throbber
REVIEW
`Antibody-Enzyme Conjugates for
`Cancer Therapy
`
`Roger G. Melton, Roger F. Sherwood*
`
`Downloaded from
`
`http://jnci.oxfordjournals.org/
`
` by guest on November 24, 2014
`
`The use of antibody-enzyme conjugates directed at tumor-
`associated antigens to achieve site-specific activation of
`prodrugs to potent cytotoxic species, termed "antibody-
`directed enzyme prodrug therapy" (ADEPT), has attracted
`considerable interest since the concept was first described in
`1987. Prodrug forms of both clinically used anticancer
`agents and novel cytotoxic compounds have been developed
`to take advantage of potential prodrug-generating technol-
`ogy employing a variety of enzymes with widely differing
`substrate specificities. A particular advantage of the ADEPT
`approach is that it may allow the use of extremely potent
`agents such as nitrogen mustards and palytoxin, which are
`too toxic to be readily used in conventional chemotherapy.
`Preliminary studies using an antibody-enzyme conjugate
`constructed with a bacterial enzyme and a murine mono-
`clonal antibody not only have established the value of the
`ADEPT technique, but also have highlighted the potential
`problem of immunogenicity of proteins of nonhuman origin.
`This problem has been tackled in the first instance by the
`use of immunosuppressive agents, but long-term solutions
`are being investigated in the development of second-genera-
`tion ADEPT systems, including the development of human
`antibody-human enzyme fusion proteins and catalytic an-
`tibodies. Such improvements, coupled with further refine-
`ment of the prodrug-drug element of the system and the
`wide variety of antibody-enzyme-drug combinations avail-
`able, should mean that ADEPT-based approaches will form
`an important element of the search for the anticancer drugs
`of the future. [J Natl Cancer Inst 1996;88:153-65]
`
`The advent of monoclonal antibody technology in the 1970s
`was thought at the time to herald the fruition of the search for
`the "magic bullet" first proposed by Ehrlich in the early years of
`this century (/). In practice, however, this goal has remained
`elusive, inasmuch as many obstacles have presented themselves
`and have proved to be very difficult to overcome. To date, there
`have been numerous attempts to improve the cytotoxicity of the
`antibody "missiles" by attaching a variety of "warheads" to
`them—e.g., cytotoxic drugs such as doxorubicin or metho-
`trexate, toxins such as ricin A-chain or Pseudomonas exotoxin,
`and radioisotopes (2). Recently, enzymes capable of activating
`
`prodrugs to active drugs have been the focus of considerable in-
`terest and form the subject of this review.
`For any form of antibody-mediated targeting to be successful,
`it is axiomatic that there must be selective expression of the tar-
`get antigen by the tumor cells. This provides the first barrier to
`overcome, for the only well-characterized tumor-specific an-
`tigens described to date are the idiotypic determinants on the
`surface immunoglobulins of B-cell lymphomas (3). Tissue-
`specific antigens are also known, e.g., prostate-specific antigen
`(PSA), but have yet to be exploited to any great extent in tar-
`geted immunotherapy regimens. Many antigens, e.g., oncofetal
`antigens such as carcinoembryonic antigen (CEA) or placental
`alkaline phosphatase, and other potential targets such as growth
`factor receptors are present in elevated levels in tumor tissue,
`but most, if not all, appear to be found to a greater or lesser ex-
`tent in other tissues. These limitations have not prevented ex-
`ploitation of the better characterized tumor-associated antigens
`(e.g., CEA) as targets for therapy, even though it is also well
`documented that tumor cells exhibit considerable antigenic
`heterogeneity (4-6). Tumor cells may express varying levels of
`antigen, and some cells may not express the target antigen at all,
`such that a single monoclonal antibody conjugate targeted at a
`tumor mass will not bind to all the cells present, although other
`antibodies may be used to target alternative antigens on such
`cells. This is a disadvantage for immunoconjugates constructed
`using cytotoxic moieties that need to bind to the target cells and
`be internalized to exert their effect; however, the problem may
`be overcome to some extent by the use of antibody "cocktails"
`in which a mixture of antibodies aimed at two or more discrete
`antigens is used in noncompetitive combination (7).
`The ability of conjugates to gain access to the target cells is
`also essential, yet there is ample evidence that antibodies
`penetrate tumors poorly (8,9). Thus, the best access is at the
`tumor periphery, and cells located near the center of the tumor
`mass may be inaccessible to immunoconjugates, even though
`
`* Affiliations of authors: R. G. Melton, Centre for Applied Microbiology and
`Research, Porton Down, Salisbury, U.K.; R. F. Sherwood, Duramed Europe
`Ltd., Magdalen Centre, Oxford Science Park, Oxford, U.K.
`Correspondence to: Roger G. Melton, Ph.D., Centre for Applied Microbiol-
`ogy and Research, Porton Down, Salisbury SP4 OJG, U.K.
`See "Notes" section following "References."
`
`Journal of the National Cancer Institute, Vol. 88, No. 3/4, February 21, 1996
`
`REVIEW
`
`153
`
`IMMUNOGEN 2172, pg. 1
`Phigenic v. Immunogen
`IPR2014-00676
`
`

`

`less levels of the latter are particularly high (11). Another source of
`antigen may arise from the lysis of tumor cells following the active
`drug release from the prodrug by the action of the enzyme in the
`antibody-enzyme conjugate bound to cell surfaces. However, it is
`unlikely that there is any antibody-enzyme conjugate in the cir-
`culation at that time; thus, such antigen release may not be
`problematic except in situations where repeat cycle(s) of therapy
`are administered before this antigen is cleared from the body.
`A further advantage of ADEPT is that a single enzyme
`molecule has the potential to cleave many prodrug molecules—
`up to 800 mol/mol of enzyme per second in the case of the ben-
`zoic acid mustard substrates of carboxypeptidase G2 (13)—
`providing an amplification effect giving high levels of drug lo-
`calized at the tumor; this may be an important advantage in the
`clinic, in view of the typically low localization of immunocon-
`jugates in humans (14). There is also some evidence that high
`levels of drug generated at the surface of tumor cells are more
`effective than equivalent concentrations of free drug (75). The
`prodrug is an integral component of ADEPT systems and re-
`quires careful design in its own right. An ideal prodrug would
`be one with a large differential in cytotoxicity between drug and
`prodrug, which is a good substrate for the enzyme under
`physiologic conditions and for which there is no mammalian
`homologue capable of performing the same reaction. The use of
`human enzymes requires qualification of the latter statement, as
`discussed below. Equal cytotoxicity of the released active drug
`toward proliferating and quiescent cells is also desirable if
`residual deposits of viable but nonproliferating cells with the
`potential for outgrowth are to be eradicated. Once formed, it
`would be desirable for the drug to have a very short half-life,
`limiting the possibility of escape of active drug back into the cir-
`culation and access to healthy tissue. Development of drug
`resistance may limit the effects of conventional active drugs
`produced from prodrugs; thus, the nitrogen mustard group of
`compounds has tended to be the most commonly used group of
`active drugs for ADEPT because these compounds are not cell
`cycle specific, can be hydrolyzed to nontoxic forms, can kill
`both well-oxygenated and hypoxic cells, and cells develop only
`low levels of resistance to them (16). The potential release of
`the active drug into the circulation is conceivable after tumor
`cell lysis, but most drugs proposed for ADEPT systems are
`potent alkylating or intercalating agents and are likely to remain
`bound irreversibly to intracellular targets.
`In selecting an enzyme, one would look for activity under
`physiologic conditions, low immunogenicity, and little or no
`equivalent endogenous enzyme in humans. The enzyme needs
`to have a high specific activity, because the amounts that can be
`delivered by antibody vectors are limited; however, the defini-
`tion of optimal enzyme kinetics is clear cut and, as we shall see,
`under certain circumstances, a low kcat (i.e., the turnover rate of
`enzyme, which is expressed as number of moles of substrate
`utilized per mole of enzyme per second) may be preferable to a
`higher kcat (17). If a human enzyme is selected in an attempt to
`develop a nonimmunogenic conjugate, it will probably need to
`be an intracellular enzyme so that endogenous enzyme does not
`give rise to activation of the prodrug in the blood. When
`coupled to an antibody, the enzyme must localize efficiently at
`the tumor, and the development of immune responses to non-
`
`Downloaded from
`
`http://jnci.oxfordjournals.org/
`
` by guest on November 24, 2014
`
`they may express antigen. Cells in such inaccessible anoxic
`zones may be quiescent but possess the ability to regrow if ex-
`posed to a supply of oxygen and nutrients as a result of more
`vulnerable cells in oxic zones being killed. For this reason,
`radioimmunoconjugates have been extensively studied, because
`the emissions from the isotopes used clinically are capable of
`penetrating several cell layers, killing cells distal from where the
`antibody is localized. Although this approach is attractive, in
`practice the relatively low tumor uptake of the radioimmuno-
`conjugate may require large patient exposures, giving rise to
`dose-limiting myelosuppression and necessitating the use of
`autologous bone marrow grafts {10).
`
`Antibody-Directed Enzyme Prodrug Therapy as
`an Alternative Strategy
`
`An alternative strategy that has recently been the subject of
`considerable interest is the use of antibodies as vectors for en-
`zymes capable of activating a nontoxic drug precursor, termed a
`"prodrug," to a potently cytotoxic moiety. This approach, which
`has been termed "antibody-directed enzyme prodrug therapy"
`(ADEPT) (11), or "antibody-directed catalysis" (72), offers the
`potential to overcome most of the problems associated with the
`use of drug or toxin immunoconjugates described above. The
`system is shown by the diagram in Fig. 1. An antibody-enzyme
`conjugate is injected and allowed to localize at the tumor while
`clearing from the rest of the tissues. After a suitable interval, the
`prodrug is administered; while remaining innocuous to the nor-
`mal tissues, it is converted to a cytotoxic form by enzyme local-
`ized within the tumor tissue. Being of low molecular weight, the
`active drug can diffuse to adjacent tissues, providing a bystander
`effect, although this may be of questionable benefit if the
`released drug is relatively stable and can diffuse back into the
`circulation. It is implicit in the ADEPT approach that the an-
`tigen to which the antibody-enzyme conjugate is targeted must
`remain extracellular; thus, secreted antigen that accumulates in
`the tumor interstitial spaces may assist in achieving high levels
`of conjugate at the tumor site. Although shed antigen present in
`the circulation will act as a competitor with the tumor site for
`binding of target antigen, there is evidence that tumor localiza-
`tion of conjugate can occur in the face of circulating antigen un-
`
`Anti body-enzyme
`conjugate
`
`Fig. 1. Schematic representation of antibody-directed enzyme prodrug therapy.
`
`154 REVTEW
`
`Journal of the National Cancer Institute, Vol. 88, No. 3/4, February 21. 1996
`
`IMMUNOGEN 2172, pg. 2
`Phigenic v. Immunogen
`IPR2014-00676
`
`

`

`human enzymes is undesirable, since localization of the con-
`jugate is likely to be drastically reduced upon readministration
`after the onset of an immune response (18) and there will be a
`risk of hypersensitive reactions. However, immunosuppressive
`drug regimens designed to extend the time window before the
`onset of the human immune response observed in patients (19)
`have been developed to counteract this problem (20). The gains
`obtained using such approaches in animal models have been
`modest, but they may be sufficient to allow two or three repeat
`courses of an ADEPT cycle.
`An important factor in early stages of the development of an
`ADEPT system is the production of an antibody-enzyme con-
`jugate without loss of antigen-binding and enzymatic activities.
`The techniques and heterobifunctional agents typically used are
`analogous to those developed for use in the construction of im-
`munotoxins. An important requirement for conjugates prepared
`for use in ADEPT is that the linkage should be stable, since a
`period of up to or greater than 72 hours may elapse between the
`administration of conjugate and the administration of prodrug,
`in order to allow unbound conjugate to clear from the circula-
`tion. For this reason, conjugation chemistry based on thioether
`linkages has been virtually universally adopted.
`A number of experimental ADEPT systems have been de-
`veloped, with a range of different enzymes, and it is thus con-
`venient to categorize them by class of enzyme (Table 1).
`
`Enzymes used for ADEPT systems to date include carboxypep-
`tidases G2 and A, alkaline phosphatase, glycosidases, penicillin
`amidase, P-lactamase, and cytosine deaminase. All are at a very
`early stage of development, with only the carboxypeptidase G2
`system having been tested clinically to date. Typically, these
`systems have been tested in nude mouse models with human
`xenograft targets. These represent probably the best models cur-
`rently available, but they have clear limitations. For example,
`the subcutaneous tumor implant is nonmetastatic and, therefore,
`does not represent the typical clinical situation, where the treat-
`ment of advanced metastatic disease is likely to be the early tar-
`get, with the eventual prospect of moving to minimal residual
`disease following surgery to debulk the tumor. Moreover, the
`levels of tumor localization achievable in mice far exceed those
`that have been found clinically with radiolabeled antibodies. The
`fact that .nude mice are immunodeficient also prevents considera-
`tion of the problem of immunogenicity, an issue of considerable
`importance, as has been revealed by early clinical studies (21,22).
`Once a workable system has been established,.there is likely to be a
`lengthy development stage while all aspects of the system are re-
`evaluated and optimized. Typically, the necessary development
`work will include the synthesis of improved prodrugs, substitution
`of human enzymes for bacterial enzymes where possible, the adop-
`tion of chimeric or human antibodies and fragments thereof, the
`generation of nonimmunogenic clearing systems, and the develop-
`
`Table 1. Some potential antibody-directed enzyme prodrug therapy (ADEPT) systems described in the literature
`
`Downloaded from
`
`http://jnci.oxfordjournals.org/
`
` by guest on November 24, 2014
`
`Reference No.
`
`(13,27)
`
`(13.19,26.27)
`
`(I9.21.28J0)
`
`(153437)
`
`(42,44)
`
`(42)
`
`(45)
`(48)
`
`(46)
`
`(47)
`
`(5839)
`
`(61)
`(4930)
`
`(54)
`
`(79)
`
`(12.67.68)
`
`(65.66)
`
`(69-72)
`
`Enzyme
`
`Antibody
`
`Antigen
`
`Prodrug
`
`Active drug
`
`Carboxypeptidase G:
`
`WI4
`
`Human chorionic
`gonadotropin
`
`p-N-bis (2-Chloroethyl)benzoyl
`glutamic acid
`4-[(2-Mesyloxyethyl)(2-chloroethyl)
`amino] benzoyl glutamic acid
`
`Carboxypeptidase G2
`
`A5B7
`
`Carcinoembryonic
`antigen
`
`4-[(2-Mesyloxyethyl)(2-chloroethyl)
`amino] benzoyl glutamic acid
`
`p-N-bis (2-Chloroethyl)
`amino baaenzoic acid
`4-[(2-Mesyloxyethyl)-
`(2-chloroethyl)
`amino| benzoic acid
`4-[(2-Mesyloxyethyl)-
`(2-chloroethyl)
`amino benzoic acid
`Methotrexate
`
`Etoposide
`
`Mitomycin C
`
`Doxorubicin
`Mitomycin C
`
`In vitro cell line
`(in vivo tumor model)*
`
`CC3 choriocarcinoma
`(CC3)
`
`LS174T colon carcinoma
`(LSI74T)
`
`UCLA-P3 lung
`adenocarcinoma
`H3347 colon carcinoma
`(H3347)
`H298I lung adenocarcinoma
`(H2981)
`H298I lung adenocarcinoma
`L540 Hodgkin's lymphoma
`
`Methotrexate-a-alanine
`
`Etoposide phosphate
`
`Mitomycin C phosphate
`
`Doxorubicin phosphate
`Mitomycin C phosphate
`
`Carboxypeptidase A
`
`KS1/4
`
`Alkaline phosphatase
`
`L6
`
`L6
`
`L6
`HRS-3/API
`
`L6
`
`BW431
`
`Penicillin amidase
`
`L6
`
`p-Glucuronidase
`
`RHI
`
`323/A3
`
`BW431
`
`P-Lactamase
`
`CEM2314
`
`Cytosine deaminase
`
`L6
`
`L6
`
`Lung adenocarcinoma-
`associated antigen
`Tumor-associated
`carbohydrate
`
`CD30 HodgkirTs
`lymphoma antigen
`
`Carcinoembryonic
`antigen
`Lung adenocarcinoma-
`associated antigen
`
`Uncharacterized
`hepatocarcinoma
`antigen
`Tumor-associated
`glycoprotein
`Carcinoembryonic
`antigen
`Carcinoembryonic
`antigen
`
`*A11 cell lines tested are of human origin.
`
`/VJV-bis(2-Chloroethyl)aminophenyl
`phosphate
`Etoposide phosphate
`
`WJV-bis-(2-Chloroethyl)-
`4-hydroxyaniline
`Etoposide
`
`H2981 lung adenocarcinoma
`(H298I)
`S W1398 colon carcinoma
`
`/V-Phenylaceiamido doxorubicin
`/V-Phenylacetamido melphalan
`A'-(4'-Hydroxyphenylacetyl)palytoxin
`p-bis-2-Chloroelhylaminophenyl
`p-D-Glucopyranoside uronic acid
`(K>«-bulyl salt)
`Epimbicin-glucuronide
`
`Doxorubicin-glucuronide
`
`Cephalosporin-vinca alkaloid
`
`Cephalosporin mustard
`
`5-Fluorocytosine
`
`Doxorubicin
`Melphalan
`Palytoxin
`WJV-bis-(2-Chloroethyl)-
`4-hydroxyaniline
`
`H2981 lung adenocarcinoma
`
`H2981 lung adenocarcinoma
`HepG2 hepatoma
`
`Epirubicin
`
`Doxorubicin
`
`Desacetylvinblastine
`hydrazide
`Phenylene diamine
`mustard
`Fluorouracil
`
`MCF-7 breast cancer
`OVCAR-3 ovarian cancer
`(L0V0, Mz-Sto-l)
`colon carcinoma
`LS 174 colon carcinoma
`(LS174T)
`H298I lung adenocarcinoma
`
`H2981 lung adenocarcinoma
`(H2981.HT29 colon
`carcinoma)
`
`Journal of the National Cancer Institute, Vol. 88, No. 3/4, February 21, 1996
`
`REVIEW 155
`
`IMMUNOGEN 2172, pg. 3
`Phigenic v. Immunogen
`IPR2014-00676
`
`

`

`Downloaded from
`
`http://jnci.oxfordjournals.org/
`
` by guest on November 24, 2014
`
`ment of fusion proteins or bifunctional catalytic antibodies with
`suitable binding specificity for tumor-associated antigens. With
`such a range of parameters to be optimized, the development of
`ADEPT systems is clearly complex and likely to be time-con-
`suming and expensive.
`
`Enzymes and Prodrugs Used in Currently
`Available ADEPT Systems
`
`Carboxypeptidase G2
`
`The first ADEPT system was proposed by Bagshawe (25) in
`1987; it used carboxypeptidase G2 (CPG2), which was originally
`isolated as a methotrexate-degrading enzyme from Pseudo-
`monas species and subsequently cloned in Escherichia coli (24),
`to cleave a deactivating glutamate moiety from a benzoic acid
`mustard (25). This remains probably the best characterized sys-
`tem and is the only system to date for which pilot clinical
`studies have commenced (19,21,22). The initial prodrug to be
`synthesized was 4-[A'^V-bis(2-chloroethyl)amino]benzoyl-L-glu-
`tamic acid from which CPG2 cleaves the glutamic acid to yield
`4-[/vVV-bis(2-chloroethyl)amino]benzoic acid (Fig. 2, A). Sub-
`sequently, modified versions were developed with one or both
`of the 2-chloroethyl arms replaced by more reactive 2-mesyl-
`oxyethyl moieties (Fig. 2, B) (25). The mono-(2-mesyloxyethyl)
`(MMC1) compound was more cytotoxic and a slightly better
`substrate for the enzyme than the bis(2-chloroethyl) compound,
`but the bis(2-mesyloxyethyl) form was too reactive and rapidly
`hydrolyzed in the absence of enzyme (25,26). The enzyme con-
`jugate-prodrug combination was initially tested in vitro using
`the human chorionic gonadotropin
`(hCG)-expressing JAR
`choriocarcinoma cell line; it was found to give greater than 100-
`fold differential toxicity between drug or prodrug plus enzyme
`(IC50 [i.e., the concentration required to kill 50% of treated cells
`
`compared with untreated controls] = 20 UJW) and prodrug (IC50
`= >800 \iM) (27). In antitumor studies using the hCG-expressing
`human CC3 choriocarcinoma xenograft model in nude mice, the
`combination of W14 (anti-hCG) F(ab')2-CPG2 conjugate followed
`72 hours later by the MMC1 prodrug resulted in complete eradica-
`tion of established CC3 xenografts when prodrug was administered
`as a three-times-divided dose of 10 mg/kg (total dose, 30 mg/kg) at
`16-hour intervals from 72 hours (1327). In contrast, weekly in-
`travenous injections of daily divided doses of methotrexate (5
`mg/kg), hydroxyurea (50 mg/kg), dactinomycin (7.5 |ag/kg), or
`cytarabine (20 mg/kg) failed to retard tumor growth, whereas the
`MMC1 prodrug and the W14 F(ab')2-CPG2 conjugate produced a
`pronounced growth delay of more than 50 days (27).
`When the same two-phase system was applied to the CEA-
`expressing human LS174T colon carcinoma model, using a con-
`jugate constructed with the F(ab')2 fragment of A5B7 anti-CEA
`antibody, it proved to be less effective in that minimal growth
`delays were seen (28). This was probably because of the much
`slower clearance of conjugate from the blood in this model com-
`pared with that in the choriocarcinoma. In the latter case, high
`levels of circulating antigen probably form immune complexes,
`leading to accelerated clearance; however, much lower levels of
`antigen are present in the circulation of CEA-expressing tumors
`(29), and the clearing effect of free antigen is thus not apparent.
`As a result, a second galactosylated clearing antibody, SB43,
`which enzymatically inactivated CPG2, was used to remove un-
`bound conjugate from the circulation, making the system a
`three-phase approach (30). The galactosylated antibody has a
`relatively short half-life in the circulation because it is rapidly
`cleared by hepatic galactose receptors; consequently, it can react
`with the CPG2 conjugate in the circulation but is not ex-
`travasated sufficiently to reach the conjugate in the extravas-
`cular compartment. This situation should avoid the problem of
`
`COOH
`|
`O-NH-CH
`
`Carboxypeptidase G2
`»•
`
`OOH
`
`CICH2CH/
`
`\=
`
`4-[bis(2-chloroethyl)amino]benzoyl-
`L-gljtamic add
`
`CH2
`I
`COOH
`
`4-[bis(2-chloroethyl)amino]benzoicacid
`
`B CH3SO2O-CH2CH2
`
`CICH2CH/
`
`\=
`
`4-[(2-chloroethyl)(2-mesyloxyethyl)amino]benzoyl-L-glutamic acid (MMCI)
`
`4-[bis(2-mesyIoxyethyt)aminolbenzoyl-L-glutamicacid
`
`COOH
`O-NH-CH
`H2
`CH2
`COOH
`
`IC
`
`CH3SO2O-CH2CH2
`
`CH3SO2O-CH2CH2'
`
`Fig. 2. Benzoic acid mustard prodrugs that are substrates for carboxypeptidase G, (CPG2). A) Mechanism of cleavage of benzoic acid mustards by CPG,. B) Alterna-
`tive mustard functional groups.
`
`156 REVIEW
`
`Journal of the National Cancer Institute, Vol. 88, No. 3/4, February 21, 1996
`
`IMMUNOGEN 2172, pg. 4
`Phigenic v. Immunogen
`IPR2014-00676
`
`

`

`Downloaded from
`
`http://jnci.oxfordjournals.org/
`
` by guest on November 24, 2014
`
`clearing antibody reaching and inactivating the tumor-bound an-
`tibody-enzyme conjugate (30). When the three-phase system
`was tested against the same model, prolonged growth delays of
`up to 10-15 days, compared with untreated controls, were seen
`with the MMC1 compound and, to a lesser extent, with the
`bischloro compound (28). Using a superficially identical model,
`Blakey et al. (31) were subsequently able to achieve growth
`delay of 10-15 days without recourse to clearing antibody. The
`principal difference between the experiments lay in the method
`of tumor implantation: Solid tumor fragments were used in the
`earlier work, whereas injection of a cell suspension was done in
`the later experiments.
`In an alternative approach to the use of clearing antibody,
`Melton et al. (32) demonstrated enhanced tumor localization of
`antibody-enzyme conjugate when administered with tumor
`necrosis factor-a (TNF-a) in the murine E3 thymoma model.
`Coadministration of 1.5 (ig TNF-a with conjugate led to a
`twofold increase in tumor localization and only transient in-
`creases in normal tissue localization. It is possible that this sys-
`tem could be used in combination with the galactosylated
`clearing antibody because the two approaches are complemen-
`tary: TNF-a increases tumor localization of conjugate without
`significantly affecting blood levels, and galactosylated antibody
`markedly decreases blood
`levels without affecting
`tumor
`localization. Galactosylated antibody increases the ratios of
`tumor to normal tissue without affecting overall tumor uptake,
`whereas TNF-a provides a mechanism for achieving higher ini-
`tial tumor uptake. Similar effects can be induced by the ad-
`ministration of drugs that modify blood flow, which essentially
`appear to act by inducing localized production of TNF-a (33).
`The preclinical studies using the colon carcinoma xenograft
`model and the MMC1 compound were deemed sufficiently en-
`couraging to justify a limited clinical trial in patients with advanced
`metastatic, poorly differentiated colon or rectal cancer (1921,22).
`Initial dose escalation studies using the monomesyl benzoic acid
`mustard prodrug showed that doses from 200 up to 2500 mg/m2,
`given as six or 12 divided doses over a 3-day period, were well
`tolerated. A total of four male patients aged 55-70 years, all of
`whom had undergone extensive conventional chemotherapy (prin-
`cipally with fluorouracil), subsequently went on to receive 20 000
`U/m2 A5B7 (anti-CEA) F(ab')2-CPG2 conjugate, a small propor-
`tion of which was I25I labeled to facilitate confirmation of tumor
`localization by gamma camera imaging. SB43 clearing antibody
`was administered 36-48 hours later as a 3- to 5-hour infusion, typi-
`cally followed by MMC1 (1200-3000 mg/m2) once measured
`serum enzyme levels were less than 0.02 U/mL. The serum levels
`of CEA and a second tumor antigen, 19-9, fell in all patients by 10-
`15 days after therapy; in two cases, there were objective responses
`in the reduction in size of liver metastases. A patient who had gross
`hepatomegaly and ascites became free of jaundice and ascites, al-
`though liver size was not reduced. Other subjective responses in-
`cluded relief of pain, and three patients gained weight and reported
`improved health. Adverse effects included the development of an-
`tibodies to both components of the conjugate, and the two patients
`receiving the highest doses of prodrug developed severe myelo-
`suppression, although this was reversible once prodrug administra-
`tion was ceased. The antibody responses developed within 10-12
`days, limiting the treatment effectively to a single cycle; however,
`
`this time window may be extended by the use of the im-
`munosuppressive drug cyclosporine (20). The development of
`myelosuppression appears to be due to the relatively long half-life
`of the active drug, which can diffuse out of the tumor and back into
`the circulation, indicating that more reactive drugs with shorter
`physiologic half-lives are desirable. The results of the pilot clinical
`study are encouraging, if one bears in mind the advanced state of
`the disease of the patients in the study, and provide ample justifica-
`tion for further development of the system.
`
`Carboxypeptidase A
`
`Huennekens and co-workers (15,34-37) described a system in
`which carboxypeptidase A (CPA) is used to cleave the alpha-
`carboxyl alanine residue from methotrexate-a-alanine. When
`human lung adenocarcinoma cells were exposed to conjugate
`consisting of CPA coupled to KS1/4 antibody, followed by
`washing and addition of prodrug, the IC50 of methotrexate-a-
`alanine decreased approximately sixfold from 8.9 x 10"6 M to
`1.5 x 10"6 M after 48 hours of incubation. With prolonged in-
`cubation, the toxicity of methotrexate-a-alanine approached
`that of methotrexate (i.e., about 2 x 10"9 M) (15). It is thus clear
`that the enzyme turns over the substrate rather slowly, although
`the authors suggested that methotrexate generated in intimate
`contact with the cells in this way seems to be more cytotoxic
`than the equivalent concentration of free methotrexate. A study
`of the theoretical aspects of ADEPT by Yuan et al. (17) sug-
`gested that a low turnover rate may not be disadvantageous be-
`cause the rate-limiting step is diffusion of drug from the
`capillaries to the extravascular tumor cells; however, this obser-
`vation must be set against the need to generate sufficient drug to
`exert the desired cytotoxic effect. The most important factor in
`attaining tumor-specific activation may be the achievement of
`high tumor-to-normal tissue localization ratios and rapid reac-
`tion of the active drug to prevent its loss back into the circula-
`tion. If these issues can be resolved, then the use of an enzyme
`with a high turnover and affinity for substrate would seem to be
`ideal because the level of prodrug reaching the tumor is unlikely
`to match that present in blood. It seems likely, however, that the
`relatively poor kinetics of this enzyme with the first-generation
`prodrug are unlikely to lead to a workable system in vivo, and a
`number of groups (38J9) recently described the synthesis of
`potentially better substrates based on methotrexate. No animal
`studies have been reported for the CPA/methotrexate-a-alanine
`system, and the likelihood of activation of prodrug by endoge-
`nous CPA remains as a possible disadvantage of this approach.
`In view of the development of resistance to methotrexate and
`the high levels of drug needed to achieve maximal effect (40), it
`is questionable whether this is a suitable drug for ADEPT ap-
`plications, particularly when the kinetics of the enzyme are rela-
`tively poor.
`
`Aminopeptidase
`
`Smal et al. (41) recently described the synthesis of a series of
`2-aminoacyl methotrexate prodrugs designed to be substrates for
`pyroglutamate aminopeptidase or D-aminopeptidase. The cyto-
`toxicity of the prodrug plus enzyme in vitro did not approach
`that of methotrexate alone and required high levels of enzyme to
`achieve any selective cell kill, suggesting that this is not a prac-
`
`Journal of the National Cancer Institute, Vol. 88, No. 3/4, February 21, 1996
`
`REVIEW 157
`
`IMMUNOGEN 2172, pg. 5
`Phigenic v. Immunogen
`IPR2014-00676
`
`

`

`Downloaded from
`
`http://jnci.oxfordjournals.org/
`
` by guest on November 24, 2014
`
`tical working system in its present form. To date, the production
`and testing of antibody-enzyme conjugates have not been
`described, and it is difficult to envisage the delivery of amounts
`of enzyme activity equivalent to those used in the experiments
`described, using an antibody vector.
`
`Alkaline Phosphatase
`
`A number of groups have presented evidence that alkaline
`phosphatase can be used to activate a range of prodrugs (Fig. 3),
`taking advantage of alkaline phosphatase's relative lack of sub-
`strate specificity. Human alkaline phosphatase, in conjunction
`with a humanized monoclonal antibody, may result in a con-
`jugate with reduced immunogenicity by comparison with bac-
`terial enzyme conjugates. However, endogenous alkaline
`phosphatase is widely distributed in the body and is likely to ac-
`tivate prodrug in normal tissues, suggesting that this system is
`primarily of academic interest. To test the concept of using
`alkaline phosphatase, Senter and co-workers used calf intestinal
`alkaline phosphatase to cleave phosphate groups from etoposide
`phosphate (42-44), mitomycin C phosphate (42), doxorubicin
`phosphate (45), and phenol mustard phosphate (46) and have
`reported antigen-specific cytotoxicity in vitro and growth delays in
`vivo with human colon carcinoma xenograft models; conjugates
`constructed with an irrelevant antibody showed greatly reduced
`growth delays, indicating that nonspecific uptake of conjugate is
`relatively low compared with the uptake of conjugate with
`specificity for the appropriate antigen. The use of this enzyme does
`not address the question of possible immunogenicity, but it would
`be expected to have kinetic properties similar to those of the human
`homologue. Other researchers (47,48) described similar systems
`
`using other antibodies directed toward human colon cancer an-
`tigens. Perhaps the most interesting of these describes the use of
`a bifunctional antibody capable of binding both alkaline phos-
`phatase and the CD30 antigen; the purpose was to enable the
`production of conjugates by immunologic interaction rather than
`by chemical coupling (48). Such an approach requires an enzyme
`that is not catalytically inactivated by the antibody binding to it; in
`this instance, the authors were fortunate that the binding of alkaline
`phosphatase by bispecific intact antibody or F(ab')2 fragment did
`not adversely affect the enzyme's ability to activate the phosphory-
`lated mitomycin C prodrug.
`
`Glycosidases
`
`A number of groups have described ADEPT systems based
`on glycosidases (Fig. 4). Roffler and co-workers (49£0) used (3-
`glucuronidase from Escherichia coli to remove a glucuronide
`from hydroxyaniline mustard. This system takes advantage of
`the fact that hydroxyaniline mustard is rapidly detoxified by
`hepatic enzymes by conversion to the glucuronide form (51,52).
`In certain animal tumors, hydroxyaniline mustard glucuronide
`was activated back to hydroxyaniline mustard by elevated levels
`of endogenous (3-glucuronidase to give site-specific cytotoxicity
`(57), although attempts to use this system in humans were not
`successful (53).
`Escherichia coli (3-glucuronidase, coupled to monoclonal an-
`tibody 12.8 that bind

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