throbber
Annu. Rev. Pharmacol. Toxicol. 1992. 32:579--621
`Copyright © by Annual Reviews Inc.
`All rights reserved
`
`CYTOTOXIC CONJUGATES
`
`CONTAINING TRANSLATIONAL
`
`INHIBITORY PROTEINS
`
`
`
`S. Ramakrishnan, D. Fryxell, D. Mohanraj, M. Olson, B-Y Li.
`
`
`
`
`
`Department of Pharmacology, 3-249 Millard Hall, University of Minnesota,
`
`
`
`
`Minneapolis. Minnesota 55455
`
`
`
`KEY WORDS: targeting toxins, immunotoxins, chemotherapy, carrier molecules, growth
`
`
`
`
`
`
`
`factor conjugates
`
`INTRODUCTION
`
`Cytotoxic drugs administered during conventional chemotherapy are taken up
`
`
`
`
`
`
`by sensitive nontarget organs besides the tumor cells and this often leads to
`
`
`unwanted side effects. These problems have initiated studies during the past
`
`
`
`decade to develop novel methods in which the therapeutic reagents could be
`
`
`
`to specific cell populations to widen the therapeutic window. The
`directed
`
`
`
`concept of targeting drugs was proposed nearly a hundred years ago by Paul
`
`
`
`
`Erlich, who envisioned the possibility of transporting toxic substances to
`
`
`
`tumor cells by carrier molecules (magic bullets). This novel approach became
`
`
`
`a reality with the advent of tumor selective monoclonal antibodies (Mabs)
`
`
`produced by somatic cell hybridization. Both cytotoxic drugs and toxin
`
`
`polypeptides have been chemically linked to Mabs. During the past decade
`
`
`
`
`various molecules have been produced and tested in a number of model
`
`
`
`systems (reviewed in 1-10). The range of targets includes (a) ex vivo
`
`
`
`treatment of bone marrow grafts to remove contaminating leukemic cells and
`
`
`T cells, (b) administration of conjugates to inhibit tumor growth
`alloreactive
`
`579
`
`Annu. Rev. Pharmacol. Toxicol. 1992.32:579-621. Downloaded from www.annualreviews.org
`
`by Reprints Desk, Inc. on 05/20/14. For personal use only.
`
`IMMUNOGEN 2176, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`580 RAMAKRISHNAN ET AL
`
`in restricted anatomical spaces such as the peritoneum, and (c) systemic
`
`
`
`
`
`
`application of conjugates to inhibit diffused tumors and solid tumors. Success
`
`
`of the last two applications depends largely on the pharmacological and
`
`
`
`
`toxicological properties of the chimeric molecules. This article focuses on the
`
`
`most recent developments in targeting toxin polypeptides.
`
`
`Immunotoxins are composed of a cytotoxic molecule linked to a carrier
`
`
`
`molecule by either a reducible or a nonreducible bond. The cytotoxic moiety
`
`
`
`is often a toxin polypeptide that catalytically inhibits a vital biosynthetic
`
`
`
`pathway. In general, the cytotoxic moiety used in targeting is a translational
`
`
`inhibitory protein but some investigators have also used enzymes such as
`
`
`
`phospholipases to destabilize the integrity of the tumor cell plasma mem­
`
`
`brane. Various carrier proteins have been investigated to deliver the toxin
`
`
`
`
`molecules, including tumor selective monoclonal antibodies, growth factors,
`and lymphokines.
`
`CHOICE OF TOXIN MOLECULES
`
`Three types of toxin polypeptides are used in the preparation of immunotoxins
`
`
`
`(IT)l: (a) bacterial toxins (b) plant toxins and (c) fungal toxins. All three
`
`
`
`
`
`groups of molecules catalytically inhibit protein synthesis in eukaryotes but
`
`
`
`each at a distinct step in translation. Most of the work on the bacterial toxins
`
`
`was carried out on two highly toxic molecules, namely diphtheria toxin (DT)
`
`studies on the structure/function and pseudomonas exotoxin A (PE). Detailed
`
`
`
`
`of these two molecules are primarily responsible for the advances made in
`
`
`preparing highly effective chimeric molecules against tumor cells. Based on
`
`
`their biochemical characteristics, plant toxins can be grouped as Type I,
`
`
`
`
`
`single chain polypeptides that enzymatically inhibit translation, and Type II
`
`
`(e. g. ricin and abrin), which are heterodimers. The A chains of Type II toxins
`
`are the true toxic moieties, whereas the B chains contain binding sites for
`
`
`carbohydrates through which the A chain gains access to the interior of the
`
`
`cell. Fungal toxins such as alpha sarcin are also single chain proteins but are
`
`
`
`
`functionally different from Type I polypeptides. Alpha sarcin, for example, is
`
`a phosphodiesterase while the Type I toxins are N-glycosidases.
`
`I Abbreviations used: CDR: complementarity-determining regions; DT: diphtheria toxin;
`
`
`
`
`
`
`
`
`EGF: epidermal growth factor; Fc: fragment crystallizable; GVHD: graft versus host disease;
`
`
`
`
`
`HAMA: human anti-murine antibody; IT: immunotoxins; KDEL: endoplasmic retention signal
`
`
`
`
`
`
`sequence; LDL: low density lipid; PAP: pokeweed antiviral protein; PE: pseudomonas exotoxin
`
`
`
`
`
`A; PEG: polyethylene glycol; RIP: ribosomal inhibitory proteins; SCA: single chain antibodies;
`
`
`
`SPDP: N-Succinimidyl 3-(2-pyridyl)dithiopropionic acid; TeS: trichosanthin; TFR: transferrin
`
`
`
`receptor; tPA: tissue plasminogen activator.
`
`Annu. Rev. Pharmacol. Toxicol. 1992.32:579-621. Downloaded from www.annualreviews.org
`
`by Reprints Desk, Inc. on 05/20/14. For personal use only.
`
`IMMUNOGEN 2176, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`CYTOTOXIC CONJUGATES 581
`
`RICIN
`
`
`
`Structural Features
`
`Ricin is a toxic lectin isolated from castor beans. Its precursor is a single
`
`
`
`
`
`
`polypeptide composed of a signal peptide and two chains linked by a 1 2
`
`
`
`
`
`amino acid joining peptide (J peptide) . It is proteolytic ally cleaved to form
`
`the mature ricin of 62 kd containing an A chain (267 amino acids) and a B
`chain (262 amino acids) linked by a disulfide bond ( 11 ) . The toxin is post­
`
`
`translationally glycosylated on both A and B chains with high mannose
`
`
`branched chains containing xylose and fucose. The A chain has two N-linked
`
`
`sugars at positions 1 0 and 236. The B chain has N-linked carbohydrates
`
`
`
`
`attached to residues 95 and 1 35. Heterogeneity in composition of sugars
`
`
`has been observed in all four sites (12) . In the mature ricin molecule the
`
`A chain is linked to the B chain by a reducible bond between Cys 259 of the
`
`A chain and Cys 4 of the B chain. Ricin has been crystallized, and the three­
`
`
`
`
`
`dimensional structure has been elucidated by Robertus and colleagues ( 1 3 ,
`of 2 . 5 A. Interaction
`1 4) at a resolution
`
`between the subunits i s mainly
`
`
`
`due to hydrophobic forces among various aromatic rings and aliphatic side
`chains.
`The carboxy terminal of the B chain seems to be involved in association
`
`
`
`with the A chain. Three domains can be distinguished in theA chain by X-ray
`
`
`
`crystallography ( 14). Domain I consists of amino acids 1-11 7 dominated by a
`
`
`six-stranded B-sheet and two helices. Domain II encompasses residues 1 18-
`
`2 1 0 and is mainly made of alpha helices. The C-terminal end constitutes
`
`
`
`Domain III, and the striking feature of this part of the molecule is a stretch of
`
`
`
`
`hydrophobic residues located between positions 247 and 257. This region of
`
`
`
`the molecule is suggested to play a key role in the translocation of the toxic
`
`
`subunit into the cytoplasm.
`The A chain of ricin is a specific N-glycosidase that catalytically hydro­
`
`
`
`
`
`
`lyzes the glycosidic bond of adenosine residue 4324 in the 28S rRNA (15).
`
`
`
`
`
`Depurination results in irreversible inactivation of ribosomes and inhibition of
`
`
`
`protein synthesis. Ricin is such a potent toxin that a single A chain molecule
`
`
`
`can inactivate 1 500 ribosomes per minute and once in the cytosol can kill the
`
`cell ( 1 6) . The Km for reticulocyte ribosomes is about 0.1 -0.2 f.tM and the
`
`
`Kcat about 1 ,500 min-i. The depurination mechanism of ricin is predicted to
`
`
`
`
`
`be similar to that of adenosine monophosphate nucleosidase involving the
`
`
`
`formation of an oxycarbonium ion. Based on site-directed mutagenesis and
`
`
`
`
`
`X-ray diffraction studies, important residues of the A chain either in direct
`
`
`
`
`contact with the substrate or those involved in the actual catalysis have been
`
`
`
`identified. The catalytic site is present in a cleft; Tyr 80 and Tyr 123 are
`
`
`
`
`
`present at the top of the cleft. Neither is bclieved to be important for catalysis
`
`
`
`but both may contribute to the tight binding of the substrate. Substitution of
`
`Annu. Rev. Pharmacol. Toxicol. 1992.32:579-621. Downloaded from www.annualreviews.org
`
`by Reprints Desk, Inc. on 05/20/14. For personal use only.
`
`IMMUNOGEN 2176, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`582 RAMAKRISHNAN ET AL
`
`other residues present around the cleft such as Asn 209 and Trp 211 does not
`
`
`
`
`
`significantly change the enzymatic activity (17, 1 8) .
`Tight binding o f the substrate (28S rRNA) is responsible for the partial
`
`
`
`
`
`
`
`
`breakage of the N-C bond (18). Two important changes involving either Glu
`
`
`
`177 or Arg 1 80 have consistently been shown to dramatically decrease the
`
`
`
`biological activity of the A chain. The positive charge on Arg 1 80 is supposed
`
`
`
`to help in the partial breaking of the bond by ion pairing to the phosphate
`
`
`backbone. The destabilized N-C bond is then vulnerable to nucleophilic attack
`
`
`
`
`by a water molecule. Interestingly, intact ribosomes are better substrates than
`
`
`
`
`
`the synthetic oligomers homologous to the conserved region covering the
`
`
`
`
`
`depurination site. It is therefore likely that the ribosomal proteins may con­
`
`
`
`tribute to the proper presentation of the sensitive site in a manner facilitating
`
`
`
`
`catalysis or that some important regions of ricin A chain may interact with
`
`
`
`
`
`ribosomal proteins in stabilizing the enzyme-substrate complex. This issue is
`
`
`
`
`
`currently being investigated by genetic deletion analyses in many labora­
`tories.
`The B chain of ricin has two functions: (a) binding reversibly to galactose
`
`
`
`
`
`presented by cell surface glycopeptides and glycolipids, and (b) facilitating
`
`
`
`
`uptake of whole ricin by endocytosis and transport through the Golgi appara­
`
`
`tus before translocation of the A chain into the cytosol (19). It is still
`
`
`contentious whether the two functions of the B chain are separable. Some
`
`
`
`
`studies indicate that toxin entry could be mediated by mechanisms in­
`
`
`
`
`
`dependent of galactose recognition, but other evidence supports the galactose
`
`
`
`binding activity on the B chain as an important step in the entry to the cytosol
`(20-24).
`
`Conjugates Prepared with Ricin and Ricin A Chain (RTA)
`
`
`
`
`
`Many immunotoxins using intact ricin and monoclonal antibodies or growth
`
`
`
`
`factors have been developed (25, 26). They are extremely cytotoxic to
`
`
`
`relevant target cells, but the specificities are suboptimal since B chains may
`
`
`
`
`
`bind to surfaces on nontarget cells. Another problem is the rapid clearance of
`
`
`ricin IT from the bloodstream (27). This is due at least partially to the
`
`
`
`clearance of native ricin by oligosaccharide-mediated uptake by the liver cells
`
`
`(28, 29). To circumvent this problem, immunotoxins have been made with
`
`
`
`chemically/enzymatically deglycosylated A chain (30, 3 1 ) or recombinant A
`
`
`
`
`
`chain. Chemical treatment destroys the mannose and other sugar residues on
`
`
`
`
`native A chain while recombinant A chain expressed in bacteria is free of B
`
`
`chain and completely lacks oligosaccharide side chains. These constructs
`
`
`
`
`have better pharmacological properties than conjugates made with native ricin
`A chain.
`Several groups have obtained ricin A chain (rRT A) by genetic engineering
`
`
`
`
`
`techniques. Shire et al (32) expressed a synthetic 842 bp RT A gene in
`
`Annu. Rev. Pharmacol. Toxicol. 1992.32:579-621. Downloaded from www.annualreviews.org
`
`by Reprints Desk, Inc. on 05/20/14. For personal use only.
`
`IMMUNOGEN 2176, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`CYTOTOXIC CONJUGATES 583
`
`coli at the level of 1 .5 mg/liter of culture. O'Hare et al (33)
`
`
`Escherichia
`
`
`cloned ricin A chain cDNA using vector pDs/3 and expressed the recombinant
`
`A chain with ten extra amino acids at the N-terminus in E. coli at the level of
`
`2-3 mglliter of culture. Piatak et al (34) obtained a correct 267 amino acid A
`
`
`
`
`
`
`chain from E. coli using several different host/vector systems at the level of
`
`
`1-2% to 6-8% of total cell proteins. Frankel et al (35) used an expression
`
`
`system to produce a fusion protein containing ricin A chain linked to the
`
`
`
`enzyme f3-galactosidase via a collagen fragment. Treatment of the fusion
`
`
`
`protein with collagenase released free ricin A chain that could be recovered
`
`
`
`from the enzyme by selectively absorbing the latter on an affinity column. In
`
`
`
`
`our laboratory, the coding region of the ricin A chain gene was cloned into the
`
`
`pET3b vector. The cloned fragment is flanked by the T7 promoter and
`
`
`
`terminator in this construction and expressed in E. coli, BL21(DE3). The
`
`
`
`production of purified A chain protein with 1 1 extra amino acids at the
`
`
`N-terminus was 80-90 mg/liter of culture or 24-26% of total cell proteins.
`
`
`
`due to The specificities of the IT made from nRTA or rRTA are excellent
`
`
`
`the absence of nonspecific binding of B chain (27, 36-39). However, the
`
`
`
`cytotoxicities of these conjugates were relatively less (40) than intact ricin IT
`
`
`
`due to the loss of translocation functions asssociated with the B chain (40-
`
`
`43). Some of the immunoconjugates prepared with ricin A chain were not
`
`cytotoxic, whereas the same antibody linked to ricin was effective against
`
`
`
`
`target cells in the presence of lactose. Translocation signals associated with
`
`
`the B chain of ricin have been effectively used in potentiating ricin A chain
`
`
`
`
`conjugates. The B chain can be either delivered directly to cell bound A chain
`
`
`conjugates or indirectly by a second antibody homing onto the same cell. In
`
`
`
`
`
`both approaches however, lactose has to be included to prevent nonspecific
`toxicity
`
`
`
`. Alternatively, intact ricin could be modified (blocked ricin) by
`
`
`
`
`
`chemical modifications (41-43) that retain the translocation properties with­
`
`
`
`
`
`out the nonspecific toxicity mediated by the sugar binding sites. The cell
`
`
`
`binding, enzymatic specificity, and mechanism of cytosol entry between
`
`bacterial toxins and plant toxins are very different.
`
`SINGLE CHAIN RIBOSOMAL INHIBITORY PROTEINS
`
`
`(TYPE I RIP)
`
`preparations This class of proteins is used as an alternative in immunotoxin
`
`
`
`
`
`
`
`and has many advantages in the treatment of several clinical diseases. Perhaps
`
`
`the best-studied single chain toxins are pokeweed antiviral proteins (PAP)
`
`isolated from the plant Phytolacca
`
`
`saporin 6, isolated from the
`americana,
`
`
`
`plant Saponaria and gelonin purified from Gelonum multiflorum.
`officinalis
`
`
`
`Similar proteins have been isolated from other plants (reviewed in 44) and are
`
`Annu. Rev. Pharmacol. Toxicol. 1992.32:579-621. Downloaded from www.annualreviews.org
`
`by Reprints Desk, Inc. on 05/20/14. For personal use only.
`
`IMMUNOGEN 2176, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`584 RAMAKRISHNAN ET AL
`
`generally basic proteins of about 30 kd. Most are nonglycosylated although
`
`
`
`recent evidence suggests the presence of simple sugars. The mechanism of
`
`
`action of Type I RIP seems to be similar to ricin in depurinating a single
`
`
`
`adenine residue at position 4324 of 28S rRNA.
`A major problem in using dual chain toxins (Type II) is that they also bind
`
`
`
`to saccharide residues present on normal cells. Before they can be used in IT
`
`
`
`preparations, these toxins must either be modified to prevent binding or must
`have their B chain removed. Single chain toxins, on the other hand, do not
`bind to any cell surface components.
`and evolutionarily, Type I
`Structurally
`
`
`
`RIP are related to Type II heterodimeric toxins. They usually do not contain
`
`free sulfhydryl groups (SH); these are often introduced to facilitate conjuga­
`
`
`
`tion to antibodies. SH groups can be introduced by modifying the epsilon
`
`
`amino group of lysine that, in certain instances, has led to a reduction in
`
`
`
`biological activity. Modification of lysine residues with reagents such as
`
`
`SPDP results in the loss of a positive charge on the molecule. Alternative
`
`
`
`
`
`thiolation procedures using 2-iminothiolane retain the positive charge and do
`
`
`
`not significantly reduce the biological activity of the toxins.
`
`
`
`
`
`Pokeweed Antiviral Protein (PAP)
`
`Three different forms (PAP-I, PAP-II, and PAP-S) of RIP have been isolated
`
`
`from the plant P. americana (45, 46). PAP-I and II were found in the leaves,
`
`
`
`
`
`and PAP-S was purified from the seeds. Pokeweed plants express the various
`
`
`
`forms of PAP at different seasons (46). Amino terminal sequences of these
`
`
`
`proteins are homologous with minor variations and there is limited immuno­
`
`
`
`logical crossreactivity between these proteins. Recent studies have shown that
`
`
`
`PAP inactivates ribosomes in a manner similar to ricin A chain (47). IT
`
`
`
`prepared with PAP have been highly specific in cytotoxicity to target cells
`
`
`
`(48-50). These conjugates proved useful in bone marrow purging and showed
`
`
`
`minimal toxicity to pluripotent progenitor cells.
`
`PAP-linked conjugates could be potentiated severalfold
`by ammonium
`and monensin (51), which indicates
`chloride
`
`that the conjugates were routed
`
`
`
`
`through acidic intracellular compartments. Since PAP does not have complex
`
`
`
`sugars, it is relatively more stable than native ricin A chain conjugates (52).
`
`
`
`Free PAP, however, is rapidly excreted from the circulation. PAP-IT directed
`
`
`to CD40 proved to be effective against cionogenic cells from acute
`
`
`
`lymphoblastic leukemia and non-Hodgkin's lymphoma in the absence of
`
`
`
`potentiators. PAP-IT specific toward CD7 were only effective against a T cell
`
`
`
`
`acute lymphoblastic leukemia lineage (53). A recent study showed that Type I
`
`
`
`
`RIP including PAP are useful in inhibiting phagocytic parasites when targeted
`
`via antibodies (54).
`
`Annu. Rev. Pharmacol. Toxicol. 1992.32:579-621. Downloaded from www.annualreviews.org
`
`by Reprints Desk, Inc. on 05/20/14. For personal use only.
`
`IMMUNOGEN 2176, pg. 6
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`CYTOTOXIC CONJUGATES 585
`
`Saporin
`Saporin is resistant to proteolysis, has a pI greater than 10, and has no
`
`
`
`
`
`
`
`
`carbohydrate chains. Yoshikawa et al (55) have described the separation of
`
`
`two distinct saporins from the seeds, N(l) and P(2). When conjugated to an
`
`
`
`
`
`anti-CD4 antibody, saporin kills CD4 cells effectively. Neither CD4 nor CD8
`
`
`
`
`
`cells internalize RTA-IT made with anti-T4 or anti-T8 specificities; the cells
`
`are not killed by either of these IT (56). Another study included three saporin
`
`IT that recognized CDS, CD2, or CD3. All three IT bound T cells and were
`
`
`
`
`
`
`effective in inhibiting protein synthesis in cell-free systems. The anti-CD2 IT,
`
`
`
`however, had weak toxicity to whole cells, whereas the other two were quite
`
`
`
`
`
`potent. The anti-CDS IT was marginally potentiated by amantadine (57).
`
`
`
`
`Saporin conjugated to the transferrin receptor (TFR) suppresses leukemic
`
`
`
`
`stem cell generation. Progenitors of the erythroid lineage were more sensitive
`
`than those of the myeloid lineage. There was no effect in vitro on normal or
`
`
`
`
`leukemic human hematopoietic progenitors. Since TFR expression is associ­
`
`
`
`ated with proliferation, it would be expected that the most susceptible cells
`
`
`
`
`would be those that are actively dividing. The antibody itself did not block
`
`
`
`
`TFR function (58). Similarly. Barbieri et al (59) found that saporin IT with
`
`
`antibodies directed toward plasma cell antigens bound and killed RAJI and
`
`
`U266 cells. Both had a high toxicity toward bone marrow, but only one
`
`
`
`
`
`inhibited differentiation of myeloid precursors. Bifunctional antibodies used
`
`
`
`
`to deliver saporin to specific tissues were tenfold more potent if the portion
`
`
`binding the toxin was obtained from a polyclonal mixture rather than a
`
`
`
`monoclonal source. This was shown for guinea pig leukemic cells (60).
`
`Trichosanthin
`Trichosanthin (TCS) is characterized by high content of Asn, Asp, GIu, Gin,
`
`
`
`
`
`
`
`and no Cys residues or carbohydrates. Its sequence is homologous to that of
`
`
`
`
`RTA, as is the tertiary structure. TCS loses over 90% of its activity when
`
`
`
`
`modified with SPDP; treatment with 2-iminothiolane resulted in the addition
`
`
`
`
`of 1.5 sulfhydryl groups per molecule on average. When conjugated to an
`
`
`
`
`anti-hepatoma antibody, the IT had a 50-fold higher toxicity than native TCS.
`
`
`
`Cytotoxicity of the IT was time dependent. Trichokirin, isolated from a
`
`
`
`
`related plant, is homologous but not completely identical to TCS (61).
`
`
`
`
`
`Trichokirin linked to an anti-Thy1.2 antibody was cytotoxic against T2 cells
`
`
`
`
`(62). Conjugation with dimethyl 3, 3'-dithiobis propionimidate (DTBP) re­
`
`
`sulted in only a small loss of activity; derivatization with SPDP and 2-
`
`
`
`
`
`iminothiolane reduced the biological activity significantly. The ability to
`
`
`
`
`inhibit cell-free translation was comparable to that of RTA and had the same
`
`
`
`
`enzymatic target. NH4CI had a negative dose-related effect on toxicity,
`
`
`
`
`
`characteristic of a requirement for passage through an acidic cellular compart-
`
`Annu. Rev. Pharmacol. Toxicol. 1992.32:579-621. Downloaded from www.annualreviews.org
`
`by Reprints Desk, Inc. on 05/20/14. For personal use only.
`
`IMMUNOGEN 2176, pg. 7
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`586 RAMAKRISHNAN ET AL
`
`ment. Monensin potentiated the trichikirin-IT fivefold. Clearance of the IT
`
`
`
`
`
`
`was slower than RTA-IT and had a biphasic pattern with half-lives of 0.5 and
`8 hours.
`A closely related toxin, J3-trichosanthin, can be effective against cells in the
`
`
`
`
`
`
`
`
`
`lymphocytic and monocytic lineages and can also block HIV replication in
`
`
`
`
`acutely and chronically infected cells. A rank order potency for inhibition
`of
`
`
`
`
`protein synthesis is j3-TCS, j3-momorcharin, luffaculin > a-momorcharin
`>
`
`
`TCS > momorcochin. These toxins are homologous in amino acid composi­
`
`
`
`tion, tertiary structure, have molecular weights between 28 and 32 kd, and
`
`have N-terminal Asp residues
`(63).
`
`Other Single Chain RIP
`Anti-Thy 1.1 antibodies conjugated to momordin or bryodin with SPDP were
`
`
`
`Thy 1.1 positive
`
`effective against
`
`
`target cells in the low nanomolar range.
`
`
`Bryodin IT were slightly more toxic. A two-to fivefold
`loss of activity was
`
`
`
`
`seen following SPDP treatment. This loss can possibly be prevented by
`
`
`
`
`treatment with 2-iminothiolane instead. The positive charge in vivo (high pI)
`(64).
`
`
`
`may shield the disulfide bond and confer higher stability in circulation
`
`Alpha Sarcin
`The toxin a-sarcin is also a basic nonglycosylated protein, but has a molecular
`
`
`
`
`
`weight of 1 7 kd. The smaller size of this peptide could prove useful in
`
`
`
`
`penetration of solid tumors and be easier to use in clinical settings. It has no
`
`
`
`sequence similarity to plant RIP but is homologous to restrictocin and
`
`
`
`
`mitogellin. a-sarcin is an example of single chain toxins that hydrolyze the
`
`
`
`phosphodiester bond in the 28S rRNA. An a-sarcin-IT had similar pharmaco­
`and was stable for 4 days at
`
`kinetics as other RIP with the same antibody
`
`
`
`
`37°C, much better than RTA-IT. The activity of the a-sarcin IT was 800-fold
`
`better than for a-sardn alone and approached the potency of RTA-IT. The
`
`
`
`a-sarcin IT had a longer biological in serum, with clearance follow­
`half-life
`
`
`ing the biphasic pattern common to RTA-IT (0.8 and 6 hr half-lives). IT with
`
`gelonin or momordin behaved similarly (65).
`
`
`The gene for a-sarcin has recently been sequenced
`(66) and shown to
`
`
`encode a stretch of 27 amino acids at the amino terminus that could potentially
`
`be cleaved. This could afford the cell protection
`
`against its own product.
`
`Restrictocin is homologous to a-sarcin and its gene has also been cloned and
`
`
`
`
`
`expressed, showing a single 52-base intron, possible regions for transcription
`
`
`
`signals, and polyadenylation sites flanking the open reading frame. An im­
`
`
`mature form of restrictocin may be produced that could undergo activation
`Both genes show homology to the U2 ribonuclease.
`during secretion.
`IT made
`have been used in IT
`
`with restrictocin were toxic in vitro (67). Other proteins
`(68-72).
`
`Annu. Rev. Pharmacol. Toxicol. 1992.32:579-621. Downloaded from www.annualreviews.org
`
`by Reprints Desk, Inc. on 05/20/14. For personal use only.
`
`IMMUNOGEN 2176, pg. 8
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`CYTOTOXIC CONJUGATES 587
`
`PSEUDOMONAS EXOTOXIN A
`
`Structural Features
`
`Pseudomonas exotoxin A (PE) is a single chain protein toxin produced by
`
`
`
`
`
`The molecular weight of PE is 66,000 daltons, and
`
`Pseudomonas aeruginosa.
`
`
`in its native form PE has four intrachain disulfide bridges, no titratable
`
`
`sulfhydryl groups, and is devoid of sugars and other posttranslational mod­
`
`
`
`ification (73). Structural analysis of PE has indicated that it is composed of
`
`
`
`
`three domains when interacting with mammalian cells. Domain Ia located at
`
`
`
`the N-terminal end of the molecule (aa 1-252) is responsible for cell binding.
`
`
`
`
`Domain II located in the middle of the molecule (aa 253-364) is suggested to
`
`
`be associated with translocation of the toxin moiety across cell membranes,
`
`
`
`
`and Domain III located at the C-terminal end (aa 405-613) possesses the ADP
`
`
`
`
`ribosylation activity. ADP ribosylation of elongation factor 2 irreversibly
`
`
`
`inhibits the synthesis of proteins and leads to cell death. The role of Domain
`
`Ib (aa 365-404) has not yet been determined (74). Domain II contains
`
`
`
`
`important information for protein secretion (75); the absence of Domain I
`
`
`
`shortens the half-life of OVB3-PE immunotoxin in mice (76). Once inside the
`
`
`cell, native PE undergoes proteolytic cleavage at a site rich in arginine
`
`
`
`
`
`residues. Primary sequence analysis indicates the presence of a consensus
`
`
`
`
`
`sequence homologous to the endoplasmic retention signal sequence (KDEL)
`
`
`at the C-terminal end. Amino acid substitutions at this region affect transloca­
`
`tion of PE to the cytoplasm (77).
`
`
`Pseudomonas exotoxin is a lethal toxin with an ID50 for a 20-gram mouse
`
`in the range of 0.1-0.2 JLg. Liver toxicity is a major cause of the death in
`
`
`
`
`
`mice (78-80). Intact PE has been coupled to a variety of tumor-binding mono­
`
`clonal antibodies (for example 260F9, 454Cl l, 280D ll, JLl, 106AIO,
`245E7, 520C9) to produce potent IT, with ID50 in the 0.01-0.1nM range
`
`
`
`after a 24 hr exposure of cells to IT (8). In addition to killing cancer cells
`
`
`
`
`in culture, PE-containing IT are also effective in inhibiting tumor growth
`
`in mice. However, PE conjugates cannot be given in large amounts to
`
`
`animals or patients because of residual binding of PE-IT to normal cells
`(74).
`To minimize this undesirable side effect, Chaudhary et al (75, 81) used
`
`
`
`
`
`
`
`genetic engineering to construct a plasmid containing codons for amino acids
`
`
`252-613 of PE and changed the glutamic acid at position 252 to lysine by
`
`
`
`
`site-directed mutagenesis. This construct was expressed in E. coli and pro­
`
`
`
`duced a protein of 40,000 daltons (Lys PE40) that was 100-fold less toxic to
`mice than was intact PE and lacked Domain Ia of PE (74). An extra lysine
`
`
`
`
`residue at the N-terminus of PE40 also facilitated derivatization with cross­
`
`
`
`linking reagents (75, 81). Lys57 (in Domain la) was shown to be involved in
`
`
`
`
`the cellular binding of PE (82) and, when mutated to arginine, decreased the
`
`Annu. Rev. Pharmacol. Toxicol. 1992.32:579-621. Downloaded from www.annualreviews.org
`
`by Reprints Desk, Inc. on 05/20/14. For personal use only.
`
`IMMUNOGEN 2176, pg. 9
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`588 RAMAKRISHNAN ET AL
`
`
`
`
`toxicity to murine cells in vitro and also was less toxic to mice. Use of PEarg57
`as the toxic moiety in IT also reduced liver damage.
`
`Cytotoxic Conjugates Made with Native PE and Genetically
`
`
`
`
`Engineered P E
`Pai et al (76) coupled a monoclonal antibody OVB3, which reacts with many
`
`
`
`
`
`
`human carcinomas, to native PE, lys PE40, and PEarg57, and compared the
`
`
`characteristics of the three IT. Each was cytotoxic to human tumor cell lines
`
`
`
`expressing the OVB3 antigen on their surfaces. All three IT caused complete
`
`
`regression of 50mm3 tumors with no toxic effects to the animals at therapeutic
`
`
`doses. The half-life in blood of OVB3-PE and OVB3-PEarg57 was 20 hr,
`
`
`whereas the half-life of OVB3-LysPE40 was 4 hr. But the single dose LD50
`
`value for OVB3-lys PE40 (200lLg) was much larger than for OVB3-PE or
`
`OVB3_PEarg57 (2.5 J-Lg and 3.5 J-Lg). This means OVB3-lys PE40 can be
`
`
`
`
`
`administered in higher doses to treat cancer more efficiently (76). In a recent
`
`
`
`
`study, PE linked to an antibody directed to human cervical carcinoma was
`
`
`
`
`evaluated for tumor growth inhibition in vivo. The F(ab)z fragment of the
`
`
`
`
`or nonreducible antibody was chemically conjugated to PE via a reducible
`
`
`
`
`thioether bond. Both conjugates were tumor suppressive when injected 4 to 14
`
`days after tumor transplantation (83).
`Immunotoxins made by chemically coupling protein toxins to specific
`
`
`
`
`
`
`
`
`
`antibodies are faced with low yields, formation of heterogenous products, and
`
`
`
`
`difficulties in large-scale production. To circumvent s()me of these problems,
`
`
`
`
`various chimeric toxins have been prepared by genetically fusing the coding
`
`
`
`
`
`sequences of toxin moieties to carrier proteins (84). Chaudhary et al (85)
`
`
`
`
`constructed a single chain antibody toxin fusion protein by ligating the DNA
`
`
`
`fragment of anti-Tac (Fv) with the gene of PE40. Anti-Tac is a monoclonal
`
`
`
`
`antibody to the P55 subunit of the human interleukin-2 receptor. A variable
`
`
`domain is the smallest binding unit of an antibody. The fusion protein
`
`anti-Tac (Fv)-PE40 was highly cytotoxic
`
`to two IL-2 receptor-bearing human
`
`
`cell lines but was not cytotoxic to receptor-negative cells (85). PE-40 was also
`
`
`to create a linked to the HIV -binding portion of the human CD4 molecule
`
`
`
`
`
`hybrid protein, CD4-PE40. CD4-PE40 displayed selective toxicity toward
`
`
`
`
`cells expressing the HIV envelope glycoprotein gp120 and could therefore be
`
`
`
`used as a therapeutic agent for the treatment of Acquired Immune Deficiency
`Syndrome (86).
`In addition, many receptor-specific chimeric toxins have been constructed
`
`
`
`
`
`by fusing cDNAs encoding cytokines (TGFa, IL2, IL4, IL6) to the gene of
`
`
`
`
`PE40. Each chimeric protein was specifically cytotoxic to the appropriate
`
`
`
`
`receptor-bearing cells (87-90). Interestingly, the position of the ligand moi­
`
`
`
`ety, i.e. cell recognition element, in the chimeric molecule might be impor­
`
`
`tant. When the N-terminus of PE40 was fused to the C-terminus of TGF-a
`
`Annu. Rev. Pharmacol. Toxicol. 1992.32:579-621. Downloaded from www.annualreviews.org
`
`by Reprints Desk, Inc. on 05/20/14. For personal use only.
`
`IMMUNOGEN 2176, pg. 10
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`
`CYTOTOXIC CONJUGATES
`589
`
`(TGF-a-PE40), the conjugate was 30-fold more potent than a fusion between
`
`
`
`
`
`the C-tenninus of PE40 to the N-tenninus of TGF-a (PE40-TGF-(a). The
`
`
`fonner construct recognized and bound with about the same affinity as EGF.
`
`
`
`
`In native PE the cell recognition domain (Ia) is attached directly to the
`
`
`
`translocation domain. The close association of the receptor recognition and
`
`
`
`translocation domains may possibly facilitate movement of toxin into the
`
`cytosol (88).
`
`DIPHTHERIA TOXIN
`
`Structural Features
`
`Diphtheria toxin (DT) is a single polypeptide exotoxin secreted by
`
`
`
`
`
`
`
`and is lysogenic for bacteriophage Betatox+ that
`
`Corynebacterium diphtheriae
`
`
`
`
`carries the gene for DT (91, 92). Intact DT has two disulfide bridges. After
`
`
`
`
`mild trypsin digestion ("nicking") and reduction of the disulfide bonds, DT
`
`
`can be separated into an N-terminal 2 1 . 1 7-kd fragment A of 1 93 amino acids
`
`( 1-193) and a C-terminal 37.20-kd fragment B of 342 amino acids from
`
`position 1 94 to 535 (8, 93, 94) .
`The A chain enzymatically ADP ribosylates a unique amino acid (diptha­
`
`
`
`
`
`mide) of elongation factor-2 (EF-2), resulting in irreversible inactivation and
`
`tennination of cellular protein synthesis (95). The B chain has two functions:
`
`
`
`
`
`binding cell surface receptors and translocation of the A chain to the cytosol.
`
`
`
`
`B inding activity is located in the 8-kd C-terminal fragment of the B chain, and
`
`
`the last 50 amino acids are confirmed to be required to form the receptor
`
`
`
`
`binding domain (94, 96). The translocation activity is in the N-terminal three
`
`fourths of fragment B . From amino acids 270 to 370 and amino acids 42

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