throbber
04/03/2001
`
`04:18 PM Cancer Research-V.81
`
`PS039-03.tex
`
`PS039-03.xml
`
`LaTeX2e(2000/11/15)
`
`Textures 2.0
`
`Recombinant Immunotoxins in
`Targeted Cancer Cell Therapy
`
`Yoram Reiter
`Faculty of Biology
`Technion-Israel Institute of Technology
`Haifa 3200, Israel
`
`I. Introduction
`II. Design of Recombinant Immunotoxins
`A. The Toxin Moiety
`B. The Targeting Moiety—Recombinant Antibody Fragments
`III. Construction and Production of Recombinant Immunotoxins
`IV. Preclinical Development of Recombinant Immunotoxins
`V. Application of Recombinant Immunotoxins
`A. Recombinant Immunotoxins against Solid Tumors
`B. Recombinant Immunotoxins against Leukemias and Lymphomas
`VI. Other Applications of Recombinant Antibody Fragments
`A. Radioimaging and Radioimmunotherapy
`B. Prodrug Therapy with Fv-Enzyme Fusion Proteins
`VII. Challenges and Future Directions of Recombinant Immunotoxins
`A. Immune Responses and Dose-Limiting Toxicity
`B. Specificity
`References
`
`Targeted cancer therapy in general and immunotherapy in particular combines ratio-
`nal drug design with the progress in understanding cancer biology. This approach takes
`advantage of our recent knowledge of the mechanisms by which normal cells are trans-
`formed into cancer cells, thus using the special properties of cancer cells to device novel
`therapeutic strategies.
`Recombinant immunotoxins are excellent examples of such processes, combining the
`knowledge of antigen expression by cancer cells with the enormous developments in
`recombinant DNA technology and antibody engineering.
`Recombinant immunotoxins are composed of a very potent protein toxin fused to
`a targeting moiety such as a recombinant antibody fragment or growth factor. These
`molecules bind to surface antigens specific for cancer cells and kill the target cells by
`catalytic inhibition of protein synthesis.
`Recombinant immunotoxins are developed for solid tumors and hematological ma-
`lignancies and have been characterized intensively for their biological activity in vitro on
`cultured tumor cell lines as well as in vivo in animal models of human tumor xenografts.
`The excellent in vitro and in vivo activities of recombinant immunotoxins have lead to
`their preclinical development and to the initiation of clinical trail protocols. Recent trail
`
`Advances in CANCER RESEARCH
`0065-230X/01 $35.00
`
`93
`
`Copyright (cid:2)C 2001 by Academic Press.
`All rights of reproduction in any form reserved.
`
`IMMUNOGEN 2152, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`04/03/2001
`
`04:18 PM Cancer Research-V.81
`
`PS039-03.tex
`
`PS039-03.xml
`
`LaTeX2e(2000/11/15)
`
`Textures 2.0
`
`94
`
`Yoram Reiter
`
`results have demonstrated potent clinical efficacy in patients with malignant diseases that
`are refractory to traditional modalities of cancer treatment: surgery, radiation therapy,
`and chemotherapy.
`The results demonstrate that such strategies can be developed into a separate modality
`of cancer treatment with the basic rationale of specifically targeting cancer cells on the
`basis of their unique surface markers. Efforts are now being made to improve the current
`molecules and to develop new agents with better clinical efficacy. This can be achieved
`by development of novel targeting moieties with improved specificity that will reduce
`toxicity to normal tissues.
`In this review, the design, construction, characterization, and applications of recom-
`binant immunotoxins are described. Results of recent clinical trails are presented, and
`future directions for development of recombinant immunotoxins as a new modality for
`cancer treatment are discussed. (cid:2)C 2001 Academic Press.
`
`I. INTRODUCTION
`
`The rapid progress in understanding the molecular biology of cancer cells
`has made a large impact on the design and development of novel therapeutic
`strategies. These are developed because treatment of cancer by chemotherapy
`is limited by a number of factors and usually fails in patients whose malig-
`nant cells are not sufficiently different from normal cells in their growth and
`metabolism. Other limiting factors are the low therapeutic index of most
`chemotherapeutic agents, the emergence of drug-resistant populations, tu-
`mor heterogeneity, and the presence of metastatic disease. The concept of
`targeted cancer therapy is thus an important means to improve the thera-
`peutic potential of anticancer agents and lead to the development of novel
`approaches such as immunotherapy.
`The approach of cancer immunotherapy and targeted cancer therapy com-
`bines rational drug design with the progress in understanding cancer biology
`(1–4). This approach takes advantage of some special properties of cancer
`cells: many of them contain mutant or overexpressed oncogenes on their
`surface, and these proteins are attractive antigens for targeted therapy. The
`first cell-surface receptor to be linked to cancer was the EGF receptor, which
`is present in lung, brain, kidney, bladder, breast, and ovarian cancer (5, 6).
`Several other members of the EGF family of receptors, the erbB2, erbB3, and
`erbB4 receptors, appear to be abundant on tumors of breast and ovary and
`erbB2, for example, is the target for Phase I and II immunotherapy clinical
`trails (7, 8).
`Other promising candidates for targeted therapy are differentiation anti-
`gens that are expressed on the surface of mature cells but not on the immature
`stem cells.
`The most widely studied examples of differentiation antigens which are
`currently being used for targeted therapy are expressed by hematopoietic
`
`IMMUNOGEN 2152, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`04/03/2001
`
`04:18 PM Cancer Research-V.81
`
`PS039-03.tex
`
`PS039-03.xml
`
`LaTeX2e(2000/11/15)
`
`Textures 2.0
`
`Recombinant Immunotoxins
`
`95
`
`malignancies and include CD19, CD20, and CD22 on B-cell lymphomas
`and leukemias and the IL-2 receptor on T-cell leukemias (9–11). Differenti-
`ation antigens have also been found on ovarian, breast, and prostate cancer
`(12–14).
`Another class of antigens, termed tumor-associated antigens (TAA), are
`molecules which are tightly bound to the surface of cancer cells and are as-
`sociated with the transformed cancer cells. An example is the carbohydrate
`antigen Lewis Y, which is found in many types of solid tumors (15). An-
`other class of TAAs are cancer peptides that are presented by class I MHC
`molecules on the surface of tumor cells (16, 17).
`It should be possible to use these molecular cell-surface markers as targets
`to eliminate the cancer cells while sparing the normal cells. For this approach
`to be successful, we must generate a targeting moiety which will bind very
`specifically the antigen or receptor expressed on the cancer cell surface and
`arm this targeting moiety with an effector cytotoxic moiety. The targeting
`moiety can be a specific antibody directed toward the cancer antigen or
`a ligand for specific overexpressed receptor. The cytotoxic arm can be a
`radioisotope, a cytotoxic drug, or a toxin. One strategy to achieve this is
`to arm antibodies that target cancer cells with powerful toxins which can
`originate from both plants and bacteria. The molecules generated are termed
`recombinant immunotoxins.
`The goal of immunotoxin therapy is to target a very potent cytotoxic agent
`to cell surface molecules which will internalize the cytotoxic agent and result
`in cell death. Developing this type of therapy has attracted much interest in
`the past years. Since immunotoxins differ greatly from chemotherapy in their
`mode of action and toxicity profile, it is hoped that immunotoxins will have
`the potential to improve the systemic treatment of tumors incurable with
`existing modes of therapy.
`As shown in Fig. 1, immunotoxins can be divided into two groups: chemi-
`cal conjugates (or first-generation immunotoxins) and second-generation (or
`recombinant immunotoxins). They both contain toxins that have their cell-
`binding domains either mutated or deleted to prevent them from binding to
`normal cells, and that are either fused or chemically conjugated to a ligand
`or an antibody specific for cancer cells (Table I).
`First-generation immunotoxins, composed of whole antibodies chemi-
`cally conjugated to toxins, demonstrated the feasibility of this concept.
`Cancer cells cultured in vitro could be killed under conditions in
`which the immunotoxin demonstrated low toxicity toward cultured nor-
`mal cells. Clinical trials with these agents had some success; however, they
`also revealed several problems, such as nonspecific toxicity toward
`some normal cells, difficulties in production, and, particularly for the
`treatment of solid tumors, poor tumor penetration owing to their large
`size.
`
`IMMUNOGEN 2152, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`04/03/2001
`
`04:18 PM Cancer Research-V.81
`
`PS039-03.tex
`
`PS039-03.xml
`
`LaTeX2e(2000/11/15)
`
`Textures 2.0
`
`96
`
`Yoram Reiter
`
`Fig. 1 Immunotoxins for targeted cancer therapy. First-generation immunotoxins are whole
`monoclonal antibodies to which the toxin is chemically conjugated. Second-generation immuno-
`toxins made by recombinant DNA technology by fusing recombinant antibody fragments to
`the toxin (usually a truncated or mutated form of the toxin).
`Three types of recombinant antibody fragments are used as the targeting moiety in recom-
`binant immunotoxins. Fabs are composed of the light-chain and the heavy-chain Fd fragments
`(VH and CH1), connected to each other via the interchain disulfide bond between CL and CH1.
`ScFv fragments are stabilized by a peptide linker which connects the carboxyl terminus of VH
`or VL with the amino terminus of the other domain. The VH and VL heterodimer in dsFv
`is stabilized by engineering a disulfide bond between the two domains. The biochemical and
`biological properties described in the figure are depicted for B3-lysPE38 (LMB-1) (89) (a first-
`generation antibody–PE chemical conjugate), B3(Fv)-PE38 (LMB-7) (67) (second-generation
`recombinant scFv-immunotoxin for a scFv-immunotoxin), and B3(dsFv)-PE38 (LMB-9) (74)
`(for a second-generation recombinant dsFv-immunotoxin).
`
`Second-generation immunotoxins have overcome many of these problems.
`Progress in the elucidation of the toxins’ structure and function, combined
`with the techniques of protein engineering, facilitated the design and con-
`struction of recombinant molecules with a higher specificity for cancer cells
`and reduced toxicity to normal cells. At the same time, advances in recom-
`binant DNA technology and antibody engineering enabled the generation
`of small antibody fragments. Thus, it was possible to decrease the size of
`immunotoxins significantly and to improve their tumor-penetration poten-
`tial in vivo. The development of advanced methods of recombinant-protein
`
`IMMUNOGEN 2152, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`04/03/2001
`
`04:18 PM Cancer Research-V.81
`
`PS039-03.tex
`
`PS039-03.xml
`
`LaTeX2e(2000/11/15)
`
`Textures 2.0
`
`Recombinant Immunotoxins
`
`97
`
`Table I Examples of Recombinant Immunotoxins against Cancer
`
`Immunotoxin
`
`Antigen
`
`Toxin
`
`Cancer
`
`Clinical trail References
`
`Anti CD7-dgA
`
`CD7
`
`DAB389-IL2
`
`IL-2R
`
`Anti-Tac (Fv)-PE38 CD25
`(LMB-2)
`DT-Anti-Tac(Fv)
`
`CD25
`
`RFB4(dsFv)-PE38
`Di-dgA-RFB4
`
`CD22
`CD22
`
`B3-lysPE38
`(LMB-1)
`B3(Fv)-PE38
`(LMB-7)
`B3(dsFv)-PE38
`(LMB-9)
`BR96(sFv)-PE40
`e23(Fv)-PE38
`FRP5(scFv)ETA
`Tf-CRM107
`HB21(Fv)-PE40
`MR1(Fv)-PE38
`SSI(Fv)-PE38
`
`DT
`
`PE
`
`DT
`
`Ricin Non-Hodgkin’s
`lymphoma
`T-cell lymphoma,
`Hodgkin’s disease
`B and T lymphoma,
`leukemias
`Leukemias,
`lymphoma
`B leukemias
`PE
`Ricin Leukemias,
`non-Hodgkin’s
`lymphoma
`Carcinomas
`
`Lewis Y
`
`Lewis Y
`
`Lewis Y
`
`PE
`
`PE
`
`PE
`
`Carcinomas
`
`Carcinoma
`
`PE
`Lewis Y
`PE
`erbB2/HER2
`PE
`erbB2/HER2
`DT
`Transferrin-R
`PE
`Transferrin-R
`Mutant EGF-R PE
`Mesothelin
`PE
`
`Carcinoma
`Breast cancer
`Breast cancer
`Glioma
`Various
`Liver, brain tumors
`Ovarian cancer
`
`Phase I
`
`156
`
`Phase III
`
`122, 125
`
`Phase I
`
`66,106
`
`—
`
`Phase I
`—
`
`Phase I
`
`Phase I
`
`Phase I
`
`—
`Phase I
`—
`Phase I
`—
`—
`—
`
`78
`
`76
`157
`
`89
`
`67
`
`74
`
`95
`68
`96
`103
`55
`99
`100
`
`production enabled the large-scale production of recombinant immunotox-
`ins of high purity and quality for clinical use in sufficient quantities to per-
`form clinical trials.
`Another strategy to target cancer cells is to construct chimeric toxins in
`which the engineered truncated portion of the toxin (PE or DT) gene is fused
`to cDNA encoding growth factors or cytokines. These include transforming
`growth factor (TGF)-␣ (18), insuline-like growth factor (IGF)-1 (19), acidic
`and basic fibroblast growth factor (FGF) (20), IL2 (21), IL4 (22), and IL6
`(23). These recombinant toxins (oncotoxins) are designed to target specific
`tumor cells that overexpress these receptors.
`This review will summarize knowledge of the design and application of
`second-generation recombinant Fv immunotoxins, which utilize recombi-
`nant antibody fragments as the targeting moiety, in the treatment of cancer,
`
`IMMUNOGEN 2152, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`04/03/2001
`
`04:18 PM Cancer Research-V.81
`
`PS039-03.tex
`
`PS039-03.xml
`
`LaTeX2e(2000/11/15)
`
`Textures 2.0
`
`98
`
`Yoram Reiter
`
`and will also discuss briefly the use of recombinant antibody fragments for
`other modes of cancer therapy and diagnosis.
`
`II. DESIGN OF RECOMBINANT IMMUNOTOXINS
`
`A. The Toxin Moiety
`
`The toxins that are most commonly used to make immunotoxins are ricin,
`diphtheria toxin (DT), and Pseudomonas exotoxin (PE). The genes for these
`toxins have been cloned and expressed in Eschesichia coli, and the crystal
`structures of all three proteins have been solved (36, 41). This information,
`in combination with mutational studies, has elucidated which toxin subunits
`are involved in their biological activity and, most important, the different
`steps of the cytocidal process. DT, PE, and ricin, and their derivatives, have
`all been successfully used to prepare immunotoxin conjugates (3, 24), but
`only PE- and DT-containing fusion proteins generate active recombinant
`immunotoxins (1, 25). This is because the toxic moiety must be separated
`from the binding moiety after internalization (26, 27). PE and DT fusion
`proteins generate their free toxic moieties by proteolytic processing. Ricin
`does not possess such a proteolytic processing site, and therefore cannot be
`attached to the targeting moiety with a peptide bond without losing cytotoxic
`activity. Recently, proteolytic processing sites were introduced into ricin by
`recombinant DNA techniques to try to overcome this problem (28).
`
`1. DIPHTHERIA TOXIN AND DT DERIVATIVES
`
`DT is a 58-kDa protein, secreted by pathogenic Corynebacterium diph-
`theria, which contain a lysogenic beta phage (29). DT ADP-ribosylates eu-
`karyotic elongation factor 2 (EF2) at a “diphthamide” residue located at
`+
`His 415, using NAD
`as a cofactor (30). This modification arrests protein
`synthesis and subsequently leads to cell death (31). Only a few, and perhaps
`only one, DT molecule needs to reach the cytosol in order to kill a cell. When
`DT is isolated from the culture medium of C. diphtheria, it is composed of
`an N-terminal 21-kDa A subunit and a C-terminal 37-kDa B subunit held
`together by a disulfide bond. DT is the expression product of a single gene
`(29), which, when secreted into the medium, is processed into two fragments
`by extracellular proteases. When DT is produced as a recombinant single-
`chain protein in E. coli, it is not cleaved by the bacteria but is instead cleaved
`by a protease in the target cells (32).
`The A domain of DT contains its enzymatic activity. The N terminus of
`the B subunit of DT (or the region between A and B in single-chain DT)
`
`IMMUNOGEN 2152, pg. 6
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`04/03/2001
`
`04:18 PM Cancer Research-V.81
`
`PS039-03.tex
`
`PS039-03.xml
`
`LaTeX2e(2000/11/15)
`
`Textures 2.0
`
`Recombinant Immunotoxins
`
`99
`
`mediates translocation of the A subunit into the cytoplasm. The B domain,
`especially its C terminus, is responsible for the binding of DT to target cells.
`Deletions or mutations in this part of the molecule abolish or greatly di-
`minish the binding and toxicity of DT (33–35). DT enters cells via coated
`pits and is proteolytically cleaved within the endocytic compartment if it is
`not already in the two-chain form, and reduced. It also undergoes a con-
`formational change at the acidic pH present in endosomes, which probably
`assists translocation of the A chain into the cytosol perhaps via a porelike
`structure mediated by the B chain (36–38). Derivatives of DT that are used
`to make immunotoxins have the C terminus altered by mutations or par-
`(cid:4)
`tially deleted (DAB486
`DAB389, DT388) but retain the translocation and
`ADP-ribosylation activity of DT (39). Recombinant antibody-fusion pro-
`teins with such derivatives target only cells that bind the antibody moiety of
`the immunotoxin.
`
`2. PSEUDOMONAS EXOTOXIN (PE) AND PE DERIVATIVES
`
`Two major research studies have enabled the use and genetic manipu-
`lation of PE for the design of immunotoxins (1–3). The first is the eluci-
`dation of the crystal structure of PE, showing the toxin to be composed
`of three major structural domains; the second is the finding that these do-
`mains are different functional modules of the toxin. PE is a single-chain
`66-kDa molecule secreted by Pseudomonas aeruginosa that, like DT, ir-
`+
`reversibly ADP-ribosylates the diphthamide residue of EF2, using NAD
`as cofactor (40). As a consequence, protein synthesis is inhibited and cell
`death ensues. PE is composed of three major domains (41). Different func-
`tions have been assigned to each domain by mutational analysis (42). The
`N-terminal domain 1a mediates binding to the a2 macroglobulin receptor
`(43). Domain lb is a small domain that lies between domain II and domain
`III and has no known function (44). Domain II mediates translocation of do-
`main III, the carboxyl-terminal ADP-ribosylating domain, into the cytosol
`of target cells (45) (see Fig. 2). Translocation occurs after internalization of
`the toxin and after a variety of other steps, including a pH-induced confor-
`mational change (46–48), proteolytic cleavage at a specific site in domain II
`(27), and a reductive step that separates the amino and carboxyl fragments.
`Ultimately, the carboxyl-terminal portion of PE is translocated from the
`endoplasmic reticulum into the cytosol. Despite a similar mode of action
`of PE and DT, which is ADP-ribosylation, and a similar initial pathway
`of cell entry (internalization via coated pits and endocytic vesicles) and of
`processing (proteolytic cleavage and a reductive step), they share almost no
`sequence homology. The only similarity is the spatial arrangement of key
`residues in their active sites, which are arranged around residue Glu553 in
`PE and Glu145 in DT (49–52).
`
`IMMUNOGEN 2152, pg. 7
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`04/03/2001
`
`04:18 PM Cancer Research-V.81
`
`PS039-03.tex
`
`PS039-03.xml
`
`LaTeX2e(2000/11/15)
`
`Textures 2.0
`
`100
`
`Yoram Reiter
`
`Fig. 2 The biological activity of Pseudomonas exotoxin A. The Fv portion of the immuno-
`toxin targets domains II and III of PE to a cell surface receptor or other target molecule on the
`tumor cell (A). The immunotoxin enters the cell by internalization and is transferred into the
`endosome (B). Within the endosome, the molecule unfolds due to a fall in pH. The conforma-
`tional change exposes a proteolytic site, and a proteolytic cleavage occurs in the translocation
`domain between amino acid 279 and 280 (C). A disulfide bond is then broken, thus creating
`two fragments: the Fv moiety and a small part of domain II, and the rest of domain II connected
`to domain III (D). The carboxyl-terminal fragment containing the ADP-ribosylation domain
`(domain III) and most of the translocation domain (domain II) are carried into the endoplasmic
`reticulum (E), and translocation occurs from the endoplasmic reticulum into the cytosol (F).
`The enzymatically active domain ADP-ribosylates elongation factor 2 at a diphtamide residue
`+
`located at His 415, using NAD
`as a cofactor. This modification arrests protein synthesis and
`subsequently leads to cell death by apoptosis. In DT the poteolytic processing occurs between
`residues 193 and 194. The catalytic A-chain (amino acids 1–193) then translocates to the cytosol
`through the endosome with the help of translocation domain residues 326–347 and 358–376,
`which form an ion channel.
`
`When the whole toxin is used to make an immunotoxin, nonspecific tox-
`icity occurs mainly due to binding of the toxin portion to cells, mediated by
`the binding domain. Consequently, the goal of making improved derivatives
`of PE-based immunotoxins has been to inactivate or remove the binding
`domain. Molecules in which the binding domain has been retained but in-
`activated by mutations were made (53); however, a better alternative to
`
`IMMUNOGEN 2152, pg. 8
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`04/03/2001
`
`04:18 PM Cancer Research-V.81
`
`PS039-03.tex
`
`PS039-03.xml
`
`LaTeX2e(2000/11/15)
`
`Textures 2.0
`
`Recombinant Immunotoxins
`
`101
`
`inactivating the cell-binding domain by mutations is to remove it from PE.
`The prototype molecule with this sort of deletion is PE40 (aa 253–613, MW
`40 kDa). Because PE40 and its derivatives described below lack the bind-
`ing domain (aa 1–252), they have very low nonspecific toxicity, but make
`very active and specific immunotoxins when fused to recombinant antibod-
`ies (54, 55). Currently, almost all PE-derived recombinant immunotoxins are
`constructed with PE38 (MW 38 kDa), a PE40 derivative that has, in addi-
`tion to the deletion of domain 1a, a second deletion encompassing a portion
`of domain Ib (aa 365–379) (44). Another useful mutation is to change the
`carboxyl-terminal sequence of PE from REDLK to KDEL. This improves the
`cytotoxicity of PE and its derivatives, presumably by increasing their delivery
`to the endoplasmic reticulum, where translocation takes place (56, 57).
`
`B. The Targeting Moiety—Recombinant
`Antibody Fragments
`
`The antibody moiety of the recombinant immunotoxin is responsible for
`specifically directing the immunotoxin to the target cell, meaning that the
`usefulness of the immunotoxin depends on the specificity of the antibody
`or antibody fragment that is connected to the toxin. Consequently, for the
`construction of recombinant immunotoxins, the only antibodies that should
`be used are those that recognize antigens that are expressed on target cancer
`cells and are not present on normal cells, present at very low levels, or are
`only present on less essential cells (Table I).
`Receptors for growth factors such as the epidermal growth factor (EGF),
`interleukin 2 (IL-2), interleukin 4 (IL-4), interleukin 6(IL-6), or erbB2 are
`common targets for targeted cancer therapy because they are highly ex-
`pressed on many cancer cells. Other carcinoma-related antigens include de-
`velopmental antigens such as complex carbohydrates, which are often highly
`abundant on the surface of cancer cells. The use of antibodies for immuno-
`toxin production also requires that the antibody–antigen complex be inter-
`nalized, because the mechanism of PE-toxin killing requires endocytosis as
`a first step in the entry of the toxin into the cell.
`Recombinant immunotoxins contain antibody fragments as the targeting
`moiety. These fragments can be produced in E. coli and are the result of
`intensive research and development in recombinant-antibody technologies
`(58–60). Several antibody fragments have been used to construct recombi-
`nant immunotoxins (Fig. 1). One type contains Fab fragments in which the
`light-chain and the heavy-chain Fd fragments (VH and CH1) are connected
`to each other via an interchain disulfide bond between CL and CH1. The
`toxin moiety can be fused to the carboxyl end of either CL or CH1.
`
`IMMUNOGEN 2152, pg. 9
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`04/03/2001
`
`04:18 PM Cancer Research-V.81
`
`PS039-03.tex
`
`PS039-03.xml
`
`LaTeX2e(2000/11/15)
`
`Textures 2.0
`
`102
`
`Yoram Reiter
`
`IMMUNOGEN 2152, pg. 10
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`04/03/2001
`
`04:18 PM Cancer Research-V.81
`
`PS039-03.tex
`
`PS039-03.xml
`
`LaTeX2e(2000/11/15)
`
`Textures 2.0
`
`Recombinant Immunotoxins
`
`103
`
`Fabs can be produced in E. coli either by secretion, with coexpression of
`light chains and Fd fragments, or by expression of the chains in intracellular
`inclusion bodies in separate cultures; in the latter case, they are reconsti-
`tuted by a refolding reaction using a redox-shuffling buffer system. Several
`immunotoxins with Fab fragments have been constructed and produced in
`this way (1–4, 61).
`The smallest functional modules of antibodies required for antigen binding
`are Fv fragments. This makes them especially useful for clinical applications,
`not only for generating recombinant immunotoxins but also for tumor imag-
`ing, because their small size improves tumor penetration. Fv fragments are
`heterodimers of the variable heavy-chain (VH) and the variable light-chain
`(VL) domains. Unlike whole IgG or Fab, in which the heterodimers are held
`together and stabilized by interchain disulfide bonds, the VH and VL of Fvs
`are not covalently connected and are consequently unstable; this instabil-
`ity can be overcome by making recombinant Fvs that have the VH and VL
`←−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−
`Fig. 3 Cloning, construction, and composition of scFv- and dsFv-immunotoxins. (A) cloning
`and construction of recombinant scFv and dsFv immunotoxins. The genes encoding the VH
`and VL variable domains are cloned usually from hybridoma mRNA by reverse transcription,
`cDNA synthesis, and subsequent PCR amplification using degenerate primers that are comple-
`(cid:4)
`(cid:4)
`mentary to the 5
`or 3
`end of the VH and VL genes or by primers which are designed according
`to the amino-terminal amino acid sequence of the MAb to be cloned and conserved sequences
`at the N terminal of the heavy and light constant regions. The variable genes can be also cloned
`by constant-domain primers and using the RACE method (rapid amplification of cDNA ends).
`Restriction sites for assembling of the peptide linker sequence which connects the VH and VL
`domains, and for cloning into the expression vector, are also introduced by PCR. Construction
`of dsFv involves the generation of two expression plasmids which encode the two components
`of the dsFv VH-cys and VL-cys. The cysteines are introduced in position 44 in FR2 of VH and
`position 100 of FR4 of VL or position 105 of FR4 in VH and position 43 of FR2 in VL (num-
`bering system of Kabat et al.) by site-directed mutagenesis using as template a uracil-containing
`single-stranded DNA of the scFv construct from the F+ origin present in the expression plas-
`mid and cotransfection with M13 helper phage. In addition to the cysteines, cloning sites, ATG
`(cid:4)
`(cid:4)
`translation-initiation codons, and stop codons are introduced at the 5
`end and 5
`end of the
`VH and VL genes, as shown by site-directed mutagenesis or PCR. The antibody variable genes
`are subcloned into an expression vector which contains the gene for a truncated form of Pseu-
`domonas exotoxin. This expression vector is controlled by the T7 promoter and upon induction
`of the T7 RNA polymerase, which is under the control of the lacUV5 promoter, in E. coli BL21
`␭DE3 by IPTG, large amounts of recombinant protein are produced. (B) composition of re-
`combinant immunotoxins. In PE-derived recombinant Fv immunotoxins the Fv region of the
`targeting antibody is fused to the N terminus of a truncated form of PE which contains the
`translocation domain (domain II) and enzymatically active ADP-ribosylation domain (domain
`III). The cell-binding domain of whole PE (domain I) is replaced by the Fv targeting moiety, thus
`preserving the relative position of the binding-domain function to the other functional domains
`of PE. In the dsFv immunotoxins there are two components. In one the VH or VL domains are
`fused to the amino terminus of the truncated PE, and the other variable domain is covalently
`linked by the engineered disulfide bond. DT-derived immunotoxins are fused to the carboxyl
`terminus due to the inverse arrangement of the functional modules of PE and DT.
`
`IMMUNOGEN 2152, pg. 11
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`04/03/2001
`
`04:18 PM Cancer Research-V.81
`
`PS039-03.tex
`
`PS039-03.xml
`
`LaTeX2e(2000/11/15)
`
`Textures 2.0
`
`104
`
`Yoram Reiter
`
`covalently connected by a peptide linker that fuses the C terminus of the VL
`or VH to the N terminus of the other domain (Fig. 1). These molecules are
`termed single-chain Fvs (scFvs) (62, 63), and many retain the specificity and
`affinity of the original antibody. The cloning, construction, and composition
`of recombinant Fv fragments of antibodies and of Fv-immunotoxins are de-
`scribed in Fig. 3. Many recombinant immunotoxins have been constructed
`using scFvs, in which molecules the scFv gene is fused to PE38 to generate a
`potent cytotoxic agent with targeted specificity (1–4, 64–70) (Figs. 1 and 3).
`Until recently, the construction of scFvs was the only general method avail-
`able to make stable Fvs. However, many scFvs are unstable or have reduced
`affinity for the antigen compared with the parent antibody or Fab fragment.
`This is because the linker interferes with binding or because the linker does
`not sufficiently stabilize the Fv structure, leading to aggregation and loss of
`◦
`activity. This is particularly true at physiological temperatures (37
`C). To
`overcome these problems, an alternative strategy has been developed that
`involves generating stable Fvs by connecting the VH and VL domains by an
`interchain disulfide bond engineered between structually conserved frame-
`work residues of the Fv; these molecules are termed disulfide-stabilized Fvs
`(de Fvs) (60, 71–73). The positions at which the cysteine residues were to
`be placed were identified by computer-based molecular modeling; as they
`are located in the framework of each VH and VL, this location can be used
`as a general method to stabilize almost all Fvs, without the need for any
`structural information. Many dsFvs have been constructed in recent years
`(mainly as dsFv immunotoxins, in which the dsFv is fused to PE38), and
`they show several advantages over scFvs (60, 74–76). In addition to their in-
`creased stability (owing to a decreased tendency to aggregate), they are often
`produced in higher yields than scFvs; in several cases, the binding affinity of
`the dsFv was significantly improved over that of the scFv.
`
`III. CONSTRUCTION AND PRODUCTION OF
`RECOMBINANT IMMUNOTOXINS
`
`In the recombinant immunotoxins derived from PE, the recombinant an-
`tibody fragments are fused to the amino terminus of the truncated deriva-
`tive of PE (with the cell-binding domain deleted), e.g., PE40 or PE38. This
`restores the original domain arrangement of PE, which consists of an
`N-terminal binding domain followed by the translocation domain and the
`C-terminal ADP-ribosylation domain (Fig. 1b). Only fusions of an antigen-
`binding domain (Fv) to the amino terminus of truncated PE are active;
`carboxyl-terminal fusions are not active because the bulky antigen-binding
`domain blocks translocation of the C-terminal fragment into the cytoplasm
`(1, 25).
`
`IMMUNOGEN 2152, pg. 12
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`04/03/2001
`
`04:18 PM Cancer Research-V.81
`
`PS039-03.tex
`
`PS039-03.xml
`
`LaTeX2e(2000/11/15)
`
`Textures 2.0
`
`Recombinant Immunotoxins
`
`105
`
`DT immunotoxins are fusions of mutated DT with antigen-binding regions
`of a recombinant antibody. However, in this case the antigen-binding domain
`must be fused to the C terminus of DT (77, 78). This corresponds to the
`inverse arrangement of the functional modules of PE and DT (see Fig. 3).
`DT immunotoxins are active only when the enzymatically active N-terminal
`domain is free to translocate into the cytosol.
`The expression vectors used for DT immunotoxins are very similar to
`those used with PE, with the exception that the DNA fragments encoding
`(cid:4)
`the binding moiety are ligated to the 3
`-end of the DT coding region.
`The cloning of the antibody variable regions is performed using cloning
`techniques that are now well established (59) (Fig. 3). The plasmid vec-
`tor for the expression of scFv immunotoxins or the components of dsFv
`immunotoxins is a high-copy-number plasmid derived from vectors made
`and described by Studier and Moffatt (79). These contain the T7 promoter,
`translation-initiation signals, and a transcription terminator, as well as an
`F+ phage-replication origin to generate single-stranded DNA to be used for
`site-directed mutagenesis.
`When these plasmids are transformed into E. coli BL21/DE3 (which con-
`tain the T7 RNA-polymerase gene under the control of the lac UV5 pro-
`moter), they generate large amounts of recombinant protein upon IPTG
`induction. The recombinant scFv immunotoxin or the components of the
`dsFv immunotoxin accumulate in insoluble intracellular inclusion bodies.
`[dsFv immunotoxins require two cultures, one expressing the VH and one
`expressing the VL; the toxin moiety (PE38)

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket