texas System
Original Assignee
University of Texas System
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 University of Texas System filed Critical University of Texas System
Application granted granted Critical
Publication of CA1232838A publication Critical patent/CA1232838A/en
Expired legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G18/00Polymeric products of isocyanates or isothiocyanates
    • C08G18/06Polymeric products of isocyanates or isothiocyanates with compounds having active hydrogen
    • C08G18/28Polymeric products of isocyanates or isothiocyanates with compounds having active hydrogen characterised by the compounds used containing active hydrogen
    • C08G18/40High-molecular-weight compounds
    • C08G18/54Polycondensates of aldehydes
    • 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
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • A61K47/6817Toxins
    • A61K47/6829Bacterial toxins, e.g. diphteria toxins or Pseudomonas exotoxin A
    • 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
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • A61K47/6817Toxins
    • A61K47/6819Plant toxins
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G18/00Polymeric products of isocyanates or isothiocyanates
    • C08G18/06Polymeric products of isocyanates or isothiocyanates with compounds having active hydrogen
    • C08G18/28Polymeric products of isocyanates or isothiocyanates with compounds having active hydrogen characterised by the compounds used containing active hydrogen
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/811Test for named disease, body condition or organ function
    • Y10S436/813Cancer
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/819Multifunctional antigen or antibody
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S530/00Chemistry: natural resins or derivatives; peptides or proteins; lignins or reaction products thereof
    • Y10S530/866Chemistry: natural resins or derivatives; peptides or proteins; lignins or reaction products thereof involving immunoglobulin or antibody fragment, e.g. fab', fab, fv, fc, heavy chain or light chain

Abstract

Abstract The present invention provides a means for potentiating the cytotoxic activity of immunotoxin con-jugates. The compositions of the present invention include a selective binding agent such as an antibody coupled to a toxin B chain moiety such as ricin B chain.

Description

I

~~65~4 -1-IMPROVEMENTS IN OR RELATING TO II~.MUNOTOXIN CONJUGATES

This invention relates to im~lunoto~in coinage-gates arid their use to delete selectively a target population of cells. In particular, a toxin B chain moiety coupled to a cell surface affinity binding agent is useful in potenti2ting the cytotoxicity provided by a cell surface affinity binding agent coupled to a -toxin A chain moiety.
Ricing is one of a number of plant proteins which, in minute quantities, exhibits considerable toxicity toward eukaryotic cells. Ricing is composed of two glycoprotein chains covalently linked via a single disulfide bond. The A chain of ricing having an apparent molecular weight (Awry) of about 30,000, is responsible four the e~pressioll of toxicity, and acts enzymatic ally upon the 60S ribosomal subunit to produce irreversible abrogation of protein synthesis ~Olsnes, et at., FOBS Let 28, 48 50 ( 197G ) ] . I' gin B chain _ _ AMY 32,000) functions as a pectin with specificity for galactose and serves to bind the -toxin to the cell membrane [see, e.cJ., Banger, et at., J_ of. Chum.
254, 9'795-97'`39 ( 197C.3 ) ] .
The use of ricing or thy purified x-icin A
chain, in conjunction with antibodies, has been the subject of great interest as poten'iallv-useful reagents in tumor therapy. Antibody-ricin and antibody-A chain conjugates, an "i~munotoxins", have been used in a number of systems with varying degas of success see, e.g., Vitetta, et at., Science 219, 6~4-650 (,983);

I, X-656~ -2-Thorpe, et at., Immunol. Rev. 62, 120-158 (1982);
Seville, et at., Immunol. rev. 62, 75-91 (1982); and Jensen, et at., Immunol. Rev. 62, 185-216 (1982)].
Procedures for deleting selected populations of cells by ricing A chain-antibody conjugates are well-recognized. The antibodies of choice are those which react with antigens on tumor cells or on subsets of normal lymphocytes. By deletion of the tumor cells, one may reduce, for example, tumor burdens in viva [Krolick, et at., J. Exp. Med. 155, 1797 (1982)] and remove tumor cells from bone marrow for autologous marrow transplantation [Thorpe, et at., Nature (London) 271, 752 (1978); and Krolick, et at., Nature (London) 295, 604 (1982)].
Also, by deleting normal subsets of lymph-cites, one may be able to "up" or "down" regulate the immune response. The advantage of immuno~oxins is that they are highly specific for their target cell and that small doses can eliminate unwanted cells. Ricing A
2C chain-antibody conjugates have been used primarily to-delete normal and neoplastic B cells, both in viva and in vitro. Certain laboratories have also used coinage-gates of ricing A chain and monoclonal antibody to eliminate neoplastic cells of T cell origin and a variety of other cancerous cells.
However, ricing A chain-antibody conjugates are not active when used against certain types o tumor cells (e.g., some T cell tumors) novel, et at., Immunol. Rev. 62, 75 (1982); and Thorpe, et at., Immunol. Rev. 62, 119 ~1982)~.

In contrast, immunotoxins coupled to the whole ricing toxin are much more potent cytocidal agents.
Unfortunately, the presence of the galactose binding site of ricing s in intact ricing prevents its use in Volvo because its target cell specificity thereby is lost. Attempts to overcome the non specificity of ricing containing immunotoxins by blocking the galactose bind-in site are ongoing however their use in Volvo has not been described yet.
Others have described studies in which ricing A chain-antibody conjugates can be potentiated by the addition of free B chain to cell cultures (Seville et at., a). Researchers have postulated, therefore, that the B chain of ricing has two functions: (1) to facilitate entry of ricing into the cell by virtue of its galactose-binding properties, and (2) to allow the A chain to gain rapid access to the cytoplasm, perhaps by formation of a pore in the endocytic-vesicle rem-brine.
In a recent unpublished study, it has keen discovered that injection of mice with nontoxic ricing A
chain, followed 4-8 hours later by injection with non-toxic ricing B chain, produces ricin-induced death. This probably occurs by reformation of the intact ricing molecule in the serum or on the surface of circulating cells. It is believed, therefore, that the B chain plays an active role in potentiating the toxic activity of the ricing A chain.

12~Z1~8 In accordance with the invention, there are provided compositions and a method for potentiating the cytotoxic activity of cell surface binding agent-toxin conjugates while at the same time retaining target cell specificity. The compositions provided by the present invention include a selective binding agent coupled to a toxin B chain moiety.
The present invention, then, in one aspect, resides in a process for preparing a toxin B chain conjugate which comprises covalently coupling a cell surface affinity binding agent to a toxin B chain moiety.
Further, there is provided a composition come prosing, in combination, a first conjugate including a selective binding agent coupled to a toxin chain moiety together with a second conjugate including a cell surface binding agent coupled to a toxin A chain moiety. The selective binding agent of the first con-gigawatt can be either a cell surface binding agent or a binding agent specific for the cell surface binding agent of the second conjugate.
In one aspect of the i~Yention, there is pro-voided a conjugate which encompasses an antibody as the cell surface binding agent coupled to a ricing B chain moiety. Further, there is provided a composition come prosing a combination a first conjugate of an antibody coupled to a ricing B chain moiety together with a second conjugate of an antibody coupled to a ricing A
chain moiety.
In yet another aspect of the invention there is provided a method for eliminating target cells from a population of cells containing such target cells by contacting the population of cells with a first con-gigawatt comprising an affinity binding agent which is specific for an antigenic determinant on such target cells and which is coupled to a toxin B chain moiety ~,~

1;232~

and a second conjugate comprising a cell surface aft finite binding agent specific to a different determi-Nat on the same target cell and which is coupled to a toxin A chain moiety. The mixture of such conjugates potentates the selective cytotoxic activity provided by the conjugate comprising a cell surface affinity binding agent coupled to toxin A chain moiety alone.
In addition, there is provided a process for preparing a toxin B chain conjugate which comprises covalently coupling a toxin B chain moiety to an anti-body or cell surface binding fragment thereof.
As a preferred embodiment, this invention provides a conjugate comprising an antibody coupled to a toxin B chain moiety. Further, this invention provides a method for eliminating target cells using, in concert, a composition comprising a first conjugate containing ricing B chain moiety and a second conjugate containing ; ricing A chain moiety.
! Ricing is one of a number of toxin proteins which, in minute quantities, exhibits considerable toxicity toward cells. Ricing toxin is composed of two different glycoprotein chains covalently linked via a single disulfide bond. The A chain of ricing (AM
30,000) is responsible for the toxicity caused by irreversible abrogation of protein synthesis. Ricing B
chain (AM 32,000) functions as a pectin which binds to galactose-containing glycoproteins or glycolipids ox-posed on cell surfaces. The general structure and mode of action exhibited by ricing is present in a variety of plant toxin proteins such as auburn, modeccin, puked X-65~4 -6-mitogen factor, and viscumin, and bacterial toxin pro-loins such as cholera, E. golf. heat-labile, pertussis, tetanus, botulinum, pseudomonas, shaggily, and diphtheria toxins.
The ricing s conjugate and the ricing A conjugate used in the methods of this invention each comprise two active moieties: a cell surface or selective binding agent and a toxin A or B chain subunit covalently joined, preferably via a coupling agent. In each composition, lo one of the active moieties is represented by a molecule having binding affinity to a surface structure of a target cell or binding affinity to cell surface binding agent of the toxin A chain conjugate. Typically such molecules may be substances such as hormones, growth factors, pectins, or antibodies. The molecules of choice are antibodies or fragments thereof (in portico-far, Fob fragments) having cell surface binding affinity.
Monoclonal antibodies are preferred but not essential. Immunoglobulin fractions from serum can be used, albeit with a lesser degree of target specificity.
Since the immunoglobulin fraction of an antiserum con twins a multitude of antibodies directed to a wide range of divergent antigens, a practical usefulness of the compositions of this invention and the defined method for eliminating target cells dictates the need to isolate a desired collection of antibodies, each directed to a surface antigenic determinant present on the particular target cell.

~:232~

x-6s64 -7-An effective collection of such antibodies can be obtained by passing the immunoglobulin fraction over a column containing the respective antigen chemically coupled to a matrix. Antibody specific to the antigen will be retained on the column while unrelated immune-globulin passes through. The retained antibody then can be collected by elusion from the column using suit-able eluding agents, such as acidic buffers of chaos tropic agents. One should note that the isolated immunoglobulin, although directed to a single antigen, is not homogeneous. It comprises antibodies directed to a variety of antigenic determinants present on the antigen molecule. Consequently, the possibility exists for cross-reaction with other related antigens.
Therefore, use of monoclonal antibodies in preparing the compositions of this invention is highly preferred because they are directed to only one of posse-by many antigenic determinants present on an antigen.
Monoclonal antibodies are available by recognized methodology from hybridomas derived from lymphocytes present in the spleen or other organ Moreover, the use of monoclonal antibodies in the compositions used in the method of this invention carries the highly desirable feature of enhanced selectivity. Therefore, that both the ricing B conjugate and the ricing A conjugate be comprised of a monoclonal antibody is highly preferred. This ensures a high level of target cell specificity.

of The preceding paragraphs have reference to owe aspect of the present invention, the use of the same cell surface affinity binding agent for construction of both the ricing A chain conjugate and the ricing B chain conjugate. A greater degree of specificity, however, can be attained. Because a normal or tumor cell bears several different surface markers, one may be able to target the ricing A and the ricing s chains to such cells by coupling each to an antibody directed against a different determinant on the same cell. For example, in the case of a neoplastic B cell bearing both surface It (spa) and surface Ig(sIg), immunotoxins against the spa and the swig idiotype can be prepared with the ricing B chain and ricing A chain, respectively. In the case of T cell tumors, a number of monoclonal antibodies exist which are reactive with subsets of human T cells. By using selected combinations of antibodies, one may target the ricing A and ricing B chains to specific sub-sets of such culls. Preferably, one antibody (coupled to ricing A chain) would define the subset, and the second (coupled to ricing B chain) would be a more general marker common to many subsets of cells. The B chain immunotoxin, directed against the more common marker, would bind also to normal cells; however, they would not be deleted. In contrast, the A chain-immunotoxin would be focused only on the tumor cell and would be potentiated by B chain-containing immunotoxin.
Another approach contemplated by the present invention involves first directing a tumor cell reactive antibody-ricin A chain conjugate to tumor cells in viva.
The antibody preferably is univalent, e.g. Phoebe, ~'~32~

and, therefore, is unable to cap and modulate. After the antibody-ricin A conjugate has been injected into a cancer-bearing patient and the excess eliminated from the recipient by excretion or degradation, a ricing B
chain-containing immunotoxin directed against the antibody of the ricing A conjugate is injected. Only those cells which had bound the first immunotoxin would focus the second immunotoxin on the first. Therefore, such cells would be selectively deleted. The second - 10 immunotoxin preferably is a diva lent anti-antibody, such as a F(ab')2-B, which would not bind to macro-phases, mainstays, or other cells bearing Fc receptors.
Furthermore, since the B chain-containing immunotoxin would be innocuous if nonspecifically bound to a cell which had not previously bound the first immunotoxin, any side effects caused by the administration of the second immunotoxin would be eliminated. In contrast, cells binding both immunotoxins would be killed.
As noted, the ricing B-containing composition of this invention and the ricing A-containing compost-lion used in the method of this invention each come proses at least two separate active moieties, one of which affords binding affinity (BY) and the other of which is a ricing subunit (US), whether ricing A (RAY) or ricing B (RUB). These are joined through a coupling reagent, the requirements of the resulting composition being (a) the presence of at least one of each class of moiety, and (b) the retention of the innate activity of at least one of each class of moiety.

I

Other toxin proteins may be similarly coupled to the binding agent component for use in accordance with the present invention. Due to the similarity in their structure and mode of action, plant or bacterial toxin proteins such as auburn, modeccin, poXeweed mitogen factor, viscumin, and cholera, E. golf. heat-labile, pertussis, tetanus, botulinum, pseudomonas, shigella and diphtheria toxins may be utilized. Fur then, it may be advantageous to couple the A chain from auburn, for example, to a cell surface binding moiety to form the first conjugate of the invention and the B chain from viscumin, for example, to a selective binding agent moiety to form the second conjugate. It may be advantageous to use a plant protein toxin such as gelonin, which consists only of an A chain, as the A chain to be coupled to the cell surface binding moiety to form the first conjugate. This first conjugate may be used then with a conjugate comprising a selective binding moiety coupled to a B chain selected from any one of the toxins ricing viscumin, modeccin or auburn.
In accordance with these limitations, the compositions of this invention and those used in the method of this invention can be dim Eric (BARS), i. e., contain one of each class of moiety; trimeLic BUYERS) or (BUYERS)], lye., contain two of one class of moiety and one of the other; tetrameric [(BYPASS), (BUYERS), or (BUYERS)]; and the like.
As noted, highly preferred compositions for use in the method of this invention are those in which the binding moiety is antibody or an antigen binding I

fragment of antibody; and preferably a monoclonal anti-body or an antigen binding fragment thereof. Typical compositions may be Ab-P~B, Ab2-RB, Ab-RB2, Ab3-RB,
2 2' A RB3, Abhor, Abhor, Ab~P~2, Brie by ERA
or Abhor.
In preparing the compositions of this invent lion, the BY and US moieties are joined via a suitable coupling reagent. A wide variety of coupling agents is reported in chose, T., and Blair, A. I., J. Tuttle. Cancer lo Inst. 61, 657-676 (1980). These authors report the use of carbodiimides as well as other bifunctional reagents, such as glutaraldehyde, ~-b~nzoquinone, deflower-m,_'dinitrodiphenylsulfone, or dim ethyl adipimidate, for coupling antibody to cytotoxic agents. Because it is highly desirable to preclude formation of homopolymers, e.g., By or Run use of a heterobifunctional reagent is preferred, ensuring formation of compositions having at least one of each class of moiety. Examples of such heterobifunctional reagents may be N-succinimidyl-3-~2-pyridyldithio)propionate (SPDP), _-maleimidobenzoyl-N-hydroxy-succinimidyl ester, bromoacetyl-_-aminobenzoyl-N-hydroxy-succinimidyl ester, or iodoacetyl N-hydroxy-succinimidyl ester.
For example, using SPDP as coupling agent, a process for preparing a composition of this invention comprises (a) separately modifying both A and US by reaction with SPDP, (b) reducing the Ab-containing pro-duct, (c) causing formation of the composition by mixing the Ab-containing and RS-contailling products, and (d) separating non-reacted monomers Joy gel filtration.
3~3 The conjugates of this invention containing ricing B, when used in concert with ricing A conjugates, have general applicability in the specific and select live killing of a cell type defined by particular anti-genie markers. By appropriate selection of the anti~genic marker the cell surface binding agent can be directed to either a set of normal cells or to a subset of neoplastic cells bearing a distinguishing determinant. As such, they are useful, for example, in the immunotherapy of cancer, for treating parasitic infections, and for treating a wide range of autoirnmune diseases. Moreover, the compositions have several in vitro applications, including, for example, elimination of leukemia cells in bone marrow prior to autologous bone marrow transplantation; elimination of T cells in bone marrow prior to allogeneic bone marrow trays-plantation; and killing of wild types for selection of mutants .
The compositions of this invention can be used in a variety of pharmaceutical formulations and can be administered by a variety of conventional routes, such as intramuscular, intravenous, subcutaneous, and intraperitoneal. As used, the term "pharmaceutically acceptable" means those agents useful in the comma-therapy of warm-blooded animals.
When administering the conjugate compositions parenterally or intraperitoneally, pharmaceutically acceptable forms for injection may include sterile aqueous solutions or dispersions and sterile powders for reconstitution into sterile injectable solutions I

X-65~4 -13-or dispersions. The carrier can be a solvent or disk pursing medium containing, for example, water, ethanol, polyol for example glycerol, propylene glycol, or fig-rid polyethylene glycol) suitable mixtures thereof, and vegetable oils. Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants. van-out antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like may also be used. In many cases, including isotonic agents, for example, sugars, sodium chloride, and the like will be desirable. Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum moo-Stewart and gelatin.
Sterile injectable solutions can be prepared by associating the conjugate composition refined earlier in the required amount of the appropriate solvent with any other ingredient as desired.
If desired, for more effective distribution, the compositions can be incorporated into slow release systems such as polymer matrices, liposomes, and micro spheres. Moreover, the compositions can be administered either alone or as a mixture of a plurality of active ingredients.
Doses of the compositions are administered Jo the recipient for a period during which a therapeutic response is desired. The weight of the recipient and mode of administration will determine the size of the dose necessary to induce the desired response.

~Z3~3~

Especially advantageous is to formulate the conjugate compositions in unit dosage form for ease of administration and uniformity of dosage. Unit dosage form refers to a physically discrete unit suited as unitary dosages for the subject to be treated. Each unit contains a predetermined quantity of the compost-lion calculated to produce the desired therapeutic effect in association with the pharmaceutically-acceptable carrier. The specific unit dosage form is dependent upon (a) the unique characteristics of the particular composition and by the particular Thorpe-tic effect to be achieved.
The following non-limiting examples are pro-voided to further illustrate the invention.
I. PREPARATION OF IMMUNOTOXINS

A. RICING A AND B CHAIN
The A and B chain subunits of ricing were pun-chased from Coma Corporation, San Francisco, California Prior to use, the A and B chains were dialyzed extent lively at 4~C against phosphate buffered saline (PBS), pi 7.2. The recovery of the A and B chains were 50%
and 80%, respectively.
B. ANTIBODY
The selective and cell surface binding agent of this embodiment is affinity purified rabbit anti-human immunoglobulin (Rug) made according to protocol described in the literature tree, e q. Myriad, et at., Blood, 42, 327 (1983); Vitetta, et at., Science, 219, 644 ~19~3~).

. .

`" ~L2~2~
~-6564 -15-C. CONJUGATION
10 I of 60 my dithiothreitol (DOT) in PBS was added to each my of the dialyzed A or B chain. The mixtures were incubated at 25C for 60 minutes and the reduced chains were separated from the DOT by gel lit-traction at 25C on a Sephadex G-25 *column (18 x 1.5 cm) in PBS, put 7.2. Antibodies were coupled as described in Vitetta, et at., Immunol. Rev. 62:159-183 (1982), and Carlsson, et at., Become. J., 173, 723 (1978), In particular, the antibody in PBS is treated with SPDP, N-succinimidyl-3-(2-pyridyl)dithio)propionate at a PI of ~uccinimydyl-3-(2-pyridyl)dithio)propionate at a pi of about 7.0 to 7.5 at a temperature of about 20C to about 25C. The antibody derivative then is treated with dithiothreitol in buffer solution. The thiolated antibody may be purified by gel filtration, if desired.
The activated coupled antibodies were mixed with the freshly reduced A or B chains at an antibody chain or antibody chain molar ratio of 5:1. The mixture was incubated for 15 minute at 4C with gentle shaking and then dialyzed overnight against ABS at 4C. The immunotoxins were concentrated to 1 mg/ml by pervapora-lion, dialyzed for 2-16 hours at 4C against PBS, and centrifuged to remove insoluble material. The swooper-lion of the immunotoxin from the majority of free Chain, B chains and antibody was performed by gel lit-traction at 25C on a Sephacryl S-200*c*olumn equal-brazed with PBS, pi 7.2. Material with an apparent molecular weight of greater than 200,000 was pooled.
Reduced and alkylated bovine alpha globulin, fraction * Trade Mark for an ion exchange resin formed of highly cross linked dextran containing functional ionic groups attached to the glucose units of the polysaccharide chains by ether linkages.
** Trade Mark .

~Z3Z~
4 (Sigma), was added to a final concentration of 1 mg/ml to the pooled samples. The samples were stored 1~-20 hours at 4C prior to affinity purification.
The Rug chain (Rug) and Rugby chain (Rugby) immunotoxins were purified by affinity chromatography on Sepharose~Ig. For purification of the A chain-containing immunotoxins, the columns were equilibrated and washed in PBS, pi 7.2 and for the puff-ligation of the B chain-containing immunotoxins, the columns were equilibrated and washed in PBS, pi 7.6 con-twining elm galactose. Samples were applied to the column and the fall-through was discarded. The columns were washed extensively in PBS (or PBS-O.lM gal) lot-lowed by 0.85% Nail. Immunotoxins were eluded batch-wise at 37C with 2-3 column volumes of 3.5M McCoy.
The McCoy was removed by dialysis and the samples were concentrated by pervaporation to approximately 200-300 gel Reduced and alkylated bovine alpha globulin was added to a final concentration of 1 mg/ml. Samples were sterilized by filtration Jon a filter preheated with PBS
2% fetal calf serum (FCS)], and then allocated into sterile vials and stored at -20C. Samples stored in - this manner were stable for up to 4-6 months.
The recovery of the immunoto~in following affinity purification was 26-45% and each immunotoxin contained one or two A (or B) chain subunits per mole-cute of antibody. When immunotoxins were analyzed on SDS-PAGE slab gels using a sensitive silver staining technique, no free A or B chain in any of the immune-toxins was detected.
* Trade Mark for Agrees ~2~2~

II. USE OF RICING B CHAIN CONTAINING CONJUGATES
AND RICING A CHAIN CONJUGATES It THE KILL-IN OF NEOPLASTIC HUMAN B CELLS _ A. NEOPLASTIC CELL CULTURE
The human Berkowitz lymphoma cell line, Saudi, as described in Houston, LO Become. Boyce. Rest Commune 92:319-326 (1980), was maintained in suspension -culture in RPMI-1640 supplemented with 10% (v/v) fetal calf serum (FCS), 20 my fresh glut amine, and antibiotics.
Cultures were maintained at 37C in a humid incubator with a 95% 2/5% C2 atmosphere. All cultures were used two days following subculture.

B. TREATMENT OF SAUDI CELLS
It days after subculture, Saudi cells were harvested and washed in buffered saline solution (BUS).
105 cells in BUS were distributed into micro titer wells.
Dilutions of each of the two immunotoxins, Rug and Rugby, or combination of both, were added to triple-gate wells for 15 minutes at 4C. Cells were centric fused and washed three times in BUS. 200 I of RPMI
lacking Lawson and containing 10% FCS were added to each well and the cells were resuspended by gentle ago-station. Plates were cultured for 22 hours at 37 in a% C2 incubator. Cells were then pulsed for 6 hours at 37 in 5% COY with 5 Sue per well of Lawson (New England Nuclear). Cells were harvested onto glass fiber discs and counted in a liquid scintillation spectrometer.
Cells were treated with each of the immunotoxins India ~3Z~

visually as well as the following combinations of immunotoxins:
1. A nontoxic amount of Rug (0.3 gig plus different amounts of Rugby, or S 2. A nontoxic amount of Rugby (0.5 gel followed by different amolmts of Rug.
The results are tabulated in TABLE 1.

Table 1 - % of Cells Remaining After Treatment vs.
Control Conjugate Used Concentration gel 0.03 0.05 0.3 0.5 1.3 2.6 IT* 83 90 100 100 90 82 IT** 100 90 82 - 22 18 IT -I IT (0.5 gel 75 40 12 - 6 5 IT + IT (0.3 gel 95 64 50 - 20 10 *IT = immunotoxin B conjugate (Rugby) **IT = immunotoxin A conjugate (Rug) As indicated in the Table, wren Saudi cells were treated with 0.3 go of Rug chain/105 cells, little toxicity was observed. No concentration of Rugby was toxic. However, when 0.3 go of the Rug was mixed with various combinations of Rugby, there was significant cytoxicity. It should be noted that this treatment of the Saudi cells with the mixture of immunotoxins was performed in BUS lacking galactose.
As shown in TABLE 1, when Saudi cells were treated with 0.5 go of Rugby, no toxicity was observed. In con-tryst, the Rug willed the Saudi cells in a dose-related manner. However, treatment of cells with 0.5 go ~3;~33~

of Rugby mixed with R~Hlg-A was toxic to the cells, even at those concentrations at which Rug itself was not toxic.
Although the conjugate compositions and methods have been described in terms of preferred embodiment, those skilled in the art will recognize that various changes may be made without departing from the intended : scope of the invention.

Claims (18)

The embodiments of the invention in which an exclusive property or privilege is claimed are defined as follows:
1. A process for preparing a toxin B chain conjugate which comprises covalently coupling a cell surface affinity binding agent to a toxin B chain moiety.
2. A process according to Claim 1 for preparing a toxin B chain conjugate which comprises covalently coupling an antibody or a cell surface binding fragment thereof to a toxin B chain moiety.
3. A process for preparing a conjugate as claimed in Claim 2 in which the antibody or cell surface binding fragment thereof is specific for a cell surface antigen.
4. A process for preparing a conjugate as claimed in Claim 2 in which the antibody or cell surface binding fragment thereof is directed to a cell surface antigen of a tumor cell.
5. A process for preparing a conjugate as claimed in Claim 2 in which the antibody or cell surface binding fragment thereof is directed against a second antibody.
6. A process for preparing a conjugate as claimed in Claim 1 in which the toxin B chain is selected from ricin B chain, modeccin B chain, abrin B chain, pokeweed mitogen factor B chain, viscumin B chain, cholera toxin B chain, E.coli heat-labile toxin B chain, pertussis toxin B chain, botulinum toxin B chain, Pseudomonas toxin B chain, shigella toxin B chain or diphtheria toxin B chain.
7. A process for preparing a conjugate as claimed in Claim 5 in which the toxin B chain is ricin B chain.
8. A toxin B chain conjugate whenever prepared by a process according to any one of Claims 1 to 3, or by an obvious chemical equivalent thereof.
9. A toxin B chain conjugate whenever prepared by a process according to any one of Claims 4, 5 or 6, or by an obvious chemical equivalent thereof.
10. A toxin B chain conjugate whenever prepared by a process according to Claim 7 or by an obvious chemical equivalent thereof.
11. A process for preparing a cytotoxic composition which comprises associating a first conjugate prepared in accordance with the process of Claim 1, together with a second conjugate comprising an antibody or cell surface binding fragment thereof covalently coupled to a toxin A chain moiety.
12. A process for preparing a cytotoxic composition as claimed in Claim 11 in which the second conjugate antibody or cell surface binding fragment thereof is directed against a cell surface antigenic determinant.
13. A process for preparing a cytotoxic composition as claimed in Claim 11 or 12 in which each of the first and second conjugates comprises an antibody or cell surface binding fragment thereof, having identical specificity to a cell surface antigenic determinant.
14. A process for preparing a cytotoxic composition as claimed in Claim 11 or 12 in which the first conjugate antibody is directed against a cell surface antigenic determinant different from the cell surface antigenic determinant to which the second conjugate antibody or cell surface binding fragment thereof is directed.
15. A process for preparing a cytotoxic composition as claimed in Claim 11 in which each antibody or cell surface binding fragment thereof is specific for a tumor cell antigenic determinant.
16. A process for preparing a cytotoxic composition as claimed in Claim 11 in which the second conjugate comprises an antibody or cell surface binding fragment thereof which is specific for a cell surface antigenic determinant and the first conjugate comprises an antibody or cell surface binding fragment which is specific for the antibody of the second conjugate.
17. A process for preparing a cytotoxic composition as claimed in Claim 11 in which the toxin A chain is selected from the A chain moiety of ricin, abrin, modeccin, gelonin, pokeweed mitogen factor, viscumin, cholera toxin, E. coli heat-labile toxin, pertussis toxin, botulinum toxin, Pseudomonas toxin, shigella toxin and diphtheria toxin.
18. A process for preparing a cycotoxin composition as claimed in Claim 17 in which the toxin A
chain moiety is ricin A chain and the toxin B chain moiety is ricin B chain.
CA000456816A 1983-06-21 1984-06-18 Immunotoxin conjugates Expired CA1232838A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US06/506,540 US4664911A (en) 1983-06-21 1983-06-21 Immunotoxin conjugates employing toxin B chain moieties
US506,540 1983-06-21

Publications (1)

Publication Number Publication Date
CA1232838A true CA1232838A (en) 1988-02-16

Family

ID=24015007

Family Applications (1)

Application Number Title Priority Date Filing Date
CA000456816A Expired CA1232838A (en) 1983-06-21 1984-06-18 Immunotoxin conjugates

Country Status (18)

Country Link
US (1) US4664911A (en)
EP (1) EP0129434B1 (en)
JP (1) JPH0639387B2 (en)
KR (1) KR860001803B1 (en)
AT (1) ATE46084T1 (en)
AU (1) AU570049B2 (en)
CA (1) CA1232838A (en)
DE (1) DE3479633D1 (en)
DK (1) DK297184A (en)
ES (2) ES8604425A1 (en)
FI (1) FI842420A (en)
GB (1) GB2142032B (en)
GR (1) GR82368B (en)
HU (1) HUT34348A (en)
IL (1) IL72117A0 (en)
PL (1) PL248202A1 (en)
PT (1) PT78752A (en)
ZA (1) ZA844512B (en)

Families Citing this family (84)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4925922A (en) * 1983-02-22 1990-05-15 Xoma Corporation Potentiation of cytotoxic conjugates
FR2550944B1 (en) * 1983-08-23 1985-12-06 Sanofi Sa PHARMACEUTICAL ASSOCIATION BASED ON TUMOR ANTI-CELL ANTIBODIES AND CYTOTOXIC CONJUGATES, SUITABLE IN PARTICULAR FOR THE TREATMENT OF CANCERS
US4590071A (en) * 1984-09-25 1986-05-20 Xoma Corporation Human melanoma specific immunotoxins
EP0232447A1 (en) * 1986-02-13 1987-08-19 Xoma Corporation Lectin immunotoxins
US6103243A (en) * 1985-05-15 2000-08-15 Biotechnology Australia Pty, Ltd Oral vaccines
US5283323A (en) * 1985-08-07 1994-02-01 The United States Of America As Represented By The Department Of Health And Human Services Method of producing immune response
AU609380B2 (en) * 1985-11-29 1991-05-02 Consolidated Pharmaceuticals Limited Ricin-antibody conjugates
US4911911A (en) * 1985-12-20 1990-03-27 Sanofi Ribosome-inactivating glycoproteins, modified by oxidation of their osidic units and formation of a schiff's base and in-vivo prolonged action immunotoxins containing such a glycoprotein
FR2602682B1 (en) * 1986-08-12 1988-12-02 Sanofi Sa LONG-DURING IN VIVO IMMUNOTOXINS COMPRISING A RIBOSOME INHIBITING GLYCOPROTEIN MODIFIED BY OXIDATION OF OSID PATTERNS AND FORMATION OF A SCHIFF BASE
US4831122A (en) * 1986-01-09 1989-05-16 Regents Of The University Of Minnesota Radioimmunotoxins
ATE128361T1 (en) * 1986-03-20 1995-10-15 Dana Farber Cancer Inst Inc LECTIN COMPLEX, AND METHOD AND PROBE FOR THE PRODUCTION THEREOF.
US5239062A (en) * 1986-03-20 1993-08-24 Dana-Farber Cancer Institute, Inc. Blocked lectins, methods and affinity support for making same using affinity ligands, and method of killing selected cell populations having reduced nonselective cytotoxicity
US5395924A (en) * 1986-03-20 1995-03-07 Dana-Farber Cancer Institute, Inc. Blocked lectins; methods and affinity support for making the same using affinity ligands; and method of killing selected cell populations having reduced non-selective cytotoxicity
US4806494A (en) * 1986-07-24 1989-02-21 The United States Of America As Represented By The Department Of Health & Human Services Monoclonal antibody against ovarian cancer cells (OVB-3)
FR2604092B1 (en) * 1986-09-19 1990-04-13 Immunotech Sa IMMUNOREACTIVES FOR TARGETING ANIMAL CELLS FOR VISUALIZATION OR DESTRUCTION IN VIVO
AU616161B2 (en) * 1986-10-09 1991-10-24 Neorx Corporation Methods for improved targeting of antibody, antibody fragments, hormones and other targeting agents, and conjugates thereof
US4808705A (en) * 1986-12-19 1989-02-28 Cetus Corporation Stable formulations of ricin toxin a chain and of RTA-immunoconjugates and stabilizer screening methods therefor
US4962048A (en) * 1987-02-19 1990-10-09 Scripps Clinic & Research Foundation Monoclonal antibodies to human pancreatic cancer
US5320942A (en) * 1987-02-19 1994-06-14 Vito Quaranta Method of diagnosing the presence of abnormal epithelial tissue using monoclonal antibodies to the A6 B4 cell surface protein
US5981278A (en) * 1987-05-29 1999-11-09 Tanox, Inc. Chimeric monoclonal antibodies which neutralize HIV-1 infection and their applications in therapy and prevention for AIDS
US6657050B1 (en) 1987-05-29 2003-12-02 Tanox, Inc. Chimeric viral-neutralizing immunoglobulins
US5834599A (en) * 1987-05-29 1998-11-10 Tanox Biosystems, Inc. Immunoconjugates which neutralize HIV-1 infection
CA1335227C (en) * 1987-06-22 1995-04-11 Wylie W. Vale, Jr. Crf analog conjugates
IE890206L (en) * 1988-02-02 1989-08-02 Akzo Nv Immunotoxins from barley toxin
WO1989006967A1 (en) * 1988-02-03 1989-08-10 Xoma Corporation Immunosupression with anti-pan t-cell immunotoxin compositions
US5827934A (en) * 1988-03-08 1998-10-27 The University Of Wyoming Cytotoxic diphtheria toxin fragments
AU631545B2 (en) * 1988-04-15 1992-12-03 Protein Design Labs, Inc. Il-2 receptor-specific chimeric antibodies
US5149782A (en) * 1988-08-19 1992-09-22 Tanox Biosystems, Inc. Molecular conjugates containing cell membrane-blending agents
US5045451A (en) * 1988-10-26 1991-09-03 Board Of Regents Methods for screening antibodies for use as immunotoxins
US5621078A (en) * 1989-03-22 1997-04-15 Merck & Co., Inc. Modified pseudomonas exotoxin PE40
US6207798B1 (en) 1989-08-03 2001-03-27 Merck & Co., Inc. Modified PE40 toxin fusion proteins
WO1991004050A1 (en) * 1989-09-14 1991-04-04 Board Of Regents, The University Of Texas System Therapeutic compositions; methods for treatment of hiv infections
IL98528A0 (en) * 1990-06-21 1992-07-15 Merck & Co Inc Pharmaceutical compositions containing hybrid for killing bladder cancer cells
US5401243A (en) * 1990-08-21 1995-03-28 Associated Synapse Biologics Controlled administration of chemodenervating pharmaceuticals
US5183462A (en) * 1990-08-21 1993-02-02 Associated Synapse Biologics Controlled administration of chemodenervating pharmaceuticals
WO1992007574A1 (en) * 1990-10-25 1992-05-14 Tanox Biosystems, Inc. Glycoproteins associated with membrane-bound immunoglobulins as antibody targets on b cells
US6605712B1 (en) * 1990-12-20 2003-08-12 Arch Development Corporation Gene transcription and ionizing radiation: methods and compositions
EP0495376B1 (en) * 1991-01-15 1997-05-28 Bayer Corporation Cell surface receptor complimentation
US7070991B2 (en) 1991-02-08 2006-07-04 Progenics Pharmaceuticals, Inc. Cells expressing a CD4-IgG2 chimeric heterotetramer
AU660662B2 (en) 1991-02-08 1995-07-06 Progenics Pharmaceuticals, Inc. CD4-gamma2 and CD4-IgG2 chimeras
US5112607A (en) * 1991-06-05 1992-05-12 The United States Of America As Represented By The Secretary Of The Army Potentiation of immunotoxin action by Brefeldin A
WO1993004191A1 (en) * 1991-08-15 1993-03-04 Neorx Corporation Noncytolytic toxin conjugates
WO1993015113A1 (en) * 1992-01-24 1993-08-05 Tanox Biosystems, Inc. An immunotoxin including a cytotoxin with an unpaired cysteine residue in or near its receptor-binding site
US5686072A (en) * 1992-06-17 1997-11-11 Board Of Regents, The University Of Texas Epitope-specific monoclonal antibodies and immunotoxins and uses thereof
AU2593192A (en) 1992-09-14 1994-04-12 Oystein Fodstad Detection of specific target cells in specialized or mixed cell population and solutions containing mixed cell populations
GB9305735D0 (en) * 1993-03-19 1993-05-05 North John R Novel agent for controlling cell activity
EP1099445A3 (en) 1993-06-10 2001-09-05 Allergan, Inc. Treatment of neuromuscular disorders and conditions with different botulinum serotype
US5876438A (en) * 1993-08-02 1999-03-02 Houston Biotechnology Incorporated Polymeric device for the delivery of immunotoxins for the prevention of secondary cataract
US7214787B1 (en) * 1993-09-21 2007-05-08 United States Of America As Represented By The Secretary Of The Army Recombinant vaccine against botulinum neurotoxin
EP2027872B1 (en) 1993-12-28 2010-07-14 Allergan, Inc. Neurotoxic component of a botulinum toxin for treating tardive dyskinesia
US5759546A (en) 1994-02-04 1998-06-02 Weinberg; Andrew D. Treatment of CD4 T-cell mediated conditions
NO180658C (en) 1994-03-10 1997-05-21 Oeystein Fodstad Method and Device for Detecting Specific Target Cells in Specialized or Mixed Cell Populations and Solutions Containing Mixed Cell Populations
US6967088B1 (en) * 1995-03-16 2005-11-22 Allergan, Inc. Soluble recombinant botulinum toxin proteins
GB9508204D0 (en) * 1995-04-21 1995-06-07 Speywood Lab Ltd A novel agent able to modify peripheral afferent function
US5830478A (en) * 1995-06-07 1998-11-03 Boston Biomedical Research Institute Method for delivering functional domains of diphtheria toxin to a cellular target
US6497881B1 (en) * 1995-11-30 2002-12-24 New York University High efficiency tissue specific compound delivery system using streptavidin-protein a fusion protein
US6312700B1 (en) * 1998-02-24 2001-11-06 Andrew D. Weinberg Method for enhancing an antigen specific immune response with OX-40L
US6960652B2 (en) * 1999-03-30 2005-11-01 Board Of Regents, The University Of Texas System Compositions and methods for modifying toxic effects of proteinaceous compounds
GB9909796D0 (en) * 1999-04-28 1999-06-23 Plant Bioscience Ltd Pesticidal fumes
US7838008B2 (en) 1999-12-07 2010-11-23 Allergan, Inc. Methods for treating diverse cancers
US7838007B2 (en) 1999-12-07 2010-11-23 Allergan, Inc. Methods for treating mammary gland disorders
US6139845A (en) 1999-12-07 2000-10-31 Allergan Sales, Inc. Method for treating cancer with a neurotoxin
US6641820B1 (en) 2000-01-19 2003-11-04 Allergan, Inc. Clostridial toxin derivatives and methods to treat pain
US6500436B2 (en) 2000-01-19 2002-12-31 Allergan, Inc. Clostridial toxin derivatives and methods for treating pain
US7138127B1 (en) * 2000-01-19 2006-11-21 Allergan, Inc. Clostridial toxin derivatives and methods for treating pain
ES2380007T3 (en) * 2001-04-06 2012-05-07 The University Of Chicago Induction by chemotherapeutic agents of the activity of the Egr-1 promoter in gene therapy
US20040242523A1 (en) * 2003-03-06 2004-12-02 Ana-Farber Cancer Institue And The Univiersity Of Chicago Chemo-inducible cancer gene therapy
US8034791B2 (en) 2001-04-06 2011-10-11 The University Of Chicago Activation of Egr-1 promoter by DNA damaging chemotherapeutics
US7022329B2 (en) * 2002-02-25 2006-04-04 Allergan, Inc. Method for treating neurogenic inflammation pain with botulinum toxin and substance P components
US20030211470A1 (en) * 2002-03-15 2003-11-13 Olson William C. CD4-IgG2-based salvage therapy of HIV-1 infection
US20050142139A1 (en) * 2003-03-21 2005-06-30 Norbert Schulke CD4-IgG2 formulations
EP1684785A4 (en) * 2003-11-14 2009-08-12 Jay W Chaplin B cell-targeted toxins for humoral immune response reduction
US7358334B1 (en) 2003-11-14 2008-04-15 Jay W Chaplin B cell-targeted toxins for humoral immune response reduction and methods of use thereof
US20110166319A1 (en) * 2005-02-11 2011-07-07 Immunogen, Inc. Process for preparing purified drug conjugates
CA2597407C (en) * 2005-02-11 2013-09-10 Immunogen, Inc. Process for preparing stable drug conjugates
CA2597244A1 (en) * 2005-03-03 2006-09-14 Universite Catholique De Louvain Methods and compositions for the treatment of cancer
WO2006110623A2 (en) * 2005-04-09 2006-10-19 University Of Florida Research Foundation, Inc. Targeted cholera toxin for treatment of persistent or chonic pain
RS52470B (en) * 2005-08-24 2013-02-28 Immunogen Inc. Process for preparing maytansinoid antibody conjugates
WO2007109733A2 (en) 2006-03-21 2007-09-27 The Johns Hopkins University Diagnostic and prognostic markers and treatment strategies for multiple sclerosis
JP2011514307A (en) * 2007-10-23 2011-05-06 アラーガン、インコーポレイテッド Method of treating chronic neurogenic inflammation using modified Clostridial toxin
EP2350131B1 (en) 2008-11-07 2017-06-07 Research Development Foundation Compositions and methods for the inhibition of cripto/grp78 complex formation and signaling
SG10201810743WA (en) 2009-06-03 2018-12-28 Immunogen Inc Conjugation methods
EA201991268A3 (en) 2011-03-29 2020-01-31 Иммуноджен, Инк. OBTAINING MAYTANSINOID-ANTIBODIES CONJUGATES IN ONE-STEP METHOD
NZ707091A (en) 2012-10-04 2018-12-21 Immunogen Inc Use of a pvdf membrane to purify cell-binding agent cytotoxic agent conjugates

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2437213A1 (en) * 1978-09-28 1980-04-25 Cm Ind CYTOTOXIC PRODUCTS FORMED BY COVALENT BINDING OF THE CHAIN TO RICIN WITH AN ANTIBODY AND THEIR PREPARATION METHOD
JPS55136235A (en) * 1979-04-09 1980-10-23 Teijin Ltd Antitumor protein complex and its preparation
JPS5616417A (en) * 1979-07-19 1981-02-17 Teijin Ltd Antitumor protein complex and its preparation
JPS5616418A (en) * 1979-07-20 1981-02-17 Teijin Ltd Antitumor protein complex and its preparation
JPS5686121A (en) * 1979-12-14 1981-07-13 Teijin Ltd Antitumor proten complex and its preparation
US4322495A (en) * 1980-03-11 1982-03-30 Minnesota Mining And Manufacturing Co. Immunoassay
EP0044167A3 (en) * 1980-07-14 1982-04-21 The Regents Of The University Of California Antibody targeted cytotoxic agent
US4440747A (en) * 1980-09-30 1984-04-03 The United States Of America As Represented By The Department Of Health And Human Services Monoclonal antibody-ricin hybrids as a treatment of murine graft-versus-host disease
US4359457A (en) * 1980-09-30 1982-11-16 Neville Jr David M Anti Thy 1.2 monoclonal antibody-ricin hybrid utilized as a tumor suppressant
JPS5764617A (en) * 1980-10-08 1982-04-19 Teijin Ltd Cytotoxic proteinic complex and its preparation
US4356117A (en) * 1980-10-23 1982-10-26 U.S. Govt., Dept. Of Health & Human Services Chemical modifications of proteins which induce new receptor specificities and therefore elicit new effects in cells
US4397843A (en) * 1980-10-23 1983-08-09 The United States Of America As Represented By The Department Of Health And Human Services Mannose-6-phosphate-low density protein reagent effective against hypercholesterolemia
JPS57106626A (en) * 1980-12-22 1982-07-02 Teijin Ltd Cytotoxic protein complex and its preparation
JPS57106625A (en) * 1980-12-22 1982-07-02 Teijin Ltd Cytotoxic protein complex and its preparation
FR2498192A2 (en) * 1981-01-22 1982-07-23 Pasteur Institut NOVEL POLYPEPTIDE THIOLE COMPOUND FROM A FRAGMENT OF TETANIC TOXIN, PROCESS FOR OBTAINING SAME AND APPLICATIONS THEREOF
FR2504010B1 (en) * 1981-04-15 1985-10-25 Sanofi Sa ANTI-CANCER MEDICINAL PRODUCTS CONTAINING THE RICIN-ASSOCIATED CHAIN ASSOCIATED WITH ANTIMELANOMA ANTIBODY AND PROCESS FOR THEIR PREPARATION
JPS5874614A (en) * 1981-10-30 1983-05-06 Teijin Ltd Conjugate protein, its preparation, and selective cellucidal process with the same
FR2516794B1 (en) * 1981-11-20 1985-10-25 Sanofi Sa NOVEL CANCER DRUGS FOR THE TREATMENT OF LEUKEMIA T CONTAINING RICIN AND A SPECIFIC MONOCLONAL ANTIBODY
FR2536997B1 (en) * 1982-03-11 1987-08-07 Sanofi Sa CYTOTOXIC DRUGS COMPRISING AT LEAST ONE IMMUNOTOXIN AND METHYLAMINE
FR2523445A1 (en) * 1982-03-17 1983-09-23 Sanofi Sa NOVEL CONJUGATES ASSOCIATING, BY COVALENT BINDING, AN ENZYME AND ANTIBODY, AND DRUG ASSOCIATIONS USING THE SAME
US4500637A (en) * 1982-07-19 1985-02-19 The United States Of America As Represented By The Department Of Health And Human Services Prevention of graft versus host disease following bone marrow transplantation

Also Published As

Publication number Publication date
FI842420A0 (en) 1984-06-14
DK297184D0 (en) 1984-06-18
GB2142032B (en) 1987-05-13
PL248202A1 (en) 1985-03-26
EP0129434A2 (en) 1984-12-27
KR850000248A (en) 1985-02-26
JPH0639387B2 (en) 1994-05-25
ES8604425A1 (en) 1986-02-01
GB8415507D0 (en) 1984-07-25
GR82368B (en) 1984-12-13
US4664911A (en) 1987-05-12
AU570049B2 (en) 1988-03-03
FI842420A (en) 1984-12-22
ZA844512B (en) 1986-02-26
ES544846A0 (en) 1986-04-16
EP0129434A3 (en) 1985-09-18
PT78752A (en) 1984-07-01
ATE46084T1 (en) 1989-09-15
ES533510A0 (en) 1986-02-01
GB2142032A (en) 1985-01-09
KR860001803B1 (en) 1986-10-23
JPS6069033A (en) 1985-04-19
EP0129434B1 (en) 1989-09-06
AU2948084A (en) 1985-01-03
DK297184A (en) 1984-12-22
DE3479633D1 (en) 1989-10-12
ES8606884A1 (en) 1986-04-16
IL72117A0 (en) 1984-10-31
HUT34348A (en) 1985-03-28

Similar Documents

Publication Publication Date Title
CA1232838A (en) Immunotoxin conjugates
JP3589459B2 (en) Novel antibody delivery system for biological response modifiers
Dohlsten et al. Monoclonal antibody-targeted superantigens: a different class of anti-tumor agents.
Ramakrishnan et al. Comparison of the selective cytotoxic effects of immunotoxins containing ricin A chain or pokeweed antiviral protein and anti-Thy 1.1 monoclonal antibodies
Letvin et al. In vivo administration of lymphocyte-specific monoclonal antibodies in nonhuman primates. In vivo stability of disulfide-linked immunotoxin conjugates.
HU208161B (en) Process for producing cytotoxic active ingredient - antibody conjugates and pharmaceutical compositions
Ghetie et al. The GLP large scale preparation of immunotoxins containing deglycosylated ricin A chain and a hindered disulfide bond
Flavell et al. Preclinical studies with the anti-CD19-saporin immunotoxin BU12-SAPORIN for the treatment of human-B-cell tumours
JP3340127B2 (en) Antibody conjugates for the treatment of hyperproliferative diseases
US5578706A (en) Methods and compositions concerning homogenous immunotoxin preparations
US5690935A (en) Biotherapy of cancer by targeting TP-3/P80
Kishida et al. Ricin A-chain conjugated with monoclonal anti-L1210 antibody: In vitro and in vivo antitumor activity
US4906469A (en) Appropriate cytotoxic pharmaceutical combination especially for the treatment of cancers
Wawrzynczak et al. Molecular and biological properties of an abrin A chain immunotoxin designed for therapy of human small cell lung cancer
Anderson et al. In vitro and in vivo cytotoxicity of an anti-osteosarcoma immunotoxin containing pokeweed antiviral protein
US5798097A (en) Immunogobulin conjugates
Vallera et al. Toxicity and efficacy of anti-T-cell ricin toxin A chain immunotoxins in a murine model of established graft-versus-host disease induced across the major histocompatibility barrier
Villemez et al. Preparation of an immunotoxin for Acanthamoeba castellanii
Letvin et al. In vivo administration of lymphocyte-specific monoclonal antibodies in nonhuman primates. Delivery of ribosome-inactivating proteins to spleen and lymph node T cells.
Schroff et al. Induction and enhancement by monocytes of antibody-induced modulation of a variety of human lymphoid cell surface antigens
Obrist et al. Enhancement of macrophage invasion of tumors by administration of chemotactic factor-antitumor antibody conjugates
Flavell et al. Effectiveness of combinations of bispecific antibodies for delivering saporin to human acute T-cell lymphoblastic leukaemia cell lines via CD7 and CD38 as cellular target molecules
Flavell et al. Comparison of the potency and therapeutic efficacy of the anti-CD7 immunotoxin HB2-saporin constructed with one or two saporin moieties per immunotoxin molecule
JPS6069034A (en) Immunological toxin, drug of same and use out vivo
Derbyshire et al. Monoclonal antibodies recognising the cluster 2 antigen associated with human small cell lung cancer mediate the toxic effects of ricin A chain in an indirect assay of immunotoxin cytotoxicity.

Legal Events

Date Code Title Description
MKEX Expiry