CA2110799A1 - Microbially-produced antibody fragments and their conjugates - Google Patents

Microbially-produced antibody fragments and their conjugates

Info

Publication number
CA2110799A1
CA2110799A1 CA 2110799 CA2110799A CA2110799A1 CA 2110799 A1 CA2110799 A1 CA 2110799A1 CA 2110799 CA2110799 CA 2110799 CA 2110799 A CA2110799 A CA 2110799A CA 2110799 A1 CA2110799 A1 CA 2110799A1
Authority
CA
Canada
Prior art keywords
fab
preparing
molecule
enzyme
chain
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA 2110799
Other languages
French (fr)
Inventor
Arnold H. Horwitz
Marc D. Better
Steve Carroll
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Xoma Corp
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2110799A1 publication Critical patent/CA2110799A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Recombinant DNA methods are described for the production of antibody F(ab')2 and Fab' fragment molecules from transformed microorganisms such as bacteria and yeast. The recombinant F(ab')2 and Fab' fragments are secreted from the host cells and may be recovered directly from the fermentation culture. These direct methods of F(ab')2 and Fab' production are advantageous over existing art methods by avoiding proteolytic digestions and obtaining homogeneous molecules. Methods are described for the conjugation of these fragments, by methods targeting either available lysine amino acid residues or selectively-reducible cysteine residues, to active moieties such as ricin toxin A chain. The microbially-produced F(ab')2 and Fab' fragment molecules and their immunoconjugates have a wide variety of therapeutic and diagnostic uses.

Description

WO 92/22324 PCl/IJS92/04976 9 :~

TlTLE OF THE lNVENT~ON
MICROBIALLY-PRODUCED ANTIBODY
FRAGMENTS AND THEIR CONJUGATES

BACKGROUND OF THE INVENTION

Field of the Invention .
This invention relates to methods for pro.iucing antibody F(ab')2 and Fab' fragments by recombinant DNA manipulations and microbia]
fermentation, and methods for their use in the preparation of protein conjugates, including immunoconjugates.

Brief Desc~iption of the Background Art The development of monoclonal antibody technology has resulted in the proliferation of antibodies for use in research, di~nostic and therapeutic applications. Many of these antibodies originate from mouse hybridomas. Such non-human antibodies can be problematic for use as in vivo diagnostics and as therapeutic agents in bumans due to immunogenicity. This is an especially important consideration when these products are used for repeated treatment over extended periods of time.
One solution to the problem of mouse antibody immunogenicity is to use recombinant DNA technology to produce mouse-human chimeric antibodies in which gene sequences encoding mouse V regions are fused to gene sequences encoding human const~n~ regions (reviewed by ~,
2 1 1 lJ ~ ~ 9 PCI /US92/04~?~i Morrison et al., Adv. Imm~nol. 44: 65-92 (1989)). In addition to partially ~'humanizing" the mouse-derived antibody, the same genetic engineering technology used for producing chimeric antibodies can also be used to permit easy class switching (Shaw e~ aL J. IVa~l. Cancer Ins~itute 80: 1553 (1988)).
Concurrent with the production and engineering of monoclonal antibodies directed towards a variety of tumor antigens has been the development of technologies for the use of these antibodies for in vivo diagnosis of tumor metastases (i~., tumor imaging) and for therapy, either as naked antibodies or coupled with therapeutically efficacious agents such as radionuclides, drugs and toxins. However, the whole antibody, whether it be all mouse. or mouse-human chimeric, may have disadvantages for certain diagnostic and/or therapeutic applications where greater tumor penetration, less liver interaction, or more rapid clearance time is desired.
Instead, antibody fragments such as Fab, Fab' and F(ab')2, which consist of the antigen-binding subunits but lack regions containing the effector functions (Fc), have been demonstrated to be efficacious in animal models (Colapinto el al. Cancer R~.s. 48: 5701 (1988); Wahl et al. J. Nl~L Med. 24:
316 (1983)), and in human clinical studies (Delaloye e~ al. J. Clin. Invest.
77: 301 (1986)). These fragments, which are generated by proteolytic digestion of whole antibody~ have been shown to give higher tumor/blood ratios for imaging and proms)te deeper penetra~ion into tumors ~or therapeutic applications than whole antibody.
F(ab'), molecules. which consist of two Fab units linked by the interchain disulfide bonds in the hinge region, retain the binding affinity achieved by the whole antibody. Unconjug?.ted F(ab')~ molecules may be useful therapeutic agents and have been shown to have equ~l or higher efficacy as whole antibodies in immunosuppression models (Carteron e~
al. Clin. lmml~no. Immun(Jpa~ll. 5~): 373-383 (1990)l and Hirsch el al.
Transplan~atiCJn Pr~JC~edin~ s ?3: 270-271 (1991)). Furthermore, W~ 92/22324 ~ 7 9 9 PCI/US92/04976
- 3 immunoconjugates of F(ab')~ may be superior to those containing whole antibodies for imaging applications or for delivery of therapeutic agents to tumors (Colapinto, sl~pra; Delaloye, s~pra; Wahl, supra).
F(ab')2 molecules are also useful since the individual antigen-binding units (Fab') can be separated by using mild reducing conditions (Johnstone e~ al. Immunochemistry in Practice (Blackwell, Oxford), page 52 (1982)). The resulting Fab' fragments contain reactive sulfl~ydryl groups which can be used for covalent attachment of label, or other clinically or experimentally useful molecules. They also can serve as the starting point for the construction of heterobifunctional F(ab'), molecules consisting of individual Fab' units with different specificities. Examples of such heterobifunctional F(ab'), molecules include those which combine specificity toward a tumor antigen with a chelating agent for a radionuclide, or toward an antigen and an Fc receptor.
A number of clinically important antibodies~ or their F(ab')2 and Fab fragments. have been used as immunoconjugates with cytotoxic organic compounds, protein toxins, and radionuclides (Yang et al. Proc.
^ Na~l. Acad. Sci. USA 85: 1189 (1988); Blakey et a1. Pro~. Aller~ 115: 50 (1988); Delaloye el al., sl~pra)). Typically, the crosslinking ~eagents are coupled to the Iysine residues of the antibody molecules which have been first derivatized with bifunctional cross-linking reagents such as N-succinimidyl-3 (~-pyridyldithio)propionate (SPDP~, which modified Iysine residues throughout the molecule.
We have recently described two systems for producing genetically engineered Fab fragments by secretion from Saccharomvces cere~isiae (Holwitz el ~11. Prc)c. Na~L Aca~i. Sci. USA 8~: 8678-8682 (1988)) and E~scherichia c~li (Better e~ al. Science 24(~: 1041-1043 (1988)). While microbially-produced Fab molecules are useful as starting points for the development of certain reagents. they do not retain the same degree of .

-, :`
4 PCI`/USg2/04~.16 binding affinity of the whole antibody or F(ab'?2. Furthermore, Fab fragments lack the specific thiol conjugation properties of Fab'.
Ourrent methods for F(ab')2 production involve pepsin digestion of whole antibody (Johnstone, sl~pra, at 53-55). This approach can be problematic for several reasons. First~ not all antibodies are equivalent in their susceptibility to pepsin cleavage. Second, digestion with pepsin can result in partial degradation of the fragments which may not be apparent when analyzed in non-reducing gels. Finally, additional purification steps are required beyond that for the whole antibody.
Approaches to the direct production of F(ab'), molecules have been taken with engineered animal cells. ~Neuberger el al. Nalure 312:
604 (1984)) were able to obtain production of a F(ab')~-like fragment by modification of the Fd' gene to contain a 21-amino-acid tailpiece from an immunoglobulin secretion exon. Bodmer el al. (WO 8901783 (1989~
reported obtaining only Fab' from COS cells, which could be subsequently associated to form F(ab')~. Also, Gillies et al. (Hum. Antibod. Ifvbrid. 1 47 (1990)), failed in their attempt to directly make F(ab'), fragments from animal cells. Given the difficulties associated with their p~roduction in animal cells, it is unpredictable as to whether functional F(ab')~ molecules could be directly made in a properly folded and assembled state b~
microorganisms such as bacteria or yeast.
Fab' fragments are useful for thiol-directed conjugation, and ma~
be generated by selective reduction of F(ab'), fragments (Johnstone.
sl~pra, at 53-S5). Fab' produced by selective thiol reduction of F(ab'), may however have the same problems mentioned above for F(ab'), generated by classical enzymatic` methods.
There is therefore a subst~ntial need for the establishment of methods for the direct production of functional F(ab'), and Fab' molecules from microorganisms. There is also a need to establish conditions for their use in the ~eneration of effective immunoconjugates.

,~.
, ~ .

W~ 92/22324 2 1 1 ~ 7 ~3 9 PCI/US92/04976 The present invention demonstrates that, surprisingly, microorganisms can secrete such antibody fragments. Methods are described for their use in the production of active F(ab')~ or Fab' immunoconjugates and heterobifunctional F(ab')~ molecules.

SUMMARY OF THE INVENTION
. .

Thisinvention providesimmunoglobulin fragments, such asF(ab')~5 which retain the full binding ~ffinity of whole antibody. This invention also provides immunoglobulin fragments, such as F~b', which have the useful feature of convenient and selective thiol conjugation to polypeptide and chemical moieties. These fragments are produced directly from microorganisms, such as bacteria and yeast, which are engineered to secrete intact recombinant F(ab') ~rF(ab'),) and recombinant Fab' (rFab') fTagments. This production system is advant~geous over traditional proteolysis methods which have additional steps and often give nicked or partially degraded antibody fragments. Various ~ene constructs are detailed which show truncated versions of a chimeric, mouse variable-human constant, immunoglobulin IgG1 heavy chain gene (Fd) encoding 1 or 2 cysteine residues to the carboxyl side of the light chain-Fd chain disulfide bond. These modified genes were assembled into vectors and transformed into either bac~eri~ or yeast that also expressed the homologous chimeric light chain gene. Intact F(ab'), and Fab' molecules were recovered by purification from fermentation broths.
Methods are provided for the conjugation of microbially-produced F(ab')~ molecules with protein toxins such as ricin toxin A ch~in to form immunotoxins. These immunotoxins are advanta~eous in that they have reduced levels of heterogeneity caused by proteolytic nicks introduced by standard art procedures for rF(ab') gener~tion. These immunotoxins are also advantageous because they provide high affinity binding and the wo g2/22324 2 1 1 U 7 9 9 ~ PCI`/US92/04976 rF(ab')2 molecules lack Fc receptors which may cause non-specific uptake by macrophages and other cells of the immune system.
The purified rF(ab')2 and rFab' fragments from microbial fermentation have blocked cysteine thiol groups, which required the discovery of methods to unblock the cysteines for Fab' thiol conjugation.
The invention provides reducing agents and conditions that achieve the selective reduction of the cysteine residue(s) nearest the carboxy terminus of Fd, without reducing the interchain disulfide linkage of light chain to Fd. These reduced Fab' fragments are conjugated to other proteins, polypeptides, or chemical moieties reactive with the free thiol group.
The invention provides methods for the conjugation of a reduced Fab' fragment by mixing it with a second po1ypeptide which contains free thiols under sufficient oxidizing conditions to form a disulfide linkage.
The examples show that reduced Fab' fragments can be successfully conjugated to similarly reduced Fab' fragments to form either homodimeric or bifunctional, heterodimeric F(ab'), molecules. Other useful proteins containing free thiol groups can be similarly conjugated to the Fab' fragments to form mixed-function molecules.
The invention atso provides methods for the chemical modification of reduced microbially-produced Fab' fragments to achieve directed conjugation of the activated immunoglobulin fragment. A free thiol of the reduced Fab' fragment is reacted with activating moieties such as dithiobis(pyridine-N-oxide), and the activated Fab' is mixed with a polypeptide containing a free thiol. This results in efficient conjugation to form mixed-function molecules~ such as a Fab'-ricin toxin A molecule.
or a bifunctional heterodimer F(ab')~ molecule.

WOg2122324 PCI/US92/04976 ~U7~!~

BRIEF DESCRIPTION OF FIGURES

Figure l(a) is a physical representation of the structure of IgG.
Shown are the positions of endopeptidase cleavages for papain and pepsin used to generate F(ab')2 and Fab fragments, respectively. The position of both intrachain and intérchain disulfide bonds are shown, as are the protein domains VH, CH1, CH2, and CH3 on the heavy chain and VL
and CL on the light chain.
Figure 1(b) represents the DNA and corresponding peptide sequences of the Fd' modules hinge region used for the production of F(ab')~ and Fab'.
Figure 2 represents the construction scheme for modules containing gene sequences encoding Fd' with one or both inter-heavy chain cysteines. Not drawn to scale.
Figure 3 represents the construction scheme for the gene module encoding ~Fd' with two inter-heavy chain cysteines plus 29 amino acids.
By this construction scheme, the carboxy terminal amino acid is an Asp (Glu is normally at this position). Not drawn to scale.
Figure 4 represents the construction scheme for optimized yeast expression plasmids containing the ~NG~ chimeric light chain and various Fd' genes fused to the PGK promoter (P), invertase signal sequence (S), and PGK polyadenylation signal (T). Not dr,~wn to scale.
Figure S represents the binding inhibition of yeast-derived ING-4 F(ab')2 and of ING-4 F(ab'), generated by pepsin digestlon of ING-4 IgG. The yeast and pepsin-generated ING-~ F(ab'),~ as well as ING-~Fab and IgG. were used to inhibit binding of biotinylated ING-~ IgG to the surface of antigen-positive Hl~9 colon carcinoma cells. Biotinylated IgG was incubated with HT_9 tumor cells in the presence of competing antibody at 4C. Cells were washed and further incubated with avidin-peroxidase at room temperature. The cell-bound peroxidase was ": ~ .

WO 92/22324 2 ~ 9 9 PCl /US92/04976 . ~
visualized with OPD reagent, and its OD490 was used to determine the extent of inhibition.
Figure 6 represents the construction scheme for bacterial expression plasmid containing the gene sequences encoding the H65 chimeric light chain and Fd' chain with one or two inter-heavy chain cysteines. Each gene was fused to the E carotovora pelB ribosome binding site and signal sequence. These were fused to each other and placed under the control of the SalmonelJa ~phimunum araBAD promoter and the trp trAnscription termination sequence in a plasmid containing tetR gene for selection in E. cc)li. Not drawn to scale.
Figure 7 represents the SDS polyacrylamide gel analysis of various chimeric H65 Fab and Fab' molecules secreted from bacteria. Regions of the gel which were scanned by densitometry are denoted.
Figure 8 represents the DNA sequence of pXOM1 (H65 VH).
Shown is the nu!eotide sequence including the ATG initiation codon to the JK/CK junction. Also shown is the predicted amino acid sequence of the region. Shown in bold are the regions where PCR primers bound for amplification of the V-J region.
- Figure 9 represents the DNA sequence of pXOM2 (H65 VL).
Shown is the nucleotide sequence including the ATG initiation codon to the JK/CK junction. Also shown is the predicted amino acid sequence of the region. Shown in bold are the regions where PCR primers bound for amplification of the V-J region.
Figure 10 represents the DNA sequence of the 4A2 kappa V-region. Shown is the nucleotide sequence for the ATG initiation codon to the J~VCK junction. Also shown is the predicted amino acid sequence of the region. Shown in bold are the regions where PCR primers bound for amplification of the V-J region.
Figure 11 represents the DNA sequence of the 4A2 gamma V-region. Shown is the nucleotide sequence for the ATG initiation codon ~.,,,, ,~
, ~
.

W~O g2/22324 2~ 'J~ 9 PCI`/USg2/W976 to the JHtCH junction. Also shown is the predicted amino acid sequence of the region. Shown in bold are the regions where PCR primers bound for amplification of the V-J region.
Figure 12 represents the construction scheme for bacterial expression plasmid containing the gene sequences encoding the 4A2 chimeric light and Fd' chains. Each gene was fused to the E. ~arotovora pelB ribosome binding site and signal sequence. These were fused to each other and placed under the control of the Salmonella ~phimurium araBAD promoter and the llp transcription termination sequence in a plasmid containing telR gene for selection in E. coli. Not drawn to scale.
Figure 13 represents the cytotoxicity mediated by ricin A chain immunoconjugates prepared from H6S antibodies and fragments. The human T cell line HSB~ was exposed to H6S mouse antibody conjugated to ricin toxin A (RTA) chain (-o-), or chimeric H65 Fab', either unconjugated (-A-) or conjugated (-v-) to ricin toxin 30-kd A (RTA30) chain. Cell viability was determined by the percent of leucine incorporated into acid-precipitable radioactivity.
Figure 14 represents the cytotoxicity of resting (panel a) and phytohemagglutimin-activated (panel b) human peripheral blood mononuclear cells. mediated by various H65 antibodies and fragment immunoconjugates. The samples tested were H65 mouse antibody linked by S-methyl-~-iminothiolane to RTA30 (-o-), and chimeric H65 Fab' linked to RTA30 (-O-). IND2 antibody linked to RTA30 (-A-) and RTA30 alone (- O -) were included as additional controls.

DEFINITIONS

The following definitions are supplied in order to provide clarity in the description of the invention.

WO 92/22324 ~ 9 Pcr/us92/o49l~

By the term "V domain" is intended the variable region polypeptide sequence of an immunoglobulin light chain, as shown by Kabat et al., Sequences r Proteins of Immunological Inleres~, 4th ed. (U.S. Dept. of Health and Human Services, NIH) (1987).
By the term "CL domain" is intended the constant region polypeptide sequence of an immunoglobulin light chain, as shown by Kabat et al., supra.
By the term "CHI domain" is intended the first constant region polypeptide sequence of an immunoglobulin heavy chain that is carboxy to the V domain, as shown by Kabat et aL, supra.
By the term "hinge domain" is intended the constant region polypeptide sequence of an immunoglobulin heavy chain that is on the carboxyl side of the CH1 domain, as shown by Kabat e~ al., supra.
By the term "rFab"' is intended an antigen-binding immunoglobulin fragment or its equivalent containing an intact light chain and a truncated heavy chain, linked by an interchain disulfide bond, and which includes at least one cysteine residue in the hinge domain which is carboxy to the light chain-Fd interchain disulfide bond.
By the tçnn "Fab"' is intended recombinantly produced Fab'.
By the term "F(ab') " is intended a dimer of Fab' molecules linked by at least one disulfide bond involving a cysteine residue in the hinge domain which is carboxy to the light chain-Fd interchain disulfide.
By the term "rF(ab')1" is intended recombinantly produced F(ab') By the term "Fv" is intended an antigen-binding immunoglobulin fragment or its equivalent containing only the V domains of light and heavy chains.
By the term "Fd"' is intended the heavy chain of a Fab' molecule.
By the term "RTA" is intended ricin toxin A chain.
By the term "RTA30" is intended the Mr 30.000 form of ricin toxin A.
- :
,~

wo 92/22324 ~ 1 ~ P~/US92/04g76 Recognizing that conventional amino acid numbering is from left to right; and that the amino terminus is conventionally shown on the left, with the carbo~yl terminus on the right; the term "cysteine residue with the highest residue number" is the cysteine residue with the highest amino acid number, or, stated another way, the cysteine closest to the carboxy terminus.
By the term "thiol-containing active moiety" is intended immunoglobulin Fab' molecules, enzymes, polypeptides, radionuclides, and organic or inorganic compounds containing a reactive sulfhydryl group.
By the term "culture medium" is intended a nutritive solution for culturing or growing cells. The ingredients that compose such media may vary depending on the type of cell to be cultured. In addition to nutrient composition, osmolarity and pH are considered important parameters of culture media.
By the term "tumor-associated antigen" is intended a tumor bearing antigen(s) recognized by the Fab' or F(ab'), of the present invention.
Specific examples of tumor associated antigens are disclosed ~n European Patent Application Number 8730600.
: :
DESCRIPTION OF PREFERRED EMBODIMENTS

Various genes which encode immunoglobulin fragments are provided by the present invention. The preferred genes encode both light and heav,v chains, and retain complete variable regions for the light and heavy chains to provide at least an active immunoglobulin Fv binding domain. In addition. the heavy and light chain genes encode additional peptide sequences which have at least one cysteine residue which is not located within the immunoglobulin Fv binding domain. These peptide sequences are prefeMbly a CL (kappa or lambda) region for light chain, , ~

WO 92/22324 ~ ~ 9 9 ~ PCI /US92/049 and a CH1 and hinge region for Fd chain. The genes are preferab]y linked to a secretion signal appropriate for the- host, such as the pectate Iyase B signal peptide for bacterial hosts, or the yeast invertase signal peptide for yeast hosts.
A preferred embodiment of the invention is a host transformed with a complete light chain gene and a truncated heavy chain gene (Fd) which encode an Fab' fragment molecule. These molecules are capable of spontaneous assembly into F(ab')2 fragments. The gene sequences encoding light chain CL and Fd chain CHl and hinge domains may be derived from either human or non-human immunoglobulins of any isotype. Forin Vi~!O human uses, human CL, CHI. and hinge domains are preferred; they are more compatible with the human body than non-human domains. The invention exemplifies the use of human IgGl isotype sequences as the source of CHl and hinge domains For the human IgG1 iso~pe, purification of homogeneous F~ab')~ or Fab' molecules from microbial fermentations is enhanced by including more than one, preferably two, cysteine residues in the Fd hinge region on the carboxyl side of the light chain Fd disulfide bond. A polypeptide tail may - follow on the carboxyl side of the two cysteine residues.
Other immunoglobulin fragments may be generated within the practice of this invention. For example, a selectively reducible cysteine residue may be located on the light chain of an Fab-like molecule by adding a polypeptide sequence to the carboxy terminus of the light chain.
Another embodiment of the invention is the modification of the heavy chain gene so that A partial deletion is used to genera~e a modified immunoglobulin, containing at least an Fab region, and having a selectively reducible cysteine residue in the hinge domain. Another embodiment of the invention is the modification of Fv fragment genes by including sequences encoding ~n additional polypeptide region (or re~ions) on either the li~ht or heavy chain (or both), with such additional wQ g2~22324 2 1 1 ~ 7 9 ~ Pcr/usg2/o4976 polypeptide (or polypeptides) encoding a selectively reducible cysteine residue (or residues).
The F(ab')2 and Fab' fragments of the present invention may be produced from a variety of host cells. Preferred hosts are bacteria and yeast. Vectors are exemplified for the bacterium E. coli, and the yeast 5.
cere~nsiae. Other hosts may be utilized in the practice of this invention, including gram-negative bacteria such as Salmonella nftphimlmum or Serratia marcescens, and various Pseudomonas species; gram-positive bacteria; other yeasts and fungi, and plant, insect, and animal cells.
Preferable features for hosts include the ability to be practically grown in industrial fermenters and bioreactors, and the capability of the secretion of intact immunoglobulin fragments. A method for the preparation of F(ab')2 and Fab' fragments is the culturing of host cells on culture media followed by isolation of active fragment from the fermentation broth, preferably after removal of cells from the broth.
Reducing agents useful for the selective reduction of the cysteine residues used for conjugation include dithiothreitol, cysteine, beta-mercaptoethanol, and the like. As shown in the examples, different , concentrations of reducing agents may be required to achieve the desired selective reduction. For F(ab'), molecules secreted from bacteria or yeast where the Fab' molecules are joined by a single disulfide bond, ~.0 mM
cysteine selectively reduces the single heavy ch~in-heavy chain disulfide and is preferred. However, ~.0 mM cysteine is insufficient to reduce two hinge region heavy ch~in-heavy chain disulfide bonds. which require approximately 5.0 to 15.0 mM cysteine for selective reduction.
For Fab' molecules secréted from bacteria or yeastt the desired hinge region cysteine residue (or residues) are blocked by small molecular weight adducts. These residues may be unblocked by reduction with dithiothreitol at concentrations of about 0.1 to ~.0 mM~ or by cysteine at concentrations of about 5.0 to 15.0 mM.

, ~ ~
, .

wog2/22324 2i1~99` PCI/US92/0497~

The selectively-reduced Fab' fragments can be conjugated to useful thiol-containing moieties such as enzymatic and non-enzymatic polypeptides, Fab' fragments, radionuclides, and other compounds. In one method, selectively-reduced Fab' molecules are placed in oxidizing conditions to form disulfide-linked F(ab')~ fragments. In a second method, the Fab' molecule is reactèd by disulfide exchange with an acthating compound such as dithiobis(nitrobenzoate), dithiobis (pyridine-N-oxide) or the like, creating an excellent leaving group for directed disulfide formation. The activated Fab' is then reacted with a thiol-containing moiety for the formation of heteroconjugates such as a heterobifunctional F(ab')~ or a Fab'-enzyme.
An alternate method for heteroconjugate form~tion is the reaction of the reduced Fab' molecule with linker compounds which have functional grou~s reactive with Fab' thiols such as maleimide and the like, to form a Fab'-linker conjugate. The Fab'-linker conjugate is then reacted with a useful active moiety to form a heteroconjugate. Generally, any sulfhydlyl linking compound may be utilized. For example, the linker compound may have an S-acetyl functional Proup. The Fa~-linker, S-ace~l, is reacted with a thiol-containing enzyme or other polypeptide which has been activated with an appropriate compound, such as dithiobis (nitrobenzoate) or dithiobis(pyridine-N-oxide). Such a heteroconjugate has a disulfide linkage to the active moiety. For Fab'-enzyme heteroconjugates with cytotoxic enzymes such as ricin A chain, a disulfide linkage is preferred for maximal cytotoxic activity. For other uses, a linker compound which forms other than a disulfide linkage to the active moiety may be used. Such uses include. for example. a thioether linkage to enhance stability of a Fab'-conjugate.
Both homodimeric and heterodimeric F(ab')~ molecules are provided by the invention. Homodimeric F(ab'), are preferred when a single specificity with bifunc~ional binding is desired. Bispecific, ~o g2/22324 ~ 1 1 u 7 ~ ~ PCI/US92/04976 heterodimeric F(ab'), fragments are preferred when the separate bindiDg functions of heterobifunctional antibodies are desired. The heterodimeric F(ab')2 are preferred over known bispecific antibodies in their properties of a smaller molecular weight and the deletion of the Fc region, which can be advantageous when better tissue penetration and minimization of Fc-receptor cell interactions are desired. There are different methods provided by this invention for the formation of heterodimeric F(ab')~
molecules. In one method, F(ab')2 or Fab' with different specificities are first reduced to monovalent Fab' forms. They are then either mixed and oxidized to form heterodimeric F(ab'),~ or are reacted with linker compounds known in the art and subsequently mixed and reacted to form F(ab')2. Another method is to use the hosts and vectors of this invention to separately express the genes encoding the two different Fab' molecules within the same host cell. Heterodimeric F(ab'), may then be purified from the fermentation culture.
Enzymes may be conjugated to microbially-produced F(ab'), and Fab' molecules to form immunoconjugates with therapeutic or diagnostic use. Examples of therapeutically useful enzymes include pr~otein toxins, such as the ribpsome-inhibitor ricin A chain~ to achieve therapeutically targeted killing of cells, and other enzymes such as alkaline phosphatase.
to achieve therapeutic effects by prodrug conversion into active drug, Pseudomonas toxin, Diphtheria toxin, and Tumor Neucrosis Factor (TNF). Such F(ab'), or Fab'-enzyme conjugates can also be used in in vit~o diagnostic assays to convert a substrate into a detectable form, in a similar way as immunoassays known in the art.
The F(ab'), fragments or activated Fab' fr~gments of the invention may be conjugated to non-enzymatic polypeptides that interact with cellular receptors. such as interleukin-~. epidermal growth factor~
immunoglobulin Fc re~ions. and the like. Such molecules could be useful in activating cells to accomplish desired effector functions. such as the ...... .. ... ..

wog2~22324 2 1 ~ U 7 g 9 PCI/USg2/04976 selective activation or killing of targeted cells, and thereby achieve a therapeutic e~fect.
F(ab'), fragments may be conjugated to enz~nnes and other polypeptides by current art methods that rely on the derivatization of amino acid residues with linker compounds such as N-succinimidyl 3-(2-pyridyldithio)propionate (SPDP), or preferably sterically hindered linkers such as the substituted 2-iminothiolanes (Goff e~ al., Bioconjugate Chem.
1: 381-386 (1990)). Other examples of linker compounds may be found in U.S. Patent No. 4.970,303. The derivatized F(ab')~ is then reacted with a thiol-containing polypeptide to form a stable conjugate.
The number and placement of polypeptides or enzymes which may be conjugated to the cysteine thiol(s) of a Fab' molecule is limited by the number and placement of selectively reducible cysteine residues. The preferred location of cysteines is away from the variable regions which define and accomplish the Fab' binding activity. The number of selectively reducible cysteines is from 1 to about 10, preferably 2, to achieve targeted conjugation and avoid interference of the enz~nnatic activity with the binding activity. ,~
A functionally derivatized F(ab')~ or a selectively-reduced Fab' may also be conjugated to chemical moieties (radionuclides and organic compounds) that confer a desired second function on the Fab'. Such chemicals include cvtotoxic compounds such as trichothecenes, and metal-che]ating compounds such as diethylene triamine pentaacetic acid (DTPA), daunorubicin, doxorubicin, me~hotrexate. Mitomycin C and others that are known in the art. S~e Goodman el al., Goodman and Gilman's THE PHARMACOL051CAL BASIS OF THERAPEUTICS~
7th Ed., Macmillan Publishing Co., (19S3). Examples of radionuclides include -1-Bi, 13~ Re, and 90Y~ which list is not intended to be exhaustive. Also. a selectivelv-reduced Fab' may be directly bonded to 2il~7g9 certain diagnostic or therapeutic radionuclides, such as 99Tc or 186Re, by methods that are known in the art.
It is further recognized that in-frame fusions, between the 3' end of the Fd or Fd' gene and other usef;ul sequences may be constructed.
Having generally described the invention, a further understanding can be obtained by reference to certain specific examples which are provided herein for the purpose of illustration only and are not intended to be limiting unless otherwise specified.

EXAMPLES

Example 1: Construction of Fd' Gene Modules Cellular production ot F(ab')~ or Fab' molecules requires the co-expression of genes encoding the light chain and a heavy chain fragment designated as Fd'. The human IgG1 Fd' fragment contains at least one of the two c3~steines involved in heavy chain interchain disulfide bond fonnation (Figure la).
Six different Fd' gene modules were constructed. The DNA and corresponding peptide sequences at the carboxy terrninus of these modules are shown in Figure lb. The first of these (Figure lb-A), encoding an Fd' with a single inter-heavy chain cysteine was constructed as olltlined in Figure 2, generating the plasmid plNG1624. This construction makes use of the fact that the Fd 8ene (on pING1412) previously engineered for Fab production contains a unigue Bc~I site at the stop codon which had been introduced in the DNA sequence encoding the first of two interchain cysteines. Consequently, digestion of plNG1412 with BclI. followed by treatment with mung bean nuclease, leaves a blunt end consisting of a TA base pair. Fusion of the linker:
, .
~ ~ 5'-GTCCACCATGATCACTCGA-3' ;, WO 92/22324 2 ~ ;1 U 7 9 9 PCr/US92/0491~6~

3'-CAGGTGGTACTAGTGAGCT-5' to this blunt end regenerates the codon (TGT) for the first of two cysteines in hinge plus the codons (CCA) for the two prolines that follow.
Association of this Fd' with a light chain will result in a Fab' molecule with a single Cys carboxy to the light chain-Fd disulfide or a F~ab')2 with a single disulfide bond between the two Fab' moieties. As with the construction of the Fd gene module for Fab production, a BclI site was incorporated at the stop codon to facilitate subsequent gene fusions.
Two Fd' modules (Figure Ib-E~ and C) were constructed using pING1624 as a starting point and the same strategy used for construction of plNG1624. These modules contained both of the cysteine codons for heavy chain-interchain disulfide bond formation plus codons for either t~vo or nine additional amino acids 3' to the cysteine codons (Figure 1) and were designated pING1~73 and plNG1684, respectively (Figure 2c).
A fourth Fd' module (Figure Ib-D) was constructed as outlined in Figure 3. This module (plNGI695, Figure 3c) encodes both inter-heavy chain cysteines plus 29 additional amino acids 3' to the cysteines.
Two additional Fd' modules (Figure lb-E and F~ were also constructed. In these modules. the first of two inter-heavy chain cysteines was changed to serine by site-directed mutagenesis. These novel modules were constructed as part of an attempt to decrease the heterogeneity of Fab' molecules containing a single interchain tail and secreted from microorganisms as described in Example 3. All of the above Fd' gene modules contain unique Bc/l sites at the stop codon.
The approaches described above can be generally applied to accomplish the constructioniof in-frame fusions between the 3' end of the Fd or Fd' gene modules and other useful gene sequences. Examples of such constructions include various other types of truncated heavy chain modules and fusions between Fd or Fd' and a variety of molecules of diagnostic, therapeutic or experimental importance~ such as ~ . ": ~
.,,"~

~1VO g2t22324 PCr/US92/04976 immunoglobulin heavy chain domains, receptor-binding proteins, or cytotoxic polypeptides.

Example 2: Secretion of Functional F(?~b')~ and Fa~b' bv Yeast Yeast cells are capable of secreting functional Fab and IgG
molecules such as mouse-human cbimeric IgG and Fab molecules (Holwitz et al., sl~pra). Secretion of IgG is accomplished by co-expressing genes encoding the mature forms of light chain and heavy chain each fused to yeast invertase signal seguence and PGK promoter and polyadenylation si~nals. Fab secretion is accomplished by co-expressing light chain with a genetically en~ineered Fd chain containing a stop codon introduced at the gene sequences encoding the first of two cysteines responsible for IgG1 heavy chain interchain disulfide bonds. As for the light chain gene~ the Fd chain gene is fused to the yeast invertase signal sequence-PGK promoter and polyadenylation signal.
In the present example, yeast serves as a host for the production of mouse-human chimeric F(ab')~ and Fab' molecules. The chimeric antibody fragments in this example contain the light and heavy chain variable re~ions of a monoclonal antibody designated Me4. which binds to an antigen expressed on the surface of cells from many melanomas and carcinomas. The chimeric version of Me~ is designated as ING-4 and the production of ll~G-~ IgG1 by mouse Sp/20 cells and of Fab by yeast and bacterial cells have been previously described (Better e~ a/., PCr US8903~52).

a. Yeasl strains and ~ro~h conditions S. cerelisiae strain PS6 (ura3 leu~ MATa) was developed and çubsequently used as a host for yeast tr~nsformations performed as WO92/22324 ~ 7 g`~ PCl/US92/04976 described by Ito e~ al. J. Bacteriol. 153: 163-168 (1983). Yeast transformants were selected on SD + leu agar (2% glucose, 0.67% yeast nitrogen base, 2% agar) and grown in SD + leu broth buffered with S0 mM sodium succinate, pH 5.5.

b. Construction of yeast expression plasmids containing antibody genes Production of F(~b')~ or Fab' molecules by yeast requires the simultaneous production of both the light chain and Fd' chain proteins.
This can be accomplished by co-transforming a leu2- and ura3- strain with plasmids containing the light and Fd' ch7in genes with the leu2 and ura3 genes, respectively. Optimal production is achieved by placing both the light and Fd chain genes on the same plasmid. Accordingly, the Fd' gene module from plNG1624 (module A, Figure 1 ) described in Example 1 was ligated to a ura3 expression vector to form plasmid pING1636 (Figure 4b). The yeast expression plasmid plNG1697 was then constructed as shown in Figure 4c. plNGl697 contains the gene sequences encoding the mature forms of the ING-~ chimeric light chain plus the chimeric Fd' chain module A gene fused to tlle DNA sequences encoding the invertase signal sequence and the PGK promoter and polyadenylation signals.
Using this method, the plasmids pING1673, pTl~GI684 and plNG1695 were used to construct e~pression plasmids pINGl698 (module B).
plNG1800 ~module C), and plNG1699 (module D).

c. Yeast secretion of c.himeric ING-4 F~ab') and Fab' The plasmids plNG1697, pING1698, plNG1699, and plNG1800 were transformed into S. ~ere~ e PS6 by selection for Urat colonies in SD+ leu agar. Transformants were grown in SD broth lacking uracil and the levels of secreted F(ab')~ or Fab' assessed by EL~SA. The cultures WO 92/22324 2 1 1 U 7 '~ 9 PCI/US92/04976 secreting the highest levels of Fab' (pII~G1697, 0.6 ~ug/ml; pING1698, 1.9 l; pING1699, 1.9 ~g/ml; pING1800, 2.5 ~Lg/ml) were grown in 10 liters of SD broth for 60 hours and F(ab') and Fab' proteins were purified from the culture supernatant.

d. Isolation of chimeric F(ab')~ and Fab' from yeast and production of F(ab'), from chimeric IgG

A mixture of F(ab'), and Fab' was purified from 10 liters of culture supernatant. The culture supernatants were first concentrated by a DC10 concentrator (Amicon) using a SlOY30 cartridge (Amicon), washed with 20 liters of distilled water, reconcentrated, and then washed with 10 mM sodium phosphate buffer at pH 8.0, and concentrated again.
The concentrate was then loaded on a DE5~ (Whatman) column pre-equilibrated with 10 mM sodium phosphate buffer at pH 8Ø The flow-through from the DEAE column was collected and adjusted to a pH of 6.8 and a conducti~i~ity of 1.0 ms/cm. The sample was next loaded onto a CM52 column. equilibrated with 10 mM sodium phosphate at pH 6.8.
The CM52 column was eluted with ~5 mM NaCl in 10 mM NaP04, pH
6.8. The eluant was collected and diluted with water to a conductivity of 1.0 ms/cm and lo~ded onto a second CM5~ column equilibrated with 10 mM sodium phosphate at pH 6.8 and the bound antibody was eluted with a linear salt gradient from O to ~5 mM NaCl in 10 mM sodiurn phosphate, pH 6.8. The fractions were analyzed by ELISA and SDS-PAGE and those that contained a mixture of F(ab')~ and Fab' were pooled and concentrated. The F(ab'~ and Fab' were purified away from eac}l other using a TSK-1'5 gel filtr~tion HPLC column. The purified Fab', and ~(ab')7 ran at -~8 Kd ,and ~ 100 kd, respeetively, in non-reducing SDS
polyaclylamide gels. Samples from both the F(ab'), and Fab' fractions resolved into light chain and Fd chain bands on reducing SDS
polyac~ylamide gels. During the purifications. it was observed that those WO 92/22324 ,~ 7? 9 9 PCI/US92/04976 constructs which contained two hinge cysteines carboxy to the light chain-Fd disulfide bond (plNG1698, pING1699, and pING1800) had a lower level of heterogeneous forms than the construct with the single cysteine (plNG1697), as evidenced by protein bands at other than ~48 kd and ~ 100 kd on non-reducing SDS polyacrylamide gels.
F(ab')2 was generated by pepsin digestion of ING-4 IgG as follows.
25 ul of S0% slurry of immobilized pepsin (Pierce) was equilibrated with 400 ~l of digestion buffer (20 mM sodium acetate, pH 4.5), and centrifuged at 1000 x g, 5 min. The immobilized pepsin was resuspended in S0 ~l of digestion buffer. 50 ~l of ING-4 (1 mg) was added to the pepsin suspension and incubated 4 hours, 30C on a shaker. The digested IgG was extracted by adding 1~0 ~l of 10 mM Tris, pH 7.5 followed by centrifugation at 1000 x g, 5 min. The supernatant, containing F(ab')2, was separated from undigested IgG and Fc fragment by passage through a 1 ml protein A-sepharose column. SDS-PAGE analysis revealed the presence of F(ab'), plus two lower molecular weight bands. F(ab'), was further purified on a TSK-125 HPLC column.

.
e. Binding characteristics of F(ab'), and Fab' secreted by yeast . --The purificatior; from yeast culture supernatant of proteins of the expected size of F(ab')~ and Fab' sug~ests th~t yeast secrete correctly associated molecules. Direct and competition binding assays were used to confirm that these molecules are functional in binding ~o antigen. In direct binding assays, the various chimeric ING-4 F(ab'), and Fab' molecules bound to the same target cell ~s the chimeric ING-4 IgG and Fab (data not shown). In a competiti~e bindin_ assay using Hl~9 cells and biotlnylated ING-~ IgG. the yeast-produced pING1800 F(ab'), protein containing two interchain cysteines was equivalent to the chimeric lNG-~ IgG ~Figure 5). The F(ab')~ proteins from plNG1698 and W~) 92/22324 ~ l 1 U 7 ~ ~ PCI`/US92/04976 plNG1699 were also tested; their competitions of biotinylated ING-4 binding were also equivalent to ING-4 IgG (data not shown). By contrast, the competition of F(ab')2 prepared by pepsin digestion of ING-4 was equivalent to ING-4 Fab, rather than to IgG (Figure 5), suggesting that pepsin digestion adversely affected the binding characteristics of the F(ab')2. Monovalent Fab' proteins containing either one (pING1697) or two (plNG1698, pING1699, pI~G1800) Fd' interchain cysteines competed in a manner similar to that of monovalent ING-4 Fab (data not shown).

Example 3: Secretion of Functional H6~ F(ab')2 and Fab' bv E.scherichia coli - Bacteria such as E. coti are capable of secreting functional mouse-human chimeric Fab (Better ~l al.~ supra). In the present example, E. coli serves as the host for the production of mouse-human chimeric F(ab')~
and Fab' molecules containing Fd' modules with either one or two inter-heavy chain Fd' cysteines in the hinge domain carboxy to the cysteine that normally forms the light chain-he~vy chain disulfide bond (see Figure lb).
The gene modules used for these experiments include Fd' module C
(Figure lb) encoding two inter-heavy chain cysteines plus nine additional , .
amino acids on the carboxyl side of the l~st cysteine and several different Fd' modules A, E. and F (Figure Ib), with one inter-heavy chain cysteine.
The chimeric antibody fragments in this example contain the light and heavy chain variable regions of a monoclonal ~ntibody, designated as H65.
which binds to the CD5 antigen on human T cells (Kernan el al. J.
Immunolo~v 133: 137-1~6 (198~)3.

a. Dicistronic expression system for ligllt chain and Fd' genes ,~ .
Secretion of F(ab')~ or Fab' by bacteria requires the simultaneous ~: expression of the ~enes encod:ng both the li~ht chain and Fd' chain each .:, WO92/22324 ~ 9 PCl/USg2/049 fused to a bacterial ribosome binding site and signa! sequence. This is optimally achieved by fusing both genes to each other and placing the dicistronic operon under the control of a strong, inducible promoter. This example describes a system using the pelB signal sequence from Erwinia carotovora (Lei et al. J. Bac~enol. 169: 4379-4383 (1987)) and the araBA;D
promoter from Salmonella lvphiml~rium (Horwitz e~ al. Gene 14: 309-319 (1981)) to produce chimeric H65 F(ab')~ and Fab' in E. coli.
For production of H65 F(ab')2 and Fab' containing Fd' modules A, C, E, and F (see Figure 1), the expression vectors plNG3217, plNG3219, plNG3518, and plNG3519 were constructed as shown in Figure 6 for plNG3217.
To provide the variable-region sequences. H65 hybridoma cells secreting a mouse IgGl. kappa (Kernan et al., sllpra) were used for RNA
isolation and cDNA preparation. After determination of heavy chain and light chain sequences, oligonucleotides were designed, synthesized, and used to prime the amplification of the VH-JH1 and V~-Jkl coding sequences by polymerase chain reaction (PCR) using standard methods ~: (PCR Pr~ocols, Innis. ed., Academic Presst (1990)). The PCR-amplifiedV gene modules were ligated to Sacl-digested plasmid pUC18 to generate pXOM1 (VH) and pXOM? (VL~. Figures 8 and 9 show the DNA
sequences of pXOMI and pXOM2. Subsequently, new primers were designed, synthesized and used to amplify the V region sequences encoding the fully processed VH and VL domains. The PCR primers were designed so that a blunt end would be present at the 5 end to join to a pelB leader peptide-encoding sequence prepared by treatment with Sstl-restriction endonuclease and T4 polymerase to generate a b1unt end at its 3' end. The PCR primers were also designed to include a BstEII
site (JH1) or a Hindlll site (Jkl) at the 3' end of the V region to match those of the plT106 expression vector of Better ~I al.. supra. and Robinson al., PCT US880 '51~. The primers used for VH PCR amplification are , :
; ~ , ,.-:

WO 92/22324 ~ .1.1 i3 7 ~ 9 Pcr/usg2/o4976 H65G1 (5'-AAC ATC CAG TTG GTG CAG TCr G-3') and H65G2 (5'-GAG GAG ACG GTG ACC GTG GT-3'), and for VL amplification were H65K1 (5'-GAC ATC AAG ATG ACC CAG T-3') and JK1-HindIII (5'-GTI TGA l~T CAA GCT TGG TGC-3').
The Fd' gene module A from Example 1 was used to assemble first pXlSF, then plNG3217 (Figure 6). Fd' gene module C was used in the same way to construct pING3219. For pING3518 and pI~G3519, Fd' gene modules E and F were generated by PCR gene amplification using Fd' gene modules A and C as templates. Synthetic oligonucleotides to introduce the desired sequence changes were synthesized and used in a PCR reaction to generate Sall toX71~1 Fd' gene fragments for ligation to DNA fragments from the chimeric Fd' vector pXlSF. The remainder of the assembly is the same as for pING3217 (Figure 6).

Descriptions of other starting plasmids in Figure 6 are:
1. plNG1500 is identical to pRR187 described in Figure 38 of Robinson el al., PCr US880~514. except for the insertion of aXlloI linker oligonucleotide into the unique SalI site.
2. p~NG3215 is identical to pING1500 except that it has the Fd' gene module A (Example 1).
3. plNG3104 is described in Figure 13 of Better et al., PCT
US8903852.
4. pS2D contains the human Ck HindlII to X7~I sequence present in plNG1431 described in Figure 27 of Robinson e~ al.. PCT
US880251~.
5. pLE10 contains tlle human CH1 BstElI to BstEII fragment of pIT106 described by Better e~ al., .s~pra.

WO 92/22324 ~ ,~ 9 9 PCl`/US92/0~9~7.

b. Production of chimeric H65 F(ab') and Fab' in bacteria The plasmids pING3217, pING3219, plNG3518, and pING3519 were transformed into E. cc~li strain E104. These strains were cultured in a Chemap 14-liter fermenter in 10 liters of minimal medium supplemented with 0.7% glycerol as a carbon source and induced by adding arabinose gradually over a period of 16 hours to a final concentration of 0.2% when an A600=100 was attained. The cells were incubated for 32 hours following the initiation of induction.
F(ab')~ and Fab' were purified from 10 liters of culture supernatant using a process simil?lr to that described for the yeast Fab'.
The fermentation broth was passed through a 0.2 ,I~M filter and concentrated five-fold by ultrafiltration (Amicon YM10 membrane). The media was replaced with 10 mM sodium phosphate buffer, pH 6.8, by diafiltration. After passing through a second 0.1 ~M membrane, the concentrate was bound to a CM cellulose column and eluted with 0.10M
sodium chloride in pH 6.8 sodium phosphate buffer The eluate is concentrated by ultrafiltration. and F(ab')~ and Fab' fra,gments are separated by size exclusion chromatography. The concentratep eluate containing no more than 500 mg total protein is loaded onto a 3.2 cm .
diameter x 120 cm high Sephacryl-S200(~ column preequilibrated in pH
7 d, sodium phosphate buffer. The sample volume is less than 2.5C~ of the total column volume. The resulting protein fractions were analyzed on a non-reducing SDS polyacrylamide gel stained with coomassie b]ue. The Fab' fractions containing the two selectively-reducible cysteine/Fd' module C (pING3219) resolved into the expected Fab' b~nd plus small-molecul~r-wei~ht proteolytic digestion products of the li~ht and Fd ch~ins (Figure 7.
lanes 5 and 6). The F~b' band comprised greater than 50'~c of the relative area within these lanes as measured bv densitometry scans of the coomassie blue-stained gel (.fC~' Table 1 below) and could be re7dily ~' :

j~ ~ f '- !' ~ 9 separated from the small-molecular-weight light and Fd' chain fragments by standard chromatographic methods.
The various Fab' proteins containing the single selectively reducible cysteine Fd' modules A, E, and F (plNG3217, pING3518, and pING3519) also resolved into the expected Fab' band and the proteolytic digestion products of unassociated light and Fd chain bands (Figure 7). Depending on the isolate, the Fab' bands comprised 30-40% of the relative area - within each lane as determined by densitometry scans of the Coomassie blue-stained gel (Table l ).

¦ Table 1 SDS PAGE/Densitometry Analysis of Purified Bacterial Fab' ¦Percent of Lane Total(b) ; ¦ PlasmidI Lane I Peak 1 I Peak 2 I Peak 3 I Peak 4 .
pING3217 2 ND(a) ND ND ND
_ ~ ~plNG3518 3 15 41 21 22 -~ ~NG3519 4 28 31 20 2~
plNG3219 5 -- 57 8 2S
plNG3219 6 1 56 3 35 . -(a) ND = not done (b) Values are reported as the percent of the total densitorneter scan of each lane of the SDS gel shown in Surprisingly. these preparations contained three additional bands not observed with the Fab' containing the two cysteines (Figure 7, lanes '.
3 and 4). Densitometry scal7s of these lanes of the gel established that the additional band~ comprised approximately ~oc~ Of the relative area ~- (Table l). N-terminal amino acid analysis established that the largest of these bands consisted of li~ht chain, while the other two consisted of two , ~ , WO92/22324 ~ 99` PCI/US92/0497 different forms of processed Fd chain. The purification of Fab' protein was unusual and problematic because intact Fab' could only be separated from these additional non-disulfide-bonded forrns by treatment with guanidine hydrochloride followed by urea exchange prior to separation by ion exchange chromatography. Further experiments established that only the light chain component of the three bands contained a selectively-reducible thiol available for conjugation with RTA-301 suggesting that the Fd' bands contained no selectively-reducible thiol groups. These results demonstrate that, unexpectedly, human IgG1 Fab' constructs with a single hinge cysteine carboxy to the cysteine normally bonded to light chain result in the production of a mixture of non-covalently associated Fab' as well as Fab' with correct disulfide bonds.
The F(ab')~ fractions gave the expected higher molecular weight band, indicating the presence of properly forrned, bivalent F(ab'),. As found in Example 1, there was a higher yield and less heterogeneity observed for F(ab')~ from the two cysteine plNG3219 construct than from any of the single cysteine constructs.

- ~ ~ c. Bindin characteristics of H65 Fab' and F(ab')~ produced bv E. coli The bacterially-produced Fab' containing the single selectively-reducible hinge cysteine (pING3~17) was assessed for function in a competitive binding assay using Molt-4 cells and FlTC-labeled H65 IgG.
In this assay. CD5+ Molt ~ cells (10~ ml) were mixed with varying concentrations (1 to 4 ~g/ml) of the H65 IgG or Fab' together with a fixed concentration (0.~ ~g/ml) of H65-FITC. Following a 1-hour incubation at 4C in the dark. cell-associated H65-FITC was measured by flow cytometry. Binding of the Fab' fragments relative to H65 IgG was then quantitated by comparing the slopes of the competitive binding culves for each sample. Intact H65 IgG would therefore gjve a value of WO 92/22324 2 ~ 1 ~ 7 9 9 PCl/US92/04976 100Yo in this assay. On a weight basis, H65 Fab' exhibited a binding affinity equivalent to that of the parental murine H65 IgG. On a molar basis, this value is roughly 30% that of the parental IgG. Identical results were obtained with the Fab' containing two selectively reducible hinge cysteines. In separate experiments, E. coli-produced H65 F~ab')2 was found to have approximately 80~o of the binding affinity of the parental murine H65 IgG, as measured on a molar basis.

Example 4: Secretion of Functional 4A2 F~b' bv E. c~li Bacterial expression vectors for 4A2 Fab' were constructed in a similar manner to those for H65 Fab' (Example 3). In this example, the mouse hybridoma cell line producing the 4A2 antibody served as the starting point for isolation of the antibody genes.

a. Construction of expression vectors One liter of hybridoma cells which produce antibody 4A2 were collected and polyA RNA was prep~red. A cDNA library was prepared from this RNA. and individual cDNA clones containing the full length light chain (p~A2K-13) and heavy ch~in (p4A2g-12) were identified by hybridization to mouse const~nt region probes. The DNA sequence of each variable region was determined and is shown in Figures 10 and 11.
The junction region of the 4A2 light chain (k~ppa) was JK1 and that of the heavy chain was JH3. Specific primers were used to PCR amplify the variable and;junction (V-J) segments from each of the genes from p4A2K-13 and p4A2g-12 for subsequent cloning into expression vectors. The N-terminal amino acid sequence for both the light and heavy chain proteins was also delermined to assure that the correct genes were identified and .

WO 92/22324 2 1 1 ~ 7 9 9 PCI/US92/049~7~

used for subsequent cloning steps. For cloning into bacterial expression vectors, PCR amplification of the light chain V-J region was with primers:
5'-GACATl GTGCI`CACCCAATC-3' and 5'-GTl~GATl lCAAGCl~GGTGC-3', hile the heavy chain V-J region was amplified with:
5'-GAAGTGCAGCI`GGTGGAGTC-3' and 5'-GAGACGGTGACCAGAGTCCCr-3'.

The DNA fragment containing the light chain V-J region was digested with HindIII and cloned as ~ blunt to HindlII fragment into pING1500 along with the human k~ppa constant region to generate pZlG. Similarly, the DNA fragment containing the heavy chain V-J
region was digested with B.slEII and cloned as a blunt to BslEII fragment into pING3215 along with a Bs~EII fragment from CH1 to generate pD28H. Plasmids pZlG and pD28H were used to assemble the 4A~ Fab' expression vector, pING3218. This cloning scheme is outlined in Figure 12.
Plasmid p~NG3218 contains Kappa and Fd', where the C-t~rminus of Fd' is ~s shown in Figure 1 gene module A. A derivative of this plasmid, plNG3197~ was constructed that has an Fd' C-terminus as shown in Figure 1, gene module C. Pl~smid plNG3197 was derived from pZ1G
and pD28H where the genes were assembled in the order kappa followed by Fd (opposite order to pING3~18), and the Fd' sequence illustrated on Figure 1 gene module C w~s introduced as a Saul ~oX~oI fragment from plN(i3~19 (Example 3).

WO 92/22324 2 1 1 ~ 7 9 `9 PCr/US92/04976 b. Production of chimeric 4A2 F(ab')2 and Fab' in bacteria Plasmids pING3218 and pING3197 were transformed into E. coli strain E104. These strains were cultured in a Chemap 14-liter fermenter in 10 liters of minimal medium supplemented with 0.7% glycerol as a carbon source and induced by adding arabinose to 0.05% when an A600=50 was attained. The ce]ls were incubated for 24 hours following induction.
F(ab')~ and Fab' from either plNG321~ or plNG3197 were purified from 10 liters of culture supernat~snt using a process similar to that described for the H6~ F(ab'), and Fab' from either yeast or E. coli.
The fermentation broth was passed through a 0.1 ~M filter and concentrated by ultrafiltration (Amicon SlOYIO membrane) and adjusted to pH 6.~. This concentrate was loaded onto a CM52 column at pH 6.S
in 10 mM phosphate buffer. The CM52 column was eluted with 0.02 N
NaCl, and concentrated to 50 ml. SDS-PAGE and Western blot analysis of the concentrate from plNG3218 (two hinge cysteines) fermentations showed three major bands containing light chain: F(ab') " F,ab', and free light chain. However, plNG3197 (1 hinge cysteine) gave about~7 major bands, indicating a higher degree of heterogeneity of products than obtained from p~G3218. This material was loaded in b~tches to a CBX
PreplS HPLC column and eluted with a O to 0.15 N NaCl gradient in 10 mM phosphate pH 6.S. The purified F(ab'), and Fab' are prepared by pooling appropriate fr~ctions, as assessed by SDS-PAGE.

Example~ 5: Est?blishment of Conditions for Reduction of F(ab'!, The Fab' molecules described in Examples '~ 3~ and 4 should contain cysteine thiol groups which can be used for in vitro coup~ing to a variety of molecules. Initial attempts to couple two Fab' molecules to WO92/22324 PCI/US92/049~6 i 7 ~3 9 ` 3~
_ form a F(ab')2 were unsuccessful, suggesting that the cysteine thiol groups on these molecules were blocked, possibly due to adduct formation. This hypothesis was confirmed by testing with Ellman's reagent.
In the present example, we establish conditions for the selective reduction of the Fd cysteine residue(s) on the carboxyl side of the Fd-light chain disulfide bond. Yeast-derived chimeric ING-4 F(ab');~ containing two inter-heavy chain cysteines plus nine additional amino acids (see Figugre 1(b)(C) and E. coli-derived chimeric H65 Fab' containing a single hinge chain cysteine carboxy to the Fd-light chAin disulfide-linked cysteine were used to establish the reduction conditions.
ING~ F(ab')~ produced by yeast and purified as described in Example 2 was incubated for 4 hours at 4C in 20 mM Tris-HCI, pH 8.0 containing dithiothreitol (Dl~) at concentrations from 0.05 to 1 mM.
The proteins subsequently were analyzed by SDS-PAGE, followed by coomassie blue staining. A DTl concentration of 0.5 mM was sufficient to reduce the F(ab')~ molecules to Fab' without affecting the Fd-light chain disulfide bond. DTT concentrations of 1 mM or higher reduced the F(ab')2 molecules to individual Fd and light chains, as asses~ed by SDS-. ~
PAGE.
`` - The h6s Fab' produced by E. c~li was tested as above and also .
required 0.5 mM 1~ in 20 mM Tris-HCl, pH 8.0 to selectively reduce the single hinge cysteine neArest the carboxy terminus of Fd without affecting the Fd-light chain disulfide bond. Following removal of excess DlT by gel filtration in O.lN acetic acid Ellman's reagent was used to establish that there were approximately 1.~ thiol groups per Fab' molecule. ' ' . .

;,.
. -~
", , ...

," ~

WO 92/22324 PCr/US92/04976 33~ 7~9 .
Example 6: Formation of F(ab')2 from Fab' Fab' molecules with selectively-reducible thiol groùps can be used to form either homodimeric or bifunction~l heterodimeric F(ab')2 molecules. Homodimeric Fab' molecules should retain the a~inity of an IgG molecule. Heterobifunctional F(ab'), molecules could include specificities toward two different cell types or a cell and a ligand.
In the present example. chimeric ING-2 and ING-4 Fab' molecules containing Fd chains with one or two cysteines at the C terminal side of the Fd-light chain disulfide bond are used for the production of homodimeric and bifunctional. heterodimeric F(ab') molecules.

I. Production of Homodimeric F(ab')~ Molecules a. Forrnation of F(ab'), from Fab' ING-4 Fab' molecules containing an Fd chain with either one or two cysteines on the carboxyl side of the Fd-light chain disulfide bond (see Example 1) were used to form F~ab') Adducts associated with these additional Fd cysteines were removed by incubating 50 ,ug of the various Fab' proteins in 20 mM Tris-HCI, pH 8.0 containing dithiothreitol at 0.5 mM Dl~ in a volume of 50 ~I for 4 hours at 4C. The reduced Fab' proteins were then diluted to 20 ~g/ml into a cold (~C) aqueous solution comprised of 2 to 5 mM cysteine in 50 mM Tris-HCI pH 7.8 and incubated overnight at 4C to allow reassociation to occur. The solutions were then concentrated by Centricon(E~) 10 to a ~olume of a~proximately 50 ~1 and F(ab'), formation was quantitated bv densitometric scanning of coomassie blue-stained bands following SDS-PAGE. For further characterization. the F(ab')~ can be sep~rated from Fab' by gel filtration.

WO 92/22324 PCr/lJS92/049~?6 7 ~ ~ 34 Only the Fab' molecules containing two selectively-reducible hinge cysteines carboxy to the interchain disulfide cysteine formed F(ab')~
molecules using the above conditions. Of the three Fab' constructs with two cysteines (see Example 1), the one with a nine amino acid tail appeared to give the best conversion to F~ab'),. The failure to produce F(ab')2 using the established conditions with the Fab' containing one selectively-reducible cysteine is explained by an observation that H65 F(ab')2 with one cysteine produced directly by E. coR could be reduced to Fab' by treatment with the same concentr~tion of cysteine (2 mM) as used in the reassociation buffer. Thus, 2 mM cysteine maintains a reduced state for Fab' containing a single seJectively-reducible cysteine but not for Fab' with two selectively-reducible cysteines.
, b. Binding characterist,cs of in vilro-formed F(ab')~

The in vitro production of proteins of the expected size for F(ab'), suggests that the reassociation procedure resulted in the formation of correctly associated molecules. Competition binding assays were used to confirm that these molecules are functional. In a competition binding assay using HT29 cells and biotinylated lNG-4 IgG the various in vitro-produced F(ab')~ proteins containing two inter-heavy chain cysteines competed equiv?.lently to the chimeric ING-4 IgG and F(ab')2 produced by direct secretion from yeast (data not shown).

WO g2/22324 PCI`/US92/04976 73~ ) 7 ~ 3 II. Production of Heterobifunctional F(~b')~ Molecules a. Formation of F(ab') molecules from Fab' Heterobifunctional F(ab')2 molecules can be made by reducing the Fab' of two different specificities. either alone or in equimolar mixture, followed by simultaneous addition of the reduced Fab' molecules to the reassociation buffer described above. Using this procedure, a heterobifunctional F(ab'), was constructed Witll chimeric ING-2 and ING-4 Fab' molecules. Chimeric ING-2 Fab is described by Better et al., PCr US8903852; and chimeric ING-2 Fab' was made in the same manner as described above for ING-4 F~b'. Analysis of the reassociated protein on non-reducing, Coomassie blue-stained SDS gels demonstrated the presence of F(~b'), protein of the expected molecular weight.

b. Characteristics of heterobifunctional F(ab')2 molecules The heeerobifunction~l nature of this construct was confirmed by tests which make use of the fact that the ING-2 antibody contains a lambda light chain while the ING -4 ~ntibody contains a kappa light chain. .
A sandwich ELISA using antisera against human l~mbda as the coat and against human kappa as the second antibody revealed the presence of F(ab')~ with both kappa and lambda light chains. Direct binding assays can be per~ormed with the F(ab')2 using HT29 and BT20 cells or membrane preparations derived from these cells, which are specifically recognized by ING-4 (kappa light ch~in) and lNG-2 (lambda light chain).
respectlvely.

WO 92/22324 PCI`/US92/04g76 21107~)9 36 -Example 7: Construction and Activitv of Fab'-RTA30 Conjugates Immunotoxins (antibody-toxin conjugates) are frequently prepared by randomly derivatizing an antibody with the heterobifunctional crosslinker SPDP, followed by a disulfide-exchange reaction with the free thiol of ricin toxin A chain (RTA). The disulfide linkage so produced is essential for maximal conjugate cytotoxic activity ~Blakey et al. Prog.
Allergy 115: 50 (1988)). Since the microbially-produced Fab' fragments described in the present invention contain a selectively-reducible thiol on the Fd chain. the process of disulfide conjugation is greatly simplified and highly specific.
Similarly, the procedures outlined below could be used to generate conjugates between a Fab' and any other thiol-containing toxin (viz., abrin A chain), or to create monospecific or bispecific F(ab')~ fragments linked by a disulfide bridge. In conjunction with appropriate linkers, such as SPDP or 2-iminothiolane, conjugates could also be prepared between Fab' fragments and compounds that do not contain selectively-reducible thiols, such as the type I ribosome-inactivating proteins (gelonin, saporin, polkweed antiviral protein, the barley RIP, etc.), enzymes (alkaline phosphatase), or drugs (daunomycin, etc.).
The number of RTA30 molecules conjugated to a Fab' is dependent on the reaction conditions and the number of selectively reducible thiols on the Fab'. For example, the Fab' from construct plNG3219 has two selectively reducible thiols on the Fd chain~ and was conjugated by the methods shown below to make an active Fab'-immunoconjugate witll 2 RTA molecules per Fab.

WO 92/22324 ~ 1 1 D 7 9 ~ PCI/US92/04g76 a. Preparation of H65 Fab'-RTA30 In this example, the H65 Fab' containing the single selectively-reducible hinge cysteine described in Example 3 and the aromatic disulfide dithiobis(pyridine-N-oxide) were used to prepare a disulfide-crosslinked conjugate with the 30 kD form of RTA (RTA30). H65 Fab' (86 mg, 2.4 mg/ml in PBS, pH 7.0) was reduced by the addition of dithiothreitol to a final concentration of 2 mM with rapid stirring.
Following incubation for 1 hour at 25C, dithiobis(pyridine-N-oxide) was added to 7 mM and incubation was continued for ] additional hour. This process both deblocks the Fab' thiol ?nd activates this thiol in preparation for conjugation. The thiol-activated H65 Fab' was then recovered by - desalting on a S x 30 cm column of Trisacrylt~ GF-05LS equilibrated in PBS, pH 7Ø The number of activated thiol groups per Fab' was found to be 1.0~, as measured spectrophotometrically following the addition of Ellman's reagent.
Prior to conjugation, RTA30 (300 mg, 6 mglml in PBS) was reduced by adding DTT to 50 mM for 30 minutes at 25C, divided in half, and each half was desalted on a 5 x 30 cm column of Trisacryl GF-OSLS
equilibrated in PBS. The RTA30-SH contained 0.91 SHfmol, and was concentr~ted by ultrafiltr71tion on ~n Amicon YMIOg) membrane to 6 mg/ml. The thiol-activated H65 Fab' (8~ mg) was then mixed with a five-foid molar excess (246 mg) of the freshly-reduced RTA30 and incubated at 25C for 3 hours. The final concentrations of activated Fab' and RTA30 were 1.0 and 3.0 mg/ml, respectively.
Following an additional incubation ~or 15 hours at 4C, the Fab'-RTA30 conju~ate was purified from the reaction mixture by sequential affinity chromatography on protein G and Cibacl1ron Blue(~ F3GA resins.
First, residual-free RTA30 was removed by applying the mi~ture to a 10 ml column of GammaBind Plus(~ (Genex) previously equilibrated in PBS.

wo 92/22324 ` Pcr/lJS92/o4976 The column was washed with PBS until the absorbance at 280 nm approached zero; Fab'-RTA30 and any remaining unconjugated Fab' were eluted with 0.5 M ammonium acetate, pH 3Ø This material was immediately neutralized with saturated Trizm~ base and dialyzed against 10 mM Tris-HCI, 150 mM NaCI, pH 8Ø Next, the small amount of contaminating free Fab' was removed by diluting the dialyzed protein G
eluate 1:1 with 10 mM Tris-HCI, pH 8.0, and then applying the mixture to a 10 ml column of Blue ToyoPearl~ (TosoHaas) equilibrated in 10 mM Tris-HCI, 75 mM NaCI. pH 8Ø After washing the column with equilibration buffer, purified H65 Fab'-RTA30 was batch eluted with 1 M
NaCI in equilibration buffer. The final conjugate (6S.4 mg) exhibited only a single, symmetrical peak when analyzed by size-exclusion HPLC, and contained one RTA30/Fab'.

b. Evaluation of H65 Fab'-RTA30 The antigen binding and cytotoxic properties of the H65 Fab'-RTA30 conjugate were evaluated in several systems. Antigen reactivity was assessed by using the competitive binding assay described in Example 3(c). Relative to unconjugated H65 Fab', binding of the Fab-RTA30 conjugate was virtuall~ 100% (30C/~, vs. 27C~, of control on a molar basis, respectively). Although these values are lower (roughly 1/3 on a molar basis) than that obtained with the parental, bireactive H65 antibody, these results indicate that the process of conjugation has little effect on the reactivity with antigen.
The cytotoxicity of the H65 Fab'-RTA30 conjugate was examined on several different cell types. Figure 13 shows the results of a representative experiment in whicll cells from the CD5 T-cell line HSB2 (5 x 10~ per ml) were incubated with increasing concentrations of conjugate for up to 2~ hours, at which time the ability of the cells to . - ~ .. ..

WO 92/22324 2 1 1 0 7 9 9 PCI/USg2/04976 s~,rnthesize protein was quantitated by measuring the incorporation of 3H-leucine. On a weight basis, the Fab'-RTA30 conjugate inhibited protein synthesis in these cells by 50% (the ICS0) at a concentration of 43 ng/ml.
Based upon a molecular weight of 80,000 daltons, this translates to an IC50 of 0.5 nM. The curves obtained for the free H65 Fab', as well as for the intact IgG H65-RTA30 conjugate are also shown for comparison.
H65 Fab'-RTA30 was also tested against human peripheral blood mononuclear cells (PBMC) by an assay which measures the inhibition of DNA synthesis as an index of cellular cytotoxicity. PBMC obtained from nonnal healthy donors were isolated over Ficoll-Hypaque and washed extensively. Phytohemagglutinin (PHA) blasts were obtained by activating PBMC in bulk culture for 3 days with an optimal concentration of PHA.
- Both cell populations were incubated with samples for a total of 90 hours:
3H-thymidine was added 16 hours prior to cell harvest.
The cytotoxicity of H65 Fab'-RTA30 for resting and PHA-activated PBMCs is shown in Figure 14. In these assays, both H6S Fab'-RTA30 and the control H65 IgG-RTA30 exhibited similar ICS0 values, which averaged ca. 100 ng/ml for each of the t~vo cell populations. Control IgG
(IND1-RTA30) or Fab' (ING-~RTA30) conjugates were nontoxic at the concentrations tested. On a molar basis, the H65 Fab'-RTA30 conjugate was thus roughly 1/3 as active as the IgG conjugate. Interestingly, under both sets of experimental conditions (resting and activated PBMCs), the - H65 Fab'-RTA30 conjugate exhibited a greater degree of cell killing than did the corresponding H65 IgG-RTA30 conjugate. To a lesser extent, this trend to more complete killing with the Fab'-RTA30 conjugate was also noted with e~ch of the cell lines tested ~data not shown).
These results demonstrate that immunotoxin conjugates derived - from microbially-produced Fab' molecules can be similar in potency to immunotoxins derived from ~nimal cell-produced whole antibodies. The H65-RTA control whole antibody Immunotoxin in Figure 8 has been " ~ ~
.: :
,,~

WO 92/22324 PCI`/US92/W976 2110~9 40 shown to be useful in the therapy of steroid-resistant graft vs. host disease (Kernan et al., J. Amer. Med. Assoc. 259: 3154-3156 (1988)). Thus, the H65 Fab-RTA in this example and similar T-cell-reactive molecules of this invention may be therapeutically useful in the treatment of T lymphocyte diseases and disorders.

Example 8: Construction and Activitv of F(ab')2-RTA30 Conju~ates As described in the previous example, immunotoxins are typically composed of an antibody linked to a ribosome-inactivating protein (such as RTA) via a reducible disulfide bond. In this format advantage is taken of the intrinsic bivalency of the intact antibody molecule, thereby providing ?~ targeting moiety with an affinity for target cells generally higher than is typical for monovalent antibody fragments. The F(ab')2 fragments described in the present invention offer an alternative to intact antibodies for the preparation of immunotoxins. ln addition to providing high affinity binding, F(ab')~ fragments lack Fc receptors which may cause non-specific uptake by macroph~ges and other cells of the immune system. Although the present example employs an F(ab')2 molecule composed of Fab' fragments with identical antigen specificity, similar methodologies could be used to create bivalent immunotoxins with heterologous Fab' fragments. Such conjugates could be targeted to different cell-surface antigens, or to different epitopes on the same antigen. Moreover, non-disulfide linked conjugates to F(ab')~ fragments could also be prepared by appropriate linker selection.

WO 92/22324 PCI`/USg2/04976 '3 11~7~9 a. Preparation of H65 F(ab')2-RTA30 In order to prepare the desired conjugate, the F(ab')2 fragments must first be derivatized with a crosslinking reagent so as to introduce a reactive thiol group necessary for conjugation. In this example, the H65 F(ab')2 molecule described in Example 3 was first reacted with the heterobifunctional crosslinking reagent 5-methyl-2-iminothiolane (M2IT;
(see Goff et al., Bioconjugnle Chem. 1: 381-386 (1990); and U.S. Patent No. 4,970,303) and the aromatic disulfide dithionitrobenzoic acid (DTNB).
This reaction both introduces a free SH group via the M2IT linker, and then activates this thiol with a thionitrobenzoic acid (TNB) leaving group in preparation for the disulfide-exchange reaction with RTA30-SH.
H65 F(ab')~ (65 m8; ~.9 mglml in 25 mM triethanolamine, 150 mM
NaCl, 2.5 mM DTNB, pH 8.0) was reacted with a 12-fold molar excess of M2IT at 25C. Under.these conditions, the F~ab')~ fragments are first derivatized with M2IT, and then the newly exposed linker thiol becomes activated with TNB. After 70 min with constant stirring, the thiol-activated H65 F(ab'),-(M~IT)-TNB was recovered by desalting on a 2.5 cm x 40 cm column of Trisac~ ) GFOSLS equilibrated in 0.1 M NaP04, 0.1 M NaCl, pH 7.5, at 4C. Spectrophotornetric analysis of DTT-treated samples indicated that 1.9 a~tivated thiols had been introduced into the H65 F(ab')~ fragments.
For conjugation, the thiol-activated H65 F(ab') -(M21T)-TNB (62 mg) was mixed with a three-fold molar excess (58 mg) of RTA30-SH
(prepared as described in Example 7) and the mixture was allowed to incubate at ~5C for 2 hr. The final concentrations of F(ab')~ and RTA30 were 1.4 and 1.3 mg/ml, respectively. Following an additional 16 hr at 4C, the H65 F(ab'),-(M21T)-RTA30 conju~ate was purified away from residual unreacted F(ab'), fragments and RTA30 by sequential affinity chromatography on Protein G~) and Cibacron Blue~ F3GA resins WO 92/22324 PCI /US92/04~76 as described in the preceding example. The final conjugate (15 mg) contained a mixture of F(ab')2 fragments containing 1 or 2 RTA30 molecules, with an average RT~ F(ab')2 ratio of 1.3.

b. Evaluation of H65 F(ab')2-RTA30 Antigen reactiv'ity of the H65 F(ab')2-(M2IT)-RTA30 conjugate was examined in the competitive binding assay as described in Examp]e 3(c). Relative to the intact H65 antibody, binding of the F(ab')~-RTA
conjugate was actually gre~ter (147Y~,) on a molar basis. This result is similar to that seen for the intact H65 antibody conjugate (153% re]ative to antibody). Thus. the F(~b'),-RTA conjugate binding was 97% that of the ~orresponding int~ct antibody conjugate.
Simil~rly, the cytotoxic ~ctivity of the F(ab') conjugate was examined against several cell types, as described in Ex?mple 7(b). Against HSB2 cells, the lC50 w~s 1.8 ng/ml (13 pM), rel?ltive to 23 ng/ml (112 pM) for the intact antibody H65-RTA conjugate. In addition, as was noted with the Fab'-RTA30 conjugate of Ex~mple 7. the F(ab'), conjugate killed the target cells to a ~reater extent (92%) than did H65-RTA (78~c). Ag?inst CEM cells, the IC50 oE H65 F(ab'),-(M~IT)-RTA30 was 22 ng/ml (15~ pM), vs. 43 ng/ml (214 pM~ for H65-RTA. For PBMCs, the IC50 w~s 9.~ ng/ml (68 pM) and the extent of kill was S95~, relative to ~6 ng/ml (12~ pM) and 66C~f~ for H65-RTA. Thus, the RTA30 immunoconjuga~e of microbially-produced F(~b'), shows a more potent and complete killing of ~ v~riety of t?~rget CD5 T cells than does the immunoconjugate of p~rental mouse antibody.
Having fullv described the invention, it will be apparent to one of ordinary skill in the art th~t many challges 7~nd modifications can be m~de here~o without departing from the spil it or scope of the invention as set forth herein.

W O 92t22324 ~ 1 1 0 7 ~ 9 PCT/USg2/04976 SEQUENCE LISTING

(1) GENERAL INFORMATION:

(i) APPLICANT: Horwitz, Arnold Better, Marc D
Carroll, Steve (ii) TITLE OF INVENlION: MICROBIALLY-PRODUCED ANTIBODY FRAGMENTS
AND THEIR CONJUGATES

(iii) NUMBER OF SEQUENCES: 25 (iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Sterne, Kessler, Goldstein & Fox (B) STREET: 1225 Connecticut Ave. NW Suite 300 (CJ CITY: Washington (D) STATE: D.C.
(E) COUNTRY: USA
(F) ZIP. 20036 (v) COMPUTER READABLE FORM: d (A) MEDIUM TYPE: Floppy disk (B) COMPUTER: IBM PC compatible (C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SDFTWARE: PatentIn Release #1.0, Version #1.25 (vi) CURRENT APPLICATION DAJA:
(A) APPLICATION NUMBER: US
(B) FILING DATE: 14-JUN-1991 (C) CLASSIFICATION:

(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Goldstein, ~orge A

WO g2/22324 PCI`/US92/04976 2`110799:
~ .
(B) REGISTRATIGN NUMBER: 29,021 (C) REFERENCE/DOCKET NUMBER: 0610.0830000 (ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: 202 833-7533 (B) TELEFAX: 202 833-8716 (2) INFORMATION FOR SEQ ID NO:1:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 14 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear -.:.
(ii) MOLECULE TYPE: peptide (xi) SEQUENCE DESCRIPTION: SEQ ID NO:1:

: Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro (2) INFORMATION FOR SEQ ID NO:2:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 12 base pairs : (B) TYPE: nucleic acid (CJ STRANDEDNESS: both (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA

. , ~,., W O 92/22324 ~ 7 9 9 PCT/U592/04976 -4~-(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:

(2) INFORMATION FOR SEQ ID NO:3:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 21 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both (D) TOPOLOGY: linear (iiJ MOLECULE TYPE: DNA

,, , :

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:

(2) INFORMATION FOR SEQ ID NO:4:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 42rbase pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both (D) TOPOLOGY: linear (ii3 MOLECULE TYPE: DNA
.

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:4:

:::

wo g2/22324 Pcr/us92/o4g76 ~ ~ lU ~9 -46-(2) INFORMATION FOR SEQ ID NO:5:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 42 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both (D) TOPOLOGY linear (ii) MOLECULE TYPE: DNA

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:5:

(2) INFORMATION FOR SEQ ID NO:6:
;
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 amino acids (B) TYPE: amin`o acid (D) TOPOLOGY: linear " .
. , .
(ii) MOLECULE TYPE: peptide (xi) SEQUENCE DESCRIPTION: SEQ ID NO:6:

Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Asp 1' - ~ : :

.
2~ U 7~9 -~7-(2) INFORMATION FOR SEQ ID NO:7.

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 14 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide (xi) SEQUENCE DESCRIPTION: SEQ ID NO:7:

Thr Ser Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro ~-: (2) INFORMATION FOR SEQ ID NO:8:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 15 base pairs B) TYPE: nucleic acid (C) STRANDEDNESS: both (D) TOPOLOGY: linear .

(ii) MOLECULE TYPE: DNA

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:8:
;

(2) INFORMATION FOR SEQ lD NO:9:

~:~ (i) SEQUENCE CHARACTERISTICS:

,:, .~ .

,,i .
~lu799 4~
(A) LENGTH: 42 base pairs (B) TYPE: nucl-eic acid (C) STRANDEDNESS: both (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:9:

(2) INFORMATION FOR SEQ ID NO:10:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 15 base pairs (BJ TYPE: nucleic acid (C) STRANDEDNE5S: both (D) TOPOLOGY: linear -(ii) MOLECULE TYPE: DNA

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:10:

(2) INFORMATION FOR SEQ ID NO:11:

(i) SEQUENCE CHARACTERISTICS:
~A) LENGTH: 19 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both " ~ ~ .

WO 92/22324 PCI~/US92/04976 - 211~799 (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:11:

(2) INFORMATION FOR SEQ ID NO:12:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 21 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both (D) TOPOLOGY: linear :
(ii) MOLECULE TYPE: DNA

: (xi) SEQUENCE DESCRIPTION: SEQ ID NO:12: ' ,TCCAGCTTG ATCACTCGAG G 21 (2) INFORMATION FOR SEQ ID NO:13:

(i) SEQllENCE CHARACTERISTICS:
(A) LENGTH: 25 base pairs (B) TYPE: nucleic acid ~C) STRANDEDNESS: both (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA

, -, ~; ': ' W O 92/22324 ` PCT/US92/04976 $11U799 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:13:

(2) INFORMATION FOR SEQ ID NO:14:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 10 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide (xi) SEQUENCE DESCRIPTION: SEQ ID NO:14:

Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro (2) INFORMATION FOR SEQ ID NO:15: ' (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 42 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA

~ (xi) SEQUENCE DESCRIPTION: SEQ ID NO:15:

"

W o 92/22324 2 1 1 ~ 7 9 9 PCT/US92/04g76 (2) INFORMATION FOR SEQ ID NO:16:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 46 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:16:

: AATTCCTCGA GTGATCATGG ACCACCCAAC AATTCAGGAG CTGGAC 46 (2) INFORMATION FOR SEQ ID NO:17:

i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 15 base pairs (B) TYPE: nucleic acid ~C) STRANDEDNESS: both (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA

(xi) SEQUENCE DESCRIPTION: 'FQ ID NO:17:

~: (2) INFORMATION FOR SEQ ID NO:18:
,~
", ; ' WO 92/22324 PCl`/US92/04976 9 - 5~ -( i ) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 414 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both (DJ TOPOLOGY: li near (ii) MOLECULE TYPE: DNA

(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 18:

GGAAAGGGTT TAAGGTGGAT GGGCTGGATA AACACCCACA CTGGAGAGCC AACATATGC~ 240 GATGACTTCA AGGGACGGTT TGCCTTCTCT TTGGAAACGT CTGCGAGCAC TGCC~ATTTA 300 îACTGGTACT TCGATGTCTG GGGCGCAGGG ACCACGGTCA CCGTCTCCTC AGCC 414 (2) INFORMATION FOR SEQ ID NO:l9:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 138 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein W 0 92/22324 2 1 1 ~ 7 ~ 9 PCT/US92~04976 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:19:

Met Ala Trp Val Trp Thr Leu Leu Phe Leu Met Ala Ala Ala Gln Ser Ala Gln Ala Gln Ile Gln Leu Val Gln Ser Gly Pro Glu Leu Lys Lys Pro Gly Glu Thr Val Lys Ile Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Asn ~yr Gly Met Asn Trp Val Lys Gln Ala Pro Gly Lys Gly Leu
6~

Arg Trp Met Gly Trp Ile Asn Thr His Thr Gly Glu Pro Thr Tyr Ala Asp Asp Phe Lys Gly Arg Phe Ala Phe Ser Leu Glu Thr Ser Ala Ser 9~

hr Ala ~yr Ley Gln Ile Asn Asn Leu Lys Asn Glu Asp Thr Ala Thr Tyr Phe Cys Thr Arg Arg Gly Tyr Asp Trp Tyr Phe Asp Val Trp Gly Ala Gly Thr Thr Val Thr Val Ser Ser Ala ~2) INFORMATION FOR SEQ ID NO:20:

(i3 SEQUENCE CHARACTERISTICS:
(A) LENGTH: 388 base pairs WO 92/22324 ~ 9 PCI/US92/04976 (B) TYPE: nucleic acid (C) STRANDEDNESS: both (D) TOPOLOGY: linear .
(ii) MOLECULE TYPE: DNA

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:20:

~

(2J INFORMATION FOR SEQ ID NO:21:

(i) SEQUENCE CHARACTERISTICS:
(AJ LENGTH: 129 amino acids (B) TYPE: amino àcid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein ..;:
" ~, "',' "

W o 92~22324 PCT/US~2/04976
7 ~ ~

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:21:

Met Asp Met Arg Thr Pro Ala Gln Phe Leu Gly Ile Leu Leu Leu Trp Phe Pro Gly Ile Lys Cys Asp Ile Lys Met Thr Gln Ser Pro Ser Ser Met Tyr Ala Ser Leu Gly Glu Arg Val Thr Ile Thr Cys Lys Ala Ser Gln Asp Ile Asn Ser Tyr Leu Ser Trp Phe Gln Gln Lys Pro Gly Lys Ser Pro Lys Thr Leu Ile Tyr Arg Ala Asn Arg Leu Val Asp Gly Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Gln Asp Tyr Ser Leu Thr . 85 90 95 Ile Ser Ser Leu Asp Tyr Glu Asp Met Gly Ile Tyr Tyr Cys Gln Gln Tyr Asp Glu Ser Pro Trp Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys (2) INFORMATION FOR SEQ ID NO:22:

- (i) SEQUENCE CHARACTERISTICS:
(Al LENGTH: 393 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both WO 92/22324 PCI`/VS92/0497 ~lli)'l~9 - 56 -(D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:22:

, ~2~ INFORMATION FOR SEQ ID NO:23:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 131 amino acids (B) TYPE: amino acid (D) TOPOIOGY: linear (ii) MOLECULE TYPE: protein (xi) SEqUENCE DESCRIPTION: SEQ ID NO:23:

WO 92/22324 PCl /US92/04976 -~7-Met Glu Ser Asp Thr Leu Leu Leu Trp Yal Leu Leu Leu Trp Val Pro Gly Ser Thr Gly Asp Ile Val Leu Thr Gln Ser Pro Ala Ser Leu Ala Val Ser Leu Gly Gln Arg Ala Thr Ile Ser Cys Arg Ala Ser Glu Ser Val Glu Tyr Tyr Gly Thr Ser Leu Met Gln Trp Tyr Gln Gln Lys Pro Gly Gln Pro Pro Lys Leu Leu lle Tyr Ala Ala Ser Asn Val Glu Ser Gly Val Pro Ala Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Ser g5 ~, Leu Asn Ile His Pro Val Gly Glu Glu Asp Ile Ala Met Tyr Phe Cys ;' -- Gln Gln Ser Arg Lys Val Pro Trp Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys (2) INFORMATION FOR SEQ ID NO:24:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 426 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both (D) TOPOLOGY: linear .~
.~ , ,j W O 92/22324 PCT/U~92/04976 ~llU'`~)9 5~
(ii) MOLECULE TYPE: DNA

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:24:

. - --.
: GACAGT6TGA TGGGGCGATT CACCATCTCC AGAGACAATG CCAAGAACAA CCTGTACCTG 300 .

(2) INFORMATION FOR SEQ ID NO:25:

(i) SE~UENCE CHARACTERISTICS:
(A) LENGTH: 142 amino acids (B~ TYPE: amino acid .; (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:25:

.
. ,;.., ., .; ., .

WO 92/22324 PCl`/US92/04976 7 ~ 9 Met Asn Phe Gly Leu Ser Leu Ile Phe Leu Val Leu Val Leu Lys Gly Val Gln Cys Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Lys Pro Gly Gly Ser Leu Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Asp Phe Tyr Met Tyr Trp Val Arg Gln Thr Pro Glu Lys Arg Leu S5 ~0 Glu Trp Val Ala Thr Ile Ser Asp Gly Gly Ile Tyr Thr Tyr Tyr Ser Asp Ser Val Met Gly Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Asn Leu Tyr Leu Gln Ile Ser Ser Leu Lys Ser Glu Asp Thr Ala Met ~, Tyr Tyr Cys Ala Arg Asp Pro Tyr Ser Tyr Asp Ser Ser Pro Ala Trp 115 lZ0 125 Phe Ala Tyr Trp Gly Gln Gly Thr Leu Yal Thr Val Ser Ala

Claims (44)

WE CLAIM:
1. A polynucleotide molecule comprising a bacterial promoter sequence operably linked to a sequence encoding the Fd polypeptide of an immunoglobulin Fab' or F(ab')2 fragment molecule.
2. The molecule of claim 1 wherein the Fd sequence encodes two or more cysteine residues in the Fd hinge region on the carboxyl side of the disulfide bond formed between the light chain and the Fd chain.
3. The molecule of claim 2 wherein there are less than 30 amino acid residues between the cysteine residue with the highest amino acid number and the carboxyl terminal.
4. The molecule of claim 1 wherein the Fd polypeptide encodes a variable region, which when part of an Fab' or F(ab')2, molecule, is reactive with T cells.
5. The molecule of any of claims 1, 2, 3 or 4 wherein the Fd polypeptide is chimeric.
6. A vector comprising the molecule of claim 1.
7. The vector of claim 6 which also encodes the light chain polypeptide of an immunoglobulin Fab' or F(ab')2 fragment molecule.
8. A bacterium transformed with the vector of any of claims 6 or 7.
9. A polynucleotide molecule comprising a yeast promoter sequence operably linked to a sequence encoding the Fd polypeptide of a Fab' or F(ab')2 immunoglobulin fragment.
10. The molecule of claim 9 wherein the Fd sequence encodes two or more cysteine residues in the Fd hinge region on the carboxyl side of the disulfide bond formed between the light chain and the Fd chain.
11. The molecule of claim 9 wherein there are less than 30 amino acid residues between the cysteine residue with the highest amino acid number, or the cysteine residue closest to the carboxyl terminal, and the carboxyl terminal.
12. The molecule of claim 9 wherein the Fab' or F(ab')2 molecule is reactive with T cells.
13. The molecule of any of claims 9, 10, 11, or 12 wherein the Fab' or F(ab')2 molecule is chimeric.
14. A vector comprising the molecule of claim 9.
15. The vector of claim 14 which also encodes the light chain polypeptide of an immunoglobulin Fab' or F(ab')2, fragment molecule.
16. A yeast transformed with the vector of any of claims 14 or 15.
17. A method for preparing an immunoglobulin Fab' molecule, comprising the steps of:

a) culturing a bacterium transformed with the vector of claim 7; and b) isolating the Fab' from the culture medium.
18. A method for preparing an immunoglobulin F(ab')2 mole-cule, comprising the steps of:
a) culturing a bacterium transformed with the vector of claim 7; and b) isolating the (Fab'), from the culture medium.
19. A method for preparing an immunoglobulin Fab' molecule.
comprising the steps of:
a) culturing a yeast transformed with the vector of claim 15:
and b) isolating the Fab' from the culture medium.
20. A method for preparing an immunoglobulin F(ab')2 mole-cule, comprising the steps of:
a) culturing a yeast transformed with the vector of claim 15;
and b) isolating the F(ab')2, from the culture medium.
21. A method for preparing an immunoglobulin F(ab')2 mole-cule, comprising the steps of:
a) preparing a first Fab' by the method of claim 17 or claim 19:
b) prepaling a` second Fab' by the method of claim 17 or claim 19; and c) combining the first Fab' and the second Fab' such that an F(ab')2, is formed.
22. A method of preparing an immunoconjugate comprising:
a) preparing a Fab' by the method of either claim 17 or claim 19;
b) reducing the selectively reducible hinge cysteine of the Fab';
and c) linking the reduced Fab' to a thiol-containing active moiety.
23. A method of preparing an immunoconjugate comprising:
a) preparing a Fab' by the method of either claim 17 or claim 19;
b) reducing the selectively reducible hinge cysteine of the Fab':
c) linking the reduced Fab' to a thiol-containing activating compound: and d) further linking the thiol-containing activating compound previously linked to reduced Fab' to a thiol-reactive active moiety.
24. The method of claim 23 wherein the activating compound is dithiobis(pyridine-N-oxide) or dithiobis(nitrobenzoate).
25. A method of preparing an immunoconjugate comprising:
a) preparing a F(ab')2, by the method of claim 18 or claim 20:
and b) linking the F(ab')2 to an active moiety.
26. A method of preparing an immunoconjugate comprising:
a) preparing a F(ab')2, by the method of claim 21; and b) linking the F(ab')2 to an active moiety.
27. A method for preparing Fab'-enzyme conjugate comprising:
a) preparing a Fab' by the method of claim 17 or claim 19: and b) linking the Fab' to an enzyme.
28. The method for preparing the Fab'-enzyme conjugate of claim 27 wherein the enzyme is a ribosome-inactivating protein.
29. The method for preparing the Fab'-enzyme conjugate of claim 27 wherein the enzyme is ricin toxin A chain.
30. The method for preparing the Fab'-enzyme conjugate of claim 27 wherein the Fab' is specific for a human T cell.
31. The method for preparing the Fab'-enzyme conjugate of claim 27 wherein the Fab' is specific for CD5 or CD7.
32. A method for preparing F(ab')2-enzyme conjugate compris-ing:
a) preparing an F(ab')2 by the method of claim 18 or claim 20; and b) linking the F(ab')2 to an enzyme.
33. The method for preparing the F(ab')2-enzyme conjugate of claim 32 wherein the enzyme is a ribosome-inactivating protein.
34. The method for preparing the F(ab')2-enzyme conjugate of claim 32 wherein the enzyme is ricin toxin A chain.
35. The method for preparing the F(ab')2-enzyme conjugate of claim 32 wherein the F(ab')2 is specific for a human T cell.
36. The method for producing the F(ab')2-enzyme conjugate of claim 32 wherein the F(ab')2 is specific for CD5 or CD7.
37. A method for preparing F(ab')2-enzyme conjugate comprising:
a) preparing an F(ab')2 by the method of claim 21; and b) linking the F(ab')2 to an enzyme.
38. The method for preparing the F(ab')2-enzyme conjugate of claim 37 wherein the enzyme is a ribosome-inactivating protein.
39. The method of preparing the F(ab')2-enzyme conjugate of claim 37 wherein the enzyme in ricin toxin A chain.
40. The method for preparing the F(ab')2-enzyme conjugate of claim 37 wherein the F(ab')2 is specific for a human T cell.
41. The method for producing the F(ab')2-enzyme conjugate of claim 37 wherein the F(ab')2 is specific for CD5 or CD7.
42. An F(ab')2 with specificity for two different antigens.
43. The F(ab')2 of claim 42 wherein said antigens are tumor-associated antigens.
44. A method of producing an F(ab')2 with specificity for two different antigens wherein said method comprises:
a) preparing a first Fab' having a specificity for a first tumor-associated antigen by the method of claim 17 or claim 19;
b) preparing a second Fab' having a specificity for a second tumor-associated antigen by the method of claim 17 or claim 19; and c) combining the first Fab' and the second Fab' such that an F(ab')2 with specificity for two different tumor-associated antigens is formed.
CA 2110799 1991-06-14 1992-06-15 Microbially-produced antibody fragments and their conjugates Abandoned CA2110799A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US71417591A 1991-06-14 1991-06-14
US07/714,175 1991-06-14

Publications (1)

Publication Number Publication Date
CA2110799A1 true CA2110799A1 (en) 1992-12-23

Family

ID=24869022

Family Applications (1)

Application Number Title Priority Date Filing Date
CA 2110799 Abandoned CA2110799A1 (en) 1991-06-14 1992-06-15 Microbially-produced antibody fragments and their conjugates

Country Status (5)

Country Link
EP (1) EP0590067A1 (en)
JP (1) JPH07503124A (en)
AU (1) AU2238292A (en)
CA (1) CA2110799A1 (en)
WO (1) WO1992022324A1 (en)

Families Citing this family (391)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5618920A (en) 1985-11-01 1997-04-08 Xoma Corporation Modular assembly of antibody genes, antibodies prepared thereby and use
WO1993006217A1 (en) 1991-09-19 1993-04-01 Genentech, Inc. EXPRESSION IN E. COLI OF ANTIBODY FRAGMENTS HAVING AT LEAST A CYSTEINE PRESENT AS A FREE THIOL, USE FOR THE PRODUCTION OF BIFUNCTIONAL F(ab')2 ANTIBODIES
US5837491A (en) * 1991-11-04 1998-11-17 Xoma Corporation Polynucleotides encoding gelonin sequences
US6146850A (en) * 1991-11-04 2000-11-14 Xoma Corporation Proteins encoding gelonin sequences
US5621083A (en) * 1991-11-04 1997-04-15 Xoma Corporation Immunotoxins comprising ribosome-inactivating proteins
US5869619A (en) * 1991-12-13 1999-02-09 Xoma Corporation Modified antibody variable domains
CA2507749C (en) * 1991-12-13 2010-08-24 Xoma Corporation Methods and materials for preparation of modified antibody variable domains and therapeutic uses thereof
DE4312916C2 (en) * 1993-04-14 1995-03-23 Fresenius Ag Medicines used to treat immune reactions
JPH08510642A (en) * 1993-05-12 1996-11-12 ゾマ コーポレイション Immunotoxin consisting of gelonin and antibody
US7429646B1 (en) 1995-06-05 2008-09-30 Human Genome Sciences, Inc. Antibodies to human tumor necrosis factor receptor-like 2
US7888466B2 (en) 1996-01-11 2011-02-15 Human Genome Sciences, Inc. Human G-protein chemokine receptor HSATU68
US7964190B2 (en) 1996-03-22 2011-06-21 Human Genome Sciences, Inc. Methods and compositions for decreasing T-cell activity
US6635743B1 (en) 1996-03-22 2003-10-21 Human Genome Sciences, Inc. Apoptosis inducing molecule II and methods of use
EP1093457B8 (en) 1998-03-19 2011-02-02 Human Genome Sciences, Inc. Cytokine receptor common gamma chain like
WO2000050620A2 (en) 1999-02-26 2000-08-31 Human Genome Sciences, Inc. Human endokine alpha and methods of use
US7291714B1 (en) 1999-06-30 2007-11-06 Millennium Pharmaceuticals, Inc. Glycoprotein VI and uses thereof
US20040001826A1 (en) 1999-06-30 2004-01-01 Millennium Pharmaceuticals, Inc. Glycoprotein VI and uses thereof
EP1242457B1 (en) 1999-12-28 2004-08-11 Esbatech AG Intrabodies with defined framework that is stable in a reducing environment and applications thereof
WO2002043660A2 (en) 2000-11-28 2002-06-06 Mediummune, Inc Methods of administering/dosing anti-rsv antibodies for prophylaxis and treatment
AU2001266557A1 (en) 2000-04-12 2001-10-23 Human Genome Sciences, Inc. Albumin fusion proteins
US20030031675A1 (en) 2000-06-06 2003-02-13 Mikesell Glen E. B7-related nucleic acids and polypeptides useful for immunomodulation
AU2001282856A1 (en) 2000-06-15 2001-12-24 Human Genome Sciences, Inc. Human tumor necrosis factor delta and epsilon
CA2407910C (en) 2000-06-16 2013-03-12 Steven M. Ruben Antibodies that immunospecifically bind to blys
JP2004529849A (en) 2000-09-01 2004-09-30 インターナショナル バイオイムン システムズ,インコーポレーテッド Identification and development of specific monoclonal antibodies against squamous cell carcinoma
GB0022978D0 (en) 2000-09-19 2000-11-01 Oxford Glycosciences Uk Ltd Detection of peptides
US7090843B1 (en) 2000-11-28 2006-08-15 Seattle Genetics, Inc. Recombinant anti-CD30 antibodies and uses thereof
PT1355919E (en) 2000-12-12 2011-03-02 Medimmune Llc Molecules with extended half-lives, compositions and uses thereof
CN1564826A (en) 2001-02-09 2005-01-12 人类基因组科学公司 Human G-protein chemokine receptor (CCR5) HDGNR10
US6709655B2 (en) 2001-02-28 2004-03-23 Instituto Bioclon, S.A. De C.V. Pharmaceutical composition of F(ab1)2 antibody fragments and a process for the preparation thereof
DK1411962T3 (en) * 2001-03-15 2011-04-04 Neogenix Oncology Inc Pancreatic cancer therapy with monoclonal drug
US8231878B2 (en) 2001-03-20 2012-07-31 Cosmo Research & Development S.P.A. Receptor trem (triggering receptor expressed on myeloid cells) and uses thereof
US8981061B2 (en) 2001-03-20 2015-03-17 Novo Nordisk A/S Receptor TREM (triggering receptor expressed on myeloid cells) and uses thereof
EP2228389B1 (en) 2001-04-13 2015-07-08 Human Genome Sciences, Inc. Antibodies against vascular endothelial growth factor 2
US6867189B2 (en) 2001-07-26 2005-03-15 Genset S.A. Use of adipsin/complement factor D in the treatment of metabolic related disorders
EP2261250B1 (en) 2001-12-21 2015-07-01 Human Genome Sciences, Inc. GCSF-Albumin fusion proteins
US7317091B2 (en) 2002-03-01 2008-01-08 Xencor, Inc. Optimized Fc variants
US20040132101A1 (en) 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
US8093357B2 (en) 2002-03-01 2012-01-10 Xencor, Inc. Optimized Fc variants and methods for their generation
US8188231B2 (en) 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
GB0207533D0 (en) 2002-04-02 2002-05-08 Oxford Glycosciences Uk Ltd Protein
US7371383B2 (en) 2002-04-12 2008-05-13 Medimmune, Inc. Recombinant anti-interleukin-9 antibodies
CN100418981C (en) 2002-06-10 2008-09-17 瓦西尼斯公司 Gene differentially expressed in breast and bladder cancer and encoded polypeptides
US7425618B2 (en) 2002-06-14 2008-09-16 Medimmune, Inc. Stabilized anti-respiratory syncytial virus (RSV) antibody formulations
DK1534335T4 (en) 2002-08-14 2015-10-05 Macrogenics Inc FCGAMMARIIB-SPECIFIC ANTIBODIES AND PROCEDURES FOR USE THEREOF
US7335759B2 (en) 2002-12-02 2008-02-26 Universidad Nacional Autónoma de Méxica (UNAM) Recombinant immunogens for the generation of antivenoms to the venom of scorpions of the genus Centruroides
DE10303974A1 (en) 2003-01-31 2004-08-05 Abbott Gmbh & Co. Kg Amyloid β (1-42) oligomers, process for their preparation and their use
CA2516455C (en) 2003-02-20 2012-05-01 Seattle Genetics, Inc. Anti-cd70 antibody-drug conjugates and their use for the treatment of cancer and immune disorders
US8388955B2 (en) 2003-03-03 2013-03-05 Xencor, Inc. Fc variants
US8084582B2 (en) 2003-03-03 2011-12-27 Xencor, Inc. Optimized anti-CD20 monoclonal antibodies having Fc variants
US20090010920A1 (en) 2003-03-03 2009-01-08 Xencor, Inc. Fc Variants Having Decreased Affinity for FcyRIIb
RS54160B1 (en) 2003-03-19 2015-12-31 Biogen Idec Ma Inc. Nogo receptor binding protein
EP2316487B1 (en) 2003-04-11 2014-06-11 MedImmune, LLC Recombinant IL-9 antibodies & uses thereof
US9051373B2 (en) 2003-05-02 2015-06-09 Xencor, Inc. Optimized Fc variants
JP2008500005A (en) 2003-07-15 2008-01-10 バロス リサーチ インスティテュート Compositions and methods for immunotherapy of cancer and infectious diseases
EP1651266B1 (en) * 2003-07-25 2010-03-03 Laboratorios Silanes, S.A. de C.V. Administration of anti-tnf-alpha f(ab')2 antibody fragments
WO2005014795A2 (en) 2003-08-08 2005-02-17 Genenews Inc. Osteoarthritis biomarkers and uses thereof
JP4934426B2 (en) 2003-08-18 2012-05-16 メディミューン,エルエルシー Antibody humanization
US8101720B2 (en) 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
US9714282B2 (en) 2003-09-26 2017-07-25 Xencor, Inc. Optimized Fc variants and methods for their generation
HUE027902T2 (en) 2004-02-09 2016-11-28 Human Genome Sciences Inc Corp Service Company Albumin fusion proteins
KR101239542B1 (en) 2004-06-24 2013-03-07 바이오겐 아이덱 엠에이 인코포레이티드 Treatment of conditions involving demyelination
US20150010550A1 (en) 2004-07-15 2015-01-08 Xencor, Inc. OPTIMIZED Fc VARIANTS
CA2576193A1 (en) 2004-08-03 2006-02-16 Biogen Idec Ma Inc. Taj in neuronal function
US7700720B2 (en) 2004-09-21 2010-04-20 Medimmune, Llc Antibodies against and methods for producing vaccines for respiratory syncytial virus
EP2422811A2 (en) 2004-10-27 2012-02-29 MedImmune, LLC Modulation of antibody specificity by tailoring the affinity to cognate antigens
US8367805B2 (en) 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
US8546543B2 (en) 2004-11-12 2013-10-01 Xencor, Inc. Fc variants that extend antibody half-life
US8802820B2 (en) 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
EP2332985A3 (en) 2004-11-12 2012-01-25 Xencor, Inc. Fc variants with altered binding to FcRn
GB0426146D0 (en) 2004-11-29 2004-12-29 Bioxell Spa Therapeutic peptides and method
CA2599589A1 (en) 2005-02-07 2006-08-17 Genenews,Inc. Mild osteoarthritis biomarkers and uses thereof
CA2597924C (en) 2005-02-15 2018-10-02 Duke University Anti-cd19 antibodies and uses in oncology
EP1858545A2 (en) 2005-03-04 2007-11-28 Curedm Inc. Methods and pharmaceutical compositions for treating type 1 diabetes mellitus and other conditions
JP5153613B2 (en) 2005-03-18 2013-02-27 メディミューン,エルエルシー Antibody framework shuffle
US7381802B2 (en) 2005-04-15 2008-06-03 Universidad Nacional Autónoma De México (UNAM) Human antibodies that specifically recognize the toxin Cn2 from Centruroides noxius scorpion venom
US7393919B2 (en) 2005-05-25 2008-07-01 Cure Dm, Inc. Peptides, derivatives and analogs thereof, and methods of using same
KR20080025174A (en) 2005-06-23 2008-03-19 메디뮨 인코포레이티드 Antibody formulations having optimized aggregation and fragmentation profiles
TW201444869A (en) 2005-06-30 2014-12-01 Abbvie Inc IL-12/P40 binding proteins
RS53058B (en) 2005-07-08 2014-04-30 Biogen Idec Ma Inc. Sp35 antibodies and uses thereof
EP1920057A4 (en) 2005-08-03 2009-03-18 Grains Res & Dev Corp Polysaccharide synthases
EP1928506A4 (en) 2005-08-19 2009-10-21 Abbott Lab Dual variable domain immunoglobin and uses thereof
EP2500353A3 (en) 2005-08-19 2012-10-10 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
JP2009510002A (en) 2005-09-30 2009-03-12 アボット ゲゼルシャフト ミット ベシュレンクテル ハフツング ウント コンパニー コマンディトゲゼルシャフト Binding domains of proteins of the repulsion-inducing molecule (RGM) protein family, and functional fragments thereof, and uses thereof
EP1931709B1 (en) 2005-10-03 2016-12-07 Xencor, Inc. Fc variants with optimized fc receptor binding properties
CA2625998C (en) 2005-10-06 2015-12-01 Xencor, Inc. Optimized anti-cd30 antibodies
EP2319941A3 (en) 2005-10-21 2011-08-17 GeneNews Inc. Method and apparatus for correlating levels of biomarker products with disease
CA2628451A1 (en) 2005-11-04 2007-05-18 Biogen Idec Ma Inc. Methods for promoting neurite outgrowth and survival of dopaminergic neurons
AU2006311661B2 (en) 2005-11-07 2011-05-26 The Scripps Research Institute Compositions and methods for controlling tissue factor signaling specificity
DK1963369T3 (en) 2005-11-28 2013-06-03 Zymogenetics Inc IL-21 Antagonists
RU2442793C2 (en) 2005-11-30 2012-02-20 Эбботт Лэборетриз ANTISUBSTANCES AGAINST GLOBULOMER Aβ, THEIR ANTIGEN-BINDING PARTS, CORRESPONDING HYBRIDOMAS, NUCLEIC ACIDS, VECTORS, HOST CELLS, WAYS OF PRODUCTION OF MENTIONED ANTISUBSTANCES, COMPOSITIONS CONTAINING MENTIONED ANTISUBSTANCES, APPLICATION OF MENTIONED ANTISUBSTANCES AND WAYS OF APPLICATION OF MENTIONED ANTISUBSTANCES
SG2014013437A (en) 2005-11-30 2014-07-30 Abbott Lab Monoclonal antibodies and uses thereof
EP2543384A3 (en) 2005-12-02 2013-04-10 Biogen Idec MA Inc. Treatment of conditions involving demyelination
CA2913655A1 (en) 2006-01-27 2007-08-09 Biogen Ma Inc. Nogo receptor antagonists
EP2650306A1 (en) 2006-03-06 2013-10-16 Aeres Biomedical Limited Humanized Anti-CD22 antibodies and their use in treatment of oncology, transplantation and autoimmune disease
GB0611116D0 (en) 2006-06-06 2006-07-19 Oxford Genome Sciences Uk Ltd Proteins
BRPI0713426A2 (en) 2006-06-14 2012-10-09 Macrogenics Inc methods of treating, slowing the progression, or ameliorating one or more symptoms of a disorder, and preventing or delaying the onset of a disorder
LT2029173T (en) 2006-06-26 2016-11-10 Macrogenics, Inc. Fc riib-specific antibodies and methods of use thereof
US7572618B2 (en) 2006-06-30 2009-08-11 Bristol-Myers Squibb Company Polynucleotides encoding novel PCSK9 variants
USRE47123E1 (en) 2006-07-18 2018-11-13 Sanofi EPHA2 receptor antagonist antibodies
WO2008022152A2 (en) 2006-08-14 2008-02-21 Xencor, Inc. Optimized antibodies that target cd19
JP5209625B2 (en) 2006-08-28 2013-06-12 協和発酵キリン株式会社 Antagonist human LIGHT specific human monoclonal antibody
MX349810B (en) 2006-09-08 2017-08-14 Abbvie Bahamas Ltd Interleukin -13 binding proteins.
AU2007299843B2 (en) 2006-09-18 2012-03-08 Xencor, Inc Optimized antibodies that target HM1.24
EP2407548A1 (en) 2006-10-16 2012-01-18 MedImmune, LLC Molecules with reduced half-lives, compositions and uses thereof
EP1914242A1 (en) 2006-10-19 2008-04-23 Sanofi-Aventis Novel anti-CD38 antibodies for the treatment of cancer
US8785400B2 (en) 2006-11-22 2014-07-22 Curedm Group Holdings, Llc Methods and compositions relating to islet cell neogenesis
US8455626B2 (en) 2006-11-30 2013-06-04 Abbott Laboratories Aβ conformer selective anti-aβ globulomer monoclonal antibodies
EP2687232A1 (en) 2006-12-06 2014-01-22 MedImmune, LLC Methods of treating systemic lupus erythematosus
EP2740744B1 (en) 2007-01-09 2018-03-28 Biogen MA Inc. Sp35 antibodies and uses thereof
US8128926B2 (en) 2007-01-09 2012-03-06 Biogen Idec Ma Inc. Sp35 antibodies and uses thereof
US8685666B2 (en) 2007-02-16 2014-04-01 The Board Of Trustees Of Southern Illinois University ARL-1 specific antibodies and uses thereof
US8114606B2 (en) 2007-02-16 2012-02-14 The Board Of Trustees Of Southern Illinois University ARL-1 specific antibodies
WO2008104803A2 (en) 2007-02-26 2008-09-04 Oxford Genome Sciences (Uk) Limited Proteins
EP3118221B1 (en) 2007-02-26 2019-08-21 Oxford BioTherapeutics Ltd Proteins
US20100311767A1 (en) 2007-02-27 2010-12-09 Abbott Gmbh & Co. Kg Method for the treatment of amyloidoses
AU2008232902B2 (en) 2007-03-30 2013-10-03 Medlmmune, Llc Antibody formulation
US7807168B2 (en) 2007-04-10 2010-10-05 Vaccinex, Inc. Selection of human TNFα specific antibodies
US20080279851A1 (en) 2007-05-07 2008-11-13 Medlmmune, Llc Anti-icos antibodies and their use in treatment of oncology, transplantation and autoimmune disease
SG10201503254TA (en) 2007-05-14 2015-06-29 Medimmune Llc Methods of reducing eosinophil levels
US8816047B2 (en) 2007-08-30 2014-08-26 Cure DM Group Holdings, LLC Compositions and methods of using proislet peptides and analogs thereof
GB0719231D0 (en) 2007-10-03 2007-11-14 Oxford Genome Sciences Uk Ltd Protein
PT2219452E (en) 2007-11-05 2016-01-26 Medimmune Llc Methods of treating scleroderma
US9308257B2 (en) 2007-11-28 2016-04-12 Medimmune, Llc Protein formulation
SI2235059T1 (en) 2007-12-26 2015-06-30 Xencor, Inc. Fc variants with altered binding to fcrn
AU2009203350B2 (en) 2008-01-11 2014-03-13 Gene Techno Science Co., Ltd. Humanized anti-alpha9 integrin antibodies and the uses thereof
CN102083460A (en) 2008-01-18 2011-06-01 米迪缪尼有限公司 Cysteine engineered antibodies for site-specific conjugation
JP5608100B2 (en) 2008-02-08 2014-10-15 メディミューン,エルエルシー Anti-IFNAR1 antibodies with reduced Fc ligand affinity
US8962803B2 (en) 2008-02-29 2015-02-24 AbbVie Deutschland GmbH & Co. KG Antibodies against the RGM A protein and uses thereof
US20110020368A1 (en) 2008-03-25 2011-01-27 Nancy Hynes Treating cancer by down-regulating frizzled-4 and/or frizzled-1
EP2276509B1 (en) 2008-04-11 2016-06-15 Seattle Genetics, Inc. Detection and tratment of pancreatic, ovarian and other cancers
AU2009238897B2 (en) 2008-04-24 2015-03-19 Gene Techno Science Co., Ltd. Humanized antibodies specific for amino acid sequence RGD of an extracellular matrix protein and the uses thereof
CN102076355B (en) 2008-04-29 2014-05-07 Abbvie公司 Dual varistructure domain immunoglobulins and uses thereof
BRPI0911758A8 (en) 2008-05-09 2017-10-10 Abbott Lab ANTIBODIES FOR RECEPTOR OF ADVANCED GLYCATION END PRODUCTS (RAGE) AND USES THEREOF
EP3002299A1 (en) 2008-06-03 2016-04-06 AbbVie Inc. Dual variable domain immunoglobulins and uses thereof
US9109026B2 (en) 2008-06-03 2015-08-18 Abbvie, Inc. Dual variable domain immunoglobulins and uses thereof
RU2559525C2 (en) 2008-07-08 2015-08-10 Эббви Инк Proteins binding prostaglandin e2 and using them
KR20110031369A (en) 2008-07-08 2011-03-25 아보트 러보러터리즈 Prostaglandin e2 dual variable domain immunoglobulins and uses thereof
DK2982695T3 (en) 2008-07-09 2019-05-13 Biogen Ma Inc COMPOSITIONS CONCERNING ANTIBODIES AGAINST LINGO OR FRAGMENTS THEREOF
CA2741523C (en) 2008-10-24 2022-06-21 Jonathan S. Towner Human ebola virus species and compositions and methods thereof
EP2358392B1 (en) 2008-11-12 2019-01-09 MedImmune, LLC Antibody formulation
AU2009334498A1 (en) 2008-12-31 2011-07-21 Biogen Idec Ma Inc. Anti-lymphotoxin antibodies
EP2389195B1 (en) 2009-01-20 2015-05-20 Homayoun H. Zadeh Antibody mediated osseous regeneration
WO2010084408A2 (en) 2009-01-21 2010-07-29 Oxford Biotherapeutics Ltd. Pta089 protein
US8852608B2 (en) 2009-02-02 2014-10-07 Medimmune, Llc Antibodies against and methods for producing vaccines for respiratory syncytial virus
WO2010093993A2 (en) 2009-02-12 2010-08-19 Human Genome Sciences, Inc. Use of b lymphocyte stimulator protein antagonists to promote transplantation tolerance
EP2810652A3 (en) 2009-03-05 2015-03-11 AbbVie Inc. IL-17 binding proteins
WO2010100247A1 (en) 2009-03-06 2010-09-10 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research Novel therapy for anxiety
KR101667227B1 (en) 2009-03-10 2016-10-18 가부시키가이샤 진 테크노 사이언스 Generation, expression and characterization of the humanized k33n monoclonal antibody
EP2241323A1 (en) 2009-04-14 2010-10-20 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Tenascin-W and brain cancers
KR101736076B1 (en) 2009-04-20 2017-05-16 옥스포드 바이오테라퓨틱스 리미티드 Antibodies specific to cadherin-17
US20120213705A1 (en) 2009-06-22 2012-08-23 Medimmune, Llc ENGINEERED Fc REGIONS FOR SITE-SPECIFIC CONJUGATION
IE20090514A1 (en) 2009-07-06 2011-02-16 Opsona Therapeutics Ltd Humanised antibodies and uses therof
DK2464664T3 (en) 2009-08-13 2016-01-18 Crucell Holland Bv ANTIBODIES AGAINST HUMAN RESPIRATORY SYNCYTIAL VIRUS (RSV) AND METHODS FOR USING IT
EP2292266A1 (en) 2009-08-27 2011-03-09 Novartis Forschungsstiftung, Zweigniederlassung Treating cancer by modulating copine III
PE20121647A1 (en) 2009-08-29 2012-12-31 Abbvie Inc THERAPEUTIC BINDING PROTEINS TO DLL4
NZ598929A (en) 2009-09-01 2014-05-30 Abbvie Inc Dual variable domain immunoglobulins and uses thereof
WO2011028952A1 (en) 2009-09-02 2011-03-10 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
EP2480573A1 (en) 2009-09-22 2012-08-01 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Treating cancer by modulating mex-3
AR078470A1 (en) 2009-10-02 2011-11-09 Sanofi Aventis ANTIBODIES THAT SPECIFICALLY JOIN THE EPHA2 RECEIVER
WO2011047083A1 (en) 2009-10-13 2011-04-21 Oxford Biotherapeutics Ltd. Antibodies against epha10
EP2488548A1 (en) 2009-10-14 2012-08-22 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung A low density lipoprotein-related protein 1 splice variant as cancer marker
CN102666875A (en) 2009-10-15 2012-09-12 雅培制药有限公司 Dual variable domain immunoglobulins and uses thereof
WO2011045352A2 (en) 2009-10-15 2011-04-21 Novartis Forschungsstiftung Spleen tyrosine kinase and brain cancers
UY32979A (en) 2009-10-28 2011-02-28 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
US20120213801A1 (en) 2009-10-30 2012-08-23 Ekaterina Gresko Phosphorylated Twist1 and cancer
WO2011053707A1 (en) 2009-10-31 2011-05-05 Abbott Laboratories Antibodies to receptor for advanced glycation end products (rage) and uses thereof
WO2011054007A1 (en) 2009-11-02 2011-05-05 Oxford Biotherapeutics Ltd. Ror1 as therapeutic and diagnostic target
WO2011056073A2 (en) 2009-11-04 2011-05-12 Erasmus University Medical Center Rotterdam Novel compounds for modulating neovascularisation and methods of treatment using these compounds
US9244063B2 (en) 2009-11-11 2016-01-26 Gentian As Immunoassay for assessing related analytes of different origin
MX2012006560A (en) 2009-12-08 2012-10-05 Abbott Gmbh & Co Kg Monoclonal antibodies against the rgm a protein for use in the treatment of retinal nerve fiber layer degeneration.
US8362210B2 (en) 2010-01-19 2013-01-29 Xencor, Inc. Antibody variants with enhanced complement activity
RU2605928C2 (en) 2010-03-02 2016-12-27 Эббви Инк. Therapeutic dll4-binding proteins
WO2011107586A1 (en) 2010-03-05 2011-09-09 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research, Smoc1, tenascin-c and brain cancers
MX360403B (en) 2010-04-15 2018-10-31 Abbvie Inc Amyloid-beta binding proteins.
WO2011131611A1 (en) 2010-04-19 2011-10-27 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Modulating xrn1
US9011852B2 (en) 2010-04-30 2015-04-21 Alexion Pharmaceuticals, Inc. Anti-C5a antibodies
PE20130205A1 (en) 2010-05-14 2013-03-24 Abbvie Inc IL-1 BINDING PROTEINS
US20110293629A1 (en) 2010-05-14 2011-12-01 Bastid Jeremy Methods of Treating and/or Preventing Cell Proliferation Disorders with IL-17 Antagonists
EP2580239A1 (en) 2010-06-10 2013-04-17 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Treating cancer by modulating mammalian sterile 20-like kinase 3
WO2012006500A2 (en) 2010-07-08 2012-01-12 Abbott Laboratories Monoclonal antibodies against hepatitis c virus core protein
UY33492A (en) 2010-07-09 2012-01-31 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
NZ603488A (en) 2010-07-09 2015-02-27 Crucell Holland Bv Anti-human respiratory syncytial virus (rsv) antibodies and methods of use
BR112013002578A2 (en) 2010-08-03 2019-05-14 Abbvie Inc. double variable domain immunoglobins and their uses
US20130177555A1 (en) 2010-08-13 2013-07-11 Medimmune Limited Monomeric Polypeptides Comprising Variant FC Regions And Methods Of Use
JP6147665B2 (en) 2010-08-14 2017-06-14 アッヴィ・インコーポレイテッド Amyloid beta-binding protein
WO2012022734A2 (en) 2010-08-16 2012-02-23 Medimmune Limited Anti-icam-1 antibodies and methods of use
AU2011291462A1 (en) 2010-08-19 2013-03-14 Zoetis Belgium S.A. Anti-NGF antibodies and their use
AU2011293253B2 (en) 2010-08-26 2014-12-11 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
WO2012032143A1 (en) 2010-09-10 2012-03-15 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research Phosphorylated twist1 and metastasis
EP2640738A1 (en) 2010-11-15 2013-09-25 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Anti-fungal agents
SG10201604699VA (en) 2010-12-21 2016-07-28 Abbvie Inc Il-1 -alpha and -beta bispecific dual variable domain immunoglobulins and their use
TW201307388A (en) 2010-12-21 2013-02-16 Abbott Lab IL-1 binding proteins
US9505826B2 (en) 2010-12-22 2016-11-29 Teva Pharmaceuticals Australia Pty Ltd Modified antibody with improved half-life
DK2668210T3 (en) 2011-01-26 2020-08-24 Celldex Therapeutics Inc ANTI-KIT ANTIBODIES AND USES THEREOF
RU2625034C2 (en) 2011-04-20 2017-07-11 МЕДИММЬЮН, ЭлЭлСи Antibodies and other molecules binding b7-h1 and pd-1
KR101833465B1 (en) 2011-05-06 2018-02-28 넥스베트 오스트레일리아 피티와이 리미티드 Anti-nerve growth factor antibodies and methods of preparing and using the same
KR20170070272A (en) 2011-05-06 2017-06-21 넥스베트 오스트레일리아 피티와이 리미티드 Anti-nerve growth factor antibodies and methods of preparing and using the same
GB201114858D0 (en) 2011-08-29 2011-10-12 Nvip Pty Ltd Anti-nerve growth factor antibodies and methods of using the same
HRP20211869T1 (en) 2011-05-06 2022-03-04 Zoetis Services Llc Anti-nerve growth factor antibodies and methods of preparing and using the same
WO2012168259A1 (en) 2011-06-06 2012-12-13 Novartis Forschungsstiftung, Zweigniederlassung Protein tyrosine phosphatase, non-receptor type 11 (ptpn11) and triple-negative breast cancer
US9561274B2 (en) 2011-06-07 2017-02-07 University Of Hawaii Treatment and prevention of cancer with HMGB1 antagonists
US9244074B2 (en) 2011-06-07 2016-01-26 University Of Hawaii Biomarker of asbestos exposure and mesothelioma
EP3385280A1 (en) 2011-06-10 2018-10-10 MedImmune Limited Anti-pseudomonas psl binding molecules and uses thereof
LT2726094T (en) 2011-06-28 2017-02-10 Oxford Biotherapeutics Ltd Therapeutic and diagnostic target
PE20140756A1 (en) 2011-06-28 2014-07-04 Oxford Biotherapeutics Ltd ANTIBODIES THAT JOIN BST1
WO2013009648A2 (en) 2011-07-08 2013-01-17 Momenta Pharmaceuticals, Inc. Cell culture process
AU2012283039A1 (en) 2011-07-13 2014-01-30 Abbvie Inc. Methods and compositions for treating asthma using anti-IL-13 antibodies
GB201112056D0 (en) 2011-07-14 2011-08-31 Univ Leuven Kath Antibodies
CA2845536A1 (en) 2011-08-15 2013-02-21 Amplimmune, Inc. Anti-b7-h4 antibodies and their uses
KR20140063752A (en) 2011-08-30 2014-05-27 엔브이아이피 피티와이 리미티드 Caninised tumour necrosis factor antibodies and methods of using the same
WO2013034660A1 (en) 2011-09-09 2013-03-14 Medimmune Limited Anti-siglec-15 antibodies and uses thereof
BR112014009799A2 (en) 2011-10-24 2017-06-13 Abbvie Inc tnf directed immunoligants
SG11201401791WA (en) 2011-10-24 2014-08-28 Abbvie Inc Immunobinders directed against sclerostin
EP2773664A1 (en) 2011-11-01 2014-09-10 Bionomics, Inc. Anti-gpr49 antibodies
JP2014533247A (en) 2011-11-01 2014-12-11 バイオノミクス インコーポレイテッド Antibodies and methods of treating cancer
ES2697674T3 (en) 2011-11-01 2019-01-25 Bionomics Inc Procedures to block the growth of cancer stem cells
AU2012332593B2 (en) 2011-11-01 2016-11-17 Bionomics, Inc. Anti-GPR49 antibodies
WO2013068431A1 (en) 2011-11-08 2013-05-16 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research New treatment for neurodegenerative diseases
US20140294732A1 (en) 2011-11-08 2014-10-02 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute Early diagnostic of neurodegenerative diseases
US10118958B2 (en) 2011-12-14 2018-11-06 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
WO2013090633A2 (en) 2011-12-14 2013-06-20 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
CA3111357A1 (en) 2011-12-23 2013-06-27 Pfizer Inc. Engineered antibody constant regions for site-specific conjugation and methods and uses therefor
TW201333035A (en) 2011-12-30 2013-08-16 Abbvie Inc Dual specific binding proteins directed against IL-13 and/or IL-17
WO2013102825A1 (en) 2012-01-02 2013-07-11 Novartis Ag Cdcp1 and breast cancer
PL2807192T3 (en) 2012-01-27 2019-02-28 Abbvie Deutschland Composition and method for diagnosis and treatment of diseases associated with neurite degeneration
CN104254778A (en) 2012-02-10 2014-12-31 西雅图遗传学公司 Detection and treatment of cd30+ cancers
CN104203977A (en) 2012-02-15 2014-12-10 诺和诺德A/S(股份有限公司) Antibodies that bind peptidoglycan recognition protein 1
US9550830B2 (en) 2012-02-15 2017-01-24 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
EP2814844B1 (en) 2012-02-15 2017-08-02 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (trem-1)
EP2831112A1 (en) 2012-03-29 2015-02-04 Friedrich Miescher Institute for Biomedical Research Inhibition of interleukin- 8 and/or its receptor cxcrl in the treatment her2/her3 -overexpressing breast cancer
EP2841452B1 (en) 2012-04-25 2023-04-12 Momenta Pharmaceuticals, Inc. Modified glycoproteins
NZ702178A (en) 2012-05-14 2017-01-27 Biogen Ma Inc Lingo-2 antagonists for treatment of conditions involving motor neurons
MX362497B (en) 2012-05-15 2019-01-21 Eisai Inc Methods for treatment of gastric cancer.
CN104603149B (en) 2012-05-24 2017-06-30 万机集团有限公司 The composition related to prevention and treatment rabies infection and method
EP2859018B1 (en) 2012-06-06 2021-09-22 Zoetis Services LLC Caninized anti-ngf antibodies and methods thereof
WO2014001482A1 (en) 2012-06-29 2014-01-03 Novartis Forschungsstiftung, Zweigniererlassung, Friedrich Miescher Institute For Biomedical Research Treating diseases by modulating a specific isoform of mkl1
WO2014006114A1 (en) 2012-07-05 2014-01-09 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research New treatment for neurodegenerative diseases
EP2869818A1 (en) 2012-07-06 2015-05-13 Novartis AG Combination of a phosphoinositide 3-kinase inhibitor and an inhibitor of the il-8/cxcr interaction
AR091755A1 (en) 2012-07-12 2015-02-25 Abbvie Inc PROTEINS OF UNION TO IL-1
GB201213652D0 (en) 2012-08-01 2012-09-12 Oxford Biotherapeutics Ltd Therapeutic and diagnostic target
US9365641B2 (en) 2012-10-01 2016-06-14 The Trustees Of The University Of Pennsylvania Compositions and methods for targeting stromal cells for the treatment of cancer
EP2906241B1 (en) 2012-10-12 2020-01-08 The Brigham and Women's Hospital, Inc. Enhancement of the immune response
MY194330A (en) 2012-11-01 2022-11-28 Abbvie Inc Anti-dll4/vegf dual variable domain immunoglobulin and uses thereof
WO2014100542A1 (en) 2012-12-21 2014-06-26 Abbvie, Inc. High-throughput antibody humanization
JP6359031B2 (en) 2012-12-21 2018-07-18 メディミューン,エルエルシー Anti-H7CR antibody
GB201302447D0 (en) 2013-02-12 2013-03-27 Oxford Biotherapeutics Ltd Therapeutic and diagnostic target
WO2014129895A1 (en) 2013-02-19 2014-08-28 Stichting Vu-Vumc Means and method for increasing the sensitivity of cancers for radiotherapy
US9217168B2 (en) 2013-03-14 2015-12-22 Momenta Pharmaceuticals, Inc. Methods of cell culture
CN105209616A (en) 2013-03-14 2015-12-30 雅培制药有限公司 HCV NS3 recombinant antigens and mutants thereof for improved antibody detection
EP2968526A4 (en) 2013-03-14 2016-11-09 Abbott Lab Hcv antigen-antibody combination assay and methods and compositions for use therein
US8956830B2 (en) 2013-03-14 2015-02-17 Momenta Pharmaceuticals, Inc. Methods of cell culture
CN113549148A (en) 2013-03-14 2021-10-26 雅培制药有限公司 HCV core lipid binding domain monoclonal antibodies
CN105324396A (en) 2013-03-15 2016-02-10 艾伯维公司 Dual specific binding proteins directed against il-1 beta and il-17
KR20220041957A (en) 2013-03-15 2022-04-01 아비에 도이치란트 게엠베하 운트 콤파니 카게 Anti-egfr antibody drug conjugate formulations
US9469686B2 (en) 2013-03-15 2016-10-18 Abbott Laboratories Anti-GP73 monoclonal antibodies and methods of obtaining the same
EP2990485B1 (en) * 2013-04-25 2019-09-11 Kaneka Corporation Fd chain gene or l chain gene each capable of increasing secretion amount of fab-type antibody
US20160108450A1 (en) 2013-05-02 2016-04-21 Momenta Pharmaceutcals, Inc. Sialylated glycoproteins
AU2014268298B2 (en) 2013-05-24 2019-01-17 Medlmmune, Llc Anti-B7-H5 antibodies and their uses
SG11201509982UA (en) 2013-06-06 2016-04-28 Igenica Biotherapeutics Inc
EP3003372B1 (en) 2013-06-07 2019-10-09 Duke University Inhibitors of complement factor h
US20160151486A1 (en) 2013-06-13 2016-06-02 Fast Foward Pharmaceuticals B.V. CD40 Signalling Inhibitor and a Further Compound, Wherein the Further Compound is a Bile Acid, a Bile Acid Derivative, an TGR5-Receptor Agonist, an FXR Agonist or a Combination Thereof, for the Treatment of Chronic Inflammation, and the Prevention of Gastrointestinal Cancer or Fibrosis
US20160178610A1 (en) 2013-08-07 2016-06-23 Friedrich Miescher Institute For Biomedical Research New screening method for the treatment Friedreich's ataxia
DK3041507T3 (en) 2013-08-26 2021-07-26 Biontech Res And Development Inc NUCLEIC ACIDS ENCOODING HUMAN ANTIBODIES TO SIALYL-LEWIS A
WO2015050959A1 (en) 2013-10-01 2015-04-09 Yale University Anti-kit antibodies and methods of use thereof
AU2014329609B2 (en) 2013-10-02 2019-09-12 Humabs Biomed Sa Neutralizing anti-influenza A antibodies and uses thereof
WO2015057622A1 (en) 2013-10-16 2015-04-23 Momenta Pharmaceuticals, Inc. Sialylated glycoproteins
WO2015073575A2 (en) 2013-11-12 2015-05-21 Centre For Probe Development And Commercialization Residualizing linkers and uses thereof
CN106062005A (en) 2013-12-30 2016-10-26 医药生命融合研究团 Anti-krs monoclonal antibody and use thereof
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
NZ711451A (en) 2014-03-07 2016-05-27 Alexion Pharma Inc Anti-c5 antibodies having improved pharmacokinetics
US9738702B2 (en) 2014-03-14 2017-08-22 Janssen Biotech, Inc. Antibodies with improved half-life in ferrets
CA2944649C (en) 2014-04-04 2022-06-21 Bionomics, Inc. Humanized antibodies that bind lgr5
US20170267780A1 (en) 2014-05-16 2017-09-21 Medimmune, Llc Molecules with altered neonate fc receptor binding having enhanced therapeutic and diagnostic properties
RS61678B1 (en) 2014-05-28 2021-05-31 Agenus Inc Anti-gitr antibodies and methods of use thereof
KR102614642B1 (en) 2014-06-04 2023-12-19 바이오엔테크 리서치 앤드 디벨롭먼트 인코포레이티드 Human monoclonal antibodies to ganglioside gd2
US20170137824A1 (en) 2014-06-13 2017-05-18 Indranil BANERJEE New treatment against influenza virus
US10308935B2 (en) 2014-06-23 2019-06-04 Friedrich Miescher Institute For Biomedical Research Methods for triggering de novo formation of heterochromatin and or epigenetic silencing with small RNAS
EP3164129A1 (en) 2014-07-01 2017-05-10 Friedrich Miescher Institute for Biomedical Research Combination of a brafv600e inhibitor and mertk inhibitor to treat melanoma
CN106536559B (en) 2014-07-17 2021-04-27 诺和诺德股份有限公司 Site-directed mutagenesis of TREM-1 antibodies to reduce viscosity
US20170298360A1 (en) 2014-09-24 2017-10-19 Friedrich Miescher Institute For Biomedical Research Lats and breast cancer
MX2017004196A (en) 2014-10-01 2017-07-19 Medimmune Ltd Antibodies to ticagrelor and methods of use.
PT3333191T (en) 2014-12-11 2020-12-15 Pf Medicament Anti-c10orf54 antibodies and uses thereof
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
EP3789039A1 (en) 2014-12-22 2021-03-10 The Rockefeller University Anti-mertk agonistic antibodies and uses thereof
AU2016205197B2 (en) 2015-01-08 2021-10-21 Biogen Ma Inc. LINGO-1 antagonists and uses for treatment of demyelinating disorders
BR112017015880A2 (en) 2015-03-03 2018-07-31 Kymab Ltd antibodies, uses and methods
JP2018518152A (en) 2015-03-27 2018-07-12 ユニバーシティ オブ サザン カリフォルニア CAR T cell therapy directed to LHR for treating solid tumors
KR20180002782A (en) 2015-05-06 2018-01-08 얀센 바이오테크 인코포레이티드 Prostate specific membrane antigen (PSMA) bispecific binding agents and uses thereof
EP3292152A1 (en) 2015-05-07 2018-03-14 Agenus Inc. Anti-ox40 antibodies and methods of use thereof
HUE048284T2 (en) 2015-05-29 2020-07-28 Abbvie Inc Anti-cd40 antibodies and uses thereof
JP6900323B2 (en) 2015-05-29 2021-07-07 アジェナス インコーポレイテッド Anti-CTLA-4 antibody and how to use it
WO2016197064A1 (en) 2015-06-04 2016-12-08 Epstein Alan L Lym-1 and lym-2 targeted car cell immunotherapy
TW201710286A (en) 2015-06-15 2017-03-16 艾伯維有限公司 Binding proteins against VEGF, PDGF, and/or their receptors
EP3344656A1 (en) 2015-09-01 2018-07-11 Agenus Inc. Anti-pd-1 antibodies and methods of use thereof
EP3569244A1 (en) 2015-09-23 2019-11-20 CytoImmune Therapeutics, LLC Flt3 directed car cells for immunotherapy
EP3368566A1 (en) 2015-10-28 2018-09-05 Friedrich Miescher Institute for Biomedical Research Tenascin-w and biliary tract cancers
EP3909983A1 (en) 2015-12-02 2021-11-17 STCube & Co. Inc. Antibodies and molecules that immunospecifically bind to btn1a1 and the therapeutic uses thereof
KR20180100122A (en) 2015-12-02 2018-09-07 주식회사 에스티사이언스 Antibodies specific for glycated BTLA (B- and T-lymphocyte weakening factor)
WO2017097889A1 (en) 2015-12-10 2017-06-15 Katholieke Universiteit Leuven Anti adamts13 antibodies and their use for treatment or prevention of haemorrhagic disorders due to ventricular assist device
AU2017239038A1 (en) 2016-03-22 2018-10-04 Bionomics Inc Administration of an anti-LGR5 monoclonal antibody
WO2017187183A1 (en) 2016-04-27 2017-11-02 Itara Therapeutics Methods for the identification of bifunctional compounds
SG11201810023QA (en) 2016-05-27 2018-12-28 Agenus Inc Anti-tim-3 antibodies and methods of use thereof
US11427632B2 (en) 2016-07-06 2022-08-30 Celgene Corporation Antibodies with low immunogenicity and uses thereof
CN109689685A (en) 2016-07-08 2019-04-26 斯塔滕生物技术有限公司 Anti- APOC3 antibody and its application method
EP3519824A1 (en) 2016-10-03 2019-08-07 Abbott Laboratories Improved methods of assessing uch-l1 status in patient samples
CA3037380A1 (en) 2016-10-11 2018-04-19 Agenus Inc. Anti-lag-3 antibodies and methods of use thereof
KR20190107656A (en) 2016-11-02 2019-09-20 이뮤노젠 아이엔씨 Combination treatment with antibody-drug conjugates and PARP inhibitors
EP3534947A1 (en) 2016-11-03 2019-09-11 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses & methods
KR102539159B1 (en) 2016-11-07 2023-06-02 주식회사 뉴라클사이언스 Anti-family with sequence similarity 19, member a5 antibodies and method of use thereof
CA3046082A1 (en) 2016-12-07 2018-06-14 Agenus Inc. Antibodies and methods of use thereof
KR102504605B1 (en) 2016-12-07 2023-03-02 아게누스 인코포레이티드 Anti-CTLA-4 Antibodies and Methods of Using The Same
US11016092B2 (en) 2017-03-23 2021-05-25 Abbott Laboratories Methods for aiding in the diagnosis and determination of the extent of traumatic brain injury in a human subject using the early biomarker ubiquitin carboxy-terminal hydrolase L1
US20200330590A1 (en) 2017-03-27 2020-10-22 Celgene Corporation Methods and compositions for reduction of immunogenicity
BR112019017241A2 (en) 2017-04-13 2020-04-14 Agenus Inc anti-cd137 antibodies and methods of using them
BR112019021612A2 (en) 2017-04-15 2020-05-12 Abbott Laboratories METHODS TO ASSIST HYPERAGUTE DIAGNOSIS AND DETERMINATION OF CRANIENCEPHALIC TRAUMATISM IN A HUMAN INDIVIDUAL WITH THE USE OF EARLY BIOMARKERS
MA50958A (en) 2017-04-21 2020-10-14 Staten Biotechnology B V ANTI-APOC3 ANTIBODIES AND THEIR METHODS OF USE
EP3635407A1 (en) 2017-04-28 2020-04-15 Abbott Laboratories Methods for aiding in the hyperacute diagnosis and determination of traumatic brain injury using early biomarkers on at least two samples from the same human subject
CA3062061A1 (en) 2017-05-01 2018-11-08 Agenus Inc. Anti-tigit antibodies and methods of use thereof
JP2020518673A (en) 2017-05-05 2020-06-25 フュージョン・ファーマシューティカルズ・インコーポレイテッド Enhanced pharmacokinetics of bifunctional chelates and their use
US10865238B1 (en) 2017-05-05 2020-12-15 Duke University Complement factor H antibodies
KR20200004861A (en) 2017-05-05 2020-01-14 퓨전 파마슈티칼즈 인크. IGF-1R monoclonal antibodies and uses thereof
JOP20190256A1 (en) 2017-05-12 2019-10-28 Icahn School Med Mount Sinai Newcastle disease viruses and uses thereof
CN110651190A (en) 2017-05-25 2020-01-03 雅培实验室 Method for using early biomarkers to help determine whether to perform imaging on a human subject who has suffered or may have suffered a head injury
US11129564B2 (en) 2017-05-30 2021-09-28 Abbott Laboratories Methods for aiding in diagnosing and evaluating a mild traumatic brain injury in a human subject using cardiac troponin I
KR20200015602A (en) 2017-05-31 2020-02-12 주식회사 에스티큐브앤컴퍼니 Antibodies and molecules immunospecifically binding to BTN1A1 and therapeutic uses thereof
CN111051346A (en) 2017-05-31 2020-04-21 斯特库伯株式会社 Methods of treating cancer using antibodies and molecules that immunospecifically bind to BTN1a1
EP3635007A1 (en) 2017-06-06 2020-04-15 STCube & Co., Inc. Methods of treating cancer using antibodies and molecules that bind to btn1a1 or btn1a1-ligands
GB201709379D0 (en) 2017-06-13 2017-07-26 Univ Leuven Kath Humanised ADAMTS13 binding antibodies
US11169159B2 (en) 2017-07-03 2021-11-09 Abbott Laboratories Methods for measuring ubiquitin carboxy-terminal hydrolase L1 levels in blood
EA202090247A1 (en) 2017-07-14 2020-05-12 Цитомкс Терапьютикс, Инк. ANTIBODIES AGAINST CD166 AND THEIR APPLICATION
US11707522B2 (en) 2017-10-13 2023-07-25 Boehringer Ingelheim International Gmbh Human antibodies to Tn antigen
WO2019087115A1 (en) 2017-10-31 2019-05-09 Staten Biotechnology B.V. Anti-apoc3 antibodies and methods of use thereof
WO2019089594A1 (en) 2017-10-31 2019-05-09 Immunogen, Inc. Combination treatment with antibody-drug conjugates and cytarabine
EP3706795A4 (en) 2017-11-09 2021-10-13 Pinteon Therapeutics Inc. Methods and compositions for the generation and use of humanized conformation-specific phosphorylated tau antibodies
CN111094983A (en) 2017-12-09 2020-05-01 雅培实验室 Methods of using Glial Fibrillary Acidic Protein (GFAP) and/or ubiquitin carboxy-terminal hydrolase L1(UCH-L1) to aid in the diagnosis and evaluation of patients who have suffered orthopedic injury and who have suffered or may have suffered a head injury such as mild Traumatic Brain Injury (TBI)
US11016105B2 (en) 2017-12-09 2021-05-25 Abbott Laboratories Methods for aiding in diagnosing and evaluating a traumatic brain injury in a human subject using a combination of GFAP and UCH-L1
MA51154A (en) 2017-12-15 2020-10-21 Aleta Biotherapeutics Inc CD19 VARIANTS
BR112020017701A2 (en) 2018-03-12 2020-12-29 Zoetis Services Llc ANTI-NGF ANTIBODIES AND METHODS OF THE SAME
KR20200139219A (en) 2018-04-02 2020-12-11 브리스톨-마이어스 스큅 컴퍼니 Anti-TREM-1 antibodies and uses thereof
JP2021521273A (en) 2018-04-12 2021-08-26 メディアファーマ エス.アール.エル. LGALS3BP antibody-drug conjugate and its use for cancer treatment
EP3784274A1 (en) 2018-04-27 2021-03-03 Fondazione Ebri Rita Levi-Montalcini Antibody directed against a tau-derived neurotoxic peptide and uses thereof
AU2019265888A1 (en) 2018-05-10 2020-11-26 Neuracle Science Co., Ltd. Anti-family with sequence similarity 19, member A5 antibodies and method of use thereof
WO2019222130A1 (en) 2018-05-15 2019-11-21 Immunogen, Inc. Combination treatment with antibody-drug conjugates and flt3 inhibitors
TW202016144A (en) 2018-06-21 2020-05-01 日商第一三共股份有限公司 Compositions including cd3 antigen binding fragments and uses thereof
EP3824287A1 (en) 2018-07-20 2021-05-26 Pierre Fabre Médicament Receptor for vista
JP2022504287A (en) 2018-10-03 2022-01-13 スターテン・バイオテクノロジー・ベー・フェー Antibodies specific for human and cynomolgus monkey APOC3, and methods of their use
PE20211286A1 (en) 2018-12-03 2021-07-19 Fusion Pharmaceuticals Inc COMBINATION THERAPY OF RADIOIMMUNOCONJUGATES WITH CONTROL POINT INHIBITOR
CA3130303A1 (en) 2019-02-26 2020-09-03 Rgenix, Inc. High-affinity anti-mertk antibodies and uses thereof
CN113874392A (en) 2019-03-28 2021-12-31 丹尼斯科美国公司 Engineered antibodies
JP2022545368A (en) 2019-08-12 2022-10-27 アプティーボ リサーチ アンド デベロップメント エルエルシー 4-1BB and 0X40 Binding Proteins and Related Compositions and Methods, Antibodies to 4-1BB, Antibodies to 0X40
AU2020335928A1 (en) 2019-08-30 2022-02-17 Agenus Inc. Anti-CD96 antibodies and methods of use thereof
US20220306736A1 (en) 2019-09-04 2022-09-29 Y-Biologics Inc. Anti-vsig4 antibody or antigen binding fragment and uses thereof
US20220411511A1 (en) 2019-09-26 2022-12-29 Stcube & Co. Antibodies specific to glycosylated ctla-4 and methods of use thereof
US20220356248A1 (en) 2019-10-09 2022-11-10 Stcube & Co Antibodies specific to glycosylated lag3 and methods of use thereof
CN113416258B (en) * 2019-10-24 2023-08-29 北京免疫方舟医药科技有限公司 Multispecific antibody and preparation method and application thereof
KR20220137698A (en) 2020-02-05 2022-10-12 라리마 테라퓨틱스, 인코포레이티드 TAT peptide binding protein and uses thereof
WO2021202473A2 (en) 2020-03-30 2021-10-07 Danisco Us Inc Engineered antibodies
EP4136459A1 (en) 2020-04-13 2023-02-22 Abbott Laboratories Methods, complexes and kits for detecting or determining an amount of a ss-coronavirus antibody in a sample
KR20230005268A (en) 2020-04-24 2023-01-09 밀레니엄 파머슈티컬스 인코퍼레이티드 Anti-CD19 Antibodies and Uses Thereof
WO2022031804A1 (en) 2020-08-04 2022-02-10 Abbott Laboratories Improved methods and kits for detecting sars-cov-2 protein in a sample
WO2022081436A1 (en) 2020-10-15 2022-04-21 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Antibody specific for sars-cov-2 receptor binding domain and therapeutic methods
WO2022087274A1 (en) 2020-10-21 2022-04-28 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Antibodies that neutralize type-i interferon (ifn) activity
CA3198161A1 (en) 2020-12-01 2022-06-09 Beth MCQUISTON Use of one or more biomarkers to determine traumatic brain injury (tbi) in a subject having received a head computerized tomography scan that is negative for a tbi
IL303328A (en) 2020-12-01 2023-07-01 Aptevo Res & Development Llc Heterodimeric psma and cd3-binding bispecific antibodies
WO2023102384A1 (en) 2021-11-30 2023-06-08 Abbott Laboratories Use of one or more biomarkers to determine traumatic brain injury (tbi) in a subject having received a head computerized tomography scan that is negative for a tbi
EP4271998A1 (en) 2020-12-30 2023-11-08 Abbott Laboratories Methods for determining sars-cov-2 antigen and anti-sars-cov-2 antibody in a sample
WO2022153195A1 (en) 2021-01-13 2022-07-21 Memorial Sloan Kettering Cancer Center Anti-dll3 antibody-drug conjugate
TW202237135A (en) 2021-01-13 2022-10-01 紀念斯隆凱特琳癌症中心 Antibody-pyrrolobenzodiazepine derivative conjugate
KR20230156727A (en) 2021-03-03 2023-11-14 피에르 파브르 메디카먼트 Anti-VSIG4 antibody or antigen-binding fragment thereof and uses
WO2022215054A1 (en) 2021-04-09 2022-10-13 Takeda Pharmaceutical Company Limited Antibodies targeting complement factor d and uses therof
JP2024516645A (en) 2021-04-26 2024-04-16 ミレニアム ファーマシューティカルズ, インコーポレイテッド Anti-CLEC12A antibodies and uses thereof
AR125450A1 (en) 2021-04-26 2023-07-19 Millennium Pharm Inc ANTI-ADGRE2 ANTIBODIES AND USES THEREOF
JP2024519858A (en) 2021-05-18 2024-05-21 アボット・ラボラトリーズ Methods for assessing brain injury in pediatric subjects
EP4348263A1 (en) 2021-05-28 2024-04-10 Alexion Pharmaceuticals, Inc. Methods for detecting cm-tma biomarkers
EP4355776A1 (en) 2021-06-14 2024-04-24 Argenx BV Anti-il-9 antibodies and methods of use thereof
EP4356129A1 (en) 2021-06-14 2024-04-24 Abbott Laboratories Methods of diagnosing or aiding in diagnosis of brain injury caused by acoustic energy, electromagnetic energy, an over pressurization wave, and/or blast wind
WO2023034777A1 (en) 2021-08-31 2023-03-09 Abbott Laboratories Methods and systems of diagnosing brain injury
AU2022354059A1 (en) 2021-09-30 2024-03-28 Abbott Laboratories Methods and systems of diagnosing brain injury
TW202330612A (en) 2021-10-20 2023-08-01 日商武田藥品工業股份有限公司 Compositions targeting bcma and methods of use thereof
EP4177266A1 (en) 2021-11-05 2023-05-10 Katholieke Universiteit Leuven Neutralizing anti-sars-cov-2 human antibodies
WO2023114978A1 (en) 2021-12-17 2023-06-22 Abbott Laboratories Systems and methods for determining uch-l1, gfap, and other biomarkers in blood samples
US20230213536A1 (en) 2021-12-28 2023-07-06 Abbott Laboratories Use of biomarkers to determine sub-acute traumatic brain injury (tbi) in a subject having received a head computerized tomography (ct) scan that is negative for a tbi or no head ct scan
WO2023150652A1 (en) 2022-02-04 2023-08-10 Abbott Laboratories Lateral flow methods, assays, and devices for detecting the presence or measuring the amount of ubiquitin carboxy-terminal hydrolase l1 and/or glial fibrillary acidic protein in a sample
WO2023192436A1 (en) 2022-03-31 2023-10-05 Alexion Pharmaceuticals, Inc. Singleplex or multiplexed assay for complement markers in fresh biological samples
WO2023240124A1 (en) 2022-06-07 2023-12-14 Regeneron Pharmaceuticals, Inc. Pseudotyped viral particles for targeting tcr-expressing cells
WO2024006876A1 (en) 2022-06-29 2024-01-04 Abbott Laboratories Magnetic point-of-care systems and assays for determining gfap in biological samples
WO2024015953A1 (en) 2022-07-15 2024-01-18 Danisco Us Inc. Methods for producing monoclonal antibodies
WO2024050524A1 (en) 2022-09-01 2024-03-07 University Of Georgia Research Foundation, Inc. Compositions and methods for directing apolipoprotein l1 to induce mammalian cell death
WO2024054436A1 (en) 2022-09-06 2024-03-14 Alexion Pharmaceuticals, Inc. Diagnostic and prognostic biomarker profiles in patients with hematopoietic stem cell transplant-associated thrombotic microangiopathy (hsct-tma)
WO2024059708A1 (en) 2022-09-15 2024-03-21 Abbott Laboratories Biomarkers and methods for differentiating between mild and supermild traumatic brain injury

Also Published As

Publication number Publication date
JPH07503124A (en) 1995-04-06
EP0590067A1 (en) 1994-04-06
AU2238292A (en) 1993-01-12
WO1992022324A1 (en) 1992-12-23

Similar Documents

Publication Publication Date Title
CA2110799A1 (en) Microbially-produced antibody fragments and their conjugates
US5744580A (en) Immunotoxins comprising ribosome-inactivating proteins
CA2162689C (en) Immunotoxins comprising gelonin and an antibody
JP2761012B2 (en) Recombinant DNA products and methods
US5824782A (en) Immunoconjugates II
US5877305A (en) DNA encoding biosynthetic binding protein for cancer marker
EP0856054B1 (en) Monoclonal antibody br110 and uses thereof
US5416202A (en) Materials comprising and methods of preparation and use for ribosome-inactivating proteins
US5892019A (en) Production of a single-gene-encoded immunoglobulin
US6146850A (en) Proteins encoding gelonin sequences
US5837491A (en) Polynucleotides encoding gelonin sequences
Dunn et al. Antigen binding and cytotoxic properties of a recombinant immunotoxin incorporating the lytic peptide, melittin
JPH09509043A (en) A novel ribosome-inactivating protein isolated from the plant Bryonia dioica
Ogata et al. Analysis of Pseudomonas exotoxin activation and conformational changes by using monoclonal antibodies as probes
Brüsselbach et al. Enzyme recruitment and tumor cell killing in vitro by

Legal Events

Date Code Title Description
FZDE Dead