throbber
antibody-based therapy
`
`Immunotoxins: magic bullets
`or misguided missiles?
`
`Ellen S. Vitetta, Philip E. Thorpe and
`Jonathan W. Uhr
`
`Thirteen years have passed since specific in vitro and in vivo killing of
`tumor cells by immunotoxins was first described. Why, then, has it taken
`so long to determine whether these pharmaceuticals will have a major
`impact on the treatment of cancer, AIDS and autoimmune disease? The
`answer is that the transfer of basic discoveries to the clinic is a slow,
`multistep, interdisciplinary process. Thus, immunotoxin molecules must
`be designed and redesigned by the basic scientist depending on the efficacy
`and toxicity shown in vitro and in relevant experimental models. Next,
`each version must be evaluated by clinicians in humans through a lengthy
`process (1—3 years) in which the dose regimen is optimized and in which
`new problems and issues frequently emerge. These problems must again
`be modeled and studied in animals before additional clinical trials are
`initiated. In this article, Ellen Vitetta and colleagues discuss both basic
`and clinical aspects of the development of immunotoxin therapy.
`
`mAbs can be used as intact molecules or as frag-
`ments‘. While fragments are less immunogenic,
`they
`have a shorter half-life in vivo and are often partially
`inactivated by their coupling to toxins. These problems
`should be circumvented by generating fusion proteins
`containing portions of the constant
`regions of the
`heavy chain, which confer a long half—life
`in the
`circulation.
`Growth factors Other ligands for preparing immuno-
`toxins are growth factors”. Although these bind to
`normal cells, tumor cells frequently express elevated
`levels of growth factor receptors. Advantages of using
`growth factors as ligands include their relative lack of
`immunogenicity, high affinity for their receptors, and
`the availability of cloned genes for generating fusion
`proteins. Problems include rapid in vivo clearance,
`stimulation of target cells by small amounts of bound
`immunotoxin insufficient
`to kill
`the cells, and the
`presence of circulating ligands or soluble receptors that
`compete for the immunotoxin.
`
`Immunotoxins are chimeric molecules in which cell-
`binding ligands are coupled to toxins or their subunits.
`If the ligand moiety is tumor cell-specific, the immuno-
`toxin should kill tumor cells selectively, unlike conven-
`tional chemotherapy and radiotherapy, which kill
`rapidly dividing or metabolizing cells, whether malig—
`nant or normal.
`
`Components of an immunotoxin
`The toxins used for different types of immunotoxins
`are depicted in Table 1, and their components, ligand,
`toxin and crosslinker, are discussed below'.
`
`Ligand
`Monoclonal antibodies The ligand most frequently used
`is a cell-reactive monoclonal antibody ’mAb)‘. Although
`tumor—reactive mAbs often react with some normal tis-
`sue, crossreactivity does not necessarily prohibit their
`use. Thus, low antigen density, anatomical barriers or
`poor endocytcsis could prevent the killing of a cell that
`has a crossreacting antigen”. Conversely, some cross—
`reactions not detectable by conventional
`techniques
`can damage life-sustaining tissues]. Hence, a primate
`model in which the mAb reacts with the primate anti-
`gen is desirable to test the safety of an immunotoxin to
`be used in humans.
`Only a proportion of mAbs make potent immuno-
`toxins‘. Depending on their specificity, they may not
`be internalized or, if they are, they may not be routed
`to the appropriate intracellular compartment
`for
`translocation of their attached toxin into the cytosol.
`Hence, mAbs must also be screened for effectiveness as
`carriers of toxin.
`
`Toxin
`The toxins used for immunotoxins are derived from
`bacteria and plants and all inhibit protein synthesis (as
`described below; and see Table 1). Unlike chemothera-
`peutic agents, these toxins kill both resting and dividing
`cells. Hence, as immunotoxins, they have the potential
`to kill tumor cells that are not in cycle at the time of
`treatment (dormant
`tumor cells) and that may be
`spared by conventional chemotherapy. These toxins
`share common features7:
`1 They are all synthesized as single chain proteins and
`are processed either post translationally or in the
`0 I993. Elsevier Science Publishers Ltd, UK. 0l67-569v/9JISDG.00
`
`Immunology Today 252 Vol. 14 No. 6 1993
`
`Immunogen 2144, pg. 1
`Phigenix v. Immunogen
`|PR2014-00676
`
`Immunogen 2144, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`antibody-based therapy
`
`Table 1. Structure and function of toxins and RIPs used for immunotoxins
`
`
`
`LD,0 of immunotoxins
`(mice, mg kg"
`Structure of mature form
`Toxin
`A-chain action
`Toxin receptor
`total protein)
`
`Diphtheria toxin
`(DT)
`
`o—ss—o
`
`Truncated
`diphtheria toxin
`(DAB486)
`
`Pseudomonas
`exotoxin (PE)
`
`Truncated
`Pseudomonas
`
`exotoxin (PE40)
`
`Ricin/abrin
`
`Blocked
`ricin/abrin
`
`Ricin toxin
`A-chain (RTA)
`
`Ribosome
`inactivating
`protein (RIP)
`
`3;}.
`:3
`
`.
`‘3)
`‘
`
`
`
`3”
`
`heparin-binding
`epidermal growth
`factor-like precursor
`
`none
`
`ADP-ribosylation of
`elongation factor 2
`
`ADP-ribosylation of
`elongation factor 2
`
`uz—macroglobulin
`receptor-like molecule
`
`ADP-ribosylation of
`elongation factor 2
`
`none
`
`galactose
`
`none
`
`none
`
`ADP—ribosylation of
`elongation factor 2
`
`N—glycosidase for
`285 ribosomal RNA
`
`N—glycosidase for
`285 ribosomal RNA
`
`N-glycosidase for
`285 ribosomal RNA
`
`0.3
`
`>1.0
`
`0.1
`
`2.0
`
`0.1—0.2
`
`0.4-0.8
`
`20
`
`5—20
`
`N~glycosidase for
`285 ribosomal RNA
`
`
`none
`
`A, B: different polypeptide chains; I: hydrophobic region in the polypeptide; indentations: cell-binding sites; X: partial or complete
`blockade of Iectin activity at the binding site.
`
`target cell to which they are delivered into two-chain
`molecules with interchain disulfide bonds.
`2 The disulfide bond linking the two chains is critical
`for cytotoxicity.
`3 All
`toxins have subunits or domains devoted to
`binding to cells,
`translocation across membranes,
`and the destruction of protein synthesis in the cell.
`These domains can be separated or genetically
`manipulated to delete those that are unwanted.
`Plant toxins The most widely used plant toxins, ricin
`and abrin, consist of two disulfide-linked polypeptides,
`A and B (Ref. 8). The toxin binds via the B-chain
`to galactose—containing glycoproteins and glycolipids
`that are present on the surface of all cell types. The
`toxin is then endocytosed and routed to the trans-
`Golgi network which is believed to be the site where
`the A-chain translocates to the cytosol. The A-chain
`then kills the cell by enzymatically removing a crucial
`adenine
`residue
`from the 605 ribosomal
`subunit
`which is needed for the binding of elongation factor 2
`(EF-Z) during protein synthesis”. Ribosome inacti-
`vating proteins (RIPS) are single—chain proteins found
`in many plants”, and have the same enzymatic proper—
`ties as the A-chain of ricin”.
`Bacterial toxins The active form of diphtheria toxin
`
`(DT) is a disulfide-bonded two-chain molecule”. The
`toxin binds via the B-chain to an epidermal growth
`factor-like receptor that is present on most cell types
`in DT-sensitive species”. The toxin is then endocy-
`tosed and, within an acidic intracellular compartment,
`the B-chain undergoes a conformational change to
`expose hydrophobic regions, which are thought
`to
`be important in enabling the A-chain to translocate
`across the membrane to the cytosol“. The A-chain
`then kills the cell by catalysing a modification of
`EF-Z that prevents
`its
`participation
`in
`protein
`synthesis”.
`is produced by the
`Pseudomonas exotoxin (PE)
`bacterium as a single-chain protein“. It binds via its N-
`terminal
`region (domain I)
`to an az-macroglobulin
`receptor-like molecule present on the surface of most
`cell
`types”. The toxin is
`then endocytosed and
`becomes converted through the action of proteolytic
`enzymes into a disulfide-bonded two—chain form”.
`The C-terminus of domain Ill (the equivalent of the
`A-chain) has an endoplasmic reticulum retention se—
`quence, REDLK, which causes the toxin to concentrate
`in the endoplasmic reticulum — probably the site where
`domain III enters the cytosol. Once in the cytosol, the
`toxin kills the cell in the same manner as DT.
`
`Immunology Today
`
`253 Vol. 14 No.61993
`
`Immunogen 2144, pg. 2
`Phigenix v. Immunogen
`|PR2014-00676
`
`Immunogen 2144, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`antibody-based therapy
`
`.
`Crosslinker
`The crosslinker used to join the ligand and the toxm
`must remain stable extracellularly but be labile intra-
`cellularly so that
`the toxic fragment can enter the
`cytosol. The choice of crosslinker depends on whether
`intact toxins, A-chains or RIPs are used. A-chains and
`RIPS are coupled to ligands using linkers that intro-
`duce a disulfide bond between the ligand and the
`A-chain'l". Bonds that cannot be reduced render these
`immunotoxins much less toxic or nontoxic probably
`because the A—chain must be released from the ligand
`by reduction to be cytotoxic”. Such immunotoxins
`tend to be labile in UiI/O unless hindered crosslinkers
`are used. These crosslinkers place bulky groups around
`the disulfide bond to protect it from attack by thiols
`in the blood and tissues'. Intact
`toxins are usually
`linked to ligands using nonreducible (cg.
`thioether)
`linkages to prevent release of active free toxin in UIl/O.
`Recombinant
`immunotoxins have been prepared by
`splicing the genes
`encoding truncated DT (c.g.
`DAB486) or Pseudcmonas exotoxin (e.g. PE40) to the
`gene encoding the ligand and expressing the entire
`immunotoxin as a
`fusion protein”. Recombinant
`immunotoxins are highly stable in VII/O because they
`contain nonreducible peptide bonds.
`
`Preclinical evaluation
`Cytotoxic potency and specificity
`Immunotoxins prepared from holotoxins (i.e. com-
`plete toxins containing both A- and B-chains or anal—
`ogous domains) are usually more potent than those con—
`raining A-chains (or RIPS) because the toxin moiety
`can interact with toxin receptors in or on the target
`cellu'z‘. This enables the immunotoxin to enter and kill
`the cell by the highly efficient entry pathway used
`by the native toxin. Predictably, however, holotoxin—
`containing immunotoxins are highly toxic to animals
`because they can bind to toxin receptors that are pres-
`ent on non-target cells. The problem of nonspecific tox-
`icity can be reduced with ricin-containing immunotox-
`ins by reversibly blocking the galactose-binding sites of
`the toxin either Sterically by the ligand itself or with
`galactose-based affinity labels'iz‘. Such ‘blocked’ ricin
`immunotoxins appear to act by being degraded inside
`the cell to release unblocked ricin or ricin fragments.
`Immunotoxins prepared from ricin A-chain or
`RIPS are highly specific for their designated target cells
`but vary in potency depending on the affinity of the
`ligand, the cell surface molecule and epitope that it
`recognizes and the capacity of that molecule to enter
`an intracellular compartment
`that
`is
`favorable for
`toxin translocation'l‘”. Immunotoxins prepared from
`DAB486 or PE40, which lack cell-binding domains,
`are also highly specific in their cytotoxic action on
`target cellsu'”.
`It
`is unclear whether immunotoxins
`containing DAB486 or PE40 are more uniformly
`cytotoxic than their A-chain counterparts, as might be
`expected if the truncated toxins have, for example,
`hydrophobic regions that assist the ent: y of the enzy-
`matic fragment or subunit into the cytosol. Thus far,
`the evidence with PE40—containing immunotoxins is
`that they have variability in potency similar to that of
`A-chain immunotoxins.
`
`Immunotoxins prepared with A-chains can often be
`made more potent by lysosomotropic amines and
`carboxylic ionophores, which inhibit
`the fusion of
`endosomes with lysosomes (where the A-chains are
`destroyed) or retard the transit of the immunotoxins
`through
`a
`compartment
`favorable
`for A—chain
`translocationn'”.
`
`Toxicity
`Many immunotoxins can cause hepatotoxicity‘
`(Tables 1 and 2). In the case of ricin A-chain (RTA),
`mannose- and fucose-containing oligosaccharides bind
`to liver cells leading to rapid clearance and hepatic
`damage'. This problem has been successfully circum-
`vented either by deglycosylating RTA (chemically or
`enzymatically)‘ or using recombinant RTA (expressed
`in a non-glycosylating cell)". In the case of blocked
`ricin immunotoxins,
`the oligosaccharides on the A-
`and B-chains and the affinity labels used to block the
`B-chain’s lectin sites result in liver homing and liver
`damage“. Bacterial toxins and RIPs produce hepato-
`toxicity by binding to molecules other than carbohy-
`drate receptors on liver cells or by binding to serum
`proteins that have receptors in the liverl'”.
`RTA-based immunotoxins cause vascular leak in
`humans, which is manifested by extravasation of fluids
`and proteins from the vasculature into the periphery
`causing edema and weight gain, and, occasionally, life-
`threatening pulmonary edema“. The mechanisms
`underlying vascular
`leak are not known, although
`recent evidence suggests that they may be related to the
`binding of the RTA to vascular endothelial cells”. In
`addition, these immunotoxins cause myalgias (rarely,
`rhabdomyolysis) via unknown mechanisms.
`
`Pharmacokinetics
`An effective immunotoxin must have a serum half-
`life of sufficient duration for a cytotoxic quantity of it
`to access the target cells. When the target cells are
`intravascular (for example, circulating tumor cells or
`normal lymphocytes), access is not a problem and the
`immunotoxins are highly effective, but when the target
`cells reside in large solid tumor masses with a poor
`blood supply and high interstitial pressure“, the need
`for a long serum half-life becomes critical. The half-life
`of immunotoxins prepared with mAbs is longest when
`the mAbs are intact, the crosslinker is stable and the
`toxin moiety does not bind to normal
`tissues.
`In
`contrast, when the ligand is an antibody fragment or
`growth factor,
`the crosslinker is not stable, or the
`toxin displays some nonspecific binding, the half—life is
`short”. The problem of a rapid half-life can be partially
`solved by continuous intravenous infusion of
`the
`immunotoxin, although increasing the half-life may
`also increase the likelihood that these immunotoxins
`will gain access to other tissues and cause unwanted
`toxicities.
`
`Immunogenicity
`Individuals with a functional immune system make
`antitoxin antibodies even when humanized‘antibodies
`or human growth factors are used as carriers"-“.
`Strategies to decrease such immunogenicity, such as
`
`Immunology Today
`
`254 Vol.” No.61993
`
`Immunogen 2144, pg. 3
`Phigenix v. Immunogen
`|PR2014—00676
`
`Immunogen 2144, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`antibody-based therapy
`
`immunosuppressive
`concomitant administration of
`drugs, have not yet been successful
`in humans“. In
`contrast, multiple courses of immunotoxin can be
`given to highly immunosuppressed individuals, such as
`B-cell lymphoma patients, without a resultant immune
`response”. Even in these patients, when earlier disease
`is treated,
`immunogenicity will become a problem.
`Circulating antibodies can inhibit
`the efficacy of
`immunotoxins by increasing their rate of clearance,
`and/or by blocking the binding site on the antibody or
`the enzymatic site on the toxin. Despite these consider-
`ations, immunotoxins have been administered in the
`face of serum antibody and, in some cases, have been
`effective”. With immunotoxins of a very short half-life
`(e.g.
`IL-2-DAB486; Ref. 38),
`the binding of non-
`neutralizing antibody may,
`in fact, increase the half-
`life. Nevertheless,
`immunogenicity will
`remain a
`problem until the entire immunotoxin is humanized.
`This may be possible by using human ‘toxins’, such as
`ribonuclease, attached to human antibody“. However,
`even this strategy may not avoid the formation of new
`immunogenic epitopes created by linking autologous
`proteins.
`
`Immunotoxin-resistant mutants
`In several rodent tumor models, immunotoxins have
`produced excellent tumor regressions but have failed
`to cure the animals because immunotoxin-resistant
`tumor
`cells emerge”. These are usually antigen-
`deficient mutants whose outgrowth can be prevented
`by
`administering immunotoxin cocktails directed
`against
`alternative
`tumor—associated
`antigens‘3~”.
`However, mutants have also been observed that have
`defects in intracellular transport of the endocytosed
`immunotoxin".
`Importantly, mutants with toxin-
`resistant ribosomes have not been observed, suggesting
`that such mutations may be lethal.
`
`Difficulties in evaluating immunotoxins
`From experimental studies and theoretical consider-
`ations, the optimal efficacy of immunotoxin should be
`obtained by administration of a single short course in
`patients with minimal', dormant“, or premalignant‘”
`disease. The latter is a particularly attractive state for
`intervention since
`the development of
`full-blown
`malignancy appears to require an additional
`rare,
`stochastically determined genetic event. Hence, killing
`of 100—1000 premalignant cells would probably prevent
`development of malignancy.
`However, the design of clinical trials does not allow
`this strategy to be tested readily. The initial
`trials
`(Phase I) require treatment of patients with intractable
`disease. Dose escalations of the drug are performed in
`small cohorts of patients until the maximally tolerated
`dose (MTD) is established. Side-effects, pharmacokinetics,
`and immunogenicity are analysed. As in most Phase I
`clinical
`trials, clinical benefit
`is unlikely to occur
`because the patients have far—advanced, bulky tumors
`and organ damage from previous therapy. Alterations
`in the drug or the protocol are usually not acceptable
`until completion of the trial. Therefore, lack of efficacy
`in a Phase I trial should not preclude further clinical
`testing of the drug.
`
`then administered at a safe dose to
`The drug is
`patients with less advanced disease to determine effi-
`cacy (Phase II
`trial). Generally, a response rate of
`20—40% (partial or complete remissions) must be
`observed at Phase II, or drug development is halted.
`This may be too stringent a criterion for an agent that
`is likely to be most effective when used in the treat-
`ment of minimal disease and in combination with one
`or two other immunotoxins and chemotherapy. An
`additional problem is that an MTD established in
`patients with bulky tumor may be very different from
`that in patients with minimal disease.
`Phase III trials require several hundred patients to be
`treated in multiple clinical centers along with controls
`(who usually receive placebos or the current
`‘best’
`therapy) and, therefore, requires support by a pharma-
`ceutical company. The result of the above consider-
`ations is that few immunotoxins have proceeded beyond
`the stage of Phase I or II trials. Therefore it might be
`wiser to test immunotoxins by an alternative strategy,
`for example to establish MTD (Phase I)
`in patients
`with less bulky disease and then use a safe dose in
`combinatorial therapy (Phase II) before proceeding to
`randomized Phase III
`trials in which immunotoxins
`plus or minus additional therapies are compared for
`efficacy.
`
`Clinical trials
`
`The completed or ongoing clinical trials involving
`systemic therapy with immunotoxins are summarized
`in Table 2. The major findings to emerge are:
`1 The side-effects of immunotoxin therapy are differ-
`ent from those of conventional therapy, in that there
`is no damage to rapidly dividing normal
`tissues.
`Blocked immunotoxins consisting of ricin, DT and
`Pseudomonas exotoxin routinely cause hepatotox-
`icity. All
`the
`ricin-based immunotoxins
`cause
`reversible vascular leak and myalgias. The MTD
`appears inversely related to the half life and the
`stability of the immunotoxin are directly related to
`the size of the antigenic sink. Multiple courses of
`immunotoxin therapy have been well
`tolerated,
`indicating that toxicity is not cumulative.
`2 Severe neurotoxicity was observed in two trials and
`was due to cross-reactivities of the antibody portion
`of
`the immunotoxins with neural cells‘“. This
`emphasizes the importance of carefully screening
`antibodies for unexpected cross-reactivities with life-
`sustaining tissues and, when possible,
`selecting
`mAbs which cross-react with their homologs in non-
`human primates. Conversely, administration of an
`anti-CD19 immunotoxin that was known to cross-
`react with astrocytes“ did not cause CNS lesions,
`presumably because the astrocytes were inaccessible
`to the immunotoxin, or were insensitive to it.
`3 Optimal regimens for administration of the immuno-
`toxins have not yet been devised. The half-lifes in
`trials to date have generally been shorter than would
`be predicted to induce an optimal therapeutic index.
`4 A general problem is that techniques for isolating
`and immunophenotyping the malignant progenitor
`cells have not been developed for the majority of
`tumors. The assumption usually has to be made that
`
`Immunology Today
`
`255
`
`Vol. )4 No. 6 1993
`
`Immunogen 2144, pg. 4
`Phigenix v. Immunogen
`|PR2014—00676
`
`Immunogen 2144, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Trial
`Disease
`phase
`
`
`Metastatic
`melanoma
`
`Colorectal
`carcinoma
`Metastatic
`breast
`carcinoma
`
`Ovarian
`carcinoma
`Non-Hodgkin's
`lymphoma
`
`Hodgkin‘s
`disease;
`non-Hodgkin‘s
`lymphoma
`Hodgkin's
`disease
`B—cell chronic
`lymphocytic
`leukemia
`
`T-cell
`lymphoma
`B-cell acute
`lymphoblastic
`leukemia
`
`l
`
`ll
`
`II
`
`l
`
`l
`
`l
`
`I
`
`I/Il
`
`I
`
`l
`
`I
`
`I
`
`I
`
`l
`
`l
`
`l
`
`Xomazyme-Mel
`
`Xomazyme-Mel
`
`Xomazyme-Mel
`plus cyclophosphamide
`
`Anti-gp72—ricin
`toxin A—chain
`260F9—ricin
`toxin A-chain
`(bolus)
`260F9—ricin
`toxin A-chain
`(continuous infusion)
`Anti-OVB3-
`Pseudomonas exotoxin
`Anti-CD19-
`blocked ricin (bolus)
`
`Anti-CD19—
`blocked ricin
`(continuous infusion)
`Fab‘ anti-CDZZ-
`deglycosylated
`ricin A-chain
`
`IgG anti-CDZZ-
`deglycosylated
`ricin A-chain
`
`IL-Z-truncated
`diphtheria
`toxin (DAB 486)
`
`Anti-CD30—
`saporin
`Anti-CD5 (TI-OI)—
`ricin toxin
`A-chain
`
`Anti-CD5 (H65)—
`ricin toxin A—chain
`Anti—CD19
`(B43)—PAP
`
`>3
`
`n.d.
`
`n.d.
`
`>1
`
`0.05
`
`0.4
`
`n.d.
`
`0.25
`
`0.35
`
`1.8
`
`0.7
`
`1.5
`
`n.d.
`
`n.d.
`
`3.3
`
`Not yet
`reached
`
`1.3
`
`vascular leak syndrome, myalgia
`
`vascular leak syndrome
`
`vascular leak syndrome
`
`vascular leak syndrome, aphasia
`
`vascular leak syndrome, myalgia,
`paresthesia
`
`vascular leak syndrome, myalgia,
`neuropathies
`
`SGOT/SGPT elevations, abdominal
`pain, encephalopathy
`SGOT/SGPT elevations,
`thrombocytopenia
`
`SGOT/SGPT elevations,
`thrombocytopenia, edema
`
`Vascular leak syndrome, myalgia
`
`vascular leak syndrome, myalgia
`
`hepatic transaminase elevations,
`hypoalbuminemia, hypersensitivity,
`creatinine elevations, thrombocytopenia,
`renal insufficiency
`thrombocytopenia, SGOT/SGPT
`elevations, proteinuria
`fever
`
`vascular leak syndrome, dyspnea
`
`hypoalbuminemia
`
`vascular leak syndrome, myalgia,
`hematuria, tremors
`
`
`antibody—based therapy
`
`Table 2. Summary of clinical trials of immunotoxins
`
`Maximum
`tolerated
`dose (total
`Toxicity
`Immunotoxin
`mg kg")
`t_’____~___________—»———————~——_.____._.__.a
`
`Anti-CD5 (H65)—
`Steroid-resistant
`ricin toxin A-chain
`graft-versus-host
`isease
`
`
`ll
`
`"Human antibodies made by the patient against the two components of the immunotoxin. measured by radio- or enzyme~
`linked-immunoassay.
`ARE anti-ricin A-chain antibody; AM: anti-mouse lg antibody; ADT: anti-diphtheria toxin antibody; AIL-2: anti-lL-Z
`antibody; AS: anti-saporin antibody.
`
`Immunology Today 256 Vol, 14 No. 6 1993
`
`Immunogen 2144, pg. 5
`Phigenix v. Immunogen
`|PR2014—00676
`
`Immunogen 2144, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Antibody
`production
`in response to
`toxin‘
`
`17/21
`
`n.d.
`
`13/13
`
`15/16 AR
`16/17 AM
`4/4
`
`4/5 AR
`3/5 AM
`
`n.d.
`
`12/15 ARIAM
`
`26/43 ARIAM
`
`4/14 AR
`1114 AM
`
`8/24 AR
`7/24 AM
`
`45/109 ADT
`28/109
`AIL-2 (26%)
`
`4/4 AM
`4/4 AS
`1/4 AR
`0/4 AM
`
`n.d.
`
`n.d.
`
`n.d.
`
`n.d.
`
`8.3
`
`4—6
`
`n.d.
`
`n.d.
`
`n.d.
`
`1.4
`
`7.8
`
`0.1—0.33
`
`n.d.
`
`n.d.
`
`3/43 PR
`.
`1/43 mixed
`9/43 stabilization
`
`4/20 PR
`
`5/16 mixed tumor
`regressions
`1/4 resolution of
`lung nodule
`
`0/5
`
`0/23
`
`1/25 CR
`2/25 PR
`10/25 mixed or transient
`
`2/43 CR
`5/43 PR
`11/43 transient
`
`5/13 PR
`
`6/24 PR
`1/24 CR
`
`4/109 CR
`8/109 PR
`
`3/4 PR
`
`4/5 transient
`rapid fall in circulating
`leukemic cells
`
`antibody-based therapy
`
`
`
`tm (h)
`
`Clinical
`responsef
`
`
`Reference
`
`1/22 CR
`9/22 mixed or stabilized
`
`Spitler, LE. er al. (1987)
`Cancer Res. 47, 1717-1723
`
`Spitler, LE. et al. (1988)
`in Immtmotoxins (Frankel, A.E., ed.),
`pp. 493-515, Kluwer Academic Publishers
`
`Oratz, R. et al. (1990)
`]. Biol. Resp. Med. 9, 345—354
`
`Byers, V.S. er al. (1989)
`Cancer Res. 49, 6153—6160
`Weiner, LM. et al. (1989)
`Cancer Res. 49, 4062—4067
`
`Gould, 8.]. et al. (1989)
`1. Natl Cancer Inst. 81, 775—781
`
`Pai, LH. et a1. (1991)
`J. Clin. Oncol. 9, 2095—2103
`Grossbard, ML. et al. (1992)
`Blood 79, 576—585
`
`Grossbard, M.l.. et al. (1992)
`Blood 79, 576—585
`
`Vitetta, 12.5. et al. (1991)
`Cancer Res. 15, 4052-4058
`
`Amlot, P.L. et 11]., unpublished
`
`Uckun, EM. et al., unpublished
`
`Falini, B. et al. (1992)
`Lancet 339, 1195—1196
`Hertler, A.A. et al. (1989)
`Int. ]. Cancer 43, 215—219
`
`LeMaistre, C.F. er (ll. (1991)
`Blood 78, 1173~1 182
`
`Uckun, EM. et al., unpublished
`
`10/12 ARIAM
`
`1.2—2.9
`
`4/14 PR
`
`12/14 AM
`
`4.4—5.7
`
`80—99% decrease in
`circulating B cells in 4/5
`
`6/23 AM
`6/23 AR
`
`1.5-3.9
`Byers, V.S. et al. (1990)
`9/32 CR
`Blood 75, 1426—1432
`7/32 PR
`6/32 mixedM
`
`f Measured in terms of the change in ‘tumor burden’, which is the sum of the products of perpendicular diameters of all tumor
`nodules by C1" scans. CR: complete response = tumor burden l« 100%; PR: partial response = tumor burden l 50-99%.
`.
`n.d.: not determined; SGOT: serum glutamic—oxaloacetic transaminase; SGPT: serum glutamtc—pyruVm transammase;
`PAP: pokeweed anti-viral protein.
`
`Immunology Today 257 Vol. 14 No. 6 1993
`
`Immunogen 2144, pg. 6
`Phigenix v. Immunogen
`|PR2014—00676
`
`Immunogen 2144, pg. 6
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`antibody-based therapy
`
`the malignant progenitors bear the same antigens as
`their progeny. If, however, the major population of
`tumor cells derives from a progenitor cell lacking the
`target antigen, immunotoxin therapy will be palli-
`ative, not curative.
`5 Clinical responses in lymphomas have been excellent
`considering that most trials were Phase 1, patients
`had bulky disease, and a single immunotoxin was
`used.
`In eight separate trials,
`the percentage of
`patients achieving partial or complete remissions
`ranged from 12 to 75% (Table 2). To put this
`into perspective, of those drugs which are mar—
`keted today for the treatment of cancer, the great
`majority (>90%) produced fewer
`than a 5%
`response rate in Phase I trials. In addition, clinical
`responses have also been excellent (28% complete
`responses) when accessible circulating T cells have
`been targeted as a treatment for steroid-resistant
`graft-versus~host disease or autoimmune disease“.
`By contrast, responses of large tumor masses to
`immunotoxin therapy have rarely been seen in Phase
`I
`trials in patients with solid tumors, primarily
`because of inaccessibility of the cells to the immuno-
`toxins‘w’.
`
`Future prospects
`The ideal immunotoxin should be non-immunogenic
`and cause minimal toxicity to normal tissue, yet have
`access to, and the potency to kill, 10“—10lz tumor cells
`and their progenitors
`in solid and disseminated
`tumors. Immunogenicity might be eliminated by com-
`plete humanization of the immunotoxin molecule or
`by using a short course of a potent immunosuppressive
`agent (e.g. anti-CD4). Further analysis of the struc-
`ture-function relationships of immunotoxins reveal
`ways by which to increase the therapeutic index.
`Elucidation of the mechanisms underlying side-effects
`may allow their successful treatment with conventional
`pharmaceuticals.
`is
`The problem of treating large solid tumors
`presently being approached by targeting the immuno-
`toxin to the vascular endothelial cells responsible
`for supplying blood to the growing tumor mass
`(F. Burrows and P. Thorpe, unpublished). The advan-
`tages of this approach are that endothelial cells are
`accessible, that they are normal and,
`therefore, un~
`likely to mutate. Furthermore, a single immunotoxin
`should be useful for treating a variety cf solid tumors,
`since the destruction of a single blood vessel should
`result in the death of a large number of tumor cells of
`any type.
`Immunotoxins may also prove useful in eliminating
`other undesirable cells such as those infected with
`human immunodeficiency virus and those involved in
`autoimmune disease“.
`
`Conclusions
`
`Clinical research is now focusing on refining dose
`regimens for already—developed constructs with the
`long-term goal of using cocktails of immunotoxins
`together with chemotherapy to treat minimal disease.
`Basic research is aimed at eliminating immunogenicity,
`understanding the basis for side-effects and developing
`
`new second- and third—generation immunotoxin con—
`structs with improved therapeutic indices and ability to
`attack solid tumors. Considering that
`took several
`decades after the introduction of chemotherapy before
`regimens were developed to cure patients, progress
`with immunotoxins has been substantial by the stan-
`dards of conventional drug development. The early
`clinical
`trials demonstrate considerable biological
`activity. The continued refinement in design of these
`pharmaceuticals in basic science laboratories further
`suggests that improved immunotoxins may eventually
`be useful
`in the treatment of cancer, autoimmune
`diseases and viral infections.
`
`We thank Ms Cindy Patterson for secretarial assistance.
`
`E.S. Vitetta is at the Cancer Immunobiology Center,
`RE. Thorpe and ].W. Uhr are at the Departments of
`Microbiology and Pharmacology, University of Texas
`Southwestern Medical Center, Dallas, TX 75235,
`USA.
`
`References
`1 Vitetta, E.S. and Thorpe, P.E. (1991) in Biologic
`Therapy of Cancer (DeVita, V.T., Jr, Hellman, S. and
`Rosenberg, S.A., eds), pp. 482—495, J.B. Lippincott
`Company
`2 Goldmacher, V.S. et al. (1989) ]. Cell. Physiol. 141,
`222-234
`3 Gould, 13.]. et a1. (1989) ]. Natl Cancer Inst. 81,
`775—781
`4 Bjorn, M.]., Ring, D. and Frankel, A. (1985) Cancer Res.
`45, 1214-1221
`5 Pastan, [., Willingham, M.C. and FitzGerald, D.J.P.
`(1986) Cell 47, 641—648
`6 Williams, D.P. et al. (1987) Protein Eng. 1, 493-498
`7 Olsnes, S., Kozlov, ].V., Van Deurs, B. and Sandvig, K.
`(1991) Semin. Cell Biology 2, 7—14
`8 Olsnes, S. and Pihl, A. (1982) in Molecular Action of
`Toxins and Viruses (Cohen, P. and van Heyningen, 5.. eds),
`pp. 51—105, Elsevier
`9 Endo, Y., Mitsui, K., Motizuki, M. and Tsurugi, K. (1987)
`J. Biol. Chem. 262, 5908-5912
`10 Stirpe, F. and Barbieri, L. (1986) FEBS Lett. 195, 1-8
`11 Endo, Y., Tsurugi, K. and Lambert, ].M. (1988)
`Biochem. Biophys. Res. Commun. 150, 1032—1036
`12 Collier, RJ. (1975) Bacterial. Rev. 37, 54-85
`13 Naglich, J.G., Metherall, ].E., Russell, D.W. and Eidels,
`L. (1992) Cell 69, 1051—1061
`14 Blewitt, M.C., Chung, LA. and London, E. (1985)
`Biochemistry 24, 5458—5464
`15 Pappenheimer, A.M., Jr (1977) Annu. Rev. Biochem.
`46, 69—94
`16 Allured, V.S., Collier, R.]., Carroll, S.F. and McKay,
`D.B. (1986) Proc. Natl Acad. Sci. USA 83, 1320—1324
`17 Kounnas, M.Z. et al. (1992)]. Biol. Chem. 267,
`12420—12423
`18 Ogata, M., Chaudhary, V.l(., Pastan, l. and FitzGerald,
`DJ. (1990)]. Biol. Chem. 256, 20678-20685
`19 Chaudhary, V.K., Jinno, Y., FitzGerald, D. and Pastan, l.
`(1990) Proc. Natl Acad. Sci. USA 87, 308—312
`20 Myers, D.E., Irvin, ].D., Smith, R.S., Kuebelbeck, V.M.
`and Uckun, EM. (1991)]. lmmunol. Meth. 136, 221—237
`21 Masuho, Y., Kishida, K., Saito, M., Umemoto, N. and
`Hara, T. (1982)]. Biochem. 91, 1583—1591
`22 Pastan, l., Chaudhary, V. and FitzGerald, DJ. (1992)
`Annu. Rev. Biochem. 61, 331—354
`
`Immunology Today
`
`258 Vol. 14 No.61993
`
`Immunogen 2144, pg. 7
`Phigenix v. Immunogen
`|PR2014—00676
`
`Immunogen 2144, pg. 7
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`antibody-based therapy
`
`23 Newton, D.L. et al. (1992) 1. Biol. Chem. 267,
`11917-11922
`24 Lambert, ].M., Goldmacher, V.S., Collinson, A.R.,
`Nadler, L.M. and Blattler, W.A. (1991) Cancer Res. 51,
`6236—6242
`25 Lambert, J.M. et al. (1991) Biochemistry 30, 3234—3247
`26 Youle, R.J., Uckun, F.M., Vallera, DA. and
`Colombatti, M. (1986) ]. Immunol. 136, 93—98
`27 Uckun, F.M. et al. (1992) Blood 79, 2649-2661
`28 Murphy, ].R. et al. (1987) Biochem. Soc. Symp. 53,
`9-23
`29 Casellas, P., Bourrie, BJ.P., Gros, P. and Jansen, F.
`(1984) 1. Biol. Chem. 259, 9359-9364
`30 Raso, V. and Lawrence, 1. (1984) ]. Exp. Med. 160,
`1234-1240
`31 O’Hare, M. et al. (1987) FEBS Lett. 216, 73—78
`32 Grossbard, M.L., Lambert, ].M., Goldmacher, V.S.,
`Blartler, WA. and Nadler, L.M. (1992) Cancer Res. 52,
`4200—4207
`33 Gherie, M.A., Uhr, J.W. and Vitetta, E.S. (1991) Cancer
`Res. 51, 1482-1437
`34 Byers, v.5. and Baldwin, R.W. (1988) Immunology 65,
`329—335
`35 Soler-Rodriguez, A.M., Ghetie, M.A., Oppenheimer-
`Marks, N., Uhr, ].W. and Vitetta, E.S. Exp. Cell Res.
`
`(in press)
`36 Jain, R.K. (1990) Cancer Metast. Rev. 9, 253—266
`37 Bacha, P. et al. (1990) Cancer Chemother. Pharmacol.
`26, 409—414
`38 LeMaistre, F. et al. (1990) Blood 76, 360A
`39 Vitetta, E.S. et al.

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