+ All Categories
Home > Documents > Delivery of Saporin to Human B-Cell Lymphoma …...field (5), recent trials in Hodgkin's (6) and...

Delivery of Saporin to Human B-Cell Lymphoma …...field (5), recent trials in Hodgkin's (6) and...

Date post: 24-Feb-2020
Category:
Upload: others
View: 4 times
Download: 0 times
Share this document with a friend
8
[CANCER RESEARCH 53. 3015-3021. July I. 1993) Delivery of Saporin to Human B-Cell Lymphoma Using Bispecific Antibody: Targeting via CD22 but not CD19, CD37, or Immunoglobulin Results in Efficient Killing1 M. Antonietta Bonardi, R. R. French, P. Amlot, G. Gromo, D. Modena, and M. J. Glennie2 Lvmphoma Research Unit. Tenovus Laboratory. General Hospital. Southampton SO9 4XY, United Kingdom [M. A. B.. R. R. F.. M. J. G.I; Italfarmaco Centro Ricerche. 20092 Cinisello Balsama. Ml. Italy ¡M.A. B., G. G., D. M.¡;and Department of Immunology. Royal Free Hospital. London NW3 2OG. United Kingdom ¡P. A.I ABSTRACT A panel of bispecific F(ab')2 antibodies (BsAb) have been constructed for delivering the ribosome-inactivating protein saporin to human B cell Ivmphoma. Each derivative was prepared with specificity for saporin and CD19, CD22, CD37, or immunoglobulin. In vitro studies measuring inhi bition of [3H]leucine uptake by cultured Daudi and Raji cells demon strated that, despite all BsAb capturing saporin on the cell surface, BsAb targeting through CD22 were far more cytotoxic than those functioning via CD19, CD37, or surface immunoglobulin. This exceptional activity of the CD22-specific BsAb appears to derive from its ability to deliver and accumulate saporin inside the target cells. Further studies showed that four CD22-specific BsAb all performed with equal potency and were able to increase saporin toxicity (50% inhibitory concentration) up to 1000- fold, from 2x IO-7 M to 2 x IO"10 M. Pairs of anti-CD22 BsAb which recognized different nonblocking epitopes on the saporin molecule were able to bind saporin more avidly to the target cell and, as a consequence, increased cytotoxicity by at least an additional 10-fold, resulting in 50% inhibitory concentration for protein synthesis of 2 x 1(1 " M.These results suggest that selected combinations of BsAb which bind cooperatively to a toxin and the cell surface may provide an efficient way of delivering toxins to unwanted cells in patients. INTRODUCTION Immunotoxins in which a toxin, such as a RIP,3 is chemically or genetically linked to a mAb are being investigated as potential anti- cancer agents by a number of groups (1^). Despite generally disap pointing clinical responses and some troublesome side effects in this field (5), recent trials in Hodgkin's (6) and non-Hodgkin's lymphoma (7, 8) have been encouraging and suggest that immunotoxin may yet find an application in cancer treatment. As an alternative approach for delivering toxins, we have been investigating the use of BsAb, as suggested by Raso and Griffin in the early 1980s (9). Here an antibody with dual specificity for a toxin, such as a RIP, and a suitable cell marker delivers the toxin to the unwanted cells in the form of immune complexes. The potential advantages of this approach over conventional, disulfide-bonded, im- munotoxins include a lack of chemical modification during conjuga tion that could damage the toxin or the binding site of the antibody, the ability to release the toxin moiety from the antibody without reducing a disulfide bond (the drop in pH experienced by the BsAb-toxin complexes when they enter the cell may aid their dissociation), and, finally, the use of BsAb not only as a carrier of the toxin but also to reduce the nonspecific toxicity of the toxin by blocking any natural ligand-binding activity that it may have for normal cells, e.g., the galactose-binding sites on ricin. Received 8/17/92; accepted 4/23/93. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. 1This work has been supported by Tenovus (Cardiff, UK) and Italfarmaco (Milano. Italy). 2 To whom requests for reprints should be addressed. 1The abbreviations used are: RIP, ribosome-inactivating protein; BsAb, bispecific antibody (-ies); ICW, concentration giving 50% inhibition of |'H]leucine uptake; IT, immunotoxin(s); Ig, immunoglobulin; mAb, monoclonal antibody (-ies). The major disadvantage of this approach is that, because the toxin is carried through relatively weak, reversible, noncovalent interac tions, it may be released before entering the target cells. Two maneu vers have helped us safeguard against this shortcoming; first, before administration the BsAb-toxin complexes are formed with a signifi cant molar excess of BsAb, which greatly reduces the opportunity for free toxin to exist (10), and, second, the avidity with which the toxin binds to the target cells is increased by using pairs of BsAb, selected to bind simultaneously to the toxin and the cell surface (11). To date, results have shown that in leukemic guinea pigs (10, 11) a cooperative mixture of two bispecific F(ab')2 antibodies with specificity for the RIP, saporin, and the appropriate tumor marker can deliver the toxin and achieve a significant therapeutic effect. Furthermore, preliminary data in patients with lymphoma indicate that combinations of BsAb can deliver saporin and achieve impressive clinical responses without toxic side effects (12).4 Before extending the use of BsAb in human B-cell lymphoma, we need to determine which surface antigens on the malignant cells provide suitable targets for delivering toxin. Here we have assessed the performance of bispecific F(ab')2 antibodies designed to deliver saporin to Burkitt's lymphoma cell lines via CD 19, CD22, CD37, or surface Ig. The widespread expression of these particular differentia tion antigens on B-cell lymphomas and their absence from hemopoi- etic stem cells is likely to make them especially suitable targets for therapy (13). We show that within this group of molecules only CD22 provides a suitable vehicle for delivering saporin with BsAb. Further more, the level of toxicity achieved with the appropriate pair of CD22-specific BsAb was equivalent to that reported for conventional IT directed against this surface molecule. MATERIALS AND METHODS Saporin. The RIP saporin (SO-6), purified from the seeds of Saponaria offÃ-cinalis, was kindly provided by Prof. F. Stirpe (University of Bologna, Italy) (14). Cell Lines. BsAb were tested on the Burkitt's lymphoma cell lines Daudi and Raji. Cells were maintained in supplemented RPMI [RPMI 1640 medium containing 2 m.Mglutamine, 1 ITIMpyruvate, 100 ID/ml penicillin. 100 lU/ml streptomycin. 2 fig/ml fungizone, 10 /j.g/ml ciprofloxacin, and 10% fetal calf serum (Myoclone) (GIBCO Ltd. Paisley, Scotland)]. Antibodies. The antibodies used in this study included four mAb directed against CD22. i.e.. HD6 and HD39, both gifts from Dr. B. Dorken (University of Heidelberg, Germany) (15, 16), 4KB 128, provided by Dr. D. Y. Mason, John Radcliffe Hospital (Oxford, UK) (17), and RFB-4 (18). The epitope designa tions of these are as follows: 4KB 128, CD22D; HD6. CD22B; RFB-4. CD22B; and HD39. CD22A (19, 20). Other antibodies included the ami-CD19 mAb HD37, kindly provided by Dr. B. Dorken (21). the anti-CD37 mAb WR17. provided by Dr. J. Smith, Regional Immunology (Southampton, UK) (22). the anti-CD37 mAb MB-1, provided by Dr. Richard Miller and Prof. Ron Levy, Stanford University Medical Center (Stanford, CA) (23). and the anti-K-chain antibody HB6-I, supplied by the American Type Culture Collection (Rock- ville, MD). The anti-CD 19 mAb RFB-9 was produced in the Department of 4 A. Bell, R. R. French. T. Hamblin, and M. J. Glennie, unpublished observations. 3015 on June 9, 2017. © 1993 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from
Transcript
Page 1: Delivery of Saporin to Human B-Cell Lymphoma …...field (5), recent trials in Hodgkin's (6) and non-Hodgkin's lymphoma (7, 8) have been encouraging and suggest that immunotoxin may

[CANCER RESEARCH 53. 3015-3021. July I. 1993)

Delivery of Saporin to Human B-Cell Lymphoma Using Bispecific Antibody:

Targeting via CD22 but not CD19, CD37, or Immunoglobulin Resultsin Efficient Killing1

M. Antonietta Bonardi, R. R. French, P. Amlot, G. Gromo, D. Modena, and M. J. Glennie2

Lvmphoma Research Unit. Tenovus Laboratory. General Hospital. Southampton SO9 4XY, United Kingdom [M. A. B.. R. R. F.. M. J. G.I; Italfarmaco Centro Ricerche. 20092Cinisello Balsama. Ml. Italy ¡M.A. B., G. G., D. M.¡;and Department of Immunology. Royal Free Hospital. London NW3 2OG. United Kingdom ¡P.A.I

ABSTRACT

A panel of bispecific F(ab')2 antibodies (BsAb) have been constructed

for delivering the ribosome-inactivating protein saporin to human B cell

Ivmphoma. Each derivative was prepared with specificity for saporin andCD19, CD22, CD37, or immunoglobulin. In vitro studies measuring inhibition of [3H]leucine uptake by cultured Daudi and Raji cells demon

strated that, despite all BsAb capturing saporin on the cell surface, BsAbtargeting through CD22 were far more cytotoxic than those functioningvia CD19, CD37, or surface immunoglobulin. This exceptional activity ofthe CD22-specific BsAb appears to derive from its ability to deliver and

accumulate saporin inside the target cells. Further studies showed thatfour CD22-specific BsAb all performed with equal potency and were ableto increase saporin toxicity (50% inhibitory concentration) up to 1000-fold, from 2 x IO-7 M to 2 x IO"10 M. Pairs of anti-CD22 BsAb which

recognized different nonblocking epitopes on the saporin molecule wereable to bind saporin more avidly to the target cell and, as a consequence,increased cytotoxicity by at least an additional 10-fold, resulting in 50%inhibitory concentration for protein synthesis of 2 x 1(1 " M.These results

suggest that selected combinations of BsAb which bind cooperatively to atoxin and the cell surface may provide an efficient way of delivering toxinsto unwanted cells in patients.

INTRODUCTION

Immunotoxins in which a toxin, such as a RIP,3 is chemically or

genetically linked to a mAb are being investigated as potential anti-

cancer agents by a number of groups (1^). Despite generally disappointing clinical responses and some troublesome side effects in thisfield (5), recent trials in Hodgkin's (6) and non-Hodgkin's lymphoma

(7, 8) have been encouraging and suggest that immunotoxin may yetfind an application in cancer treatment.

As an alternative approach for delivering toxins, we have beeninvestigating the use of BsAb, as suggested by Raso and Griffin in theearly 1980s (9). Here an antibody with dual specificity for a toxin,such as a RIP, and a suitable cell marker delivers the toxin to theunwanted cells in the form of immune complexes. The potentialadvantages of this approach over conventional, disulfide-bonded, im-

munotoxins include a lack of chemical modification during conjugation that could damage the toxin or the binding site of the antibody, theability to release the toxin moiety from the antibody without reducinga disulfide bond (the drop in pH experienced by the BsAb-toxin

complexes when they enter the cell may aid their dissociation), and,finally, the use of BsAb not only as a carrier of the toxin but also toreduce the nonspecific toxicity of the toxin by blocking any naturalligand-binding activity that it may have for normal cells, e.g., thegalactose-binding sites on ricin.

Received 8/17/92; accepted 4/23/93.The costs of publication of this article were defrayed in part by the payment of page

charges. This article must therefore be hereby marked advertisement in accordance with18 U.S.C. Section 1734 solely to indicate this fact.

1This work has been supported by Tenovus (Cardiff, UK) and Italfarmaco (Milano.

Italy).2 To whom requests for reprints should be addressed.1The abbreviations used are: RIP, ribosome-inactivating protein; BsAb, bispecific

antibody (-ies); ICW, concentration giving 50% inhibition of |'H]leucine uptake; IT,

immunotoxin(s); Ig, immunoglobulin; mAb, monoclonal antibody (-ies).

The major disadvantage of this approach is that, because the toxinis carried through relatively weak, reversible, noncovalent interactions, it may be released before entering the target cells. Two maneuvers have helped us safeguard against this shortcoming; first, beforeadministration the BsAb-toxin complexes are formed with a signifi

cant molar excess of BsAb, which greatly reduces the opportunity forfree toxin to exist (10), and, second, the avidity with which the toxinbinds to the target cells is increased by using pairs of BsAb, selectedto bind simultaneously to the toxin and the cell surface (11). To date,results have shown that in leukemic guinea pigs (10, 11) a cooperativemixture of two bispecific F(ab')2 antibodies with specificity for the

RIP, saporin, and the appropriate tumor marker can deliver the toxinand achieve a significant therapeutic effect. Furthermore, preliminarydata in patients with lymphoma indicate that combinations of BsAbcan deliver saporin and achieve impressive clinical responses withouttoxic side effects (12).4

Before extending the use of BsAb in human B-cell lymphoma, we

need to determine which surface antigens on the malignant cellsprovide suitable targets for delivering toxin. Here we have assessedthe performance of bispecific F(ab')2 antibodies designed to deliversaporin to Burkitt's lymphoma cell lines via CD 19, CD22, CD37, or

surface Ig. The widespread expression of these particular differentiation antigens on B-cell lymphomas and their absence from hemopoi-

etic stem cells is likely to make them especially suitable targets fortherapy (13). We show that within this group of molecules only CD22provides a suitable vehicle for delivering saporin with BsAb. Furthermore, the level of toxicity achieved with the appropriate pair ofCD22-specific BsAb was equivalent to that reported for conventional

IT directed against this surface molecule.

MATERIALS AND METHODS

Saporin. The RIP saporin (SO-6), purified from the seeds of Saponaria

offícinalis, was kindly provided by Prof. F. Stirpe (University of Bologna,Italy) (14).

Cell Lines. BsAb were tested on the Burkitt's lymphoma cell lines Daudi

and Raji. Cells were maintained in supplemented RPMI [RPMI 1640 medium

containing 2 m.Mglutamine, 1 ITIMpyruvate, 100 ID/ml penicillin. 100 lU/mlstreptomycin. 2 fig/ml fungizone, 10 /j.g/ml ciprofloxacin, and 10% fetal calfserum (Myoclone) (GIBCO Ltd. Paisley, Scotland)].

Antibodies. The antibodies used in this study included four mAb directedagainst CD22. i.e.. HD6 and HD39, both gifts from Dr. B. Dorken (Universityof Heidelberg, Germany) (15, 16), 4KB 128, provided by Dr. D. Y. Mason, JohnRadcliffe Hospital (Oxford, UK) (17), and RFB-4 (18). The epitope designations of these are as follows: 4KB 128, CD22D; HD6. CD22B; RFB-4. CD22B;and HD39. CD22A (19, 20). Other antibodies included the ami-CD19 mAbHD37, kindly provided by Dr. B. Dorken (21). the anti-CD37 mAb WR17.

provided by Dr. J. Smith, Regional Immunology (Southampton, UK) (22). theanti-CD37 mAb MB-1, provided by Dr. Richard Miller and Prof. Ron Levy,Stanford University Medical Center (Stanford, CA) (23). and the anti-K-chainantibody HB6-I, supplied by the American Type Culture Collection (Rock-ville, MD). The anti-CD 19 mAb RFB-9 was produced in the Department of

4 A. Bell, R. R. French. T. Hamblin, and M. J. Glennie, unpublished observations.

3015

on June 9, 2017. © 1993 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 2: Delivery of Saporin to Human B-Cell Lymphoma …...field (5), recent trials in Hodgkin's (6) and non-Hodgkin's lymphoma (7, 8) have been encouraging and suggest that immunotoxin may

BSAB FOR DELIVERY OF SAPORIN TO LYMPHOMA

Immunology, Royal Free Hospital (London. UK); an anti-fi-chain mAb.M15-8, and twoantisaporin mAb, anti-sap-1 and anti-sap-5, were raised in this

laboratory and have been described previously (II). All antibodies are of theIgGl isotype, except WRI7, which is a IgG2a. mAb were purified from asciticfluid by precipitation in 2 M ammonium sulfate followed by ion exchangechromatography on Trisacryl-M-DEAE (IBF-LKB) (24).

Preparation of Bispecific Antibodies. Heterodimeric F(ab')2 fragments

were constructed using the chemical cross-linker o-phenylenedimaleimide, as

described by Glennie et al. (25).Incorporation of [3H]Leucine by Cultured Lymphoma Cells. The

method of assessing pH]leucine uptake was modified from our previous studies (11, 25). BsAb at a final concentration of I fig/ml (10~8 M) and serial

dilutions of saporin were preincubated for I h at 37°Cas 100-/xl aliquots in

leucine-free supplemented RPM1 in flat-bottomed 96-well microtiter plates(GIBCO). The cells (lOVwell), also in 100 fu of leucine-free supplementedRPMI. were then added and plates were incubated for 18 h at 37°C,in a

humidified atmosphere of 5% COs in air. before pulsing each well for 12 h at37°Cwith 0.5 /iCi/well ['Hjleucine. The uptake of ['Hjleucine was assessed

by harvesting the cells onto glass fiber filters and counting radioactivity in aliquid scintillation counter (LKB). All experimental points were determined intriplicate and results are expressed as a percentage of the ['Hjleucine incor

porated in untreated cells. The experimental error within each experiment wasminimal, with SD being <10% of the mean. However, some variation was

observed between experiments, probably due to variation between batches oftarget cells, giving IC5() values for saporin toxicity in the range 5 X 10~s to10~6M.Despite this interexperimental variation, the relative performance of the

various derivatives remained the same throughout.Radioiodination of Saporin. Saporin was trace radiolabeled for binding

studies by using carrier-free I25I (Radiochemical Centre. Amersham, UK) and

lodo-Beads (Pierce Chemicals Co., Rockford. IL) as the oxidizing reagent (26).

Radioactivity was measured in a Rackgamma counter (LKB).Binding of 125I-Saporin to Cell Surfaces by BsAb. A method described

by Dower et al. (27) and modified by French et al. (11) was used to quantifythe binding of 12SI-saporin-BsAb complexes to target cells. Radiolabeled saporin was serially diluted and incubated for l h at 37°Cin 1-ml aliquots of

supplemented RPMI containing BsAb at I fig/ml. A 100-fil sample of Daudior Raji cells (5 x 107/ml) was then added to each tube and the incubation wascontinued for an additional l h at 37°Cto allow binding to reach equilibrium.

Any endocytosis of surface-bound saporin-BsAb complexes was prevented byinclusion of sodium azide ( 15 HIM)and 2-deoxyglucose (50 HIM)in the reaction

mixture. The cells were then separated from the aqueous phase by rapidcentrifugation through phthalate oils, as described previously (11, 27).

Uptake of I25l-Saporin by Cultured Daudi Cells. To distinguish betweensurface-bound and intracellular I25l-saporin in cultured Daudi cells, we used an

acid wash method to remove surface-bound BsAb-saporin complexes, as described by Press et al. (28). Fresh Daudi cells (107/ml) were first incubated

with a mixture of BsAb ( 1 u,g/ml) and radioiodinated saporin ( 1 u.g/ml) for Ih at 0°Cto allow binding at the cell surfaces. Cells were then warmed to 37°C

and cultured in a humidified atmosphere of 5% COs in air, to allow uptake ofthe labeled complexes. At intervals during the culture, cells were carefullyresuspended and either (a) duplicate 0.4-ml aliquots were rapidly centrifuged

through phthalate oils to separate the cells, as described above, or (b) duplicateI-ml aliquots were washed three times in acidic RPMI 1640 medium to removesurface-bound BsAb-saporin complexes and allow the internalized I25l-saporin

to be measured. For each of these washes. 2 ml of ice-cold RPMI 1640 medium

(pH 1.5 with HC1) were added and the samples remained on ice for 15 minbefore the cells were recovered by centrifugation at 500 x g for IO min.Radioactive counts obtained with an irrelevant BsAb were subtracted to givelevels of specific binding and uptake. Surface-bound I25l-saporin was then

deduced by subtraction of intracellular counts from the total counts and resultswere plotted as number of saporin molecules in each cellular compartment.

RESULTS

Incorporation of [ 'HjLeucine into Cells in the Presence of Sa-porin-BsAb Complexes. A panel of bispecific F(ab')2 antibodies

were prepared to deliver saporin to neoplastic B-cells via a range ofdifferentiation antigens [CDI9 (RFB-9 and HD37), CD22 (4KB128),CD37 (WR17 and MB-1), /¿-chain(M15-8), and «-chain(HB6-1)]. In

each case Fab' fragments from these antibodies were thioether-linkedto Fab' fragments from the antisaporin antibody anti-sap-1. The effi

ciency with which saporin could be delivered to Daudi or Raji cells toinhibit incorporation of [3H]leucine was investigated in vitro. Fig. 1

shows that, in the absence of BsAb, saporin was only slightly toxicand for Daudi cells inhibited the uptake of [3H]leucine by 50% (IC50)at 2 X 10~7 to UT" M. However, in the presence of anti-CD22 BsAb,

saporin toxicity was increased approximately 700-fold for both Rajiand Daudi cells, resulting in an IC50 close to 3 X 10~'°M. Surpris

ingly, no significant increase in toxicity was observed with BsAbcontaining anti-CD 19 or anti-CD37, despite the presence of these

molecules on Daudi and Raji cells (see below). Fig. la shows resultsobtained using BsAb constructed with Fab from the anti-CD19 mAbRFB-9 and the anti-CD37 mAb WR17. BsAb containing Fab from asecond anti-CD 19 mAb (HD37) and a second anti-CD37 mAb (MB-1 )

also failed to potentiate saporin toxicity significantly (Table 1). Finally, two BsAb directed to surface Ig on Daudi cells, anti-/x andanti-K BsAb, did produce modest increases in saporin toxicity, whichresulted in an approximately 10-fold reduction in the saporin IC50

(Fig. \b). Similar small increases in toxicity were obtained when theseanti-Ig reagents were used on Raji cells (data not shown). The anti-

CD22 BsAb was not effective on Namalwa cells (data not shown),which is consistent with their very low expression of CD22 (29).

To investigate whether other CD22-specific BsAb performed in amanner similar to that of 4KB 128 X anti-sap-1, BsAb were preparedusing Fab' from three more anti-CD22 mAb, HD6, HD39, and RFB-4,

and their performance was compared with that of the original 4KB 128BsAb. As shown in Fig. 2, all four anti-CD22 BsAb performed very

similarly in cytotoxicity assays, giving IC50 values between 1.5 X10-'°Mand 6 X IO'10 M for Daudi and Raji cells.

100 -

80 -

= 60

O *O

** 20

•¿�co«J 10"

g. 100 T

80

E 60o

I 40Xco' ' 20

10' 10" 10' 10" 10' 10"

10'" 10H° 10'' 10"' 10"7 10'6

Saporin (M)

10'

Fig. I. Uptake of ['H]leucine by Daudi cells cultured in medium containing saporin

and BsAb. Cells (lOVwell) were cultured for 18 h with BsAb at 1 ng/ml and saporin althe concentrations shown, before pulsing with ( 'H|leucine and harvesting of cells to assess

the level of incorporated radioactivity. Triplicate samples were assayed for each concentration of saporin investigated. Panel a: X, saporin alone; O, RFB-9 X anti-sap-1 (anti-CD19);«,4KBI28 x anti-sap-l (anti-CD22); •¿�.WR17 X anti-sap-l (anti-CD37). Panelb: X, saporin alone; •¿�.4KB 128 X anti-sap-l (anti-CD22); G. M 15-8 X anti-sap-l(anti-fi-chain); A. HB6-I x anti-sap-l (anti-Kl. At least three other experiments yieldedsimilar results.

3016

on June 9, 2017. © 1993 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 3: Delivery of Saporin to Human B-Cell Lymphoma …...field (5), recent trials in Hodgkin's (6) and non-Hodgkin's lymphoma (7, 8) have been encouraging and suggest that immunotoxin may

BSAB FOR DKUVERY OF SAPORIN TO LYMPHOMA

Table I Delivery of saporin to Duitdi cells using one or two BsAb

BsAbspecies"Saporin

aloneHD37x ami-sap-14KB

128 x anti-sap-14KB128 xanti-sap-5MB-I

x anti-sap-1HB6-1x anti-sap-1M

15-8 x anti-sap-14KB128 x anti-sap- 1+4KB

128 xanti-sap-5HD6x anti-sap- 1+4KB12X

xanti-sap-54KB128 x anti-sap-5+HB6-1

x anti-sap-14KB128 x anti-sap-5+M15-8 x anti-sap-14KB128 x anti-sap-5+RFB-9

x anti-sap-14KB128 x anti-sap-5+WRI7

x anti-sap- 1Antiger.'7CDI9CD22CD22CD37K-chain¿¿-chainCD22CD22CD22CD22CD22K

chainCD22/i

chainCD22CDI9CD22CD37IC51,

(Ml'3.1

±0.5 x IO'7(12)>3x IO'8(4/6.6±

l.8xl(r"'(IO)2.4±0.9 x IO'9(3)>3

x IO"*<3/5x l(r8(2/3x IO"8(2/1.5±0.4x

IO-"(5)*2.1

x l(r"(2)3x

ur"ID7x

ur"ID7x

l()-'"(l)3x

l(r"'<l>Molecules

atl/^d

I<-W>2().(KX)I.(XX)ND*>W.(X)07().(XX)NDI.(XX)NDNDNDNDNDFoldIncrease''<KS300-1.

(XX)150-3000-5611)HMXXMO.OUOIO.ÕXXKSO.ÜOOU).(XX)4.3(X)400I.IXX)

11Mixtures of BsAb were selected (equal quantities) to bind to saporin through different specificities (anti-sap-1 and anti-sap-5) and one or two B-cell differentiation antigens as

indicated.''Target antigen recognized by BsAb on Daudi cells.' Saporin concentrations giving half-max imum inhibition of protein synthesis in cylotoxicity experiments. Shown are mean ±SE of ICs»values determined in the number of separate

experiments given in parentheses. Where the experiment was done only once or twice, the mean value alone is given.*' Average number of molecules present/Daudi cell at the ICso concentration, as calculated from binding studies (see Fig. 3).'' Fold increase in saporin toxicity, compared with that of saporin alone, obtained in individual experiments.

fp < 0.01. compared with 4KB128 x anti-sap-1.K ND. not determined.h 0.02 < p < 0.05, compared with 4KB 128 x anti-sap-1.

IO" 10" 10-' 10""

Saporin IMI

10"'0 10"' 10-

Fig. 2. Toxicity of saporin in the presence of different CD22-specific BsAb. Using thesame conditions as described for Fig. I. we have compared the uptake of [ 'Hjleucine in

the presence of saporin and 4KB 128 X anti-sap-1 (•).HD6 X anti-sap-1 (D), HD39 Xanti-sap-1 (A>. or RFB-4 X anti-sap-1 (O). with Daudi (a) and Raji (fc) cells. The 1C,,,values for saporin alone on Daudi and Raji cells were 5.3 x IO'7 and 3 X I0~7 M,

respectively. One of two similar experiments.

Binding of I25l-Saporin to Daudi Cells in the Presence of BsAb.

We next investigated whether the performance of the BsAb derivatives was related to their ability to capture saporin on the target cellsurface. The binding of radiolabeled '-^I-saporin to Daudi cells by

BsAb was investigated using the same concentrations of saporin andantibody as those used in the toxicity assay. Cell-bound and freeI25l-saporin in the equilibrated reaction mixture were separated by

rapid centrifugation of the cells through a mixture of phthalate oils, asdescribed in "Materials and Methods." The binding curves in Fig. 3

show that all the BsAb, regardless of which differentiation antigenthey recognized, bound saporin to Daudi cells. Interestingly, the anti-

CD22 BsAb which had been most active in toxicity was probably theleast effective in capturing saporin on the cells. In contrast, the anti-CD37 BsAb containing MB-1, which had been totally inactive in

boosting saporin toxicity (Table 1), bound the most saporin to the cell.These binding data emphasize the efficiency of CD22 BsAb in delivering saporin and show that, at its 1C,,, value of 3 X 10~'°M(Fig. 1

and Table 1). this derivative binds only about 1000 molecules to eachcell surface. The anti-CD37 BsAb MB-1 X anti-sap-1, however, was

unable to affect the uptake of |'H]leucine significantly even when

90.000 molecules of saporin were bound to each cell.Uptake of I25l-Saporin by Cultured Daudi Cells. Because of

this lack of correlation between binding of I25l-saporin and cytotoxic

activity, we next investigated the ability of each BsAb to transportI25l-saporin from the cell surface into the interior of metabolically

active Daudi cells. To distinguish between surface-bound and internalized 12il-saporin, at various intervals during culture aliquots of

cells were taken and washed three times in RPMI 1640 medium at pH1.5 to remove any surface-bound '2<iI-saporin-BsAb complexes. Fig.

4 shows the results for four BsAb derivatives. anti-CD22 (4KB 128),

10 -,

vo.E 8 -

OCLiou>

oE

VIo

6 -

2 -

CD37

CD19CD22

10' 10" 10' 10'' 10" 10'

Saporin (M)Fig. 3. Binding of I2'l-saporin to Daudi cells in the presence of single BsAb. BsAb

(I ng/m\} were incubated with various concentrations of I2il-saporin for I h at 37°C.Daudi cells ( 107/tube) were then added, and the incubation was continued for an additionalI h. Any endocytosis of cell-bound complexes was inhibited by including 15 niM sodiumazide and 50 HIM2-deoxyglucose in the reaction mixture. The cells were then sedimentcdthrough phthalate oils and the pellet was counted for radioactivity. The results are expressed as the number of molecules of saporin bound/cell. O, RFB-9 X anti-sap-1(anti-CDW);«. 4KB128 x anti-sap-1 (anti-CD22|; •¿�.WRIT X anti-sap-1 (anti-CD37);A. HB6-1 X anti-sap-1 (anti-K-chain). One of three similar experiments.

3017

on June 9, 2017. © 1993 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 4: Delivery of Saporin to Human B-Cell Lymphoma …...field (5), recent trials in Hodgkin's (6) and non-Hodgkin's lymphoma (7, 8) have been encouraging and suggest that immunotoxin may

BSAB FOR DELIVERY OF SAPORIN TO LYMPHOMA

0246 02

Time in culture (h)Fig. 4. Internalizaron of I25l-saporin by Daudi cells in the presence of BsAb. BsAb and

I25l-saporin, each at 1 ng/ml. were incubated with Daudi cells for l h at 4°C.Thetemperature was then increased to 37°Cto allow uptake to occur and duplicate aliquolswere taken at the times shown. With the first sample, total cell-associated I25l-saporin

(surface and internal) was determined by measuring radioactivity after sedimenting thecells through phthalate oils. With the second sample, intracellular I25l-saporin was determined by measuring the remaining radioactivity after removing all surface-bound complexes by washing the cells three times in RPM1 1640 medium, pH 1.5. Surface radioactivity was obtained by subtraction. The results are expressed as the number of moleculesof saporin/cell. •¿�,total cell associated; •¿�.intracellular; A, surface. The four BsAb usedin these experiments were. 4KBI28 X anti-sap-1 (anti-CD22). (Panel a); RFB-9 Xanti-sap-1 (anti-CDI9), (Panel b); WR17 X anti-sap-1 (anti-CD37), (Panel c); and M15-8X anti-sap-1 (anti-n) (Panel d). Two other experiments yielded similar results.

anti-CD19 (RFB-9), anti-CD37 (WR17), and anti-K. Under the conditions used (125I-saporin and BsAb each at 1 fxg/ml), between 20,000and 65,000 molecules of 125I-saporin were bound to each cell at theoutset of culture. As expected, the efficiency of I25l-saporin-binding

was in the same order as that shown in Fig. 3, i.e., anti-CD37 > anti-K> anti-CD 19 = anti-CD22. However, once the cells were warmed to37°C,the accumulation of radioactivity inside the cells showed a

completely different hierarchy, with BsAb performing in the orderanti-CD22 anti-K > anti-CD 19 = anti-CD37. Thus, despite anti-

CD22 BsAb binding only 20,000 molecules of saporin to the surfaceof each cell at the outset, by 4 h of culture radioactive counts indicatedthat at least 15,000 molecules of saporin had been internalized(Fig. 4a). In contrast, the anti-CD 19 and anti-CD37 derivatives, which

captured close to 23,000 and 65,000 saporin molecules, respectively,accumulated <2000 molecules of saporin in each cell over the sameperiod. The anti-K reagent shown in Fig. 4d fell partway between these

two extremes, collecting counts consistent with approximately 5000molecules of saporin. Thus, the hierarchy of 125I-saporin uptake

by BsAb correlates closely with their performance in cytotoxicity(Fig. 1).

Inhibition of Protein Synthesis in the Presence of Combinationsof BsAb Directed at One or Two Differentiation Antigens. Previous work from this laboratory has shown that selected pairs of BsAbthat bind cooperatively to saporin outperform single derivatives in thedelivery of saporin to lymphoma cells (11). In the current work weinvestigated whether a similar improvement could be achieved bycombining various BsAb specific for CD22. For this investigation anew BsAb was prepared with Fab' from the anti-CD22 mAb 4KB 128

cross-linked to Fab' from anti-sap-5, an antisaporin antibody which

binds to an epitope on saporin different from that recognized byanti-sap-1. This new derivative, 4KB 128 X anti-sap-5, alone wasslightly less effective than 4KB 128 X anti-sap-1 at enhancing saporin

toxicity. However, when combined with an equal amount of 4KB 128x anti-sap-1, the mixture was 30-50-fold more potent than 4KB 128X anti-sap-1 or 4KB 128 X anti-sap-5 alone (Fig. 5), giving an IC50value for saporin on Daudi and Raji cells of about 2 X 10~" M.

Similar increases in toxicity were observed when 4KB 128 X anti-sap-5 was paired with BsAb containing anti-sap-1 and Fab' from each

of the three other anti-CD22 mAb, HD6 (Table 1), HD39, and RFB-4

(data not shown). The enhanced performance of cooperative pairs ofBsAb depends on binding to separate epitopes on saporin (e.g., sap-1and sap-5) (11) but is not dependent on whether one (CD22D) or

two (e.g., CD22D and CD22B) epitopes are recognized on CD22(Table 1).

To investigate whether toxicity could also be enhanced by delivering saporin via different target molecules, we combined pairs of BsAbwhich were specific for separate epitopes on saporin and differentantigens on the target cell. The results in Table 1 show that mixing ananti-CD22 BsAb, 4KB 128 X anti-sap-5, with either the anti-/¿oranti-K BsAb (Table 1) produced an approximately 30-fold increase intoxicity over that produced by 4KB 128 X anti-sap-5 alone and came

close to matching the performance of the most potent combination ofCD22-BsAb (Fig. 5). However, there was only a minimal increase intoxicity when 4KB 128 X anti-sap-5 was combined with either ananti-CD 19 or anti-CD37 BsAb. Here the activity of the mixtureslightly exceeded that of 4KB 128 x anti-sap-5 alone but was neverbetter than that of 4KB 128 X anti-sap-1. Thus, it appears that pairs of

BsAb can target via two different surface antigens and achieve augmented toxicity, but only if both the selected antigens (e.g., CD22 andIg) perform individually as target molecules.

Binding of I2sl-Saporin to Cells in the Presence of Combina

tions of BsAb. Finally, experiments were performed to measure thefunctional affinity with which radiolabeled saporin binds to targetcells when tethered by a single BsAb or cooperative pairs of BsAb.Previous work has shown that the main advantage of using two BsAbstems from the increased avidity with which they can capture saporinon the target cells (11). The results in Fig. 6 confirm this benefitshowing that there was an approximately 20-fold increase in aviditywhen 125I-saporin was bound to cells via a combination of 4KB 128

= 100 n

u 80 HK

co5 6<oo.k_8 40 -

1 2°H

X

—¿� O10' 10" 10" 10" 10' 10" 10'

Saporin (M)Fig. 5. Comparison of the performance of a single anti-CD22 BsAb and a pair of

anti-CD22 BsAb constructed with different antisaporin Fab' arms. Daudi cells were

cultured as described for Fig. 1, using BsAb at 1 ¿ig/ml(for the pair, 0.5 ^ig/ml of each)and saporin at the concentrations shown. The points represent the means of triplicates, x,saporin alone, •¿�,4KB128 X anti-sap-1; A, 4KB128 X anti-sap-5; •¿�4KB128 X anti-sap-1 plus 4KB128 X anti-sap-5. At least five other experiments yielded similar results.

3018

on June 9, 2017. © 1993 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 5: Delivery of Saporin to Human B-Cell Lymphoma …...field (5), recent trials in Hodgkin's (6) and non-Hodgkin's lymphoma (7, 8) have been encouraging and suggest that immunotoxin may

BSAB FOR DELIVERY OF SAPORIN TO LYMPHOMA

1(T12 10'11 10 10-

Saporin (M)Fig. 6. Binding of '2M-saporin lo Daudi cells in the presence of a single anli-CD22

BsAb or a pair of anti-CD22 BsAb. The binding of '"I-saporin to Daudi cells was

determined as described for Fig. 3. The results are expressed as the number of moleculesof saporin bound/cell. Inset, saturation curves plotted on a linear scale. The BsAb derivatives used were 4KB 128 X anti-sap-1 (•)and 4KB 128 X anti-sap-1 plus 4KB 128 Xanti-sap-5 (•).

X anti-sap-1 and 4KB 128 X anti-sap-5, compared with that of theindividual BsAb. This is based on the concentration of I25l-saporinneeded to achieve half-maximum binding (5 X 10~lu M, comparedwith >1 X 10~8 M). Interestingly, despite this significant increase in

avidity for saporin, both the single BsAb, 4KB 128 X anti-sap-1, andthe mixture of CD22-BsAb, 4KB 128 X anti-sap-1 plus 4KB 128 Xanti-sap-5, needed to capture approximately 1000 molecules of sa

porin on each cell to achieve the ICM) (Table 1). At saturation, approximately half the number of saporin molecules were bound percell with the pair of BsAb, compared with a single derivative(10,000/cell versus >20,000/cell). These data are consistent with the1:1 and 2:1 BsAb:saporin binding ratios expected when each molecule of saporin is delivered by one or two molecules, respectively, ofBsAb.

DISCUSSION

Previous studies from this laboratory have shown that F(ab')2 BsAb

can be used to target saporin in the treatment of animal (10, 11) andhuman (12) lymphoma. In both situations the derivatives were designed to target saporin, through either surface IgM idiotype on theguinea pig cells or CD22 on the human lymphoma. The resultsachieved, although preliminary, were sufficiently encouraging to justify investigation of other B-cell differentiation molecules whichcould be considered as targets for therapy in B-cell lymphoma. Here

we have compared the performance of a panel of BsAb, with specificity for saporin and CD 19, CD22, CD37, /¿-chain,or «-chain, intargeting the Burkitt's lymphoma cell lines Raji and Daudi (Fig. 1).

Surprisingly, only the anti-CD22 BsAb gave efficient potentiation ofsaporin toxicity, resulting in close to a 1000-fold reduction in the

amount of saporin required to achieve an IC50 with Raji and Daudicells. In contrast, BsAb specific for p.- and x-chain performed poorly,giving just a 10-fold increase in saporin toxicity, and, finally, twoanti-CD19 and two anti-CD37 BsAb derivatives failed to enhance

saporin toxicity significantly.Binding studies with '25I-saporin showed that there was no corre

lation between the activity of the various BsAb in toxicity assays andtheir ability to bind saporin to the target cell surface. Thus, for example, the CD37-specific reagents, which failed to potentiate saporintoxicity, bound by far the most '-^ I-sapori n to the target cells, reflect

ing the level at which CD37 is expressed by Daudi and Raji cells (30).However, the CD22-specific BsAb, which bound only 20-25% as

much saporin on the surface of cells, was a highly potent reagent intoxicity. These differences in efficiency of binding and toxicity areemphasized when we compare the number of saporin molecules required at the cell surface, for each type of BsAb derivative, to achievean IC50 for protein synthesis. With the anti-CD22 BsAb only 1000

molecules of saporin are required on each cell to reach the IC,0 (3 X10~'°M) (Figs. \A and 2), while when saporin is delivered by the

anti-Ig BsAb approximately 60,000 are needed, and with the anti-

CD37 reagents significant inhibition of protein synthesis is not seeneven when 90,000 saporin molecules are bound to each cell. Suchresults indicate that capture of saporin on the cell surface alone is notenough to determine the potency of a BsAb and suggest that perhapsa key event lies at a later stage in delivery, such as during endocytosisand/or intracellular routing of the BsAb-saporin complexes.

Uptake studies lend weight to this interpretation, showing thatCD22-specific BsAb were alone in accumulating significant levels ofradiolabeled saporin inside cultured cells. Thus, when an anti-CD22BsAb was used to capture 20,000 molecules of I25l-saporin on the

surface of each cell, the transfer of radioactivity over 4 h of culturesuggested that approximately 15,000 molecules had been internalized. By contrast, under the same conditions a BsAb functioningthrough CD37 bound >70,000 molecules to the surface of each celland yet never appeared to accumulate >3000 molecules inside eachcell. The anti-Ig reagents fell somewhere between these two extremes.

Thus, the performance of the various BsAb appeared to correlate veryclosely with their ability to accumulate saporin within the target cells.

A number of studies have shown a high correlation between thepotency of an IT and its rate of endocytosis into the target cell(31-34). Since a single BsAb is univalent for saporin and the targetcell and is therefore unable to promote internalization through cross-

linking of adjacent target molecules (35), it is likely that the entry ofBsAb-saporin complexes into the cell depends on constitutive mem

brane turnover of the surface molecule being targeted, a process whichmay not occur if the molecule being targeted is not being continuallydelivered inside the cell (36). Numerous studies have shown that,since univalent IT function efficiently, bivalent binding is not requiredfor activity, and the small loss of potency which is sometimes ( 11, 29,37) but not always (38, 39) observed on moving from a bivalent to aunivalent reagent probably reflects a drop in functional affinity (avidity) (29). A plausible explanation for our findings, therefore, is thatonly CD22 and to a lesser extent surface Ig are turning over at a ratewhich allows efficient translocation of saporin into the cell, whileCD 19 and CD37 are not. The binding and uptake data in Figs. 3 and4 would support this conclusion, and the data from Press et al. (30),which show that mAb against CD22 and surface /¿-chainare inter

nalized by lymphoid cells, while those reacting with CD37 remain onthe cell surface, are also consistent with this interpretation.

Our failure to potentiate saporin toxicity or to accumulate saporin intarget cells with either of the anti-CD 19 BsAb is more difficult to

explain. While such results are consistent with a failure to internalize the complexes, most IT containing anti-CD 19 mAb and RIPhave functioned well as both bivalent (29, 40—42)and, more impor

tantly, univalent (29) derivatives. One exception to this finding waswith an IT made with the anti-CD 19 mAb B4, which was unusual inthat it was not internalized by B-cells during extended culture (34).

Our only reluctance in accepting this explanation for our finding isthat most anti-CD 19 mAb, at least in a bivalent form, are internalized

3019

on June 9, 2017. © 1993 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 6: Delivery of Saporin to Human B-Cell Lymphoma …...field (5), recent trials in Hodgkin's (6) and non-Hodgkin's lymphoma (7, 8) have been encouraging and suggest that immunotoxin may

BSAB FOR DELIVERY OF SAPORIN TO LYMPHOMA

(30, 40), and one of the anti-CD 19 BsAb was constructed using

HD37, a mAb which is known to function as a univalent reagent (29).Among the alternative explanations which should be considered is thepossibility that these BsAb are being internalized but rapidly degradedinside the cell, perhaps in the lysosomal compartment (36). It isknown from a number of studies that, within the group of IT whichundergo internalization, intracellular routing often plays a key part indetermining their potency (43^6). For example, the activity of arange of IT directed at various T-cell antigens, CD2, CD3, and CDS,

was shown to depend more on their rate of transfer to the lysosomalcompartment than on their delivery from the surface to the interior ofthe cell (45). Likewise, differences in the route of internalization ofanti-CD22 and anti-CD 19 mAb, especially their ability to enter a"prominent tubular endocytic compartment" adjacent to the trans-

Golgi apparatus (30, 46), may explain the somewhat superior performance of IT made with anti-CD22 over those made with anti-CD 19

(29). Experiments are currently underway to investigate the fate ofCD 19 BsAb-saporin complexes on target B cells and, if internalized,

their destiny within the cell.In the current work, the performance of a single CD22-specific

BsAb could be improved 10-50-fold by using a combination of twoCD22-specific BsAb or one anti-CD22 BsAb and one anti-Ig BsAb

(see Table 1). Each pair of BsAb was constructed to bind differentnonblocking epitopes on the saporin. We have previously described asimilar improvement in performance with selected pairs of BsAbdirected towards guinea pig L2C leukemic cells (11). Similar improvements in performance have been described using cooperative bindingof two BsAb specific for a tumor-associated antigen and an indium-

labeled bivalent hapten, when imaging tumors in mice (47) and morerecently patients.5

We have found that the superior performance of cooperative pairsof BsAb derives mainly from the increased avidity with which saporinis held at the cell surface (Fig. 6) ( 11). Multivalent binding of saporinto the cell surface could also favor more rapid uptake of BsAb-saporin

complexes through antigenic modulation (35), but, as previously demonstrated in the L2C leukemia model in guinea pigs (11), this appearsto be a minor factor influencing the performance of combinations ofBsAb. This interpretation is supported by results showing that theaverage number of saporin molecules bound on each cell at the IC50of protein synthesis for Daudi and Raji cells is approximately the same( 1000) (Table 1), regardless of whether a single BsAb or a combination of anti-CD22 BsAb is used. Thus, although the mixture of BsAb

is about 20 times more efficient at capturing saporin on the cellsurface, it is no more effective than a single BsAb at delivering thatsaporin into the cell once it is bound.

In conclusion, the favorable internalization and/or intracellularrouting of anti-CD22 BsAb-saporin (and to a lesser extent anti-IgBsAb-saporin) complexes have resulted in high efficiency killing bysaporin and make CD22 a good target for delivering toxin to neoplas-tic B-cells by BsAb. Two other potential B-cell targets, CD19 and

CD37, have not proved suitable. While lack of potency with CD37derivatives probably arises from a failure to enter the cell, we have yetto determine whether the surprising lack of cytotoxic activity usinganti-CD 19 BsAb results from a lack of internalization or rapid deg

radation in the lysosomal compartment of the cell. Finally, the use ofa cocktail of two CD22-specific BsAb to obtain optimum delivery of

toxin could provide an important benefit for therapeutic applicationsand make BsAb an interesting alternative to conventional IT in thetreatment of neoplastic disease.

ACKNOWLEDGMENTS

We wish to thank Drs. Andrew George and Andrew Lane and members ofthe Tenovus Laboratories for helpful discussions during this project and Professor Fiorenzo Stirpe for the supply of saporin.

REFERENCES

14.

15.

5 J. M. Le Doussal, personal communication.

Vitella, E. S., Fulton. R. J.. May. R. D., Till, M., and Uhr, J. W. Redesigning nature's

poisons to create anti-tumor reagents. Science (Washington DC), 238: 1098-1104,

1987.Blakey. D. C., and Thorpe. P. E. An overview of therapy with immunotoxins containing ricin or its A-chain. Antibody Immunoconjugales Radiopharmacol., I: 1-16,

1988.Hertler, A. A., and Frankel. A. E. Immunotoxins: a clinical review of their use in thetreatment of malignancies. J. Clin. Oncol.. 7: 1932-1942, 1989.

FitzGerald. D., and Pastan. I. Targeted toxin therapy for the treatment of cancer. J.Nati. Cancer Inst., 81: 1455-1463. 1989.Byers. V. S., and Baldwin. R. W. Therapeutic strategies with monoclonal antibodiesand ¡mmunoconjugates. Immunology, 65: 329-335, 1988.

Falini, B., Bolognesi, A., Flenghi. L., Tazzari, P. L., Broe, M. K., Stein, H.. Burkop,H., Aversa, F., Comeli, P., Pizzolo, G., Barbabietola, G., Sabattini. E., Pilen, S.,Martelli, M. F., and Stirpe. F. Response of refractory Hodgkin's disease to monoclonal

anti-CD30 immunotoxin. Lancet, 339: 1195-1196. 1992.

Vitella. E. S.. Stone, M., Amlot, P., Fay, J., May, R., Till. M., Newman, J., Clark, P.,Collins. R.. Cunningham, D., Ghetie, V, Uhr, J. W., and Thorpe, P. E. Phase Iimmunotoxin trial in patients with B-cell lymphoma. Cancer Res., 51: 4052-4058,

1991.Grossbard. M. L., Freedman, A. S., Ritz, J., Coral, F.. Goldmacher, V. S., Eliseo, L.,Spector. N., Dear, K., Lambert, J. M., Blattler, W. A., Taylor, J. A., and Nadler, L. M.Serotherapy of B-cell neoplasms with anti-B4-blocked ricin: a phase I trial of dailybolus infusion. Blood, 79: 576-585, 1992.

Raso, V, and Griffin. T. Hybrid antibodies with dual specificity for the delivery ofricin to immunoglobulin-bearing target cells. Cancer Res., 41: 2073-2078, 1981.

Glennie, M. J.. Brennand, D. M., Bryden, F., McBride, H. M., Stirpe, F., Worth, A. T.,and Stevenson, G. T. Bispecific F(ab')2 antibody for the delivery of saporin in the

treatment of lymphoma. J. Immunol., 141: 3662-3670, 1988.

French, R. R., Courtenay, A. E., Ingamells, S., Stevenson, G. T., and Glennie, M. J.Cooperative mixtures of bispecific Ftab'h antibodies for delivering saporin to lymphoma in vitro and in vivo. Cancer Res., 5/.- 2353-2361, 1991.

Bonardi, M. A.. Bell. A., French. R. R.. Gromo, G., Hamblin. T., Modena, D.. Tu»,A. L., and Glennie, M. J. Initial experience in treating human lymphoma with acombination of bispecific antibody and saporin. Int. J. Cancer, 7: 73-77, 1992.Dorken, B., Moller, P.. Pezzullo, A., Schwartz-Albiez. R., and Moldenhauer. G. B-cell

antigens: section report. In: W. Knapp, B. Dorken, W. R. Gilks, E. P. Rieber, R. E.Schmidt, H. Stein, and A. E. G. K. von dem Borne (eds.), Leukocyte Typing IV: WhiteCell Differentiation Antigens, pp. 15-224. Oxford, UK: Oxford University Press,

1989.Stirpe, F., Gasperi-Campani, G., Barbieri, L., Falasca, A., Abbondanza, A., andStevens, W. A. Ribosome inactivating proteins from the seed of Saponaria officinalisL. (soapwort), of Agrostemma githago L. (com cockle) and of Asparagus officinalis(asparagus) and from the latex of Hura crépitonsL. (sandbox tree). Biochem. J., 216:617-625, 1983.Moldenhauer, G., Dorken, B.. Schwartz, R., Pezzullo, A., and Hammerling, G. J.Characterization of a human B lymphocyte-specific antigen defined by monoclonalantibodies HD6 and HD39. In: E. L. Reinherz, B. F. Haynes, L. M. Nadler, and I. D.Bernstein (eds.). Leukocyte Typing II, Vol. 2: Human B Lymphocytes, pp. 97-107.New York: Springer-Verlag, 1986.Dorken, B., Moldenhauer, G., Pezzullo, A., Schwartz, R., Feller, A., Kiesel, S., andNadler, L. M. HD39 (B3), a B lineage-reslricled antigen whose cell surface expressionis limited to resling and activated human B lymphocyles. J. Immunol., 136: 4470-

4479. 1986.Mason, D. Y., Stein, H., Gerdes, J., Pulford, K. A. F., Ralfkiaer, E., Falini, B., Erber,W. N., Micklem, K., and Gatter, K. C. Value of monoclonal anti-CD22 (pl35)antibodies for the detection of normal and neoplastic B lymphoid cells. Blood. 69:836-840, 1987.Campana, D., Janossy, G., Bofill, M., Trejdosiewicz, L. K., Ma, D., Hoffbrand, A. V.,Mason, D. Y., Lebacq, A. M., and Forster, H. K. Human B development I. Phenotypicdifferences of B lymphocytes in the bone marrow and peripheral lymphoid tissue. J.Immunol.. ¡34: 1524-1530, 1985.Pezzullo, A., Dorken, B., Moldenhauer, G., Rabinovitch, P. S., and Clark, E. A.Amplification of B-cell activalion by Workshop monoclonal anlibodies to the CD22B-cell antigen. In: A. J. McMichael (ed.), Leukocyle Typing III: White Cell Differentiation Antigens, pp. 361-363. Oxford, UK: Oxford University Press, 1989.Li, J. L., Shen, G. L., Ghelie, M. A., May, R. D., Till, M., Ghetie, V., Uhr, J. W.,Janossy, G., Thorpe, P. E., Amlol, P., et al. The epilope specificity and tissue reaclivilyof four murine monoclonal anli-CD22 antibodies. Cell. Immunol., 118: 85-99. 1989.

Pezzullo, A., Dorken, B., Feller, A., Moldenahuer, G., Schwartz, R., Wemet, P., Thiel.E., and Hunstein. W. HD37 Monoclonal antibody: a useful reagent for further char-aclerizalion of "non-T, non-B" lymphoid malignancies. In: E. L. Reinherz, B. F.

Haynes, L. M. Nadler, and I. D. Bernstein (eds.), Leukocyle Typing II, Vol. 2: HumanB Lymphocyles, pp. 391-402. New York: Springer-Verlag. 1986.Moore, K., Cooper, S., and Jones, D. B. Use of Ihe monoclonal anlibody WR17,identifying the CD37 gp 40-45 kd antigen complex, in diagnosis of B-lymphoidmalignancy. J. Palhol., 152: 13-21, 1987.

3020

on June 9, 2017. © 1993 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 7: Delivery of Saporin to Human B-Cell Lymphoma …...field (5), recent trials in Hodgkin's (6) and non-Hodgkin's lymphoma (7, 8) have been encouraging and suggest that immunotoxin may

BSAB FOR DELIVERY OF SAPORIN TO LYMPHOMA

23. Link. M. P., Bindl, J.. Meeker, T. C, Carswell, C.. Doss. C. A., Wamke. R. A., andLevy. R. A unique antigen on mature B cells defined by a monoclonal antibody. J.Immunol.. 137: 3013-3018, 1986.

24. Elliot, T. J.. Glennie, M. J., McBride, H. M., and Stevenson, G. T. Analysis of theinteraction of antibodies with immunoglobulin idiotype of neoplastic B lymphocytes:implications for ¡mmunotherapy. J. Immunol.. 138: 981-988. 1987.

25. Glennie. M. J., McBride. H. M., Worth. A. T., and Stevenson, G. T. Preparation andperformance of bispecific F(ab')2 antibody containing thioether-linked Fab' frag

ments. J. Immunol., ¡39:2367-2375, 1987.

26. Markwell, M. A. K. A new solid state reagent to iodinate protein. I. Conditions for theefficient labelling of antiserum. Anal. Biochem.. 125: 427-432, 1982.

27. Dower, S. K., De Lisi, C., Titus, J. A., and Segal. D. M. Mechanism of binding ofmultivalent immune complexes to Fc receptors. I. Equilibrium binding. Biochemistry.20: 6326-6334, 1981.

28. Press. O. W.. Martin. P. J., Thorpe. P. E., and Vitella, E. S. Ricin A chain-containing

immunoioxins directed against different epitopes on the CD2 molecule differ in theirability to kill normal and malignant T cells. J. Immunol., 141: 4410-4417, 1988.

29. Ghetie, M. A., May, R. D., Till, M.. Uhr. J. W.. Ghetie, V.. Knowles. P. P.. Reif. M..Brown, A., Wallace, P. M., Janossy, G., Amlot, P.. Vitella. E. S., and Thorpe. P. E.Evaluation of ricin A chain-containing immunotoxins directed against CD19 and

CD22 antigens on normal and malignant human B cells as potential reagents for invivo therapy. Cancer Res., 48: 2610-2617. 1988.

30. Press, O. W., Fair, A. G., Borroz, K. I., Anderson. S. K.. and Martin, P. J. Endocytosisand degradation of monoclonal antibodies targeting human B-cell malignancies.Cancer Res., 49: 4906-4912, 1989.

31. Pirker, R., FitzGerald. D. J. P.. Hamilton. T. C.. Ozols, R. F.. Laird. W.. Frankel, A.E.. Willingham, M. C., and Pastan, I. Characterization of immunotoxins active againstovarian cancer cell lines. J. Clin. Invest., 76: 1261-1267, 1985.

32. Raso, V. Mediation of toxin entry into cells via naturally occurring ligand receptorsites. In: C-W. Vogel (ed), Immunoconjugates. Antibody Conjugates in Radioimagingand Therapy of Cancer, pp. 116-152. New York: Oxford University Press, 1987.

33. Youle, R. J., and Neville, D. M. Role of endocytosis and receptor recycling inligand-toxin and antibody-toxin conjugate activily. In: C-W. Vogel (ed). Immunoconjugates. Antibody Conjugales in Radioimaging and Therapy of Cancer, pp. 153-169.New York: Oxford University Press, 1987.

34. Goldmacher, V. S„Scott, C. F.. Lambert. J. M., Mclnlyre. G. D.. Blattler, W. A.,Collinson. A. R.. Stewart. J. K.. Chong, L. D.. Cook. S.. Slayter. H. S.. Beaumont. E..and Watkins, S. Cytotoxicity of gelonin and its conjugates with antibodies is determined by the extent of their endocytosis. J. Cell Physiol., 141: 222-234. 1989.

35. Glennie, M. J., and Stevenson, G. T. Univalent antibodies kill tumour cells in vitro andin vivo. Nature (Lond.), 195: 712-714, 1982.

36. Hopkins, C. R., Gibson, A., Shipman, M.. and Miller. K. Movement of internalizedligand-receptor complexes along a continuous endosomal reticulum. Nature (Lond.),346: 335-339, 1991.

37. Engert, A.. Martin. G.. Pfreundschuh. M.. Amlot. P.. Hsu. S-M., Diehl, V.. and Thorpe.P. Antitumor effects of ricin A chain immunotoxins prepared from intact antibodiesand Fab' fragments on solid human Hodgkin's disease tumors in mice. Cancer Res..

50. 2929-2935. 1990.38. May. R. D.. Wheeler, H. T. Finkelman, F. D.. Uhr, J. W., and Vitella. E. S. Intracell-

ular routing rather than cross-linking or rate of intemalization determines the potency

of immunotoxins directed against different epitopes of slgD on murine B cells. Cell.Immunol.. US: 490-500. 1991.

39. Derocq. J. M.. Casellas, P., Laurent. G., Ravel. S.. Vidal. H., and Jansen, F. K.Comparison of the cytotoxic poiency of T101 Fab. F(ab')2 and whole IgG immuno

toxins. J. Immunol.. 141: 2837-2843. 1988.40. Uckun. F. M.. Jaszcz, W.. Ambrus. J. L.. Fauci. A. S.. Gajo-Peczalska, K.. Song. C.

W.. Wick. M. R.. Myers. D. E.. Waddick. K.. and Lcdbetter. J. A. Detailed studies onexpression and function of CD19 surface determinant by using B43 monoclonalantibody and the clinical potential of anti-CDI9 immunotoxin. Blood, 71: 13-29,

1988.41 Bregni, M.. Siena. S.. Formosa. A.. Lappi. D. A.. Martineau. D.. Malavasi. F.. Dorken.

B.. Bonadonna. G.. and Gianni. A. M. B-cell restricted saporin immunotoxins: activityagainst B-cell lines and chronic lymphocytic leukemia cells. Blood. 73: 753-762,

1989.42. Myers, D. E., Irvin. J. D., Smith, R. S.. Kuebelbech, V. M., and Uckun. F. M.

Production of a pokeweed antiviral protein (PAP)-containing immunotoxin, B43-PAP,

directed against the CD 19 human B lineage lymphoid differentiation antigen in highlypurified form for human clinical trials. J. Immunol. Methods, 136: 221-237. 1991.

43. Preijers. F. W. M. B., Tax. W. J. M.. De Witte. T.. Janssen. A.. Heijden. H. V. D.. Vidal.H., Wessel. J. M. C., and Capei, P. J. A. Relationship between internalizalion andcytotoxicity of ricin A-chain immunotoxins. Br. J. Haematol.. 70: 289-294. 1988.

44. Engert. A.. Brown. A., and Thorpe. P. Resistance of myeloid leukaemia cell lines toricin A-chain immunoioxins. Leukemia Res.. /5: 1079-1086. 1991.

45. Press. O. W., Vitella. E. S., Farr, A. G., Hansen. J. A., and Martin. P. J. Evaluation ofricin A-chain immunotoxins directed against human T cells. Cell. Immunol.. 102:10-20. 1986.

46. Calafat. J.. Molthoff. C.. Janssen. H.. and Hilkens. J. Endocytosis and intracellularrouting of an antibody-ricin A chain conjugale. Cancer Res., 48: 3822-3827, 1988.

47. Le Doussal. J. M.. Gautherot. E.. Martin. M.. Barbet. J.. and Delaage. M. Enhancedin vivo targeting of an asymmetric bivalent hapten to double-antigen-positive mouseB cells with monoclonal antibody conjugate cocktails. J. Immunol., 146: 169-175,1991.

3021

on June 9, 2017. © 1993 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 8: Delivery of Saporin to Human B-Cell Lymphoma …...field (5), recent trials in Hodgkin's (6) and non-Hodgkin's lymphoma (7, 8) have been encouraging and suggest that immunotoxin may

1993;53:3015-3021. Cancer Res   M. Antonietta Bonardi, R. R. French, P. Amlot, et al.   Immunoglobulin Results in Efficient KillingBispecific Antibody: Targeting via CD22 but not CD19, CD37, or Delivery of Saporin to Human B-Cell Lymphoma Using

  Updated version

  http://cancerres.aacrjournals.org/content/53/13/3015

Access the most recent version of this article at:

   

   

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  [email protected] at

To request permission to re-use all or part of this article, contact the AACR Publications

on June 9, 2017. © 1993 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from


Recommended