WO2009017823A2 - A fold-back diabody diphtheria toxin immunotoxin and methods of use - Google Patents
A fold-back diabody diphtheria toxin immunotoxin and methods of use Download PDFInfo
- Publication number
- WO2009017823A2 WO2009017823A2 PCT/US2008/009321 US2008009321W WO2009017823A2 WO 2009017823 A2 WO2009017823 A2 WO 2009017823A2 US 2008009321 W US2008009321 W US 2008009321W WO 2009017823 A2 WO2009017823 A2 WO 2009017823A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- immunotoxin
- diabody
- cells
- toxin
- antibody
- Prior art date
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal 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
- A61K47/51—Medicinal 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
- A61K47/68—Medicinal 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
- A61K47/6801—Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
- A61K47/6803—Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
- A61K47/6811—Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
- A61K47/6817—Toxins
- A61K47/6829—Bacterial toxins, e.g. diphteria toxins or Pseudomonas exotoxin A
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal 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
- A61K47/51—Medicinal 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
- A61K47/68—Medicinal 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
- A61K47/6835—Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
- A61K47/6849—Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal 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
- A61K47/51—Medicinal 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
- A61K47/68—Medicinal 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
- A61K47/6835—Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
- A61K47/6851—Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
- A61K47/6855—Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from breast cancer cell
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal 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
- A61K47/51—Medicinal 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
- A61K47/68—Medicinal 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
- A61K47/6835—Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
- A61K47/6851—Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
- A61K47/6869—Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from a cell of the reproductive system: ovaria, uterus, testes, prostate
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal 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
- A61K47/51—Medicinal 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
- A61K47/68—Medicinal 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
- A61K47/6835—Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
- A61K47/6875—Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody being a hybrid immunoglobulin
- A61K47/6879—Medicinal 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 the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody being a hybrid immunoglobulin the immunoglobulin having two or more different antigen-binding sites, e.g. bispecific or multispecific immunoglobulin
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/02—Antineoplastic agents specific for leukemia
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
- A61P37/06—Immunosuppressants, e.g. drugs for graft rejection
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
Definitions
- the present invention relates to an immunotoxin.
- the invention further relates to a method of treating T cell leukemias or lymphomas, graft-versus-host diseases, cancer, and autoimmune diseases by administering an immunotoxin.
- CD3 and TFR can perform the same routing function as the DT receptor, thus obviating the requirement of a DT receptor interaction for the binding site mutant conjugates anti-CD3-CRM9 and TFR-CRM9 (Intracellular routing of ricin based immunotoxins via the ricin receptor leading to enhanced efficacy has also been reported. Youle et al. (1981) Cell 23:551; Marsh and Neville (1986) Biochem. 25:4461; Youle and Colombatti (1987) J. Biol.Chem. 262:4676).
- the invention is one aspect relates to an immunotoxin comprising a diphtheria toxin moiety and a diabody targeting moiety. It is a further object of the invention to provide a method of treating cancers such as prostate cancer, breast cancer, and T cell leukemias or lymphomas.
- Figure 1 shows that the epitopes involved in human serum's inhibition of its toxicity lie in the last 150 amino acids of DT.
- a schematic diagram of the DT mutants CRM9, CRMl 97 and MSP ⁇ 5 is presented (A). The A- and B-subfragments and their relative size and position are shown. The filled circle represents a point mutation as described in the text.
- Goat (B) or human (C) serum human serum was a pool from all samples with positive ELISA for anti-DT antibodies
- UCHT1-CRM9 was added to a final concentration of 1 X 10 "10 M. This mixture was then diluted 10-fold onto Jurkat cells in a protein synthesis inhibition assay as described in the Materials and Methods. Immunotoxin incubated with medium only inhibited protein synthesis to 4% of controls. The results are representative of two independent assays.
- Figure 2 shows the schematic flow sheet for generation of the single chain antibody scUCHTl gene construct.
- PCR polymerase chain reaction
- L linker
- SP signal peptide.
- Pl to P6, SPl, and SP2 are primers used in PCR.
- Figure 3 shows one clone expressing the divalent immunotoxin fusion protein shown in Figure 5.
- Figure 4 shows another clone expressing a divalent immunotoxin fusion protein shown in Figure 7.
- Figure 5 is a schematic of a divalent fusion immunotoxin.
- Figure 6 is a schematic of a divalent fusion immunotoxin.
- Figure 7 is a schematic of a divalent fusion immunotoxin.
- Figure 8 shows the cloning scheme used to obtain scUCHTl fusion protein with DTMl and DT 483.
- Figure 9 shows the cloning scheme used to obtain scUCHTl fusion protein with DT 390.
- Figure 10 shows the cloning scheme used to obtain scUCHTl fusion protein with DT 370.
- Figure 11 shows the sizing of the diabody fusion-protein constructs shown in Table 5.
- the constructs are sized by plotting their elution times versus log MW on a calibrated Superdex 200 size exclusion column.
- the column has been calibrated with BSA that contains in addition to monomers (68 kDa) dimers, trimers and tetramers (open circles, solid line). Additionally, the column has been calibrated with scFv(M20), 27.5 kDa, and A- dmDT390bisFv(M20), 98.3 kDa, dashed line.
- the diabody constructs fall on the latter line when given the following molecular weight values: Db(C207), (Table 5, B) solid triangle, 53.7 kDa, calculated MW of [Db(C207)]2 ; A-dmDT390-scfbDb(C207), (Table 5, E) solid square, 97.1 kDa calculated monomer MW; A-dmDT390-Db(C207), (Table 5, F) inverted solid triangle, 141.1 kDa, calculated MW of [A-dmDT390-Db(C207)]2.
- Figure 12 shows a SDS 12% acrylamide bis-tris non-reducing gel of the fold-back immunotoxin A-dmDT390-scfbDb(C207) (Table 5, G), after the 3-step purification procedure, is shown in third lane from right alongside molecular weight markers in the first lane with marker kDa values printed at the left. (The trace band in the second lane is spillover from lane 3.)
- Figure 13 shows the % inhibition of tracer FITC-labeled FN 18 binding to HSC-F monkey T cells plotted versus the concentration of binding competitors consisting of scFv(C207), (Table 5, A) biscFv(C207) , the diabody of C207, Db(C207), (Table 5,B) and the single-chain fold-back diabody of C207, scfbDb(C207) (Table 5, C).
- the relative functional binding affinity for any two competitors can be estimated from the ratio of their concentrations at equal % inhibition values. Estimates are considered to be reliable when comparing parallel curves or parallel curve regions.
- the rank order of competitor binding from highest to lowest is scfbDb(C207), Db(C207), biscFv(C207) and scFv(C207).
- FNl 8 competition for FITC-labeled FNl 8 is shown for a comparative standard. In replicate assays the ratio of concentrations at equal displacement for Db(C207)/ scFv(C207) is 7 ⁇ 2 while the ratio for biscFv(C207)/ scFv(C207) is 2.5 ⁇ 0.1 (errors are SD). See Table 5 for structural schematics of these fusion proteins.
- Figure 14 shows another binding competition assay like Figure. 20 except that the competitors of FITC-labeled FNl 8 binding to HSC-F monkey T cells are now immunotoxin fusion proteins that use a diabody, A-dmDT390-Db(C207) (Table 5, F), a fold-back single- chain diabody, A-dmDT390-scfbDb(C207) (Table 5, G) and biscFv antibody moiety, A- dmDT390-biscFv(C207) (Table 5, E).
- the diabody immunotoxin is devoid of detectable binding to the T cells.
- the fold-back single-chain diabody shows 10-fold increase in binding to T cells compared to A-dmDT390-biscFv(C207).
- the data points for A- dmDT390-biscFv(C207) are for comparative purposes and were previously reported (Wang et al., 2007).
- Figure 15 shows the % control inhibition of protein synthesis for three immunotoxins on HSC-F monkey T cells is plotted versus the immunotoxin concentrations on the x-axis after a 24 h exposure. Each point is the mean of 6 replicates and 1 SD error bars are shown.
- Figure 16 shows the loss of resting monkey T cells from the lymphocyte gate by flow cytometry.
- PBMC peripheral blood mononuclear cells
- anti-T cell immunotoxins made in various formats.
- the fold-back diabody immunotoxin, solid squares, is the most potent, exceeding the biscFv immunotoxin, empty circles, by 7 ⁇ 4-fold determined on two replicate assays.
- the diabody immunotoxin is inactive.
- Chemically conjugated FN18-CRM9 is provided as a reference.
- Figure 17 shows the per cent of monkey lymph node CD3 ⁇ + T cells present within the forward scatter/side scatter lymphocyte gate determined by flow cytometry after the administration of the fold-back diabody immunotoxin A-dmDT390-scfbDb(C207) (Table 5,G).
- CX3X dashed line
- Monkey 32972 solid line
- Lymph nodes were surgically removed under anesthesia, macerated and stained prior to flow cytometry.
- the time 0 point represents a mean monkey lymph node T cell percentage from 11 individual monkeys obtained prior to immunotoxin treatment.
- Figure 18 shows the depletion of blood CD3 ⁇ + T cells, solid line, in monkey 32972 and changes in the CD20 + B cell population enumerated by flow cytometry and total blood lymphocyte counts following intravenous administration of the fold-back diabody immunotoxin A-dmDT390-scfbDb(C207) (Table 5, G) [see Figure 23 legend].
- the blood T cell count was 19/mm 3
- Figure 19 shows the fold back format anti-PSMA immunotoxin was 18-30 fold more potent than the bisFv format immunotoxin.
- Figure 20 shows the optimized variable chain amino acid and DNA sequences for the J591 variable heavy chain (Vh) (A), and J591 variable light chain (Vl) (B).
- de J591 is deimmunized J591. Characters highlighted in gray color indicate changes of DNA sequence.
- Vh de J591, Vh aa-deJ591, Vh-optimized, and Vh optimized J591 are SEQ ID NOs:16-19, respectively.
- Vl de J591, Vl aa-deJ591, Vl-optimized, and Vl optimized J591 are SEQ ID NOs:20-23, respectively.
- Ranges can be expressed herein as from “about” one particular value, and/or to "about” another particular value. When such a range is expressed, another embodiment includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent "about,” it will be understood that the particular value forms another embodiment. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. It is also understood that there are a number of values disclosed herein, and that each value is also herein disclosed as “about” that particular value in addition to the value itself. For example, if the value “10” is disclosed, then “about 10" is also disclosed.
- immunotoxins comprising a mutant diphtheria toxin moiety linked to a targeting moiety, wherein the targeting moiety is a single chain variable region diabody.
- the immunotoxins disclosed herein in one aspect comprise diabodies.
- a "diabody” is a single chain antibody fragment comprising two scFv regions wherein one scFv region associates with a second scFv to form a dimer. Where the first scFv and the second scFv share the same antigen specificity, the diabody is referred to as "bivalent" (also called divalent).
- the diabody is referred to as "bispecific.”
- the diabodies disclosed herein can be bivalent or bispecific.
- the disclosed diabodies can act as a targeting moiety for the toxin moiety.
- the disclosed diabodies can be specific for any antigen provided translocation of the toxin moiety is still possible.
- the targeting moiety can be specific for the CD3 molecule on T cells and thus T-cell specific.
- the targeting moiety can be specific for a cancer antigen including but not limited to PSMA or viral antigen such as, for example, GP 120.
- the diabody comprises the V 1 V H region directed against CD3.
- the V L V H region is derived from the anti-CD3 antibody UCHTl or C207.
- the diabody comprises the V L V H region directed against PSMA.
- the V L V H region is derived from the anti-PSMA antibody J591
- the immunotoxin can be a fusion protein produced recombinantly. Derivatives of this immunotoxin are designed and constructed as described herein.
- the toxin moieties of the immunotoxins disclosed herein are mutant diphtheria toxin moieties. It is understood that other toxin moieties such as ricin and pseudomonas exotoxin A (PeA) can be used with the disclosed methods and compositions. It is further understood that the disclosed mutant toxin moieties can comprise substitution mutants. Examples of such substitution mutants include but are not limited to, for example, CRM9. It is also understood that the disclosed mutant toxin moieties can comprise a truncation mutant.
- the deletion mutants can comprise C-terminal deletions of between 140 and 150 C-terminal amino acids.
- toxin moieties wherein the truncation mutant comprises a deletion of 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, or 150 C-terminal amino acids of the full length diphtheria toxin. Therefore, disclosed herein are immunotoxins wherein the diphtheria toxin is a deletion mutant with 140-150 C- terminal amino acids deleted. It is understood herein that the disclosed truncation mutants can be referred to either by designating the number of deleted C-terminal acids or by designating the number of remaining amino acids counting from the N-terminus.
- diphtheria toxin moiety comprising a deletion of 145 amino acids of the complete diphtheria toxin protein (535 amino acids) can be referred to as DT390.
- DT390 a deletion of 145 amino acids of the complete diphtheria toxin protein
- DT389 a deletion of 146 amino acids is DT389.
- Protein toxins of the A-B type have 3 distinct structural and functional domains.
- an enzymatic domain called the A chain that is highly cytotoxic after transfer or translocation to the cytosol compartment.
- a translocation domain located within the B chain that facilitates translocation of the a chain into the cytosol; and (3) a receptor binding domain that binds to a cell surface receptor (also in the B chain).
- the toxin A and B chain is linked by a protease cleavage site spanned by a disulfide bond. This site must be cleaved to achieve translocation into the cytosol compartment and full specific toxicity.
- Toxin receptors have several special properties. One property is that they are efficiently endocytosed into endosomal vesicles. A second property is that the toxin receptor routes the toxin along an intracellular pathway to its site of efficient translocation to the cytosol compartment. For diphtheria toxin this site is in the late endosomal compartment.
- Immunotoxins are toxins that have been altered by replacing the toxin receptor binding domain with an alternate receptor binding domain chosen to target the toxin to a specific cell type in order to eliminate that cell type.
- the new alternate receptor must also route the toxin to its efficient translocation site.
- the translocation efficiency of diphtheria toxin is 10 to 30- fold greater than that of ricin and pseudomonas exotoxin A, conferring considerable advantage to immunotoxins based on diphtheria toxin.
- the disclosed immunotoxins can comprise linkers between the toxin moiety and the diabody targeting moiety. It is understood and herein contemplated that any linker known by those of skill in the art can be used to make the disclosed immunotoxins. The choice of which linker to use can be determined by those of skill in the art based on the desired length and flexibility of the linker between each V H and V L domain as well as between the toxin and targeting moieties. Thus, it is understood that the linker can be a chemical linker. It is also understood that the linker (L) can be a Gly-Ser linker. The Gly-Ser linker can be but is not limited to (Gly4Ser)n or (Gly3Ser)n.
- the linker between the toxin and the diabody can be a chemical linker and the linker between the diabody domains can be a Gly-Ser linker. It is further understood that the for greater flexibility, a longer linker is used; whereas, a shorter linker is used for application where more rigidity is needed.
- the linker between domains of a diabody will be between 3 and 12 amino acids long. More typically, the diabody linker is between 3 and 5 amino acids long.
- specifically contemplated are immunotoxins where the linker between the V H and V L domains of the targeting diabody moiety are G4S. It is also understood that more than one of the disclosed linkers can be used in a single chain immunotoxin.
- immunotoxins wherein the linker between the toxin moiety and a first V L of the targeting diabody is G4S, between the first V L and V H is G4S, between the first V H and second V L is (G4S)3 and between the second V L and V H is G4S.
- immunotoxins where from left to right the immunotoxin comprises:
- the disclosed immunotoxins can be placed in a pharmaceutically acceptable carrier for administration to a subject.
- a chemical linker is used to fuse the toxin and diabody moieties
- both acid-cleavable and non-cleavable protein cross-linking reagents can be used in the construction of antibody-diphtheria toxin (Neville et al. (1989) J. Biol. Chem. 264:14653-14661); preferred are non-cleavable crosslinkers, such as bismaleimidohexane and m-maleimidobenzoyl-N-hydroxysuccinimide ester.
- cross-linking reagents react with proteins via heterobifunctional groups (maleimide and N-hydroxysuccinimide ester) or homobifunctional groups (bis- maleimide).
- the maleimide cross-linking is accomplished by prior protein thiolation with iminothiolane.
- Cross-linked proteins exhibit first-order dissociation under acid conditions. The ti/ 2 at pH 5.5 varies between 0.1 and 130 h for a series of six different cleavable cross- linkers (Srinivasachar and Neville (1989) Biochemistry 28:2501-2509).
- the disclosed diabodies can be formed in a "fold-back" formation. Such formation typically occurs through the use of rigid linkers from 3 to 12, more typically from 3 to 5 amino acids long such as the G4S linker.
- the Toxin-Ll-V L -L2- V H -L3-V L -L4-V H ; wherein Ll, L2, and L4 comprise G4S and L3 comprises (G4S)3 is an example of a fold-back diabody immunotoxin.
- immunotoxin fusion proteins comprising a diphtheria toxin moiety and a targeting moiety, wherein the targeting moiety is a diabody, wherein the sequence from the amino terminus from left to right is toxin moiety, Li, V L i, L 2 , V m , L 3 , V L2 , L 4 , V H2 , wherein the toxin moiety comprises a truncation mutation, Li, L 2 , and L 4 are (G4S) linkers, and V L and V H are the variable light and heavy domains.
- immunotoxin fusion proteins comprising a diphtheria toxin moiety and a targeting moiety, wherein the targeting moiety is a diabody, wherein the sequence from the amino terminus from left to right is toxin moiety, Li, Vn, L 2 , V H l , L 3 , V L2 , L 4 , V H2 , wherein the toxin moiety comprises a truncation mutation, Li, L 2 , and L 4 are (G4S) linkers, and V L and V H are the variable light and heavy domains, wherein V L i and V H i can be the same as V L2 and V H2 .
- fold-back diabodies comprising toxin moieties wherein the truncation mutant comprises a deletion of 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, or 150 C-terminal amino acids of the full length diphtheria toxin.
- diphtheria toxin immunotoxins comprising a truncation mutant and a fold-back diabody specific for an antigen or cell surface determinant such as CD3 or PSMA.
- diphtheria toxin immunotoxins comprising a truncation mutant and a fold-back diabody specific for an antigen or cell surface determinant such as CD3 or PSMA, wherein the V L and V H regions of the diabody are the V L and V H regions of an anti-CD3 or anti- PSMA antibody, respectively.
- diabodies described herein have the V L and V H regions originating from the anti-CD3 antibodies UCHTl and C207, and the anti-PSMA antibody J591, and the resulting immunotoxins are A-dmDT390scfbDb(UCHTl), A- dmDT390scfbDb(C207), and A-dmDT390scfbDb(J591), respectively.
- A-dmDT390scfbDb(C207) wherein C207 is an anti-CD3 diabody affinity matured mutant of FNl 8 (wherein dm indicates a deletion mutant and scfbDb indicates a single chain fold-back diabody).
- A- dmDT390scfbDb(C207) immunotoxins wherein the furin site amino acid sequence cleavage site RVRRSV has been replaced with the MMP cleavage site GPLGMLSQ or the PSA cleavage site HSSKLQ.
- A-dmDT390scfbDb(UCHTl) wherein UCHTl is an anti-CD3 diabody clone.
- A- dmDT390scfbDb(J591) wherein J591 is an anti-PSMA diabody clone.
- immunotoxins disclosed herein include but are not limited to A-dmDT395scfbDb(J591); A-dmDT394scfbDb(J591); A-dmDT393scfbDb(J591); A-dmDT392scfbDb(J591); A- dmDT391scfbDb(J591); A-dmDT389scfbDb(J591); A-dmDT388scfbDb(J591); A- dmDT387scfbDb(J591); A-dmDT386scfbDb(J591); A-dmDT385scfbDb(J591); A- dmDT395scfbDb(anti-PSMA); A-dmDT394scfbDb(anti-PSMA); A-dmDT393scfbDb(anti- PSMA); A-d
- the disclosed immunotoxins can be produced recombinantly as fusion proteins.
- fold-back diabody immunotoxins formed with A- dmDT390scfbDb(J591) or its derivatives in which the amino acids of the furin protease cleavage RVRRSV that begins at residue 191 from the amino terminus is replaced by a matrix metalloprotease cleavage site GPLGMLSQ (Liu, S et al., Expert Opinion Biol Ther 3,843-853, 2003) or a prostatic specific antigen cleavage site HSSKLQ (Denmeade SR et al., Cancer Research 57, 4924-30, 1997) accomplished by PCR mutagenesis.
- GPLGMLSQ matrix metalloprotease cleavage site
- HSSKLQ prostatic specific antigen cleavage site
- the bivalent immunotoxin production in a EF-2 mutant CHO cell expression system was limited to 5 mg/L and could not be increased by selection for multiple gene insertions. Due to this limitation, with three exceptions (12, 20, 25) all recombinant immunotoxin production for therapeutic uses has been limited to E. coli production necessitating denaturation and refolding from inclusion bodies (6). However, refolding of the multi-domain structure of the bivalent immunotoxin from E. coli was inefficient and full bioactivity was not recovered (25). Also, the multi-domain structure of the bivalent immunotoxin hinders efficient production in Escherichia coli. Therefore, the attempt to develop a robust Pichia pastoris production system for the bivalent or bispecific immunotoxin was driven by the inadequacy of the existing productions systems.
- Pichia pastoris is a good expression system for the bivalent and bispecific immunotoxins disclosed herein.
- Pichia was good for the bivalent anti-T cell immunotoxin A-dmDT390-bisFv as it provides optimal protein folding compared to prokaryotic expression systems and provides higher yields compared to mammalian cell expression (CHO cells).
- Antibody fusion proteins require correct disulfide bridges and the endoplasmic reticulum of yeast provides an oxidizing environment like that of eukaryotic antibody producing cells.
- the multi-domain structure of the bivalent or bispecific immunotoxin requires a eukaryotic expression system to properly fold this complex protein. Yet most eukaryotes are sensitive to the effects of protein synthesis inhibition upon expression of the immunotoxin.
- a budding yeast Pichia pastoris has a certain degree of tolerance to DT (Neville et al., 1992; Woo et al., 2002) and yielded the immunotoxin at a level of 40 mg/L in fermentor culture.
- the immunotoxin was produced by fermentation of genetically engineered Pichia pastoris (JW 102, renamed from pJHW#2 (Woo et al., 2002)) via the secretory route.
- JW 102 genetically engineered Pichia pastoris
- the present method provides a yield of 120 mg/1 after a 163 h induction period and the purified yield is 90.8 mg/L.
- Pichia pastoris was sensitive to the toxic effects of cytosolic expressed diphtheria toxin A chain which ADP ribosylates elongation factor 2 (EF-2) leading to cessation of protein synthesis. Toxicity to expression of A-dmDT390-bisFv by the secretory route was indicated by a continuous fall in methanol consumption after induction. A mixed feed of glycerol and methanol was provided to the cells. Expression of the catalytic domain (A chain) of DT in the cytosol is lethal to Pichia pastoris.
- EF-2 ADP ribosylates elongation factor 2
- EF-2 elongation factor 2
- the sensitivity of the eukaryotic EF-2 to ADP-ribosylation by these toxins lies in the structure of protein.
- EF-2 is a single polypeptide chain of about 850 amino acids and is composed of two domains.
- the N-terminal G domain is responsible for binding and hydrolysis of GTP that promotes translation, and the C-terminal R (or diphthamide) domain is thought to interact with the ribosome (Kohno et al., 1986; Perentesis et al., 1992).
- the diphthamide domain contains a histidine residue in a region of 22 residues that are well conserved in the EF-2 of all eukaryotes. This conserved histidine is specifically modified post-translationally to the derivative, diphthamide, which is the unique target for ADP-ribosylation by DT (Van Ness et al., 1880). In S.
- the conserved histidine can be mutated and substitutions with some other 2 amino acids yielded functional EF-2s that were resistant to ADP-ribosylation (Phan et al., 1993; Kimata and Kohno 1994). However, cells with EF-2 mutated at diphthamide grew more slowly than those expressing wild-type EF-2.
- the expression level of the immuntoxins disclosed herein including for example the diphtheria toxin diabody immunotoxins and diphterhia toxin bivalent antibody immunotoxins disclosed herein such as A-dmDT390-scfbDb and A- dmDT390-bisFv could be further increased by rendering Pichia pastoris insensitive to toxin, the EF-2 gene of Pichia pastoris has been mutated so that the GIy at position 701 was changed to Arg, which has been shown to prevent ADP-ribosylation of EF-2 in other organisms.
- the EF-2 mutagenesis required cloning of the gene, introduction of the in vitro mutated sequence with a selection marker, URA3, to the genome and PCR identification of mutated clones.
- the entire EF-2 gene of Pichia pastoris has been cloned and sequenced.
- the coding sequence of Pichia pastoris EF-2 is 2526 nucleotides coding for 842 amino acids.
- the Pichia pastoris EF-2 is the same as the EF-2 of S. cerevisiae and S. pombe in length and shares 88% and 78% of identity in amino acid sequence with these two, respectively. In contrast to these two yeasts, Pichia pastoris has only one copy of the EF-2 gene that contains a short intron.
- a pBLURA- ⁇ 5'mutEF-2 was constructed that targets the Pichia pastoris EF-2 gene and introduces a mutation of GIy 701 to Arg in the gene by homologous recombination.
- the construct contains the 3' end 1028 nucleotides of EF-2 that has been mutagenized in vitro to contain the amino acid substitution and the auxotrophic marker URA3.
- a PCR detection method was also developed for fast and accurate identification of mutant clones after uracil selection.
- the targeted mutation strategy with construct pBLURA- ⁇ 5'mutEF-2 allowed mutation of the EF-2 gene of Pichia pastoris with about 40% of uracil positive clones being found to contain the introduced mutations.
- EF-2 mutants were developed with different auxotrophic markers, (specifically mutEF2JC308 (adel arg4 his4), mutEF2JC303 (arg4 his4) and mutEF2JC307 (his4)) and demonstrated that the GIy 701 to Arg mutation in EF-2 confers resistance to the cytosolic expression of DT A chain.
- A-dmDT390-bisFv was not toxic to the mutant strain.
- the EF-2 mutants allowed expression of A-dmDT390-bisFv under the control of the constitutive GAP promoter (P GAP )- In shake-flask culture, the production of A-dmDT390-bisFv under P GAP was about 30% higher than that under P AOXI - The increase in production under PQ AP may be more significant in fermentation cultures since fermentation allows cells to grow to very high density.
- the expression level of an exemplary bivalent or bispecific immunotoxin was improved by 4-fold in bioreactor culture compared to shake flask culture by optimizing the fermentation conditions in Pichia pastoris as follows: (1) use of Soytone Peptone and yeast extract based complex medium, (2) use of methanol/glycerol mixed feed (4:1) to supplement the energy source during methanol induction, (3) continuous feeding of PMSF and yeast extract during induction, and (4) lowering temperature to 15 0 C during methanol induction. The lowered temperature resulted in a 2-fold increase in secretion relative to using 23 0 C during methanol induction.
- AOXl alcohol oxidase
- Bivalent immunotoxin expression was increased at low induction temperatures, reaching a maximum at 17.5 0 C, and secretion of bioactive immunotoxin reached a maximum at 15 0 C, in spite of the fact that methanol consumption rate at 15 0 C fell to 75% of its 23 0 C value. Because continuous feeding of PMSF and yeast extract during induction effectively inhibited protease activity in supernatants, it appears unlikely that a reduction in protease activity with lower induction temperature accounts for the nearly 2-fold increase in bivalent immunotoxin secretion seen at 15 0 C. The limitation in Pichia pastoris secretion of bivalent immunotoxin previously described may actually represent an overexpression at 23 0 C that is reduced at 15 0 C achieving a better balance of input and output within the secretory compartment.
- the immunotoxin was produced in Pichia pastoris (JW 102) via the secretory route under control of the AOXl promoter in the fermentor using methanol as a carbon source.
- the toxicity towards Pichia pastoris resulted in a decrease in the metabolic rate of methanol consumption, a cell growth rate reduction and very low productivity in a defined medium during methanol induction.
- the method can comprise methods of enhancing codon preference that are known and exemplified.
- any of the disclosed immunotoxins are useful for the treatment of various conditions or diseases. Therefore, disclosed herein in one aspect are methods of treating a cancer in a subject comprising administering to the subject an immunotoxin or a derivative thereof, under conditions such that the cancer is treated. Further disclosed herein are methods of treating a cancer in a subject comprising administering to the subject an immunotoxin that is a truncation mutant, wherein the truncation mutant comprises a deletion of 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, or 150 C-terminal amino acids of the full length diphtheria toxin.
- the diabody is an anti-PSMA diabody such as, for example, a diabody wherein the diabody comprises the V L V H regions of J591.
- PSMA or prostate specific membrane antigen is a transmembrane antigen expressed on prostate cells and highly over-expressed in prostate tumors. It is weakly expressed on some other tissues and is considered highly tissue specific and thus is considered a prime target for immunotoxin mediated therapy of prostatic cancer (Fracasso, G et al, The Prostate 53, 9-23, 2002 and Huang , X. et al, The Prostate 61,1-11, 2005).
- PSMA is over-expressed on the neovasculature that feeds a variety of human primary cancers and their metastatic counterparts such as breast, lung and kidney (Milowsy, MI et al.J. Clin Oncol 25,540-47,2007).
- PSMA is a homodimer with a 2-fold axis of symmetry.
- a divalent anti- PSMA diabody immunotoxin with binding sites oriented 180 degrees apart is ideally suited to complex PSMA.
- PSMA is a functional glutamate carboxylase metalloprotease.
- toxin moiety can be a truncation mutant such as, for example, DT395, DT394, DT393, DT392, DT391, DT390, DT389, DT388, DT387, DT386, and DT385.
- the immunotoxin that is administered to the subject is A- dmDT390scfbDb(J591); A-dmDT389scfbDb(J591); A-dmDT380scfbDb(J591); A- dmDT395scfbDb(J591), or any other DT truncation mutant J591 fold-back diabody combination.
- the disclosed methods of treating a cancer have utility for treating any cancer where PSMA is expressed on cancerous tissue, such as, for example, prostate cancer or in a cancer where PSMA is expressed in the neovasculature, breast cancer, lung cancer, and renal cancer.
- any cancer with a targetable antigen can be treated with the disclosed methods providing that the targeted epitope routes DT to its efficient translocation site.
- the cancer can comprise a T-cell leukemia.
- methods of treating T cell leukemias or lymphomas in an animal comprising administering to the animal one of the disclosed diphtheria toxin diabody immunotoxins, or derivatives thereof, under conditions such that the T cell leukemias or lymphomas are treated.
- the immunotoxin for treating a T cell leukemia can comprise a diabody wherein the diabody is an anti-CD3 diabody. It is further understood that any anti-CD3 diabody can be used in the disclosed methods.
- the diabody can comprise the VLVH regions of UCHTl.
- any of the disclosed anti-CD3 diabodies can be used with any of the disclosed toxin moieties including, for example, DT390, DT389, DT395, DT394, DT393, DT392, DT391, DT388, DT387, DT386, and DT385.
- the immunotoxin can comprise A- dmDT390scfbDb(UCHTl) or A-dmDT390scfbDb(C207).
- Treatment means a method of reducing the effects of a disease or condition.
- Treatment can also refer to a method of reducing the disease or condition itself rather than just the symptoms.
- the treatment can be any reduction from native levels and can be, but is not limited to, the complete ablation of the disease, condition, or the symptoms of the disease or condition. Therefore, in the disclosed methods, treatment” can refer to a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% reduction in the severity of an established disease or the disease progression.
- a disclosed method for reducing the effects of prostate cancer is considered to be a treatment if there is a 10% reduction in one or more symptoms of the disease in a subject with the disease when compared to native levels in the same subject or control subjects.
- the reduction can be a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to native or control levels. It is understood and herein contemplated that "treatment” does not necessarily refer to a cure of the disease or condition, but an improvement in the outlook of a disease or condition.
- compositions and immunotoxins disclosed herein are useful in transplantation related conditions and disease such as graft versus host disease, graft rejection, and the induction of immune tolerance.
- T cells The role of T cells in the aforementioned transplantation related diseases and conditions makes the CD3 molecule (i.e., the T cell receptor), which is only present on T cells, a viable target for the action of the disclosed immunotoxins. Therefore, disclosed herein are methods of treating graft- versus-host disease in an animal, comprising administering to the animal an immunotoxin disclosed herein, or derivatives thereof under conditions such that the graft- versus-host disease is treated.
- the immunotoxin is an anti-CD3 diphtheria toxin immunotoxin.
- any anti-CD3 antibody derived diabody such as a diabody derived from UCHTl, C207, or FNl 8 can be used in the methods disclosed herein.
- any of the disclosed anti-CD3 diabodies can be used with any of the disclosed toxin moieties including, for example, DT390, DT389, DT395, DT394, DT393, DT392, DT391, DT388, DT387, DT386; and DT385.
- the immunotoxin can comprise A-dmDT390scfbDb(UCHTl) or A-dmDT390scfbDb(C207).
- Also disclosed are methods of inducing immune tolerance to a graft in a recipient comprising: a)administering to the recipient an anti-CD3 immunotoxin disclosed herein, thereby reducing the recipient's T-cell population; and b) administering to the recipient an agent that inhibits dendritic cell maturation.
- any anti-CD3 antibody derived diabody such as a diabody derived from UCHTl, C207, or FNl 8 can be used in the methods disclosed herein.
- any of the disclosed anti-CD3 diabodies can be used with any of the disclosed toxin moieties including, for example, DT390, DT389, DT395, DT394, DT393, DT392, DT391, DT388, DT387, DT386; and DT385.
- the immunotoxin can comprise A-dmDT390scfbDb(UCHTl) or A-dmDT390scfbDb(C207).
- Also disclosed herein are methods of inhibiting a rejection response in a primate recipient to a foreign mammalian graft comprising the steps of: a) administering to the recipient an anti-CD3-DT immunotoxin, wherein the immunotoxin comprises scfbDb-DT, so as to reduce the recipient's T-cell lymphocyte population by at least 80%, as compared to the recipient's T-cell lymphocyte population prior to administration of the immunotoxin; and b) transplanting the graft into the recipient, such that a rejection response by the recipient to the graft is inhibited.
- any anti-CD3 antibody derived diabody such as a diabody derived from UCHTl, C207, or FNl 8 can be used in the methods disclosed herein. It is further contemplated that any of the disclosed anti-CD3 diabodies can be used with any of the disclosed toxin moieties including, for example, DT390, DT389, DT395, DT394, DT393, DT392, DT391, DT388, DT387, DT386; and DT385.
- the immunotoxin can comprise A-dmDT390scfbDb(UCHTl) or A-dmDT390scfbDb(C207).
- “Inhibit,” “inhibiting,” and “inhibition” mean to decrease an activity, response, condition, disease, or other biological parameter. This can include but is not limited to the complete ablation of the activity, response, condition, or disease. This may also include, for example, a 10% reduction in the activity, response, condition, or disease as compared to the native or control level. Thus, the reduction can be a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to native or control levels.
- the disclosed methods of treating graft- versus-host disease, inducing immune tolerance to a graft, and inhibiting a rejection response can be complemented by the use of an immunosuppressive agent in addition to the methods already disclosed.
- the immunotoxin for treating graft- versus-host disease can comprise a diabody wherein the diabody is an anti-CD3 diabody. It is further understood that any anti-CD3 diabody can be used in the disclosed methods.
- the diabody can comprise the VLVH regions of UCHTl.
- any of the disclosed anti-CD3 diabodies can be used with any of the disclosed toxin moieties including, for example, DT390, DT389, DT395, DT394, DT393, DT392, DT391, DT388, DT387, DT386, and DT385.
- the immunotoxin can comprise A-dmDT390scfbDb(UCHTl) or A-dmDT390scfbDb(C207). Therefore, disclosed herein are methods of treating graft- versus-host disease, inducing immune tolerance to a graft, and inhibiting a rejection response, further comprising administering an immunosuppressive agent.
- suitable immunosuppressive agents include but are not limited to cyclosporine, rapamycin, and mycophenolate mofetil and the anti-B cell antibody (anti-CD20) rituximab. It is further contemplated that the disclosed immunosuppressive agent can be an agent that inhibits dendritic cell maturation. Such action can occur, for example through the inhibition of nuclear translocation, such as, inhibition of nuclear translocation of NFKB.
- immunosuppressive agents that are inhibitors of dendritic cell maturation that are inhibitors of nuclear translocation of NFK B such as, for example, deoxyspergualin and analogs or derivatives of deoxyspergualin including, for example, methyl-deoxyspergualin or a deoxyspergualin analog lacking a chiral center (e.g., LF 08-0299) (Andoins et al., 1996, which is incorporated herein by reference).
- Other derivatives or analogs of deoxyspergualin can be used that include, for example, those identified in U.S. Pat. No. 4,518,532; U.S. Pat. No. 4,518,532; U.S. Pat. No.
- immunosuppressive agents that inhibit dendritic cell maturation wherein the agent that inhibits dendritic cell maturation activates one or more NF-AT dependent Th2 cytokines. Also disclosed are methods wherein the immunosuppressive agents that inhibit dendritic cell maturation wherein the agent that inhibits dendritic cell maturation inhibits one or more NFKB dependent ThI cytokines. It is further contemplated that the disclosed immunosuppressive agent that is an inhibitor of dendritic cell maturation can function through other means.
- the inhibitor of dendritic cell maturation is a soluble IL- 17 receptor Fc fusion protein; a glucocorticoid; blocker of tumor necrosis factor alpha binding; blocker of granulocyte macrophage colony stimulating factor binding; blocker of IL-12p70 binding; blocker of EL- I ⁇ binding; and/or an anti-CD 154 ligand.
- the graft used in the methods disclosed herein can comprises donor cells, tissue or organ. It is further contemplated that the donor cells, tissue, or organ is selected from the group consisting of kidney, liver, heart, pancreas, lung, skin, and isolated cell transplants of pancreatic islets, hepatocytes, stem cell, and differentiated precursors cells. It is understood and herein contemplated that the donor graft (tissue, cell, or organ) can be from an allogeneic, syngeneic, or xenogeneic donor. It is further contemplated that the donor source can be a cadaver.
- the present invention relates to a pharmaceutical composition
- a pharmaceutical composition comprising a diabody-DT mutant in an amount effective to treat T cell leukemias or lymphomas, which carry the CD3 epitope, graft- versus-host disease or autoimmune diseases, or treat a cancer such as prostate cancer or breast cancer which carries PSMA and a pharmaceutically acceptable diluent, carrier, or excipient.
- T cell leukemias or lymphomas which carry the CD3 epitope, graft- versus-host disease or autoimmune diseases, or treat a cancer such as prostate cancer or breast cancer which carries PSMA and a pharmaceutically acceptable diluent, carrier, or excipient.
- one embodiment of the invention provides a method of treating an autoimmune disease in an animal comprising administering to the animal an imrnunotoxin comprising a mutant diphtheria toxin moiety and an antibody moiety which routes by the anti-CD3 pathway, or derivatives thereof, under conditions such that the autoimmune disease is treated.
- an imrnunotoxin comprising a mutant diphtheria toxin moiety and an antibody moiety which routes by the anti-CD3 pathway, or derivatives thereof, under conditions such that the autoimmune disease is treated.
- the immunotoxin for treating an autoimmune disease can comprise a diabody wherein the diabody is an anti-CD3 diabody. It is further understood that any anti-CD3 diabody can be used in the disclosed methods. Thus, for example, the diabody can comprise the VLVH regions of UCHTl. It is further contemplated that any of the disclosed anti-CD3 diabodies can be used with any of the disclosed toxin moieties including, for example, DT390, DT389, DT395, DT394, DT393, DT392, DT391, DT388, DT387, DT386, and DT385. Thus, for example, the immunotoxin can comprise A- dmDT390scfbDb(UCHTl) or A-dmDT390scfbDb(C207).
- Also disclosed are methods for treating acquired immunodeficiency syndrome in an animal comprising administering to the animal an immunotoxin comprising a diphtheria toxin binding mutant moiety and an antibody moiety which routes by the anti-CD3 pathway, or derivatives thereof under conditions such that the acquired immunodeficiency syndrome is treated.
- a method of treating acquired immunodeficiency syndrome in an animal comprising administering to the animal a non-toxic mutant of diphtheria toxin followed by an antibody-diphtheria toxin immunotoxin which routes by the anti-CD3 pathway or derivatives thereof under conditions such that the acquired immunodeficiency syndrome is treated is provided.
- any anti-CD3 antibody derived diabody such as a diabody derived from UCHTl, C207, or FNl 8 can be used in the methods disclosed herein. It is further contemplated that any of the disclosed anti-CD3 diabodies can be used with any of the disclosed toxin moieties including, for example, DT390, DT389, DT395, DT394, DT393, DT392, DT391, DT388, DT387, and DT386.
- the immunotoxin can comprise A-dmDT390scfbDb(UCHTl) or A- dmDT390scfbDb(C207).
- Conjugation of anti-V/3 and anti-V ⁇ lgG monoclonal antibodies to CRM9 is performed by the same methods used to conjugate anti-CD3 to CRM9 using a non-cleavable linker such as bismaleimidohexane and previously described in detail (Neville et al. (1988) J. Biol. Chem. 264:14653-61).
- the monoclonal antibody FNl 8 is the monkey equivalent of the human anti-CD3 (UCHTl) and is known to bind the same CD3 receptor epitopes (e and ⁇ ) as bound by the human CD3 antibody and is the same isotype as the human CD3 antibody.
- the present CD3-CRM9 conjugate and FN18-CRM9 are expected to have the same activity.
- Conjugates can be administered as an LV. bolus in a carrier consisting of 0.1M Na 2 SO 4 + 0.01M phosphate buffer, pH 7.4 plus 1 part in 50 of serum previously obtained from the subject.
- the dose schedule is every other or third day for 3 to 6 days.
- the total dose is preferably from 25 to 200 micrograms of toxin per kg of body weight.
- the actual dose of FN18-CRM9 used was equal to 0.167 of the minimum lethal dose (MLD) in guinea pigs. Since the estimation of the MLD was performed in an animal lacking an immunotoxin target cell population (guinea pigs), the true MLD of FN18-CRM9 and anti-CD3-CRM9 is expected to be higher in monkeys and humans than in guinea pigs.
- MLD minimum lethal dose
- FN18-CRM9 The specificity of FN18-CRM9 toward T cells can be seen by comparing the total white blood cell (WBC) count in the same two monkeys. WBCs fell, but only to 45% of baseline value on day 2 compared to 6% of baseline values for the CD4+ T cell subset. Most of the fall in WBC values can be accounted for by the T cell component of the WBC population ( «40%). However, B cells are initially depleted after FN18-CRM9 although these cells recover more quickly. FNl 8 is an IgG, isotype and as such is known to bind to Fc 11 receptors present on B cells and macrophages with low affinity. The FN18-CRM9 depletion of B cells indicates that significant interactions between the Fc portion of the FNl 8 antibody and B cells is taking place.
- WBC white blood cell
- peripheral T cell depletion induced by unconjugated FNl 8 at a dose known to produce immunosuppression 0.2mg/kg/day was compared to the immunotoxin FN18-CRM9 administered at l/9th the FN 18 dose.
- Peripheral CD4+ T cell depletion is more pronounced and more long-lasting with the conjugate.
- the demonstration that FN18-CRM9 reduces the peripheral helper T cell subset (CD4+) to levels less than or equal to 200 cell/mm 3 for a period as long as 21 days demonstrates that this immunotoxin and its anti-human analogs are effective immunosuppressive reagents.
- FN18-CRM9 is a potent agent for inducing T cell depletion in non-human primates demonstrates that an anti-human homolog of FN18-CRM9, UCHT1-CRM9 (Oxoid USA, Charlotte, NC) for example, is a potent agent for inducing T cell depletion in humans.
- the Fc binding region of anti-TCR/CD3 monoclonals may or may not be needed to induce T cell depletion when the anti-TCR/CD3 monoclonals are conjugated to CRM9.
- the Fen binding regions can be removed, for example, by forming the conjugates with F(ab') 2 derivatives as is indicated in the literature (Thorpe et al. (1985) J. Nat'l. Cancer Inst. 75:151-159).
- anti-TCR/CD3 IgA switch variants such as monoclonal antibody T3.A may be used (Ponticelli et al. (1990) Transplantation 50:889-892). These avoid rapid vascular clearance characteristic of F(ab')2 immunotoxins.
- F(ab') 2 and IgA switch variants of anti-TCR/CD3-CRM9 immunotoxins are therefore derivative anti-TCR/CD3 immunotoxins. These derivatives will avoid the B cell interaction noted and can increase specificity. However, IgG 23 switch variants will maximize T cell activation through the Fci, receptor and may be useful in certain situations where T cell activation aids immunotoxin induced toxicity.
- antibody can mean the entire antibody or any portion of the antibody sufficient for specific antigen or receptor binding.
- TCR/CD3 Since receptor recycling is a requirement for effective CRM9 based immunotoxins and since TCR/CD3 recycles as a unit, antibodies directed at other epitopes on TCR/CD3 will constitute effective derivatives, in particular antibodies directed at the approximately 50 VjS subset families or the approximately equal number of Va subsets can be used to conjugate CRM9 and ablate specific V/3 or Va subsets in vivo. In addition, in some cases it will be desireable to develop specific monoclonal antibodies reacting with unique rearrangements of either the Va or VjS subset families.
- the target cells deplete the effective dose and this depletion decreases as target burden decreases. Since the log kill is exponential in effective dose, much higher increases in log kill than 50-fold on changing the target from T cells to a VjS subset can occur. The expected increase in log kill will only occur if the immunotoxin is specific for the defined target. Extraneous interactions with other cell types via the antibody Fc piece is preferably eliminated.
- V ⁇ n HIV has been shown to preferentially infect one (V ⁇ n) or a few of the 20 V/3 subset families providing a small T cell reservoir of HIV replication, and because HIV infection apparently involves an unknown superantigen
- CRM9 based immunotoxins directed at these specific Vj3 subsets such as anti-VjSi 2 -CRM9 can reduce the HIV virus load.
- total ablation of a VjS subset in the presence of an endogenous superantigen can lead to long-term ablation of the subset since maturing T cells are negatively selected in the presence of endogenous superantigens. Since the specific VjS subset responding to the superantigen is eliminated, infection cannot take place.
- the two strategies that can be utilized for using anti-V/3i 2 -CRM9 immunotoxins to treat HIV infections are (1) treatment depleting the susceptible VjS subset to an extent where continued infection cannot be maintained and (2) treatment to the extent that all or nearly all of the VjS 12 subset is eradicated.
- Anti-human VjS monoclonal antibodies such as S5-11 are available (T Cell Sciences, Cambridge, MA) and can be conjugated to CRM9 by standard methodologies. Briefly, as in Example 5, conjugation of anti-V/3 and anti-V ⁇ lgG monoclonal antibodies to CRM9 is performed by the same methods used to conjugate anti-CD3 to CRM9 using a non-cleavable linker such as bismaleimidohexase and previously described in detail (Neville et al. (1988) J. of Biol. Chem. 264:14653-61).
- a non-cleavable linker such as bismaleimidohexase
- Conjugates can be administered as an LV. bolus in a carrier consisting of 0.1M Na 2 SO 4 + 0.01M phosphate buffer, pH 7.4 plus 1 part in 50 of serum previously obtained from the patient.
- the dose schedule is every other or third day for 3 to 6 days.
- the total dose is preferably from 25 to 200 micrograms of toxin per kg of body weight, but may be increased if anti-diphtheria toxin antibodies are present in the patient's sera in significant amounts.
- V ⁇ or Va subsets which may be found to be associated with HIV infection can be treated in the same manner described herein by conjugating the CRM9 to the antibody specifically reactive with the appropriate V/3 or Va subset.
- CRM9 is a diphtheria toxin (DT) binding site mutant and forms the basis of the anti- T cell immunotoxin anti-CD3-CRM9.
- This immunotoxin has been constructed against human and rhesus T cells and has shown above to kill 3 logs of human T cells in a nude mouse xenograft system.
- the present example demonstrates a 2 log kill of T cells in rhesus monkey lymph nodes that is also shown to produce prolongation of skin allograft rejection in monkeys.
- ELISA ELISA assays were performed in order to determine the levels of anti-DT titers existing in 9 individuals in a population ages 27 to 55. There were 3 individuals with titers of 1 : 100 (low) and 6 with titers of 1 : 1000 (moderate).
- Rhesus monkeys were screened by the same assay and a 1 : 1000 titered monkey was selected.
- Monkeys were treated by LV. route 5 min prior to the immunotoxin dose with a 100 fold excess of CRM 197 over the CRM9 content of the immunotoxin to be administered.
- a Hl histamine blocking agent such as Benadryl or Tagevil was given LV. to minimize any possibility of an anaphylactic reaction (for Benadryl 4 mg/kg). No histaminic reaction was detected.
- Anti-CD3-CRM9 was given at a total dose between 0.1 and 0.2 mg/kg (toxin weight) in 3 equally divided doses (approximately 0.033 mg/kg) on 3 consecutive days. In these monkeys, the total dose of immunotoxin was 0.1 mg/kg.
- a comparison of the efficacy of anti-CD3-CRM9 in monkeys was conducted by comparing the decrease in the lymph node T/B cell ratio (a measure of lymph node T cell depletion) and the immunosuppressive effect of the immunotoxin as judged by prolongation of mismatched skin graft survival. Effects on the survival of skin grafts is a clear indicator of the general effect a given treatment has on the subject's immune system.
- the monkey with the preexisting anti-DT titer that was pretreated with CRM 197 shows the same level of T/B cell inversion as in the negative titered monkey.
- Skin graft survival was significantly prolonged over the titered monkey treated without CRMl 97.
- the failure to achieve a prolongation of graft survival equal to the negatively titered monkey is likely due to the lower weight of this monkey which causes T cells to repopulate faster, in this case 3-4 days faster, due to the larger thymic T cell precursor pool in younger animals.
- Age related effects such as these can be compensated for by modification of dosage levels and timing of administration.
- Immunotoxin UCHT1-CRM9 for the Treatment of Steroid Resistant Graft- Versus- Host Disease Treatment protocols for this type of disease can be expected to last a year, with Patients being followed for at least 5 years.
- UCHT1-CRM9 is a covalent 1 :1 conjugate of anti-human CD3 IgGl monoclonal antibody and CRM9.
- the conjugate is synthesized, purified, sterile filtered and assayed for concentration, biological efficacy toward target cells and non-target cell toxicity by standardized culture assays.
- the method of synthesis, purification assay are identical to that used for FNI8-CRM9 which was used in the pre-clinical monkey studies described in Examples 5 - 7.
- the patient population consists of individuals suffering from steroid resistant GVHD whose prognosis is poor. Patients are assayed for anti-CRM9 (anti-DT) titers and antibodies to murine immunoglobulin. Patients having anti-CRM9 titers of 1 : 1000 and below are treated according to the present protocol. Patients who have a history of receiving murine immunoglobulins or who exhibit positive anti-Ig titers may require special consideration.
- anti-CRM9 anti-DT
- UCHT1-CRM9 is administered at a dose which is 1/10 or less of the estimated minimum lethal dose (MLD) in a T lymphopenic patient.
- MLD estimated minimum lethal dose
- the MLD is expected to be at least 0.15 mg/kg (CRM9 content) based on the MLD of 0.15 mg/kg of IgGl-CRM9 in guinea pigs which lack a target cell population for the IgGl.
- the optimal dose schedule has been found in monkeys to be administration on 3 consecutive days in 3 equally divided doses, and this schedule can be used throughout the treatment period.
- a second dosage group can include patients selected for antitoxin titers of 1 :330 or less to whom CRM197 will be given at 1.0 mg/kg.
- a third dosage group can include patients with 1:1000 antitoxin titers or less will be given CRM197 at 3.3 mg/kg, a dose expected to be tolerable in humans, because it is easily tolerated by monkeys (see Example 7).
- the monkey MLD data should be very similar to humans on a per weight basis. However, GVHD patients are expected to be more like guinea pigs, because they have a smaller target cell population compared to non-GVHD patients.
- Dose escalation can be tested by increasing the dose by a factor of 1.5.
- Table 1 exemplifies such a dose escalation test. For example three patients are used in each dosage group. There is a 3 to 4 week delay between each patient so that any late toxicity is detected before a dosage group is completed:
- the first dosage group will consume 2.6 mg of the CRM9 immunotoxin, and will be supplied as a pool of two 2 mg batches.
- the second group will consume 3.9 mg and will also be supplied as 2 pooled batches.
- the third group will require 5.9 mg and will be supplied as three pooled batches.
- the fourth group will require 8.9 mg and will be supplied as three pooled batches and an additional two pooled batches.
- a Hl histamine blocking agent such as Benadryl or Tagevil is given LV. to minimize any possibility of an anaphylactic reaction (for Benadryl 4 mg/kg).
- the CRMl 97 is administered LV. in a 5 mg/ml sterile filtered solution in phosphate buffered saline pH 7.4 (PBS) over a 5 min time period.
- PBS phosphate buffered saline pH 7.4
- the immunotoxin is then given LV. at 0.2 mg/ml over 2 min time period in a sterile filtered solution of 0.9OmM sodium sulfate and 1OmM sodium phosphate pH 7.4.
- E Immune function assays ELIS A assays of serum to monitor antibody responses to UCHTl (primary response) and CRM9 (secondary response). ELISA assays to monitor antibody responses to polio and DPT reimmunizations done at 1 year following bone marrow transplantation.
- An anti-CD3 single-chain immunotoxin with a truncated diphtheria toxin avoids inhibition by pre-existing antibodies in human blood
- the present Example examines the effect of human serum with pre-existing anti-DT antibodies on the toxicity of UCHTl -CRM9, an immunotoxin directed against CD3 molecules on T-lymphocytes. Sera with detectable anti-DT antibodies at 1 : 100 or greater dilutions inhibited the immunotoxin toxicity.
- Experiments with radiolabeled-UCHTl-CRM9 indicate that anti-DT antibodies partially block its binding to the cell surface as well as inhibit the translocation from the endosome to the cytosol.
- the inhibitory effect could be adsorbed using a full-length DT mutant or B-subfragment. A C-terminal truncation mutant could not adsorb the inhibitory effect, suggesting that the last 150 amino acids contain the epitope(s) recognized by the inhibitory antibodies.
- an anti-CD3 single-chain immunotoxin sFv-DT390
- the IC 50 of sFv-DT390 was 4.8 X 10 "n M, 1/16 the potency of the divalent UCHTl -CRM9. More importantly, sFv-DT390 toxicity was only slightly affected by the anti-DT antibodies in human sera. "sFv” and “scUCHTl” both are singe chain antibodies containing the variable region.
- DT diphtheria toxin
- FN18-CRM9 A rhesus monkey analog of UCHTl -CRM9, FN18-CRM9 was capable of not only depleting circulating T-cells but also depleting resident T-cells in the lymph nodes 1. This immunotoxin also delayed skin allograft rejection as compared to antibody treatment and non-treatment controls. FN18-CRM9 has also been used as an adjunct in inducing tolerance to mismatched kidney transplants (24).
- a DT point-mutant, a truncation mutant and DT-subfragments were used in an attempt to neutralize the anti-DT effect in human sera. Based on the neutralization data, a single-chain immunotoxin was constructed with a C-terminal deletion mutant of DT which is expected to bypass the inhibitory effect of the pre-existing anti-DT antibodies.
- Jurkat cells (ATCC) were maintained in RPMI 1640 supplemented with 10% fetal calf serum, 25 mM sodium bicarbonate and 50 ⁇ g/ml of gentamycin sulfate.
- Serum and adsorbing molecules Serum and adsorbing molecules.
- variable light (V L ) and variable heavy (V H ) chain regions of the UCHTl antibody were amplified by a two step protocol of RT-PCR using primers based on the published sequence (13).
- the 5' V L primer added a unique Ncol restriction enzyme site while the 3' V H primer added a termination codon at the J to constant region junction and an EcoRI site.
- the V L region was joined to the V H region by single-stranded overlap extension and the two regions are separated by a (Gly 3 Ser) 4 linker that should allow for proper folding of the individual variable domains to form a function antibody binding site (14).
- genomic DNA was isolated from a strain of C. diphtheriae producing the DT mutant CRM9 (07[JS*' 0* - 20 "*- 911' ]) as described (15). This DNA was used for PCR.
- the 5' primer was specific for the toxin gene beginning at the signal sequence and added a unique Ndel restriction site.
- the 3' primer was specific for the DT sequence terminating at amino acid 390 and added an Ncol site in frame with the coding sequence.
- the PCR products were digested with the appropriate restriction enzymes and cloned into the E. coli expression plasmid pET-17b (Novagen, Inc., Madison, WI, USA) which had been linearized with Ndel and EcoRI.
- the resulting plasmid was used to transformed E. coli BL21/DE3 cells.
- Cells were grown to an OD 590 of 0.5, induced with 0.5 M IPTG (Invitrogen, San Diego, CA, USA) and incubated for an additional 3 hours.
- the sFv-DT390 protein was isolated in the soluble fraction after cells were broken with a French Press and the lysate subjected to centrifugation at 35,000 X g.
- Immunotoxins were incubated for 30 minutes with the indicated serum sample or leucine 1 free medium at room temperature prior to addition to cells. In some experiments the serum was pre-incubated for 30 minutes with an adsorbing molecule at the given concentrations to bind the antibodies.
- the immunotoxin/serum mixture was incubated with Jurkat cells (5 x 10 4 cells/well in 96 well plate) for 20 hours. A 1 hour pulse of [ 3 H]-leucine (4.5 ⁇ Ci/ml) was given before cells were collected onto filters with a Skatron harvester. Samples were counted in a Beckman scintillation counter. Each experiment was performed in 4 replicates.
- Results were calculated into a mean value, and recorded as a percentage of control cells.
- Anti-DT antibodies were detected in human serum by ELISA.
- CRM9 (10 ⁇ g/ml) was adsorbed to Costar 96-well EIA/RIA flat bottom plates (Costar, Cambridge, MA, USA) for 2 hours and then washed in phosphate buffered saline (PBS) containing 0.1% Tween 20. Each well was then incubated with PBS containing 3% gelatin to prevent non-specific binding of antibodies to the plastic. Serum samples were diluted in PBS containing 0.1% Tween 20 and 0.3% gelatin prior to addition to the plate.
- PBS phosphate buffered saline
- UCHT1-CRM9 was iodinated using the Bolton-Hunter reagent (NEN Dupont, Wilmington, DE, USA) as described (16).
- Jurkat cells were washed twice with binding medium (RPMI 1640 supplemented with 0.2% bovine serum albumin, 10 mM Hepes (pH 7.4) and without sodium bicarbonate).
- Cells (1.5 x 10 6 ) were incubated for 2 hours on ice with 125 I-UCHTl -CRM9 (1 x 10 "9 M) that had been pre-incubated with serum or binding medium. Unbound antibody was removed by washing the cells twice in PBS (pH 7.4) with centrifugation and resuspension.
- Serum with anti-DT antibodies inhibits UCHT1-CRM9 toxicity.
- aELISA was performed in triplicate for each serum sample as described under "Materials and Methods.” The O.D. values were derived from 1:100 dilutions and presented as a mean value ⁇ SD. The background value was 0.060 ⁇ 0.02. titers are recorded as the highest serum dilution that showed a positive reaction in ELISA.
- bUCHTl-CRM9 (2 x 10 '10 ) was incubated with different dilutions of serum for 30 min. The mixture was then added to cells as described under "Materials and Methods.” Four replicates were performed for each sample. Data are presented as a mean value ⁇ S. C. in percentage of the control counts.
- UCHTl -CRM9 inhibited protein synthesis to 2.0% of controls.
- the goat anti-DT serum could be diluted to 1 : 10,000 and still completely inhibited the toxicity of UCHTl -CRM9. 0ND, not done
- Sera do not inhibit endocytosis of UCHT1-CRM9.
- the inhibitory effect of serum on UCHT1-CRM9 toxicity could be due to prevention of the immunotoxin binding to the cell surface or the endocytosis of UCHTl -CRM9 into the cell.
- Endocytosis assays were conducted using ' 25 I-UCHT 1-CRM9 to determine if either of these processes were affected by anti-DT antibodies present in sera.
- the results indicate that the presence of serum (goat anti-DT or human) reduces as much as 80% of the immunotoxin counts binding to the cell surface (Table 3). While this is a significant reduction in binding, limiting 90% of input immunotoxin (one log less UCHT1-CRM9) in toxicity assays reduces protein synthesis to ⁇ 25% of controls (see Figure 1).
- [ 12 1]-UCHTl -CRM9 (2 x 10-9M) was incubated with medium or anti-DT serum (1 :4 dilution of human sample 10010 or a 1 : 1 ,000 dilution of goat serum; Table 2) for 30 minutes at room temperature. This mixture was added to Jurkat cells (1.5 X 106) for 2 hours on ice (final concentration of [ 125 I]-UCHT1-CRM9 was 1 x 10-10). The cells were then washed and endocytosis assays performed as described in Materials and Methods. The % Bound value represents the cell associated counts divided by the cell associated counts divided by the cell associated counts without serum. Non- labeled UCHT1-CRM9 was incubated with the above dilutions of sera and the resulting mixture was used in protein synthesis inhibition assays. The results shown are representative of two independent assays.
- the inhibitory effect of anti-DT antibodies can be removed by adsorption.
- CRMl 97 the full length DT-like construct, was capable of completely adsorbing the protective antibodies from both goat ( Figure IB) and pooled human serum ( Figure 1C).
- the B-subfragment of DT is also capable of complete adsorption, however —100 fold more is required.
- the A-subfragment of DT had little or no effect on either serum, although the serum samples were demonstrated to contain antibodies reactive to both the A- and the B-subfragments by Western Blot analysis. Of interest were the results seen with MSP ⁇ 5, the truncation mutant. Adsorption of goat serum with MSP ⁇ 5 gave a dose dependent removal of the serum's protecting effect (Figure IB). However, this adsorption could not bring toxicity down to levels obtained when CRMl 97 or the B-subfragment was used.
- sFv-DT390 is not inhibited by anti-DT antibodies present in human sera.
- the predicted molecular weight for the fusion protein is 71,000 Daltons and has been confirmed by Western Blot analysis of both in vitro transcribed and translated protein as well as protein isolated from E. coli using goat anti-DT antibodies.
- the IC 50 (concentration required to inhibit protein synthesis to 50% of controls) of sFv-DT390 was 4.8 X 10 "11 M compared to 2.9 X 10 ⁇ 12 M for UCHT1-CRM9, a 16-fold difference (i.e., 16-fold less toxic).
- UCHT1-CRM9 toxicity was completely inhibited with a 1:10 dilution of the human sera but at a 1 :100 dilution toxicity was equivalent to controls without serum.
- the sFv-DT390 immunotoxin is only partially inhibited with the 1:10 dilution of the human, sera and the 1 : 100 dilution no effect on the toxicity. Both immunotoxins are completely inhibited by goat anti-DT serum (1 : 1,000 dilution).
- the goat anti-DT serum which gave the highest ELISA value could be diluted 10,000 times and still completely inhibited UCHT1-CRM9 toxicity. Since this correlation exists, there is no indication that any other component of the serum inhibits the toxicity of UCHT1-CRM9. Furthermore, the data show that a titer of 1 : 1 OO dilution is necessary for an inhibition of the immuno toxin toxicity.
- Intoxication of cells by immunotoxins can be subdivided into four general stages: 1) specific binding to the cell surface, 2) endocytosis into the cell, 3) translocation of enzymatic domain of the toxin out of the endosome and 4) enzymatic inactivation of the target molecule.
- the results presented indicate that, while the amount of immunotoxin reaching the cell surface is lower in the presence of serum, the same percentage of bound immunotoxin is endocytosed. Taking into account the reduced amount of immunotoxin bound to the cell, the amount of endocytosed immunotoxin should intoxicate the cells to below 25% of controls. However, the immunotoxin had no effect on protein synthesis in the presence of serum containing anti-DT antibodies.
- the anti-DT antibodies probably affect the translocation of the A-subfragment into the cytosol.
- CRMl 97, B-subfragment, and MSP ⁇ 5 could adsorb the protecting anti-DT antibodies from the goat and rhesus monkey sera.
- MSP ⁇ 5 could not prevent the UCHTl -CRM9 toxicity in the presence of the human sera, showing a difference in the anti-DT antibody repertoire among humans, goat and rhesus monkeys. This difference does not seem to be due to immunization routes, because monkeys used in the present study were not immunized for DT and presumably acquire the antibodies after a natural infection with toxigenic strains of C. diphtheriae.
- rhesus monkeys and humans shared a similar antibody repertoire (21), but the present results suggest that the effect of antibodies from the host for whom immunotoxin treatment is intended should be useful.
- sFv-DT390 The IC 50 of sFv-DT390 is 4.8 x 10 "11 M, 1/16 as potent as UCHT1-CRM9. Like many other single-chain constructs, sFv-DT390 is monovalent as compared to immunotoxins generated with full length, bivalent antibodies. The reduced toxicity in sFv-DT390 could be explained primarily on this affinity difference. Immunotoxins generated with purified F(ab)' fragments of antibodies also show an in vitro loss in toxicity (generally a 1.5 log difference) when compared to their counterparts generated with full length antibodies (22). The toxicity of sFv-DT390 is comparable to that reported for DAB486IL-2 (23). From the present data some advantages of sFv-DT390 are expected.
- sFv-DT390 is only 1/3 of the molecular weight of UCHT1-CRM9.
- the molar concentration of sFv-DT390 will be 3 times higher than that of UCHTl -CRM9 if the same amount is given (for example, 0.2 mg/kg). Therefore, their difference in potency could be reduced to approximately 5 times.
- sFv-DT390 contains only the variable region of UCHTl, and is expected to have less immunogenicity in human anti-mouse antibody (HAMA) responses than the native murine antibody UCHTl.
- HAMA human anti-mouse antibody
- sFv-DT390 or others with similar properties, are expected to be useful in the treatment of T-cell mediated diseases in humans, especially in anti-DT positive individuals and in patients who need repeated treatments. To obtain evidence supporting this assumption, it is only necessary to construct a rhesus monkey analog of sFv-DT390, and test it in monkey models as described in previous examples. Table 4: Anti-DT antibodies present in human sera have reduced effect on sFv-DT390 toxicity.
- UCHT1CRM9 or sFv-DT390 (2 x 10 '9 M) was incubated with the indicated dilutions of serum for 30 mm. The mixture was then added to cells as desc ⁇ bed under "Materials and Methods.” The final concentration of lmmunotoxin on cells was 1 x 10 "10 M. Four replicates were performed for each sample. Data are presented as a mean value ⁇ S.D. in percentage of the control counts. UCHT1-CRM9 inhibited protein synthesis to 5% of controls while the sFv-DT390 inhibited protein synthesis to 18% of controls. The ELISA value was determined using a 1:100 dilution of serum. The results are representative of two independent expe ⁇ ments.
- Murine anti-CD3 monoclonal antibodies are used in clinical practice for immunosuppression.
- mAbs are used in clinical practice for immunosuppression.
- a chimeric anti-human CD3 single chain antibody scUCHTl was generated. It is an IgM variant of the UCHTl described in Example 9.
- scUCHTl consists of the light and heavy variable chain binding domains of UCHTl and a human IgM Fc region (CH 2 to CH 4 ). The method used was reported by Shu et al. [37] and is further described below. The following data show that the engineered chimeric anti-CD3 single chain antibody (scUCHTl) will be useful in clinical immunosuppressive treatment.
- Oligonucleotide primers and DNA amplification Oligonucleotide primers and DNA amplification.
- scUCHTl The procedures of cloning scUCHTl is schematically depicted in Fig. 2.
- mRNA isolated from UCHTl hybridoma cells (provided by Dr. P. C. Beverley, Imperial Cancer Research Fund,London was reverse transcribed into cDNA.
- the V L and V H regions of UCHTl were amplified with polymerase chain reaction (PCR) from the cDNA using primer pairs Pl, P2 and P3, P4 respectively.
- Primers P2 and P3 have a 25 bp complementary overlap and each encoded a part of a linker peptide (Gly 4 Ser) 3 .
- V L - linker- V H The single chain variable fragment (V L - linker- V H ) was created by recombinant amplification of V L and V H using primers Pl and P4.
- a mouse kappa chain signal sequence was added at the V L 5'-end by PCR, first with primers SP2 and P4, and then with primers SPl and P4.
- the human IgM Fc region (CH 2 to CH 4 ) was amplified from the plasmid pBlue-hulgM (kindly provided by Dr. S. V. S. Kashmiri, National Cancer Institute, Bethesda. This gene fragment was about 1.8 kb.
- the V L -linker-v H -CH2 region which is important for antigen recognition was confirmed by sequence analysis.
- the single chain variable fragment and the human IgM Fc region were cloned into plasmid pBK/CMV (Stratagene, La Jolla, CA, USA).
- pBK/scUCHTl plasmid as template, an in vitro transcription-translation assay yielded a product of 75 kDa, the expected size. Expression in COS-7 and SP2/0 cells.
- the gene fragment encoding scUCHTl was then cloned into an expression vector pLNCX [36].
- the scUCHTl gene construct was introduced into COS-7 cells with a calcium-phosphate method [32], and introduced into SP2/0 myeloma cells by electroporation [33].
- Cells transfected were selected with 500 ⁇ g/ml G418 (GIBCO/BRL, Gaithersburg, MD, USA) in DMEM medium.
- the drug resistant transfectants were screened for scUCHTl secretion by an anti-human IgM ELISA technique. Transfectants secreting scUCHTl were cloned by limiting dilution.
- COS-4C10 and SP2/0-7C8 Two stable clones, COS-4C10 and SP2/0-7C8, which could produce about 0.5 mg/ml scUCHTl in culture medium, were selected for further evaluation.
- the culture supernatant of COS-4C10 and SP2/0-7C8 cells was analyzed by immunoblotting using anti-human IgM antibody. Human IgM antibody was included as a control in the analysis.
- scUCHTl produced by COS-7 and SP2/0 cells had a similar electrophoretic mobility to that of the control human IgM heavy chain (75 kDa).
- scUCHTl from COS-7 cells appeared as a single band of approximately 150 kDa, which was thought to be a homodimer of the single chain antibody.
- SP2/0 cells mainly produced a protein of similar size with some higher molecular weight products.
- scUCHTl was efficiently produced by both COS-7 and SP2/0 cells. Hollow fiber culture should increase its production.
- scUCHTl the IgM-like molecule, has a secretory tailpiece with a penultimate cysteine (Cys 575) which is involved in polymerization and also provides retention and degradation of IgM monomers [41-43]. Replacing the Cys 575 with serine might also greatly improve the yield.
- scUCHTl secreted from COS-7 cells was shown to be a divalent form by immunoblotting, suggesting a disulfide bond linkage of two monovalent molecules.
- the disulfide bond is likely situated between the CH2 and CH3 regions, where the Cys 337-Cys 337 disulfide bond is thought to exist.
- Cys 337 is believed to be sufficient for assembly of IgM monomers, and was neither sufficient nor necessary for formation of polymers.
- Cys 575 was necessary for assembly of IgM polymers, and Cys 414 was not required for formation of IgM monomers or polymers [44]. This divalent form of the single chain antibody should increase its binding affinity.
- scUCHTl Western blotting analysis of scUCHTl.
- scUCHTl was precipitated from the culture supernatant using goat anti-human IgM-Agarose (Sigma, St. Louis, MO, USA), and separated on 4-20% SDS-PAGE gradient gel under reducing and non-reducing conditions. The separated proteins were transferred to ProBlottTM membrane (Applied Biosystems, Foster City, CA, USA) by electroblotting at 50 volts for 1 hour. The membrane was blocked and incubated with alkaline phosphatase labeled goat anti-human IgM antibody (PIERCE, Rockford, IL, USA) following the manufacturer's instruction. Color development was carried out with substrate NBT/BCIP (PIERCE).
- Culture supernatant was mixed with anti-human IgM-Agarose, and incubated at 4°C with shaking overnight, and then the mixture was transferred to a column.
- the column was washed with washing buffer (0.01 M Na-phosphate, pH 7.2, 0.5 M NaCl) until the OD280 of flow-through was ⁇ 0.01.
- scUCHTl was eluted with elution buffer (0.1 M glycine, pH 2.4, and 0.15 M NaCl). The fractions were neutralized with 1 M Na-phosphate (pH 8.0) immediately, and then concentrated and dialyzed against PBS.
- the parental antibody UCHTl was iodinated using Bolton-Hunter Reagent (NEN, Wilmington, DE, USA) as described previously [34].
- the 125 I-labeled UCHTl was used as tracer and diluted with DMEM medium to 0.3-0.6 nM.
- UCHTl and the purified scUCHTl from COS-7 and SP2/0 transfectant cells were used as competitors.
- Human CD3 expressing Jurkat cells were suspended in DMEM medium (2 x 10 7 /ml). 50 ⁇ l of such cell suspension (1 x 10 6 ) was incubated with 50 ⁇ l diluted tracer and 50 ml diluted competitors on ice for 2 hours.
- results were expressed as a percentage of the 125 I-UCHTl bound to cells in the absence of competitors.
- scUCHTl from both COS-7 and SP2/0 cells could specifically inhibit the binding of 125 I-UCHTl to Jurkat cells in a dose dependent way.
- concentration of the competitors (UCHTl, scUCHTl from COS-7 and SP2/0 cells) increased from 1 to 100 nM
- the tracer 125 I iodinated UCHTl bound to Jurkat cells decreased from 80% to nearly 0%. No significant difference was observed among the affinity curves of UCHTl and scUCHTl from COS-7 and SP2/0 cells.
- scUCHTl has nearly the same affinity as UCHTl .
- scUCHTl contains human IgM constant region, and is expected be less immunogenic than UCHTl.
- the degree of its immunogenicity might vary due to the murine variable region of scUCHTl .
- Humanized variable regions by CDR-grafting or human variable regions can be used to further reduce its immunogenicity [31].
- PBMCs Human peripheral blood mononuclear cells
- FCS Ficoll-Hypaque gradient
- the parental antibody UCHTl started to induce proliferation at 0.1 ng/ml, and peaked at 100 ng/ml. A small drop in CPM was observed as the concentration increased to 1,000 ng/ml.
- [ 3 H]thymidine incorporation in PBMCs incubated with scUCHTl was only slightly increased in the range of 0.1 - 10 ng/ml, and when the concentration was higher than 10 ng/ml, the incorporated counts decreased and were close to 0 counts at 1,000 ng/ml. Thus showing that scUCHTl did not induce a human T cell proliferation response.
- TNF- ⁇ and IFN- ⁇ productions of human PBMCs induced by UCHTl and scUCHTl were measured with ELISA.
- 4 x 10 5 PBMCs were cultured with serial dilutions of anti-CD3 antibodies (UCHTl, scUCHTl) in 96-well flat-bottom plates in RPMI 1640 supplemented with 10% FCS. Supernatant was collected at 36 hours for TNF- ⁇ and 72 hours for IFN- ⁇ after the start of the culture [35].
- TNF- ⁇ and IFN- ⁇ were measured with ELISA kits (Endogen Inc. Cambridge, MA, USA) following the manufacturer's instruction.
- the native antibody UCHTl induced production of both TNF- ⁇ and IFN- ⁇ in a dose dependent way. Higher concentration of UCHTl induced higher production of TNF- ⁇ and IFN- ⁇ . On the contrary, scUCHTl did not induce secretion of TNF- ⁇ at any concentration, and inhibited IFN- ⁇ production when its concentration was higher than 0.1 ng/ml.
- the PBMCs cultured with UCHTl and scUCHTl were also checked with trypan blue exclusion test. Cells were shown to be alive in both situations. In TNF- ⁇ and IFN- ⁇ ELISA assays, an unrelated human IgM was included and it did not affect the TNF-a and IFN-g production.
- Anti-CD3 mAbs can induce T cell activation and proliferation both in in vitro and in vivo situations [45]. Crossing-linking of anti-CD3 antibody between T cells and FcR expressing cells is an essential step in this process [46]. T cell activation therefore reflects an efficient interaction of the mAb with a human FcR. Previous data of in vitro study indicated that T cell activation resulted in increased production of TNF- ⁇ , IFN- ⁇ , and IL-2 [24].
- Human IgG Fc receptors Fc ⁇ R I, Fc ⁇ R ⁇ , Fc ⁇ R HT are distributed on human monocytes, T, B lymphocytes, and NK cells [47].
- Fc ⁇ R I and Fc ⁇ R II can recognize both mouse and human IgG.
- UCHTl was potent in induction of T cell proliferation and TNF- ⁇ and IFN- ⁇ release.
- Human IgM Fc receptor (Fc ⁇ R) was reported to be present mainly on a small fraction of B lymphocytes, NK cells, and possibly a helper subset of T lymphocytes [47, 48]. Pentamer form of IgM and an intact CH 3 domain are required for optimal binding to Fc ⁇ R. Monomelic or dimeric subunits of IgM are less efficient in binding to Fc ⁇ R [49, 50].
- scUCHTl can cross-link human B and T cells, and possibly T and T cells.
- scUCHTl from both COS-7 and SP2/0 cells had little effect in the T cell proliferation assay at low concentrations (below 10 ng/ml), and became inhibitory as the concentration increased. In accordance with these results, scUCHTl did not induce TNF- ⁇ production and even inhibited the basal yield of IFN- ⁇ .
- the present chimeric anti-CD3 single chain antibody scUCHTl possesses high human CD3 binding specificity and affinity, and does not induce T cell proliferation and cytokine release. Moreover, it has a human IgM Fc fragment, which should decrease the possibility of inducing human anti-mouse antibody response. Thus, scUCHTl can be used for clinical immunosuppressive treatment.
- Corynebacteriophage beta (C.diphtheriae) tox 228 gene sequence was from genebank. (Science 221, 885-858, 1983). The sequence is 2220 bp. There are 300 bp of 5' untranslated region (1 to 300) including the promoter sequence around (-180 to -10), 1682 of coding region (301- 1983) including signal peptide (301 to 376), A chain (377 to 955) and B chain (956 to 1983), and 3' untranslated region (1984 to 2220).
- the full-length DT was amplified in two fragments.
- the pelB leader sequence ( ATG AAA TAC CTA TTG CCT ACG GCA GCC GCT GGA TTG TTA TTA CTGCGCT GCC CAA CCA GCG ATG GCC 3') SEQ ID NO:1) was added to the 5' end of the DT coding sequenc to all the constructs during polymerase chain reaction by primer EcosignalDT-1 and EcosignalDT-2.
- a mutant residue was introduced at position 52.
- the glycine (GGG) at position 52 wild type DT was substituted by glutamic acid (GAG).
- the two primers p546R and p514S carried the mutant codon (GGG to GAG).
- the PCR products of these two primers contained the substituted codon (GAG) instead of codon GGG.
- the jointed double stranded DNA of the two fragments (1683bp) were cloned into pET 17b by restriction site Ndel and BamHI.
- IgM immunoglobulin M
- B cells or macrophages contain IgM Fc receptors.
- IgM Fc receptors Binding of imrnuno toxin to cells other than T cells reduces the specificity of the anti-T cell immunotoxin and this situation is purposefully avoided.
- using a bacterial expression system no carbohydrate is attached to the antibody which also eliminates Fc receptor binding.
- substituting other human IgG constant domains would be a routine modification and should be claimed.
- divalent fusion protein immunotoxins are provided. These have been expressed in E. coli, and Western blots of reduced and non-reduced SDS gels confirm that most of the immunotoxin is secreted as the dimeric (divalent) species.
- the position of the toxin has been varied in an attempt to minimize stearic hindrance of the divalent antibody site, yet provide the best interactions with the CD3 receptor to facilitate toxin translocation across the membrane.
- PCR amplification was conducted using BamHI and Ndel restriction sites and the pET17b vector.
- Figs. 3 and 4 show two different clones expressing divalent immunotoxin fusion proteins cartooned in Figs. 5 and 6, respectively. Another variation is shown in Fig. 7.
- the clone producing this consists of a clone constructed by using the single chain antibody followed by a stop codon and the single chain immunotoxin, all under one promoter (Better et al. Proc. Natl. Acad. Sci. 90:457-461, January 1993).
- 3 species are present: sc divalent antibody, divalent fusion immunotoxin, and a divalent sc antibody containing only one toxin.
- This species is isolated by size separation and is the species cartooned in Fig. 7.
- the advantage of this species is that stearic hindrance to the divalent antibody domains is limited by the presence of only one toxin domain.
- Other variations are routine to construct given the methods described herin and in the art. Those diagramed are considered to be the most likely to exhibit divalent character. Numerous orientations of toxin relative to antibody domains can be made and many are expected to be effective.
- the other mutation S to F at 508 has been added because of previous restrictions on cloning mutant DT that can revert to wild type toxin with a minimum lethal dose of 0.1 microgram/kg by means of a single base pair reversion.
- Other mutations can be routinely made in the C terminus to perform this function (Shen et al. J. Biol. Chem. 269(46):29077- 29084, 1994). They are: F530A; K526A; N524A; V523A; K516A Y514A.
- a clone having a single point mutation in DT reducing toxicity by 10-100 fold can be made providing that the clone contains an antibody fragment fusion protein, because chemical conjugation of antibody to DT has been shown to reduce systemic wild type toxin toxicity by 100 fold (Neville et al. J. Biol. Chem. 264(25): 14653-14661, 1989). Therefore, the present invention provides a full length mutant DT sequence with the 525 S to F mutation alone as well as those listed above. These same mutations are also contemplated for the B chain mutant site in DTM2 and can be made similarly. Previous data with chemical conjugation has shown that the longer the C-terminus the better the translocation function (Colombatti et al. J. Biol. Chem.
- the present invention provides an improvement on CRM 197 (a non- toxic toxin mutant described in U.S. Serial No. 08/034,509, filed September 19, 1994) referred to herein as DTM2.
- DTM2 has the same mutation as CRM 197 plus two mutations in the C- terminus which block binding (see sheet). This is expected to reduce the likelihood of immune complex disease which could result when CRMl 97 becomes bound to cells and then is further bound by circulating antitoxin. Kidneys are particularly susceptible. DTM2 can not bind to cells thereby lessening the possibility of tissue damage.
- DTM2 is made for high level production by including the pelB secretory signal for production in E.
- DTM2 a iron independent mutated promoter DT sequence cloned from CRM9 DNA for production in C. diptheriae.
- the essential feature of DTM2 is the S to F mutation at 525 and the G to E mutation at 52, and a construct containing these two mutations is provided.
- a fold-back single-chain diabody format enhances the bioactivity of an anti-CD3 recombinant diphtheria toxin based immunotoxin.
- Anti-CD3 immunotoxins induce profound but transient T cell depletion in vivo by inhibiting eukaryotic protein synthesis, and they have utility in non-human primate models of transplantation tolerance and autoimmune disease therapy (Neville et al, 1992; Hu et al, 1997; Fechner et al, 1997, Thomas et al, 1997 and Contreras et al, 2003).
- the conjugated immunotoxins were formed by chemically cross-linking the antibody or antibody fragment to a diphtheria toxin binding site mutant, CRM9. In this way the binding site of the immunotoxin was dictated by the antibody moiety (Neville et al, 1996).
- the beneficial effect of the second scFv was ascribed to the divalent character of the biscFv binding, which in the case of the anti-human CD3 antibody UCHTl was increased 5 -fold for the biscFv binding over the scFv binding toward human T cells.
- Anti-monkey CD3 immunotoxins are important tools in organ transplantation research where the primary goal is to achieve tolerance to allografted organ transplants. Rodents are easily tolerized and are not a good model for human transplantation tolerance protocols compared to monkeys (Fechner et al., 1997, Thomas et al., 1997 and Contreras et ah, 2003).
- the FN18 scFv appeared more sensitive than anti-human UCHTl to positional loss of affinity when placed distal to the DT390 toxin domains compared to the UCHTl scFv.
- the divalent character of the C207 biscFv was judged to be suboptimal as determined by the relatively low increase in binding to monkey T cells of the biscFv relative to the scFv which was only 1.7 ⁇ 0.4 fold, compared to 5-fold for the UCHTl biscFv/scFv binding ratio.
- Diabodies are recombinant non-covalent dimeric molecules with cross-over pairing of the VL and VH domains achieved by linking the VL and VH domains with a peptide linker that is too short to allow the interchain assembly of a functional Fv fragment (Holliger et al., 1993).
- the diabody will be bivalent. If the linked VL and VH domains are derived from two different Fv structures the diabody will be bispecific (Le Gall et al., 2004).
- Single-chain monomelic diabodies have also been generated by linking two diabodies through a longer linker that allows the diabody subunits to fold back head to tail (Kontermann and Muller, 1999).
- Plasmid pPICZ ⁇ was purchased from Invitrogen.
- the hybridoma secreting FN 18 was kindly provided by Dr. Margreet Jonker, Biomedical Primate Research Center Rijswijk, and was produced and purified by the National Cell Culture Center, Minneapolis, MN.
- Chemically conjugated anti-monkey CD3 immunotoxin, FN18-CRM9 was prepared as previously described (Neville et al., 1989).
- the Herpes Saimiri virus transformed cynomolgous cell T cell line HSC-F (Akari et al., 1996) was supplied by the Centralized Facility for ADDS Reagents supported by EU Programme EVA and the UK Medical Research Council. This cell line exhibits the moderate binding FNl 8 phenotype (Liu et al., 2007).
- FN18-FITC conjugate was purchased from Biosource and added to 250,000 HSC-F cells at 5 x 10 "9 M in the presence of varying concentrations of scFv and immunotoxin binding competitors at 4 0 C for 30 min.
- the displacement of FN18-FITC was measured by FACS on a Beckman-Coulter Cytomics FC 500 instrument after washing the cells two times at 400 x g at 4 0 C and resuspending in 0.5 mL of 1 X PBS containing 2.5 ⁇ g of propidium iodide to exclude dead cells from further analysis.
- MCI Mean channel fluorescent values, MCI, were corrected (MCF') by subtracting an appropriate FITC-isotope control from both the FN18-FITC tracer, and FN18-FITC tracer plus competitor MCF values.
- the % inhibition was determined as [(MCF'FNIS-F ⁇ C - MCF ⁇ 18- F 1 TC + COMPETITOR) / MCF'FNIS-F ⁇ C] X 100.
- Relative binding affinity between any two competitors was determined by dividing their respective concentrations at equal % inhibition obtained from plots of % inhibition versus concentration as previously described (Thompson et al., 2001).
- T cells from normal monkeys were isolated from blood by two centrifugations through Lymphocyte Separation Medium from BioWhittaker following the manufacturer's directions. About 400,000 cells were washed and incubated in RPMI 1640 medium containing 25 mM HEPES (Gibco) with 10% fetal calf serum + 2 g/L NaHCO3, adjusted to pH 7.4, + 0.1 mM nonessential amino acids + 1.0 mM sodium pyruvate + 50 ⁇ g/L of gentamicin sulfate with and without varying concentrations of immunotoxins for 72 h at 5% CO 2 , 37 0 C.
- HEPES Gibco
- nonessential amino acids + 1.0 mM sodium pyruvate + 50 ⁇ g/L of gentamicin sulfate with and without varying concentrations of immunotoxins for 72 h at 5% CO 2 , 37 0 C.
- Cells were washed and then stained for 30 min at 4 0 C with anti-CD3 (SP34-2- PE BD Pharmingen), anti-CD4-FITC (BD Pharmingen), anti-CD20-PE Cy7 (BD Biosciences), and the vital dye 7- ADD (BD Pharmingen) and analyzed on a Beckman- Coulter Cytomics FC 500 instrument counting 10 4 events. Cells were analyzed for uptake of the vital dye, the per cent of cells within the lymphocyte forward scatter/ side scatter gate, and the per cent of cells within the lymphocyte gate within the quadrant displaying the CD3 + and CD4 + epitopes by 2-color FACS.
- the % of cells within the lymphocyte gate at each immunotoxin concentration was divided by % of cells within the lymphocyte gate for the 72 h no-treatment control (Gl).
- the % of cells within the CD3 + CD4 + quadrant at each immunotoxin concentration was divided by % of cells within the CD3 + CD4 + quadrant for the 72 h no-treatment control (Ql).
- the % loss of CD3 + CD4 + cells was calculated as Gl x Ql x 100.
- C207 is an affinity matured anti-rhesus CD3 mutant antibody selected from a library of FN 18 sFvs randomly mutagenized with nucleotide analogs, displayed on yeast and selected by sorting flow cytometry using dye-labeled monkey CD3 ⁇ ectodomain heterodimer (Wang et al., 2007). Construction of biscFv(C207) and its production in P. pastoris has been described as has the construction of the diphtheria toxin based immunotoxin utilizing biscFv(C207) (Wang et al., 2007) shown in Table 5 schematic E.
- diabody (C207) the Db(C207) gene was PCR-amplified from the plasmid pGK1903 using the primers GP-102 and WP-052. These vectors were sequenced using two primers located in the pPICZ ⁇ , 5'-AOXl primer and 3'-AOXl primer (Invitrogen). Expression and purification of diabody (C207) and fold- back diabody (C207) was performed using a protein L affinity resin as described previously (Wang et al., 2007).
- the Db(C207) gene was PCR amplified from plasmid pGK1903 using two primer sets; the first amplification used primers GP-94 & WP-053 and second used primers GP-90 & WP-052.
- the first PCR product was digested with Ncol and BamHl and the second PCR product was digested with BamHl and EcoRI. Then the two scFv parts in the vector pGK2006 (A-dmDT390-biscFv(C207) in pwPICZ ⁇ ) were sequentially replaced with C207- Db counterparts using Ncol and BamHl for the first scFv and BamHl and EcoRI for the second scFv (see Table 5, G). The resulting plasmid (pGK2019) was transformed into Pichia pastoris JW 107 and the transformants were selected on YPD plates containing zeocin (100 ⁇ g/ml).
- A-dmDT390-scfbDb(C207) (Table 5, G) was administered as an intravenous bolus in two dosing schedules;(A) doses given on day 0 and 2 doses at a total dose of 0.15 mg/kg (monkey CX3X) and (B) 8 divided doses given over 4 days, 6 hours apart for a total dose of 0.2 mg/kg (monkey 32972). Lymphocyte phenotype levels were monitored in blood and also in lymph nodes following maceration by flow cytometry on a Beckman-Coulter Cytomics FC 500 instrument counting 10 4 events.
- Staining reagents were: CD3 + (CD3 sp34 FITC, Pharmingen, Catalog # 556611, clone SP34), CD4 + (CD4 PerCP-Cy5.5, Pharmingen, Catalog # 552838, clone L200), CD8b + (CD8beta ECD, Coulter, Catalog # 6607123, clone 2st8.5h7), CD20 + (CD20 PeCy7, BD/Pharmingen, Catalog # 335793).
- IgG isotype controls (isotype for sp34: IgG3 FITC, Southern Biotech, Catalog # 0105-02, clone BlO and isotype for fhl8: IgGl FITC, Coulter, Catalog # IM0639U, clone 679.1Mc7) were subtracted from mean fluorescent values. Blood T and B cell values were calculated by multiplying the total lymphocyte count per mm 3 by the % of T cells or B cells enumerated by flow cytometry. When the total lymphocyte count was not performed on a cytometry day, the count obtained on the previous day was used.
- the size of the diabody (Table 5, B) and the diabody immunotoxin (Table 5, F) corresponds to a dimeric protein mediated through molecular associations between the VH and VL domains on neighboring molecules since intermolecular self-associations were prevented by the short G4S linker separating these domains. This is shown to be the case in Fig. 11 where the Superdex 200 size exclusion column elution times of these constructs are plotted versus log MW. The column was calibrated using BSA monomers and oligomers (solid line) and with scFv(M20) and A-dmDT390bisFv(M20), dashed line.
- the relative binding of these constructs to the monkey T cell line HSC-F is estimated from the inhibition of the anti-monkey anti-T cell antibody FNl 8 labeled with FITC by the constructs in a competition assay.
- the ratio of the construct concentrations at equal levels of inhibition reflects the relative avidity of the constructs and is shown in Fig. 13.
- the rank order of competitor binding from highest to lowest is scfbDb(C207) (Table 5, C), Db(C207) (Table 5, B), biscFv(C207) (Table 5, D) and scFv(C207) (Table 5, A).
- FNl 8 competition for FITC-labeled FNl 8 is shown for a comparative standard.
- Anti-T cell activity of immunotoxin made in the fold-back diabody format.
- the anti-T cell activity of immunotoxin made in the fold-back diabody format was judged by two in vitro assays and by its effects in vivo.
- Fig. 15 the % control inhibition of protein synthesis for three immunotoxins on the transformed HSC-F monkey T line is shown.
- Fig. 17 the loss of lymph node T cells over time enumerated by flow cytometry following bolus intravenous administration of the fold-back diabody immunotoxin is shown for two monkeys.
- Monkey 32972 received the immunotoxin twice daily, 6 h apart, over 4 days, for a total dose of 0.2 mg/kg.
- Monkey CX3X received two intravenous bolus doses of immunotoxin, one on day 0 and one on day 2 for a total dose of 0.15 mg/kg.
- a third monkey AH44 received another biscFv immunotoxin, A- dmDT390biscFv(M20) in the same dose and schedule as monkey CX3X.
- This immunotoxin was described previously (Wang et al., 2007) and is 13.5-fold less active against resting T cells compared to A-dmDT390-biscFv(C207). Lymph nodes were not noticeably deleted of T cells 4 days after the initial immunotoxin dose. The profound loss of T cells in the blood for monkey 32972 is shown in Fig. 18. B cells are only moderately affected.
- T cells were present in the blood one month later but the per cent of SP34 + T cells in the lymphocyte gate was reduced from the pre-treatment value of 66% to 18.7% while the % of CD20 + B cells rose from 19.2 to 35.3 as did the percentage of CD2 + SP34 ' cells (probable NK cells).
- RNAase based immunotoxin targeted at B cells using the CD22 epitope.
- the results showed that the dimeric diabody format was inactive using the larger DT390 based toxin moiety, probably due to steric hindrance. Therefore a fold-back diabody format was tried. In this format two identical diabodies are linked through a longer linker that allows the diabody subunits to fold back head to tail.
- the fraction of multivalent binding never approaches the high values seen in some low molecular weight systems such as tris-vancomycin binding a trivalent D- AIa-D- Ala ligand (Rao et al., 1998).
- the trivalent KD (4 x 10 ⁇ 17 M) was approximately equal to the third power of the monovalent KD (1.6 x 10 "6 M).
- the fold-back divalent diabody shares the divalent diabody features of enhanced rigidity and a close to 180 degree angle between the two antigen sites and these features contribute to enhanced improvements over the biscFv format.
- the simultaneous and concerted interaction of all 4 domains undoubtedly stabilizes the structure and provides extra thermal stability compared to the biscFv format where only 2 concerted domain interactions occur for each part of the biscFv.
- the rigid 180 degree angle between the two antigen sites permits divalent interactions between monovalently bound diabody- immunotoxin on neighboring cells in closely packed tissue such as lymph nodes, thus expanding the fraction of divalent binding sites beyond that limited on a single cell surface. This decreases the overall kinetic off rate and enhances signaling facilitating endocytosis of immunotoxin.
- Prostate cancer is the most common non-skin neoplasm in men in the United States with 250,000 cases per year and is the second leading cause of cancer mortality in men with 30,000 deaths per year.
- One in six men will develop prostate cancer at some point in their lives.
- the disease is more common in older men and those of African- American ancestry and those with first-degree relatives with prostate cancer.
- Environmental factors including a high animal fat diet and low selenium and vitamin D may contribute to the high rate of disease in the United States.
- prostate carcinogenesis The molecular mechanisms leading to prostate carcinogenesis appear to be multiple and diverse. Disorders in cell renewal (sonic hedgehog, Notch and Wnt pathways), cell survival (PTEN and Ras signaling) and cell proliferation (MAPK pathways) may all been shown to contribute to prostate cancer formation and progression. Thus, there are multiple targets for new therapeutics, but the role of particular molecules in individual prostate cancer patients is unknown and unlikely to be solitary.
- New therapeutics agents for prostate cancer may nevertheless take advantage of the persistence of several prostate differentiation antigens in most metastatic tumor deposits.
- the cell surface membrane 100 kilodalton glycoprotein prostate specific membrane antigen (PSMA) is expressed on epithelial prostate cells on both the prostate and all stages of androgen-insensitive or metastatic disease.
- the antigen is an enzyme and forms a homodimer on the cell surface and undergoes rapid endocytosis on antibody binding.
- the protein is absent from non-prostate tissues.
- Emara M Sanfilippo F (1992) Cell Immunol; 144:143-54.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Immunology (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Pharmacology & Pharmacy (AREA)
- General Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Cell Biology (AREA)
- Epidemiology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Organic Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Toxicology (AREA)
- General Chemical & Material Sciences (AREA)
- Oncology (AREA)
- Molecular Biology (AREA)
- Reproductive Health (AREA)
- Transplantation (AREA)
- Hematology (AREA)
- Peptides Or Proteins (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
Abstract
Description
Claims
Priority Applications (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP08794978A EP2187971A2 (en) | 2007-08-01 | 2008-08-01 | A fold-back diabody diphtheria toxin immunotoxin and methods of use |
CA2695382A CA2695382A1 (en) | 2007-08-01 | 2008-08-01 | A fold-back diabody diphtheria toxin immunotoxin and methods of use |
US12/671,629 US9364557B2 (en) | 2007-08-01 | 2008-08-01 | Fold-back diabody diphtheria toxin immunotoxin and methods of use |
AU2008282729A AU2008282729B2 (en) | 2007-08-01 | 2008-08-01 | A fold-back diabody diphtheria toxin immunotoxin and methods of use |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US95341607P | 2007-08-01 | 2007-08-01 | |
US60/953,416 | 2007-08-01 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2009017823A2 true WO2009017823A2 (en) | 2009-02-05 |
WO2009017823A3 WO2009017823A3 (en) | 2009-03-19 |
Family
ID=40193537
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2008/009321 WO2009017823A2 (en) | 2007-08-01 | 2008-08-01 | A fold-back diabody diphtheria toxin immunotoxin and methods of use |
Country Status (5)
Country | Link |
---|---|
US (1) | US9364557B2 (en) |
EP (1) | EP2187971A2 (en) |
AU (1) | AU2008282729B2 (en) |
CA (1) | CA2695382A1 (en) |
WO (1) | WO2009017823A2 (en) |
Cited By (64)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2011020783A3 (en) * | 2009-08-17 | 2011-04-28 | Roche Glycart Ag | Targeted immunoconjugates |
WO2011069019A3 (en) * | 2009-12-02 | 2012-07-05 | Imaginab, Inc. | J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen |
CN103025759A (en) * | 2010-04-01 | 2013-04-03 | 安进研发(慕尼黑)股份有限公司 | Cross-species-specific psmaxcd3 bispecific single chain antibody |
WO2013059885A3 (en) * | 2011-10-28 | 2014-07-17 | Teva Pharmaceuticals Australia Pty Ltd | Polypeptide constructs and uses thereof |
US8951737B2 (en) | 1996-05-06 | 2015-02-10 | Cornell Research Foundation, Inc. | Treatment and diagnosis of cancer |
US9493578B2 (en) | 2009-09-02 | 2016-11-15 | Xencor, Inc. | Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens |
US9605084B2 (en) | 2013-03-15 | 2017-03-28 | Xencor, Inc. | Heterodimeric proteins |
US9605061B2 (en) | 2010-07-29 | 2017-03-28 | Xencor, Inc. | Antibodies with modified isoelectric points |
US9636334B2 (en) | 2014-05-01 | 2017-05-02 | Teva Pharmaceuticals Australia Pty Ltd | Combination of lenalidomide and polypeptide construct, and uses thereof |
US9650446B2 (en) | 2013-01-14 | 2017-05-16 | Xencor, Inc. | Heterodimeric proteins |
US9701759B2 (en) | 2013-01-14 | 2017-07-11 | Xencor, Inc. | Heterodimeric proteins |
US9738722B2 (en) | 2013-01-15 | 2017-08-22 | Xencor, Inc. | Rapid clearance of antigen complexes using novel antibodies |
US9745381B2 (en) | 2012-05-18 | 2017-08-29 | Scott & White Healthcare (Swh) | Bispecific scFv immunofusion (BIf) |
US9822186B2 (en) | 2014-03-28 | 2017-11-21 | Xencor, Inc. | Bispecific antibodies that bind to CD38 and CD3 |
US9850320B2 (en) | 2014-11-26 | 2017-12-26 | Xencor, Inc. | Heterodimeric antibodies to CD3 X CD20 |
US9856327B2 (en) | 2014-11-26 | 2018-01-02 | Xencor, Inc. | Heterodimeric antibodies to CD3 X CD123 |
WO2018033749A1 (en) * | 2016-08-18 | 2018-02-22 | Polytherics Limited | Anti-psma antibodies, uses thereof and conjugates thereof |
US9963515B2 (en) | 2013-04-29 | 2018-05-08 | Teva Pharmaceuticals Australia Pty Ltd. | Anti-CD38 antibodies and fusions to attenuated interferon alpha-2B |
US10106624B2 (en) | 2013-03-15 | 2018-10-23 | Xencor, Inc. | Heterodimeric proteins |
US10131710B2 (en) | 2013-01-14 | 2018-11-20 | Xencor, Inc. | Optimized antibody variable regions |
US10227410B2 (en) | 2015-12-07 | 2019-03-12 | Xencor, Inc. | Heterodimeric antibodies that bind CD3 and PSMA |
US10227411B2 (en) | 2015-03-05 | 2019-03-12 | Xencor, Inc. | Modulation of T cells with bispecific antibodies and FC fusions |
US10316088B2 (en) | 2016-06-28 | 2019-06-11 | Xencor, Inc. | Heterodimeric antibodies that bind somatostatin receptor 2 |
US10392425B2 (en) | 2015-02-05 | 2019-08-27 | Molecular Templates, Inc. | Multivalent CD20-binding molecules comprising Shiga toxin A subunit effector regions and enriched compositions thereof |
US10428155B2 (en) | 2014-12-22 | 2019-10-01 | Xencor, Inc. | Trispecific antibodies |
US10487155B2 (en) | 2013-01-14 | 2019-11-26 | Xencor, Inc. | Heterodimeric proteins |
US10501543B2 (en) | 2016-10-14 | 2019-12-10 | Xencor, Inc. | IL15/IL15Rα heterodimeric Fc-fusion proteins |
US10519242B2 (en) | 2013-03-15 | 2019-12-31 | Xencor, Inc. | Targeting regulatory T cells with heterodimeric proteins |
US10517969B2 (en) | 2009-02-17 | 2019-12-31 | Cornell University | Methods and kits for diagnosis of cancer and prediction of therapeutic value |
EP3587454A1 (en) * | 2018-06-27 | 2020-01-01 | Albert-Ludwigs-Universität Freiburg | Chimeric antigen receptors that bind to prostate specific membrane antigen |
US10526417B2 (en) | 2014-11-26 | 2020-01-07 | Xencor, Inc. | Heterodimeric antibodies that bind CD3 and CD38 |
US10544199B2 (en) | 2014-10-29 | 2020-01-28 | Teva Pharmaceuticals Australia Pty Ltd | Interferon alpha 2B variants |
US10544187B2 (en) | 2013-03-15 | 2020-01-28 | Xencor, Inc. | Targeting regulatory T cells with heterodimeric proteins |
US10787518B2 (en) | 2016-06-14 | 2020-09-29 | Xencor, Inc. | Bispecific checkpoint inhibitor antibodies |
US10793632B2 (en) | 2016-08-30 | 2020-10-06 | Xencor, Inc. | Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors |
US10815469B2 (en) | 2014-06-11 | 2020-10-27 | Molecular Templates, Inc. | Cell-targeting molecules comprising protease-cleavage resistant, Shiga toxin A subunit effector polypeptides and carboxy-terminal moieties |
US10851178B2 (en) | 2011-10-10 | 2020-12-01 | Xencor, Inc. | Heterodimeric human IgG1 polypeptides with isoelectric point modifications |
US10858417B2 (en) | 2013-03-15 | 2020-12-08 | Xencor, Inc. | Heterodimeric proteins |
US10968276B2 (en) | 2013-03-12 | 2021-04-06 | Xencor, Inc. | Optimized anti-CD3 variable regions |
US10981992B2 (en) | 2017-11-08 | 2021-04-20 | Xencor, Inc. | Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors |
US10982006B2 (en) | 2018-04-04 | 2021-04-20 | Xencor, Inc. | Heterodimeric antibodies that bind fibroblast activation protein |
EP3842461A1 (en) | 2019-12-23 | 2021-06-30 | Albert-Ludwigs-Universität Freiburg | Chimeric antigen receptors that bind to prostate specific membrane antigen |
US11053316B2 (en) | 2013-01-14 | 2021-07-06 | Xencor, Inc. | Optimized antibody variable regions |
US11084863B2 (en) | 2017-06-30 | 2021-08-10 | Xencor, Inc. | Targeted heterodimeric Fc fusion proteins containing IL-15 IL-15alpha and antigen binding domains |
US11117975B2 (en) | 2013-04-29 | 2021-09-14 | Teva Pharmaceuticals Australia Pty Ltd | Anti-CD38 antibodies and fusions to attenuated interferon alpha-2B |
US11142584B2 (en) | 2014-03-11 | 2021-10-12 | Molecular Templates, Inc. | CD20-binding proteins comprising Shiga toxin A subunit effector regions for inducing cellular internalization and methods using same |
US11225509B2 (en) | 2018-04-17 | 2022-01-18 | Molecular Templates, Inc. | HER2-targeting molecules comprising de-immunized, Shiga toxin A subunit scaffolds |
US11242400B2 (en) | 2016-09-21 | 2022-02-08 | Aptevo Research And Development Llc | CD123 binding proteins and related compositions and methods |
US11254744B2 (en) | 2015-08-07 | 2022-02-22 | Imaginab, Inc. | Antigen binding constructs to target molecules |
US11266745B2 (en) | 2017-02-08 | 2022-03-08 | Imaginab, Inc. | Extension sequences for diabodies |
US11312751B2 (en) | 2014-01-27 | 2022-04-26 | Molecular Templates, Inc. | MHC class I epitope delivering polypeptides |
US11312770B2 (en) | 2017-11-08 | 2022-04-26 | Xencor, Inc. | Bispecific and monospecific antibodies using novel anti-PD-1 sequences |
US11319355B2 (en) | 2017-12-19 | 2022-05-03 | Xencor, Inc. | Engineered IL-2 Fc fusion proteins |
US11358999B2 (en) | 2018-10-03 | 2022-06-14 | Xencor, Inc. | IL-12 heterodimeric Fc-fusion proteins |
US11365223B2 (en) | 2015-05-30 | 2022-06-21 | Molecular Templates, Inc. | De-immunized, Shiga toxin a subunit scaffolds and cell-targeting molecules comprising the same |
US11389542B1 (en) | 2016-12-07 | 2022-07-19 | Molecular Templates, Inc. | Shiga toxin a subunit effector polypeptides, Shiga toxin effector scaffolds, and cell-targeting molecules for site-specific conjugation |
US11406692B2 (en) | 2017-01-25 | 2022-08-09 | Molecular Templates, Inc. | Cell-targeting molecules comprising de-immunized, Shiga toxin a subunit effectors and CD8+ t-cell epitopes |
US11472890B2 (en) | 2019-03-01 | 2022-10-18 | Xencor, Inc. | Heterodimeric antibodies that bind ENPP3 and CD3 |
US11505595B2 (en) | 2018-04-18 | 2022-11-22 | Xencor, Inc. | TIM-3 targeted heterodimeric fusion proteins containing IL-15/IL-15RA Fc-fusion proteins and TIM-3 antigen binding domains |
US11524991B2 (en) | 2018-04-18 | 2022-12-13 | Xencor, Inc. | PD-1 targeted heterodimeric fusion proteins containing IL-15/IL-15Ra Fc-fusion proteins and PD-1 antigen binding domains and uses thereof |
US11591401B2 (en) | 2020-08-19 | 2023-02-28 | Xencor, Inc. | Anti-CD28 compositions |
US11739144B2 (en) | 2021-03-09 | 2023-08-29 | Xencor, Inc. | Heterodimeric antibodies that bind CD3 and CLDN6 |
US11859012B2 (en) | 2021-03-10 | 2024-01-02 | Xencor, Inc. | Heterodimeric antibodies that bind CD3 and GPC3 |
US11919956B2 (en) | 2020-05-14 | 2024-03-05 | Xencor, Inc. | Heterodimeric antibodies that bind prostate specific membrane antigen (PSMA) and CD3 |
Families Citing this family (9)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US9623117B2 (en) * | 2011-04-04 | 2017-04-18 | Wisconsin Alumni Research Foundation | Method for selective targeting and entry of bacterial toxins to cells |
WO2012145714A2 (en) | 2011-04-22 | 2012-10-26 | Emergent Product Development Seattle, Llc | Prostate-specific membrane antigen binding proteins and related compositions and methods |
US20160145348A1 (en) * | 2013-03-14 | 2016-05-26 | Fred Hutchinson Cancer Research Center | Compositions and methods to modify cells for therapeutic objectives |
BR112017021536A2 (en) * | 2015-04-06 | 2018-07-03 | Harvard College | compositions and methods for nonmyeloablative conditioning |
SG10202002577XA (en) | 2015-09-21 | 2020-04-29 | Aptevo Res & Development Llc | Cd3 binding polypeptides |
EA039364B1 (en) * | 2015-10-09 | 2022-01-18 | Президент Энд Феллоус Оф Гарвард Колледж | Compositions and methods for non-myeloablative conditioning |
US10188749B2 (en) | 2016-04-14 | 2019-01-29 | Fred Hutchinson Cancer Research Center | Compositions and methods to program therapeutic cells using targeted nucleic acid nanocarriers |
EP3565535A4 (en) | 2017-01-05 | 2020-12-30 | Fred Hutchinson Cancer Research Center | Systems and methods to improve vaccine efficacy |
WO2019191752A1 (en) * | 2018-03-30 | 2019-10-03 | Ohio State Innovation Foundation | Methods for pre-conditioning patients for t-cell therapy |
Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US6103235A (en) * | 1995-10-30 | 2000-08-15 | The United States Of America As Represented By The Department Of Health And Human Services | Methods of inducing immune tolerance using immunotoxins |
WO2001087982A2 (en) * | 2000-05-18 | 2001-11-22 | The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services | Immunotoxin fusion proteins and means for expression thereof |
US20040213791A1 (en) * | 2001-06-01 | 2004-10-28 | Neil Bander | Modified antibodies to prostate-specific membrane antigen and uses thereof |
WO2005012495A2 (en) * | 2003-08-01 | 2005-02-10 | The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services, National Institutes Of Health | Methods for expression and purification of immunotoxins |
US20060020258A1 (en) * | 2004-07-20 | 2006-01-26 | Medtronic, Inc. | Surgical apparatus with a manually actuatable assembly and a method of operating same |
Family Cites Families (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US7517527B2 (en) | 1995-10-30 | 2009-04-14 | The United States Of America As Represented By The Department Of Health And Human Services | Immunotoxin with in vivo T cell suppressant activity and methods of use |
US7696338B2 (en) | 1995-10-30 | 2010-04-13 | The United States Of America As Represented By The Department Of Health And Human Services | Immunotoxin fusion proteins and means for expression thereof |
CA2452288A1 (en) * | 2001-09-20 | 2003-03-27 | Cornell Research Foundation, Inc. | Methods and compositions for treating and preventing skin disorders using binding agents specific for psma |
AU2004263538B2 (en) * | 2003-08-08 | 2009-09-17 | Immunomedics, Inc. | Bispecific antibodies for inducing apoptosis of tumor and diseased cells |
WO2005070456A2 (en) * | 2004-01-09 | 2005-08-04 | Millennium Pharmaceuticals, Inc. | Diagnosing and treating female reproductive tract or chilhood cancer with pmsa antibodies |
-
2008
- 2008-08-01 CA CA2695382A patent/CA2695382A1/en not_active Abandoned
- 2008-08-01 US US12/671,629 patent/US9364557B2/en not_active Expired - Fee Related
- 2008-08-01 WO PCT/US2008/009321 patent/WO2009017823A2/en active Application Filing
- 2008-08-01 AU AU2008282729A patent/AU2008282729B2/en active Active
- 2008-08-01 EP EP08794978A patent/EP2187971A2/en not_active Ceased
Patent Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US6103235A (en) * | 1995-10-30 | 2000-08-15 | The United States Of America As Represented By The Department Of Health And Human Services | Methods of inducing immune tolerance using immunotoxins |
WO2001087982A2 (en) * | 2000-05-18 | 2001-11-22 | The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services | Immunotoxin fusion proteins and means for expression thereof |
US20040213791A1 (en) * | 2001-06-01 | 2004-10-28 | Neil Bander | Modified antibodies to prostate-specific membrane antigen and uses thereof |
WO2005012495A2 (en) * | 2003-08-01 | 2005-02-10 | The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services, National Institutes Of Health | Methods for expression and purification of immunotoxins |
US20060020258A1 (en) * | 2004-07-20 | 2006-01-26 | Medtronic, Inc. | Surgical apparatus with a manually actuatable assembly and a method of operating same |
Non-Patent Citations (2)
Title |
---|
KIM GEUN-BAE ET AL: "A fold-back single-chain diabody format enhances the bioactivity of an anti-monkey CD3 recombinant diphtheria toxin-based immunotoxin." PROTEIN ENGINEERING, DESIGN & SELECTION : PEDS SEP 2007, vol. 20, no. 9, September 2007 (2007-09), pages 425-432, XP002512188 ISSN: 1741-0126 * |
TAKEMURA SHIN-ICHI ET AL: "A mutated superantigen SEA D227A fusion diabody specific to MUC1 and CD3 in targeted cancer immunotherapy for bile duct carcinoma." CANCER IMMUNOLOGY, IMMUNOTHERAPY : CII MAR 2002, vol. 51, no. 1, March 2002 (2002-03), pages 33-44, XP002512189 ISSN: 0340-7004 * |
Cited By (130)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US8951737B2 (en) | 1996-05-06 | 2015-02-10 | Cornell Research Foundation, Inc. | Treatment and diagnosis of cancer |
US10517969B2 (en) | 2009-02-17 | 2019-12-31 | Cornell University | Methods and kits for diagnosis of cancer and prediction of therapeutic value |
WO2011020783A3 (en) * | 2009-08-17 | 2011-04-28 | Roche Glycart Ag | Targeted immunoconjugates |
US8945571B2 (en) | 2009-08-17 | 2015-02-03 | Roche GlyeArt AG | Targeted immunoconjugates |
JP2013502209A (en) * | 2009-08-17 | 2013-01-24 | ロシュ グリクアート アーゲー | Targeted immunoconjugate |
US9493578B2 (en) | 2009-09-02 | 2016-11-15 | Xencor, Inc. | Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens |
CN105085681A (en) * | 2009-12-02 | 2015-11-25 | 伊麦吉纳博公司 | J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (psma) and methods for their use |
EP3511023A1 (en) * | 2009-12-02 | 2019-07-17 | Imaginab, Inc. | J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (psma) and methods for their use |
WO2011069019A3 (en) * | 2009-12-02 | 2012-07-05 | Imaginab, Inc. | J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen |
EP2506876A2 (en) * | 2009-12-02 | 2012-10-10 | Imaginab, Inc. | J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (psma) and methods for their use |
AU2010325969B2 (en) * | 2009-12-02 | 2016-10-20 | Imaginab, Inc. | J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen |
EP2506876A4 (en) * | 2009-12-02 | 2013-07-31 | Imaginab Inc | J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (psma) and methods for their use |
EP3135302A1 (en) * | 2009-12-02 | 2017-03-01 | Imaginab, Inc. | J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (psma) and methods for their use |
CN105085681B (en) * | 2009-12-02 | 2018-12-28 | 伊麦吉纳博公司 | Target the J591 miniantibody and double antibody of human prostate-specific membrane antigen |
US11180570B2 (en) | 2009-12-02 | 2021-11-23 | Imaginab, Inc. | J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (PSMA) and methods for their use |
CN107090044A (en) * | 2010-04-01 | 2017-08-25 | 安进研发(慕尼黑)股份有限公司 | Cross-species-specific PSMAxCD3 bispecific single-chain antibodies |
CN103025759A (en) * | 2010-04-01 | 2013-04-03 | 安进研发(慕尼黑)股份有限公司 | Cross-species-specific psmaxcd3 bispecific single chain antibody |
US9605061B2 (en) | 2010-07-29 | 2017-03-28 | Xencor, Inc. | Antibodies with modified isoelectric points |
US10851178B2 (en) | 2011-10-10 | 2020-12-01 | Xencor, Inc. | Heterodimeric human IgG1 polypeptides with isoelectric point modifications |
EA034989B1 (en) * | 2011-10-28 | 2020-04-15 | Тева Фармасьютикал Австралия Пти Лтд | Polypeptide construct for use in treating cancer comprising attenuated alpha interferon |
JP2015500796A (en) * | 2011-10-28 | 2015-01-08 | テバ・ファーマシューティカルズ・オーストラリア・ピーティワイ・リミテッド | Polypeptide constructs and uses thereof |
WO2013059885A3 (en) * | 2011-10-28 | 2014-07-17 | Teva Pharmaceuticals Australia Pty Ltd | Polypeptide constructs and uses thereof |
CN104203982A (en) * | 2011-10-28 | 2014-12-10 | 特瓦制药澳大利亚私人有限公司 | Polypeptide constructs and uses thereof |
AU2012327877B2 (en) * | 2011-10-28 | 2017-10-12 | Teva Pharmaceuticals Australia Pty Ltd | Polypeptide constructs and uses thereof |
JP7111669B2 (en) | 2011-10-28 | 2022-08-02 | テバ・ファーマシューティカルズ・オーストラリア・ピーティワイ・リミテッド | Polypeptide constructs and uses thereof |
US9611322B2 (en) | 2011-10-28 | 2017-04-04 | Teva Pharmaceuticals Australia Pty Ltd | Fusions of antibodies to CD38 and attenuated interferon alpha |
AU2018200221B2 (en) * | 2011-10-28 | 2020-01-16 | Teva Pharmaceuticals Australia Pty Ltd | Polypeptide constructs and uses thereof |
US10981986B2 (en) | 2011-10-28 | 2021-04-20 | Teva Pharmaceuticals Australia Pty Ltd | Fusions of antibodies to CD38 and attenuated interferon alpha |
JP2020010697A (en) * | 2011-10-28 | 2020-01-23 | テバ・ファーマシューティカルズ・オーストラリア・ピーティワイ・リミテッド | Polypeptide construct and use thereof |
US9745381B2 (en) | 2012-05-18 | 2017-08-29 | Scott & White Healthcare (Swh) | Bispecific scFv immunofusion (BIf) |
US10472427B2 (en) | 2013-01-14 | 2019-11-12 | Xencor, Inc. | Heterodimeric proteins |
US10131710B2 (en) | 2013-01-14 | 2018-11-20 | Xencor, Inc. | Optimized antibody variable regions |
US11718667B2 (en) | 2013-01-14 | 2023-08-08 | Xencor, Inc. | Optimized antibody variable regions |
US9701759B2 (en) | 2013-01-14 | 2017-07-11 | Xencor, Inc. | Heterodimeric proteins |
US10738133B2 (en) | 2013-01-14 | 2020-08-11 | Xencor, Inc. | Heterodimeric proteins |
US10738132B2 (en) | 2013-01-14 | 2020-08-11 | Xencor, Inc. | Heterodimeric proteins |
US11053316B2 (en) | 2013-01-14 | 2021-07-06 | Xencor, Inc. | Optimized antibody variable regions |
US9650446B2 (en) | 2013-01-14 | 2017-05-16 | Xencor, Inc. | Heterodimeric proteins |
US11634506B2 (en) | 2013-01-14 | 2023-04-25 | Xencor, Inc. | Heterodimeric proteins |
US10487155B2 (en) | 2013-01-14 | 2019-11-26 | Xencor, Inc. | Heterodimeric proteins |
US9738722B2 (en) | 2013-01-15 | 2017-08-22 | Xencor, Inc. | Rapid clearance of antigen complexes using novel antibodies |
US10968276B2 (en) | 2013-03-12 | 2021-04-06 | Xencor, Inc. | Optimized anti-CD3 variable regions |
US10858417B2 (en) | 2013-03-15 | 2020-12-08 | Xencor, Inc. | Heterodimeric proteins |
US11814423B2 (en) | 2013-03-15 | 2023-11-14 | Xencor, Inc. | Heterodimeric proteins |
US10544187B2 (en) | 2013-03-15 | 2020-01-28 | Xencor, Inc. | Targeting regulatory T cells with heterodimeric proteins |
US10106624B2 (en) | 2013-03-15 | 2018-10-23 | Xencor, Inc. | Heterodimeric proteins |
US10287364B2 (en) | 2013-03-15 | 2019-05-14 | Xencor, Inc. | Heterodimeric proteins |
US10519242B2 (en) | 2013-03-15 | 2019-12-31 | Xencor, Inc. | Targeting regulatory T cells with heterodimeric proteins |
US9605084B2 (en) | 2013-03-15 | 2017-03-28 | Xencor, Inc. | Heterodimeric proteins |
US11299554B2 (en) | 2013-03-15 | 2022-04-12 | Xencor, Inc. | Heterodimeric proteins |
US9963515B2 (en) | 2013-04-29 | 2018-05-08 | Teva Pharmaceuticals Australia Pty Ltd. | Anti-CD38 antibodies and fusions to attenuated interferon alpha-2B |
US11117975B2 (en) | 2013-04-29 | 2021-09-14 | Teva Pharmaceuticals Australia Pty Ltd | Anti-CD38 antibodies and fusions to attenuated interferon alpha-2B |
US11312751B2 (en) | 2014-01-27 | 2022-04-26 | Molecular Templates, Inc. | MHC class I epitope delivering polypeptides |
US12037367B2 (en) | 2014-01-27 | 2024-07-16 | Molecular Templates, Inc. | MHC class I epitope delivering polypeptides |
US12065469B2 (en) | 2014-01-27 | 2024-08-20 | Molecular Templates, Inc. | De-immunized Shiga toxin a subunit effector polypeptides for applications in mammals |
US11142584B2 (en) | 2014-03-11 | 2021-10-12 | Molecular Templates, Inc. | CD20-binding proteins comprising Shiga toxin A subunit effector regions for inducing cellular internalization and methods using same |
US10858451B2 (en) | 2014-03-28 | 2020-12-08 | Xencor, Inc. | Bispecific antibodies that bind to CD38 and CD3 |
US11840579B2 (en) | 2014-03-28 | 2023-12-12 | Xencor, Inc. | Bispecific antibodies that bind to CD38 and CD3 |
US9822186B2 (en) | 2014-03-28 | 2017-11-21 | Xencor, Inc. | Bispecific antibodies that bind to CD38 and CD3 |
US11253591B2 (en) | 2014-05-01 | 2022-02-22 | Teva Pharmaceuticals Australia Pty Ltd. | Combination of lenalidomide and polypeptide construct, and uses thereof |
US10232041B2 (en) | 2014-05-01 | 2019-03-19 | Teva Pharmaceuticals Pty Ltd | Combination of lenalidomide and polypeptide construct, and uses thereof |
US9636334B2 (en) | 2014-05-01 | 2017-05-02 | Teva Pharmaceuticals Australia Pty Ltd | Combination of lenalidomide and polypeptide construct, and uses thereof |
US10815469B2 (en) | 2014-06-11 | 2020-10-27 | Molecular Templates, Inc. | Cell-targeting molecules comprising protease-cleavage resistant, Shiga toxin A subunit effector polypeptides and carboxy-terminal moieties |
US11319356B2 (en) | 2014-10-29 | 2022-05-03 | Teva Pharmaceuticals Australia Pty Ltd | Interferon alpha 2B variants |
US10544199B2 (en) | 2014-10-29 | 2020-01-28 | Teva Pharmaceuticals Australia Pty Ltd | Interferon alpha 2B variants |
US10913803B2 (en) | 2014-11-26 | 2021-02-09 | Xencor, Inc. | Heterodimeric antibodies that bind CD3 and tumor antigens |
US10889653B2 (en) | 2014-11-26 | 2021-01-12 | Xencor, Inc. | Heterodimeric antibodies that bind CD3 and tumor antigens |
US10259887B2 (en) | 2014-11-26 | 2019-04-16 | Xencor, Inc. | Heterodimeric antibodies that bind CD3 and tumor antigens |
US10526417B2 (en) | 2014-11-26 | 2020-01-07 | Xencor, Inc. | Heterodimeric antibodies that bind CD3 and CD38 |
US9850320B2 (en) | 2014-11-26 | 2017-12-26 | Xencor, Inc. | Heterodimeric antibodies to CD3 X CD20 |
US11225528B2 (en) | 2014-11-26 | 2022-01-18 | Xencor, Inc. | Heterodimeric antibodies that bind CD3 and tumor antigens |
US11945880B2 (en) | 2014-11-26 | 2024-04-02 | Xencor, Inc. | Heterodimeric antibodies that bind CD3 and tumor antigens |
US11352442B2 (en) | 2014-11-26 | 2022-06-07 | Xencor, Inc. | Heterodimeric antibodies that bind CD3 and CD38 |
US11673972B2 (en) | 2014-11-26 | 2023-06-13 | Xencor, Inc. | Heterodimeric antibodies that bind CD3 and tumor antigens |
US9856327B2 (en) | 2014-11-26 | 2018-01-02 | Xencor, Inc. | Heterodimeric antibodies to CD3 X CD123 |
US11859011B2 (en) | 2014-11-26 | 2024-01-02 | Xencor, Inc. | Heterodimeric antibodies that bind CD3 and tumor antigens |
US11111315B2 (en) | 2014-11-26 | 2021-09-07 | Xencor, Inc. | Heterodimeric antibodies that bind CD3 and tumor antigens |
US10428155B2 (en) | 2014-12-22 | 2019-10-01 | Xencor, Inc. | Trispecific antibodies |
US11104707B2 (en) | 2015-02-05 | 2021-08-31 | Molecular Templates, Inc. | Multivalent CD20-binding molecules comprising Shiga toxin a subunit effector regions and enriched compositions thereof |
US10392425B2 (en) | 2015-02-05 | 2019-08-27 | Molecular Templates, Inc. | Multivalent CD20-binding molecules comprising Shiga toxin A subunit effector regions and enriched compositions thereof |
US11248061B2 (en) | 2015-02-05 | 2022-02-15 | Molecular Templates, Inc. | Multivalent CD20-binding molecule comprising Shiga toxin A subunit effector polypeptides and enriched compositions thereof |
US11091548B2 (en) | 2015-03-05 | 2021-08-17 | Xencor, Inc. | Modulation of T cells with bispecific antibodies and Fc fusions |
US10227411B2 (en) | 2015-03-05 | 2019-03-12 | Xencor, Inc. | Modulation of T cells with bispecific antibodies and FC fusions |
US11365223B2 (en) | 2015-05-30 | 2022-06-21 | Molecular Templates, Inc. | De-immunized, Shiga toxin a subunit scaffolds and cell-targeting molecules comprising the same |
US11254744B2 (en) | 2015-08-07 | 2022-02-22 | Imaginab, Inc. | Antigen binding constructs to target molecules |
US11623957B2 (en) | 2015-12-07 | 2023-04-11 | Xencor, Inc. | Heterodimeric antibodies that bind CD3 and PSMA |
US10227410B2 (en) | 2015-12-07 | 2019-03-12 | Xencor, Inc. | Heterodimeric antibodies that bind CD3 and PSMA |
US11236170B2 (en) | 2016-06-14 | 2022-02-01 | Xencor, Inc. | Bispecific checkpoint inhibitor antibodies |
US11492407B2 (en) | 2016-06-14 | 2022-11-08 | Xencor, Inc. | Bispecific checkpoint inhibitor antibodies |
US10787518B2 (en) | 2016-06-14 | 2020-09-29 | Xencor, Inc. | Bispecific checkpoint inhibitor antibodies |
US10316088B2 (en) | 2016-06-28 | 2019-06-11 | Xencor, Inc. | Heterodimeric antibodies that bind somatostatin receptor 2 |
US11225521B2 (en) | 2016-06-28 | 2022-01-18 | Xencor, Inc. | Heterodimeric antibodies that bind somatostatin receptor 2 |
US12054545B2 (en) | 2016-06-28 | 2024-08-06 | Xencor, Inc. | Heterodimeric antibodies that bind somatostatin receptor 2 |
JP7039554B2 (en) | 2016-08-18 | 2022-03-22 | ポリセリックス・リミテッド | Anti-PSMA antibody, its use and its conjugate |
US11059903B2 (en) | 2016-08-18 | 2021-07-13 | Polytherics Limited | Anti-PSMA antibodies, uses thereof and conjugates thereof |
JP2019535232A (en) * | 2016-08-18 | 2019-12-12 | ポリセリックス・リミテッド | Anti-PSMA antibodies, uses thereof and conjugates thereof |
CN110049999B (en) * | 2016-08-18 | 2023-12-19 | 阿布泽纳(英国)有限公司 | anti-PSMA antibodies, uses thereof, and conjugates thereof |
EP4403226A3 (en) * | 2016-08-18 | 2024-10-16 | Abzena (UK) Limited | Anti-psma antibodies, uses thereof and conjugates thereof |
JP2022084715A (en) * | 2016-08-18 | 2022-06-07 | ポリセリックス・リミテッド | Anti-psma antibodies, uses thereof and conjugates thereof |
WO2018033749A1 (en) * | 2016-08-18 | 2018-02-22 | Polytherics Limited | Anti-psma antibodies, uses thereof and conjugates thereof |
JP7486538B2 (en) | 2016-08-18 | 2024-05-17 | ポリセリックス・リミテッド | Anti-PSMA antibodies, uses thereof and conjugates thereof |
CN110049999A (en) * | 2016-08-18 | 2019-07-23 | 宝力泰锐克斯有限公司 | Anti- PSMA antibody, its purposes and its conjugate |
US10793632B2 (en) | 2016-08-30 | 2020-10-06 | Xencor, Inc. | Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors |
US11242400B2 (en) | 2016-09-21 | 2022-02-08 | Aptevo Research And Development Llc | CD123 binding proteins and related compositions and methods |
US11939392B2 (en) | 2016-09-21 | 2024-03-26 | Aptevo Research And Development Llc | CD123 binding proteins and related compositions and methods |
US10550185B2 (en) | 2016-10-14 | 2020-02-04 | Xencor, Inc. | Bispecific heterodimeric fusion proteins containing IL-15-IL-15Rα Fc-fusion proteins and PD-1 antibody fragments |
US10501543B2 (en) | 2016-10-14 | 2019-12-10 | Xencor, Inc. | IL15/IL15Rα heterodimeric Fc-fusion proteins |
US11389542B1 (en) | 2016-12-07 | 2022-07-19 | Molecular Templates, Inc. | Shiga toxin a subunit effector polypeptides, Shiga toxin effector scaffolds, and cell-targeting molecules for site-specific conjugation |
US11857628B2 (en) | 2016-12-07 | 2024-01-02 | Molecular Templates, Inc. | Shiga toxin A subunit effector polypeptides, Shiga toxin effector scaffolds, and cell-targeting molecules for site-specific conjugation |
US11406692B2 (en) | 2017-01-25 | 2022-08-09 | Molecular Templates, Inc. | Cell-targeting molecules comprising de-immunized, Shiga toxin a subunit effectors and CD8+ t-cell epitopes |
US11266745B2 (en) | 2017-02-08 | 2022-03-08 | Imaginab, Inc. | Extension sequences for diabodies |
US11084863B2 (en) | 2017-06-30 | 2021-08-10 | Xencor, Inc. | Targeted heterodimeric Fc fusion proteins containing IL-15 IL-15alpha and antigen binding domains |
US10981992B2 (en) | 2017-11-08 | 2021-04-20 | Xencor, Inc. | Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors |
US11312770B2 (en) | 2017-11-08 | 2022-04-26 | Xencor, Inc. | Bispecific and monospecific antibodies using novel anti-PD-1 sequences |
US11319355B2 (en) | 2017-12-19 | 2022-05-03 | Xencor, Inc. | Engineered IL-2 Fc fusion proteins |
US10982006B2 (en) | 2018-04-04 | 2021-04-20 | Xencor, Inc. | Heterodimeric antibodies that bind fibroblast activation protein |
US11225509B2 (en) | 2018-04-17 | 2022-01-18 | Molecular Templates, Inc. | HER2-targeting molecules comprising de-immunized, Shiga toxin A subunit scaffolds |
US11524991B2 (en) | 2018-04-18 | 2022-12-13 | Xencor, Inc. | PD-1 targeted heterodimeric fusion proteins containing IL-15/IL-15Ra Fc-fusion proteins and PD-1 antigen binding domains and uses thereof |
US11505595B2 (en) | 2018-04-18 | 2022-11-22 | Xencor, Inc. | TIM-3 targeted heterodimeric fusion proteins containing IL-15/IL-15RA Fc-fusion proteins and TIM-3 antigen binding domains |
WO2020002015A1 (en) | 2018-06-27 | 2020-01-02 | Albert-Ludwigs-Universität Freiburg | Chimeric antigen receptors that bind to prostate specific membrane antigen |
EP3587454A1 (en) * | 2018-06-27 | 2020-01-01 | Albert-Ludwigs-Universität Freiburg | Chimeric antigen receptors that bind to prostate specific membrane antigen |
US11358999B2 (en) | 2018-10-03 | 2022-06-14 | Xencor, Inc. | IL-12 heterodimeric Fc-fusion proteins |
US11472890B2 (en) | 2019-03-01 | 2022-10-18 | Xencor, Inc. | Heterodimeric antibodies that bind ENPP3 and CD3 |
EP3842461A1 (en) | 2019-12-23 | 2021-06-30 | Albert-Ludwigs-Universität Freiburg | Chimeric antigen receptors that bind to prostate specific membrane antigen |
WO2021130042A1 (en) | 2019-12-23 | 2021-07-01 | Albert-Ludwigs-Universität Freiburg | Chimeric antigen receptors that bind to prostate specific membrane antigen |
US11919956B2 (en) | 2020-05-14 | 2024-03-05 | Xencor, Inc. | Heterodimeric antibodies that bind prostate specific membrane antigen (PSMA) and CD3 |
US11919958B2 (en) | 2020-08-19 | 2024-03-05 | Xencor, Inc. | Anti-CD28 compositions |
US11591401B2 (en) | 2020-08-19 | 2023-02-28 | Xencor, Inc. | Anti-CD28 compositions |
US11739144B2 (en) | 2021-03-09 | 2023-08-29 | Xencor, Inc. | Heterodimeric antibodies that bind CD3 and CLDN6 |
US11859012B2 (en) | 2021-03-10 | 2024-01-02 | Xencor, Inc. | Heterodimeric antibodies that bind CD3 and GPC3 |
Also Published As
Publication number | Publication date |
---|---|
CA2695382A1 (en) | 2009-02-05 |
US20110189209A1 (en) | 2011-08-04 |
WO2009017823A3 (en) | 2009-03-19 |
US9364557B2 (en) | 2016-06-14 |
AU2008282729A1 (en) | 2009-02-05 |
AU2008282729B2 (en) | 2015-07-16 |
EP2187971A2 (en) | 2010-05-26 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US9364557B2 (en) | Fold-back diabody diphtheria toxin immunotoxin and methods of use | |
US7696338B2 (en) | Immunotoxin fusion proteins and means for expression thereof | |
Hertler et al. | Immunotoxins: a clinical review of their use in the treatment of malignancies. | |
Carter et al. | Antibody-drug conjugates for cancer therapy | |
EP1015496B1 (en) | Divalent anti-t cells immunotoxins and use thereof | |
Dosio et al. | Advances in anticancer antibody-drug conjugates and immunotoxins | |
US6103235A (en) | Methods of inducing immune tolerance using immunotoxins | |
CN112189020A (en) | Use of antibody drug conjugates comprising tubulin disruptors for the treatment of solid tumors | |
EP1287147B1 (en) | Immunotoxin fusion proteins and means for expression thereof | |
AU2001261760A1 (en) | Immunotoxin fusion proteins and means for expression thereof | |
EP0830146B1 (en) | Methods of inducing immune tolerance using immunotoxins | |
US20240209080A1 (en) | Folr1 binding agents, conjugates thereof and methods of using the same | |
EP1073463B1 (en) | Use of immunotoxins to induce immune tolerance to pancreatic islet transplantation | |
US7517527B2 (en) | Immunotoxin with in vivo T cell suppressant activity and methods of use | |
US20050142117A9 (en) | Use of immunotoxins to induce immune tolerance to pancreatic islet transplantation | |
US7125553B1 (en) | Methods of inducing immune tolerance using immunotoxins | |
Zaman | Topics in Anti-Cancer Research | |
WO2022064191A1 (en) | Anti-cd45 antibodies and related therapeutics | |
JP2024521667A (en) | Combination medicine comprising an anti-CD205 antibody and an immune checkpoint inhibitor |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 08794978 Country of ref document: EP Kind code of ref document: A2 |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2008282729 Country of ref document: AU |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2695382 Country of ref document: CA |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
ENP | Entry into the national phase |
Ref document number: 2008282729 Country of ref document: AU Date of ref document: 20080801 Kind code of ref document: A |
|
REEP | Request for entry into the european phase |
Ref document number: 2008794978 Country of ref document: EP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2008794978 Country of ref document: EP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 12671629 Country of ref document: US |