17.05.2014 Views

Activation of Human Effector Cells by a Tumor Reactive ...

Activation of Human Effector Cells by a Tumor Reactive ...

Activation of Human Effector Cells by a Tumor Reactive ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

Vol. 2, 1951-1959. December 1996 Clinical Cancer Research 1951<br />

<strong>Activation</strong> <strong>of</strong> <strong>Human</strong> <strong>Effector</strong> <strong>Cells</strong> <strong>by</strong> a <strong>Tumor</strong> <strong>Reactive</strong><br />

Recombinant Anti-Ganglioside GD2 Interleukin-2<br />

Fusion Protein (chl4.18-1L2)’<br />

Jacquelyn A. Hank,2 Jean E. Surfus, Jacek Gan,<br />

Peter Jaeger, Stephen D. Gillies,<br />

Ralph A. Reisfeld, and Paul M. Sondel<br />

Department <strong>of</strong> <strong>Human</strong> Oncology. University <strong>of</strong> Wisconsin-Madison,<br />

Madison. Wisconsin 53792 Ii. A. H.. J. E. S., J. G.. P. J., P. M. 5.1:<br />

Fuji ImmunoPharmaceuticals Corporation. Lexington. Massachusetts<br />

02173 15. D. G.l: The Scripps Research Institute. La Jolla. Califrrnia<br />

92037 iR. A. RI: and Departments <strong>of</strong> Pediatrics and Medical<br />

Genetics, University <strong>of</strong> Wisconsin-Madison, Madison, Wisconsin<br />

53792 P. M. 5.1<br />

ABSTRACT<br />

Cytotoxic effector cells interact with target cells<br />

through various mechanisms. CTLs use the antigen-specific<br />

T cell receptor, whereas Fe receptor-positive natural killer<br />

cells use this receptor to interact with antibody-coated target<br />

cells. We evaluated the tumor-binding and lymphocyte-activating<br />

capability <strong>of</strong> a recombinant fusion protein consisting<br />

<strong>of</strong> a tumor-selective human/mouse chimeric anti-ganglioside<br />

GD2 antibody (chl4.18) and recombinant human<br />

interleukin-2 (1L2) (chl4.18-IL2). This fusion protein bound<br />

specifically to GD2-positive melanoma and neuroblastoma<br />

tumor cell lines, and its 1L2 component stimulated in vitro<br />

proliferation <strong>of</strong> an IL2-dependent cell line, as well as peripheral<br />

blood mononuclear cells, in healthy control individuals<br />

and in cancer patients receiving continuous infusion <strong>of</strong> 1L2.<br />

The 1L2 presented <strong>by</strong> the fusion protein, when bound to<br />

tumor cells, induced proliferation <strong>of</strong> IL2-responsive cells as<br />

well as a comparable amount <strong>of</strong> soluble 1L2 did. This suggests<br />

that localization <strong>of</strong> 1L2 at the site <strong>of</strong> contact between<br />

tumor and effector cells is an effective way <strong>of</strong> presenting this<br />

cytokine to 1L2-responsive cells. The chl4.18-1L2 fusion<br />

protein also mediated antibody-dependent cellular cytotoxicity<br />

with Fc receptor-positive effector cells to an extent<br />

similar to chl4.18. These results, together with those <strong>of</strong><br />

previous studies documenting antitumor efficacy against human<br />

tumor xenografts in SCID mice and GD2-positive murine<br />

tumors in immunocompetent syngeneic mice, suggest<br />

that the chl4.18-1L2 fusion protein should be tested in Phase<br />

I and II trials in patients with GD2-positive tumors.<br />

INTRODUCTION<br />

Immunotherapy with IL23 is <strong>of</strong> benefit to some patients<br />

with renal cell cancer and melanoma ( I ). Although multiple<br />

immune mechanisms are activated in patients receiving IL2, the<br />

immune components induced <strong>by</strong> lL2 necessary for antitumor<br />

activity have not been identified. Continuous infusion <strong>of</strong> IL2. at<br />

doses tolerated in the outpatient setting. induces systemic<br />

lymphoid activation in virtually all treated patients: however,<br />

only a minority <strong>of</strong> such patients achieve antitumor responses<br />

( 1-4). Included in this lymphoid cell activation is an expansion<br />

and activation <strong>of</strong> the CDl6, FcR NK cells (5. 6). Currently.<br />

attempts are being made clinically to target these cells to tumor<br />

through the use <strong>of</strong> tumor-specific mAbs (7. 8). These include<br />

clinical evaluation <strong>of</strong> regimens combining effector cell activation<br />

through 1L2 infusions with infusions <strong>of</strong> munine or chimeric<br />

tumor-selective mAb, such as the l4.G2a or chl4. 18 antibodies,<br />

which recognize the ganglioside GD2 expressed on neuroblastoma.<br />

melanoma, and certain other tumors (7).<br />

In an effort to augment the stimulation <strong>of</strong> the FcR antibody-directed”<br />

effector cells and activate FcR effector cells<br />

that express IL2 receptors (2, 9), a chl4.l8-IL2 fusion protein<br />

has been constructed <strong>by</strong> fusion <strong>of</strong> a synthetic sequence coding<br />

for human IL2 to the carboxyl end <strong>of</strong> the C-yl gene <strong>of</strong> the mAb<br />

ch I 4. 1 8 ( 1 0). When the antitumor variable region <strong>of</strong> this fusion<br />

protein binds to tumor, IL2 should be concentrated in the tumor<br />

microenvironment and provide activation <strong>of</strong> FcR effectors,<br />

such as NK cells, that did bind to the immunoglobulin Fe<br />

domain <strong>of</strong> the tumor-bound fusion protein. Furthermore, the IL2<br />

component <strong>of</strong> this tumor-bound fusion protein may activate<br />

IL2-responsive cells. such as cytotoxic T cells and helper T<br />

cells, that do not necessarily have FcRs and the subpopulation <strong>of</strong><br />

NK cells that express IL2 receptors but lack the FcR (9), there<strong>by</strong><br />

recruiting additional effector cells into the tumor microenvironment.<br />

The chI4.l8-1L2 fusion protein was previously shown to<br />

maintain antigen-binding activity and 1L2 activity ( 10) and has<br />

provided antitumor effects in SCID mice bearing human tumor<br />

xenografts <strong>of</strong> neuroblastoma ( I I ) and melanoma ( I 2), as well as<br />

in a syngeneic murine melanoma model ( 13). We have extended<br />

these findings <strong>by</strong> demonstrating that chl4. l8-IL2 bound to<br />

GD2 tumor cells can be visualized <strong>by</strong> flow cytornetry, detecting<br />

either the chl4.l8 or the 1L2 portion <strong>of</strong> this construct. The<br />

IL2 component <strong>of</strong> the fusion protein is able to stimulate lL2-<br />

dependent cells. The fusion protein bound to GD2 tumors<br />

Received 10/24/95; revised 8/27/96: accepted 9/18/96.<br />

‘ This research was supported <strong>by</strong> NIH grants and contracts CA-5344 I.<br />

CA-05436. CA-32685, CM-87290, CA- I 4520, CA- I 3539. CM-47669,<br />

HL-02I43. RR-03186, and American Cancer Society Grant CH-237.<br />

2 To whom requests for reprints should be addressed, at K4/454 CSC.<br />

6()0 Highland Avenue, Madison. WI 53792. Phone: (608) 263-7262:<br />

Fax: (608) 263-4226.<br />

3 The abbreviations used are: lL2. interleukin 2: NK. natural killer: FcR,<br />

Fe receptor: mAb. monoclonal antibody: ADCC. antibody-dependent<br />

cellular cytotoxicity: HLR, H<strong>of</strong>fmann LaRoche; PBMC, peripheral<br />

blood mononuclear cell: LAK. lymphokine-activated killer.


1952 Anti-GD2-IL2 Fusion Protein<br />

stimulates proliferation to the same extent as soluble fusion<br />

protein or soluble 1L2. The chl4.l8-1L2 fusion protein also<br />

stimulates cytolytic activity in vitro <strong>by</strong> cells able to mediate<br />

ADCC. FcR NK cells obtained from melanoma patients in<br />

vivo following therapy with continuous infusion 1L2 are targeted<br />

to GD2 tumors that have bound chl4.l8-IL2 in vitro, and the<br />

lytic activity was comparable to that induced <strong>by</strong> the combination<br />

<strong>of</strong> free antibody and soluble 1L2.<br />

MATERIALS AND METHODS<br />

Recombinant IL2. HLR IL2 was provided through the<br />

Cancer Treatment and Evaluation Program <strong>of</strong> the National Cancer<br />

Institute. The National Cancer Institute-Biological Response<br />

Modifiers program standard for unit dosage was used, and the<br />

specific activity <strong>of</strong> the IL2 was 15 X 106 units/mg. This unit<br />

corresponds closely with the international standard for IL2<br />

unitage (14).<br />

Chimenc Antibody and Fusion Protein. The mouse/<br />

human chimeric 14.18 antibody was constructed <strong>by</strong> combining<br />

the variable regions <strong>of</strong> the murine anti-GD2 14.18 antibody with<br />

the constant regions <strong>of</strong> human IgG1 heavy chain and K light<br />

chain (15, 16). The 14.l8-IL2 fusion protein was constructed <strong>by</strong><br />

fusion <strong>of</strong> a synthetic sequence coding for human IL2 to the<br />

carboxyl end <strong>of</strong> the human C-yl gene <strong>of</strong> the mAb chl4. 18 (10).<br />

The fused gene was inserted into the vector pdHL2-l4. 18 as<br />

described previously (15). Transfection <strong>of</strong> the expression plasmids<br />

in Sp2/0-Agl4 cells and propagation <strong>of</strong> the clones secreting<br />

chl4. 18-1L2, as well as its purification, have been described<br />

previously (10). To compare the 1L2 activity in the soluble IL2<br />

preparation and in the fusion protein, concentrations were based<br />

on weight/volume calculations <strong>of</strong> 1L2 in the two preparations.<br />

Because the fusion protein molecule consists <strong>of</strong> 80% chimeric<br />

14.18 immunoglobulin and 20% 1L2 <strong>by</strong> molecular weight, the<br />

fusion protein 1L2 concentration was based on IL2 comprising<br />

20% <strong>of</strong> the weight <strong>of</strong> the fusion protein. Thus, 50 ng/rnl <strong>of</strong><br />

fusion protein would correspond to 10 ng/ml <strong>of</strong> IL2 in the fusion<br />

protein. Because 10 ng/ml <strong>of</strong> soluble IL2 corresponds to 150<br />

units/mi <strong>of</strong> soluble HLR 1L2, we have used this conversion to<br />

describe the units <strong>of</strong> 1L2 anticipated for the fusion protein<br />

preparation based on the molecular weight <strong>of</strong> IL2 and using the<br />

specific activity <strong>of</strong> 15 X 106 units/mg for the HLR IL2.<br />

<strong>Tumor</strong> Cell Lines. The GD2-positive LA-N-S neuroblastoma<br />

target cell line, kindly provided <strong>by</strong> R. Seeger (Children’s<br />

Hospital <strong>of</strong> Los Angeles, Los Angeles, CA), was maintamed<br />

as an adherent monolayer in Leibovitz’ s medium<br />

supplemented with 15% heat-inactivated fetal bovine serum. A<br />

trypsin-EDTA solution was used to harvest the cell monolayer.<br />

The M2l human melanoma line (GD2’) was described previously<br />

(15), and the BT-20 human breast carcinoma cell line<br />

(GD2) was obtained from American Type Culture Collection.<br />

Both cell lines were maintained as adherent monolayers in<br />

RPM! 1640 supplemented with penicillin and streptomycin<br />

(P/S), L-glutamine, HEPES buffer, and 10% fetal bovine serum.<br />

Flow Cytometry. Cell-bound fusion protein and antibody<br />

were detected <strong>by</strong> standard indirect immun<strong>of</strong>luorescence<br />

methods (Becton Dickinson, San Jose, CA). Antibodies ineluded<br />

a goat antihuman IgG (Caltag, San Francisco, CA) and a<br />

rabbit antihuman IL2 (Genzyme, Cambridge, MA).<br />

Proliferation Assays. Fresh PBMCs from healthy volunteer<br />

human donors or from patients who were treated with a<br />

96-h continuous infusion <strong>of</strong> IL2 (3. 4, 17) were cultured in<br />

0.2-ml round-bottom microplates at a concentration <strong>of</strong> 1 X l0<br />

cells/well in RPMI 1640 supplemented with 10% human serum<br />

(Pel-Freez, Rogers, AR), 25 msi HEPES, 100 units/mI penicillin,<br />

and 100 p.g/ml streptomycin sulfate (RPMI-HS). Recombinant<br />

IL2 and fusion protein were added at concentrations as<br />

indicated in the “Results”. Concentrations <strong>of</strong> recombinant soluble<br />

1L2 were also tested, which corresponded to the concentration<br />

<strong>of</strong> 1L2 in the fusion protein preparation based on the<br />

molecular weight and concentration <strong>of</strong> fusion protein: I i.g <strong>of</strong><br />

the fusion protein contains approximately 3000 units <strong>of</strong> IL2<br />

(I 1). In experiments in which chl4. I 8 or fusion protein-coated<br />

M21 and LA-N-S cells were used as a proliferative stimulus, the<br />

antibody or fusion protein at S ig/ml was incubated with the<br />

tumor cells for 1 h on ice. Irradiation <strong>of</strong> these cells took place<br />

during this incubation, with LA-N-S and M2l receiving 10,000<br />

and 40,000 rads, respectively. Cultures were incubated at 37#{176}C<br />

in 5% CO2 for 48-72 h, pulsed with I pCi [3H]thymidine for<br />

18 h, and harvested with a Filterrnate 196 Packard harvester, and<br />

[3H]thymidine incorporation was quantitated with a Matrix<br />

9600 direct 13 counter with a 5-mm counting period. Informed<br />

consent forms, approved <strong>by</strong> the University <strong>of</strong> Wisconsin <strong>Human</strong><br />

Subjects Committee, were obtained prior to collection <strong>of</strong> all<br />

human blood specimens.<br />

For some proliferative studies, the 1L2-responsive cells<br />

used were the Tf-l myeloid leukemia cell line transfected with<br />

the gene for the 1L2 receptor 3 chain. This transfected line was<br />

designated Tf-l , and the mock-transfected control line contaming<br />

the LXSN vector but no IL2R3 gene was designated<br />

Tf-IL (18). This Tf-l3 cell line responded to 1L2 using intermediate<br />

affinity receptor complexes ( 1 7. I 8) and thus is<br />

analogous to the majority <strong>of</strong> NK cells in 1L2-treated patients,<br />

which also use intermediate affinity IL2 receptors (2). The Mik<br />

11 monoclonal antibody directed against the p70 IL-2 receptor<br />

1 chain was used in the blocking studies ( 19. 20).<br />

ADCC and Fusion Protein-mediated Cellular Cytotoxicity.<br />

All ADCC assays were performed in RPMI-HS. <strong>Effector</strong><br />

cells in a total volume <strong>of</strong> SO pi were plated in quadruplicate<br />

into 96-well U-bottomed microtiter plates at the indicated effector/target<br />

ratios. Just prior to the addition <strong>of</strong> target cells, SO<br />

il <strong>of</strong> antibody, antibody plus 1L2, or fusion protein were added<br />

to the effectors. While the effectors were being prepared, target<br />

cells were labeled for 2 h with 250 pCi <strong>of</strong> 5tCr in 0.2 ml <strong>of</strong><br />

RPMI-HS. Target cells were mixed every 15-30 mm during<br />

labeling to keep the cells in suspension. After being washed<br />

twice with RPMI, S x l0 target cells in 50 il <strong>of</strong> RPMI-HS<br />

were added to effector cells and centrifuged at 200 X g for S<br />

mm. In the experiments using fusion protein-coated target cells.<br />

chl4.l8-1L2 was added to the targets following one wash and<br />

incubated on ice for 1 h. Two subsequent washes removed<br />

excess fusion protein and 51Cr. The effector cells were also<br />

plated in medium and in IL2 to determine their ability to<br />

mediate lysis <strong>of</strong> target cells in the absence <strong>of</strong> antibody or fusion<br />

protein. The plates were incubated at 37#{176}Cat 5% CO2 for 4 h,<br />

and the supernatants were harvested using the Skatron Harvesting<br />

System (Skatron, McLean, VA). Maximum 5tCr release was<br />

measured <strong>by</strong> lysing target cells with the detergent cetrimide


Clinical Cancer Research 1953<br />

M21<br />

LAN-5<br />

BT-20<br />

chl4.18<br />

chl4.18-1L2<br />

JL<br />

Rabbit<br />

IgG<br />

Rabbit<br />

Anti-1L2<br />

Rabbit<br />

lgG<br />

Rabbit<br />

Anti-1L2<br />

. . . .-<br />

1u<br />

Fig. I Detection <strong>of</strong> antibody component and 1L2 component in tumor-bound fusion protein. The GD2 M2 1 melanoma and LA-N-S neuroblastoma<br />

and GD2 BT-20 breast carcinoma cell lines were each coated with I p.g <strong>of</strong> either chl4.18-1L2 fusion protein or chl4.18 antibody. To detect the<br />

antibody component (top panels). a fluorescein-conjugated goat antihuman IgG secondary antibody was used. To detect the 1L2 component (bottom<br />

panels). a rabbit anti-IL2 antibody was used, followed <strong>by</strong> a phycoerythrin-conjugated goat antirabbit antibody.<br />

(Sigma). Spontaneous 5tCr release was measured <strong>by</strong> incubating<br />

target cells in RPMI-HS alone. Percent cytotoxicity values were<br />

calculated for each effector/target ratio as follows:<br />

C/<br />

cytotoxicity<br />

experimental release - spontaneous release<br />

=l00X-- .<br />

maximum release - spontaneous release<br />

Results are expressed as percent cytotoxicity or as lytic<br />

units, where 1 lytic unit is the number <strong>of</strong> effector cells necessary<br />

to achieve 20% lysis <strong>of</strong> S X I0 targets.<br />

RESULTS<br />

Fusion Protein chl4.18-1L2 Binds to GD2 <strong>Tumor</strong><br />

<strong>Cells</strong>. The data shown in Fig. I demonstrate the binding <strong>of</strong><br />

chl4.l8-IL2 to the GD2 M2l human melanoma and LA-N-S<br />

human neuroblastoma cell lines. The fusion protein binds to the<br />

tumor cells with a fluorescence intensity similar to that <strong>of</strong> the<br />

parental ch I 4. 18 antibody (Fig. 1, top panels). The specificity <strong>of</strong><br />

ch I 4. 18-IL2 and ch I 4. 18 antibody is demonstrated <strong>by</strong> their lack<br />

<strong>of</strong> binding to the GD2 BT-20 human breast carcinoma cell<br />

line. The IL2 component <strong>of</strong> ch 14. 1 8-1L2 can be detected <strong>by</strong><br />

rabbit anti-IL2 antibody when the fusion protein is bound to<br />

M-2l and LA-N-S cells, respectively (Fig. I. bottom pane/c).<br />

These data document that the IL2 component <strong>of</strong> the fusion<br />

protein remains associated with the tumor cells that bind<br />

chl4.l8-1L2 on their surfaces and is detectable <strong>by</strong> anti-IL2<br />

antibody. Furthermore, separate flow cytometric analyses have<br />

shown that M-2l and LA-N-S cells do not express either the a<br />

or 3 chains <strong>of</strong> the 1L2 receptor (data not shown). Thus, binding<br />

<strong>of</strong> the fusion protein to these cells is mediated through the<br />

chl4.18 rather than the IL2 component <strong>of</strong> the fusion protein.<br />

Soluble chl4.18-1L2 Stimulates IL2-induced Proliferative<br />

Responses. Fig. 2 documents proliferative responses to<br />

1L2 attained with the Tf-I3 cell line and PBMCs from a melanoma<br />

patient obtained 24 h after a 96 h continuous iv. infusion<br />

<strong>of</strong> IL2. Tf-l is a GM-CSF-dependent myeloid leukemia-derived<br />

cell line that constitutively expresses the common cytokine<br />

receptor -y chain. A variant cell line bearing functional intermediate<br />

affinity IL2 receptors (3-y dimers), designated Tf- I , was<br />

obtained <strong>by</strong> stable transfection <strong>of</strong> Tf- I with eDNA encoding the<br />

1 chain <strong>of</strong> the IL2 receptor ( 1 8). The Tf- 1 3 cell line that<br />

retained responsiveness to GM-CSF also responded to IL2 in a<br />

dose-dependent fashion. The mock-transfected Tf-lL cell line<br />

did not proliferate in response to 1L2, and the Tf-l3 cell line did<br />

not respond to chl4.l8 antibody (data not shown). The results <strong>of</strong><br />

proliferative assays shown in Fig. 2 were obtained <strong>by</strong> using<br />

dilutions <strong>of</strong> IL2 and chl4.l8-1L2 to achieve equivalent molar


1954 Anti-GD2-IL2 Fusion Protein<br />

Cl)<br />

I-<br />

z<br />

0<br />

C)<br />

1200<br />

1000<br />

10000<br />

600<br />

400<br />

200<br />

8000<br />

6000<br />

4000<br />

2000<br />

0<br />

0 -<br />

400<br />

L2 UImI 1.6 3.0 00 12 25 50 100 200<br />

FP JmI.B2O 1.20 2.0 5.0 10 20 40 80 ISO<br />

Fig. 2 Proliferation induced <strong>by</strong> soluble IL2 and IL2 within the soluble<br />

fusion protein. The Tf-l3 cell line (A) and PBMCs obtained from a<br />

patient following continuous infusion lL2 (B) were stimulated with<br />

increasing concentrations <strong>of</strong> soluble IL2 (A) and soluble chl4.l8-1L2<br />

fusion protein (s). Dilutions <strong>of</strong> IL2 and fusion protein were made to<br />

achieve equivalent molar concentrations <strong>of</strong> IL2. The counts were determined<br />

for [3H]TdR incorporation <strong>by</strong> proliferating cells.<br />

concentrations <strong>of</strong> IL2. Assuming the same specific activity <strong>of</strong><br />

1L2 within the fusion protein as the recombinant HLR product,<br />

there are approximately 400 units/ml <strong>of</strong> 1L2 contained within<br />

the chl4.18-IL2 when it is applied at a concentration <strong>of</strong> 160<br />

ng/ml. When these data are expressed based on the concentration<br />

<strong>of</strong> 1L2, dose-response curves indicate that at concentrations <strong>of</strong><br />

1L2 less than or equal to SO units/rn], the fusion protein is slightly<br />

more efficient than free 1L2 at stimulating the Tf-l(3 cell line to<br />

proliferate. This finding was reproduced in two additional proliferative<br />

assays using the Tf- 13 cell line as the responding cell.<br />

PBMCs obtained from six patients with melanoma who<br />

had just completed a continuous infusion <strong>of</strong> IL2 were also tested<br />

for their responsiveness to IL2 and chl4.l8-IL2. Results from<br />

one representative patient are shown at the bottom <strong>of</strong> Fig. 2.<br />

Previous studies have shown that NK cells obtained following in<br />

vivo IL2 treatment show rapid proliferative responses to restimulation<br />

with IL2 in vitro, using primarily the intermediate<br />

affinity 13’y receptor complex (2, 21). Similar proliferation <strong>by</strong><br />

these cells was observed at most concentrations <strong>of</strong> IL2 and<br />

chl4.18-IL2 tested. These data indicate that the antibody component<br />

in the fusion protein did not adversely affect the ability<br />

<strong>of</strong> IL2 to interact with the IL2R complexes either on the Tf-l 3<br />

cells or on the PBMCs <strong>of</strong> a melanoma patients treated with IL2,<br />

nor did it affect the ability <strong>of</strong> the 1L2 component <strong>of</strong> the fusion<br />

protein to stimulate proliferation. chl4. I 8 antibody alone, at<br />

comparable concentrations, did not stimulate patient PBMCs to<br />

proliferate (data not shown). Table 1 presents data from two<br />

separate experiments indicating the in vitro proliferative response<br />

induced <strong>by</strong> 100 units <strong>of</strong> soluble IL2 and <strong>by</strong> a similar<br />

amount <strong>of</strong> IL2 contained within 40 ng <strong>of</strong> chl4.18-IL2. The<br />

responding cells were PBMCs obtained from five melanoma<br />

patients and PBMCs from one healthy control individual. The<br />

patient PBMCs were obtained 24 h after completion <strong>of</strong> a 96-h<br />

continuous infusion <strong>of</strong> IL2 in vivo. In these experiments, the<br />

fusion protein stimulated proliferation that was similar to that<br />

induced <strong>by</strong> the soluble 1L2 for patient I and for the control<br />

donor in experiment 1 and for the three patients in experiment 2.<br />

The chimeric chl4.l8 antibody itself was not stimulatory.<br />

Fusion Protein-induced Proliferation Mediated through<br />

an Intermediate Affinity #{176}y 1L2 Receptor Is Specifically<br />

Blocked <strong>by</strong> Antibody to the Chain <strong>of</strong> the IL2 Receptor.<br />

Fig. 3 demonstrates the proliferation induced <strong>by</strong> IL2 and fusion<br />

protein on cells with intermediate affinity receptors for 1L2,<br />

Tf-l 3, and PBMCs obtained from cancer patients following a<br />

4-day continuous infusion <strong>of</strong> 1L2. <strong>Cells</strong> with high-affinity aj<br />

IL2 receptors (Kit 225 cells) were also assayed. Tf- 1 3 and<br />

patient PBMCs responded to 1L2 and the fusion protein in a<br />

Table I Proliferative response to soluble IL2 and to chl4.l8-1L2 fusion protein<br />

PBMCs from a healthy volunteer individual (control) and from two patients were assayed in experiment 1. and PBMCs from three patients were<br />

assayed in experiment 2. The patient PBMCs were obtained 24 h after a 96-h continuous infusion <strong>of</strong> IL2. They were stimulated with IL2 or<br />

chl4.l8-IL2 fusion protein at concentrations containing equivalent amounts <strong>of</strong> IL2 or with an excess <strong>of</strong> chl4.I8 antibody. The cells were cultured<br />

for 4 days at 37#{176}C in 5% CO,. pulsed with 3H-thymidine for 16 h, and harvested, and counts per 5 mm were obtained with a Packard 9600 Matrix<br />

counter.<br />

3H-Thymidine incorporati on (count per 5 mm)<br />

Experiment<br />

I<br />

Control<br />

Patient 1<br />

Patient 2<br />

II Patient 16<br />

Patient 17<br />

Patient 20<br />

Medium 100 units/mI IL2 40 ng/ml chl4.18-1L2 O.5g/ml chl4.18<br />

197<br />

160<br />

184<br />

165<br />

49<br />

31<br />

14,194<br />

9,741<br />

24,381<br />

9,959<br />

2,321<br />

4,135<br />

13.387<br />

7,916<br />

7,413<br />

11,201<br />

2,700<br />

6,317<br />

190<br />

161<br />

207<br />

59<br />

46<br />

43


Clinical Cancer Research 1955<br />

120000 120000<br />

90000 90000<br />

L1<br />

I-<br />

60000 60000<br />

30000 30000<br />

0 - 0<br />

200 50 12.5 66.5 16.6 4.1<br />

16000 16000<br />

C-)<br />

0.<br />

I-<br />

a-<br />

12000<br />

8000<br />

4000<br />

0<br />

200<br />

50<br />

I 2000<br />

8000<br />

Fig. 3 Antibody to the 3 chain <strong>of</strong> the IL2 receptor<br />

blocks fusion protein induced proliferation <strong>by</strong> cells<br />

expressing intermediate affinity, but not cells expressing<br />

high-affinity lL2 receptors. Tf-l cells.<br />

PBMCs, and KIT 225 cells were cultured with lL2<br />

(leftpa,iels) or the chI4.18-1L2 fusion protein (right<br />

panels). The humanized Mik 3l antibody at a final<br />

concentration <strong>of</strong> 3 p.g/ml (A) or human serum diluted<br />

66 5 16 6 4 1 1 1/50 as the control () was added at the initiation <strong>of</strong><br />

12.5 . . a 3-day I3HITdR incorporation assay. #{149}, medium.<br />

100000<br />

100000<br />

I0<br />

C.”<br />

75000<br />

75000<br />

I-<br />

50000<br />

50000<br />

25000<br />

25000<br />

0-<br />

200<br />

‘ . 0 ‘<br />

50 12.5 66.5 16.6 4.1 1<br />

IL-2 CONCENTRATION ( U/mi) 14.18-IL-2 CONCENTRATiON (ug/mi)<br />

dose-dependent manner, and this proliferation was abrogated <strong>by</strong><br />

inclusion <strong>of</strong> the Mik 3I antibody, which recognizes the 3 chain Coated<br />

<strong>of</strong> the IL2 receptor (20). Over the dose range examined, there <strong>Tumor</strong> <strong>Cells</strong><br />

was a strong<br />

. . #{149}500<br />

proliferative response to both soluble IL2 and 60000<br />

#{149} <br />

fusion protein <strong>by</strong> the Kit 225 cell line, which expresses the U)<br />

50<br />

high-affinity afty IL2 receptor. The Mik 3 1 antibody did not I-<br />

block this proliferative response, as shown previously for IL2 z<br />

40000<br />

(19).<br />

chl4.18-1L2 Bound to GD2 <strong>Cells</strong> Stimulates Proliferation.<br />

The flow cytometric studies (Fig. 1 ) document that the<br />

chl4.18-IL2 binds to GD2 tumor cells via the antibody vanable<br />

region and that its IL2 component is recognized <strong>by</strong> rabbit<br />

antihuman 1L2 antibody. Subsequent experiments tested the<br />

functional activity <strong>of</strong> the IL2 component <strong>of</strong> the fusion protein<br />

when bound to tumor cells. In this case, the M21 and LA-N-S<br />

cell lines were coated with either chl4.18-IL2 or chl4.l8 and<br />

then washed free <strong>of</strong> any excess and cultured with the Tf-l<br />

responding cell line or the Tf-lL control cell line. The data<br />

shown in Fig. 4 demonstrate that the ch 14. 18-IL2 fusion protein.<br />

when bound to a GD2 tumor cell line, effectively presents IL2<br />

to the Tf- 1 3 cell line. When ch I 4. 18-IL2 is bound to S X 102<br />

M21 or LA-N-S cells it is able to efficiently present the bound<br />

0<br />

C)<br />

80000<br />

20000<br />

chl4.18<br />

M21<br />

FP<br />

LIt<br />

chl4.18<br />

FP<br />

LA-N-5<br />

Fig. 4 Proliferation induced <strong>by</strong> the lL2 component <strong>of</strong> the tumor-bound<br />

fusion protein. The M2l melanoma and LA-N-S neuroblastoma cell<br />

lines were coated for I h at 4#{176}Cwith either chI4.18 antibody or the<br />

chl4.18-1L2 fusion protein (FP). Irradiation took place during this<br />

incubation. These coated tumor target cells were washed twice and<br />

diluted to 500, 100, or 50 cells per well and used to stimulate l0 Tf-l<br />

cells per well. Proliferation was quantitated after 72 h <strong>by</strong> I3HITdR<br />

incorporation.


1956 Anti-GD2-lL2 Fusion Protein<br />

Table 2 Stimulatory activity <strong>of</strong> IL2 in tumor-bound fusion protein<br />

The Tf-l3 and control Tf-lL cell lines (experiment 1) or Tf-l3 cells and PBMCs from IL2-treated patients (experiment 2) were stimulated in<br />

a 3-day proliferative assay with soluble IL2 or tumor cells coated with chI4.18 antibody or the chl4.18-IL2 fusion protein. <strong>Tumor</strong> cells (106) were<br />

incubated with S i.g <strong>of</strong> chl4.18, washed twice, and diluted. Sixty % <strong>of</strong> the fusion protein used to coat the M2l cell remained bound to the cells:<br />

therefore, 100 cells have 0.3 ng <strong>of</strong> fusion protein, corresponding to approximately 1 unit <strong>of</strong> bound lL2. The total volume <strong>of</strong> the microwell is 0.2 ml,<br />

resulting in a total <strong>of</strong> 5 units <strong>of</strong> IL2/ml in the wells with chl4.18-IL2-coated tumor cells. The cells were cultured for 2.5 days at 37#{176}Cin 5% CO2.<br />

pulsed with 3H-thymidine for 16 h, and then harvested, and counts per 5 mm were obtained with a Packard 9600 Matrix counter. The proliferation<br />

<strong>of</strong> Tf- 113 cells induced <strong>by</strong> 5 units/mI 1L2 as tumor-bound fusion protein was similar to the proliferation induced <strong>by</strong> 50 units/ml <strong>of</strong> soluble IL2. and<br />

the proliferation <strong>of</strong> patient PBMCs induced <strong>by</strong> 25 units/mI 1L2 as tumor-bound fusion protein was similar to the proliferation induced <strong>by</strong> SO units/mI<br />

<strong>of</strong> soluble IL2.<br />

3H-TdR incorporation (coun<br />

Is X I0), responding cell<br />

Experiment<br />

I<br />

Experiment 2<br />

Stimulus<br />

Medium<br />

GM-CSF (5 ng/ml)<br />

1L2 (100 units/mI)<br />

IL2 (50 units/mI)<br />

1L2 (25 units/mI)<br />

100 M21 cells labeled with 0.5 ng <strong>of</strong> chl4.18-1L2 fusion protein (5.0 units <strong>of</strong> 1L2/ml)<br />

500 M2l cells labeled with 0.5 ng <strong>of</strong> chl4.l8-1L2 fusion protein (25 units <strong>of</strong> IL2/ml)<br />

‘a NT, not tested.<br />

Tf-l 3 Tf- 1L<br />

6.7 10.8<br />

67.4 91.1<br />

64.2 13.9<br />

50.7 12.1<br />

41.9 12.8<br />

56.6 14.4<br />

70.8 13.4<br />

Tf- I 3<br />

Patient<br />

0.8 .03<br />

NT’<br />

NT<br />

18.6 4.1<br />

9.3 1.7<br />

3.2 0.6<br />

1.2 0.4<br />

4.2 2.1<br />

IL2. As few as SO such cells coated with the fusion protein can<br />

still produce a significant 1L2-specific proliferative response.<br />

The control Tf-lL cell line did not respond to tumor-bound<br />

fusion protein in these experiments (data not shown).<br />

Proliferative Response to “Equivalent” Doses <strong>of</strong> <strong>Tumor</strong>-bound<br />

Fusion Protein. In an attempt to determine<br />

whether there is a dose effect <strong>of</strong> the cell-bound fusion protein on<br />

the proliferative response, we compared the proliferation induced<br />

<strong>by</strong> soluble 1L2 to that induced <strong>by</strong> an equivalent amount <strong>of</strong><br />

ch I 4. 18-1L2 coated on tumor cells. To make dose comparisons,<br />

we determined the amount <strong>of</strong> fusion protein that remained<br />

bound to the coated GD2 tumor cells, based on the measurement<br />

<strong>of</strong> free fusion protein concentration in the supernatant<br />

following incubation. This indicated that approximately 60% <strong>of</strong><br />

the chl4.l8-IL2 used to coat the cells actually remained bound<br />

to the cells. Data shown in Table 2 are from an experiment in<br />

which 106 M2l cells were coated with S ag <strong>of</strong>chl4.18-1L2 and<br />

washed twice. These cells were then diluted, and 100 or 500<br />

fusion protein-coated M2 1 cells per well were used to stimulate<br />

Tf- I 13 cells. With 60% <strong>of</strong> fusion protein bound to the M2 I cells,<br />

this corresponds to a total <strong>of</strong> 0.3 ng <strong>of</strong> fusion protein or approximately<br />

1 unit <strong>of</strong> IL2 per 100 cells in the 0.2-ml microtiter well.<br />

This corresponds to 5.0 and 25 units <strong>of</strong> IL2/ml in the proliferative<br />

assay with 100 and 500 fusion protein-coated M21 cells,<br />

respectively. Two experiments are presented in Table 2.<br />

chl4.18-IL2 fusion protein-coated M2l melanoma cells stimulated<br />

proliferative responses <strong>by</strong> the Tf-l 3 cell line (experiments<br />

1 and 2) and patient PBMCs obtained following an in vivo<br />

infusion <strong>of</strong> IL2 (experiment 2). These results indicate that the<br />

IL2 component <strong>of</strong> the fusion protein that remains coated to M2l<br />

cells is presented in a conformation able to stimulate the IL-2<br />

receptor and show that the tumor-bound IL-2 is as stimulatory as<br />

soluble 1L2.<br />

chl4.18-1L2 Fusion Protein Is Capable <strong>of</strong> Eliciting<br />

ADCC <strong>by</strong> PBMCs on GD2 <strong>Tumor</strong> Targets. PBMCs from<br />

two melanoma patients receiving IL2 were assayed for lytic<br />

L<br />

Y<br />

I<br />

C<br />

U<br />

N<br />

T<br />

S<br />

400.<br />

300.<br />

200.<br />

oo<br />

200.<br />

150.<br />

100.<br />

50.<br />

LAN 5m4 18<br />

LAN-5<br />

<br />

+112 FP<br />

c81418 +11.2<br />

cI,14.1O<br />

12<br />

chI4.18<br />

IL.2 (units) 2.5 25 ‘ 100<br />

FP (ng) I 10 40<br />

Fr<br />

Patient 1<br />

Patient 2<br />

Control<br />

M2i<br />

M24.18+1U<br />

±14 10 +112<br />

414.18<br />

18<br />

Fp<br />

11.2<br />

112 Fr<br />

2.5 25 100<br />

I 10 40<br />

Fig. 5 Soluble fusion protein facilitated ADCC on GD2 tumor cells.<br />

PBMCs obtained from two patients following a 96-h continuous infusion <strong>of</strong><br />

1L2 and from a volunteer control donor were the effector cells in a 4-h 51Cr<br />

relea.se assay. The GD2 LA-N-S and M21 cell lines were used as targets.<br />

The assay was performed in medium supplemented with 1L2 alone at 2.5.<br />

25, or 100 units/mI; with chl4.18-IL2 fusion protein (FP) alone at 1, 10, or<br />

40 ng/ml (corresponding to 2.5, 25, and 100 units <strong>of</strong> 1L2/ml): with the<br />

chI4.l8 antibody alone at 1, 10, or 40 ng/ml: or with a combination <strong>of</strong> 1L2<br />

and chl4.18 antibody at the same concentrations.


Clinical Cancer Research 1957<br />

300<br />

250<br />

M21<br />

A<br />

600 <br />

S<br />

800<br />

600<br />

8 igG<br />

200<br />

150<br />

0 IgG+iL2<br />

iichl4.18<br />

Oichl4.18+1L2<br />

U FP<br />

U)<br />

400<br />

z<br />

2000 200<br />

:<br />

, 400<br />

100<br />

. 0<br />

(I)<br />

50<br />

0<br />

#{163}<br />

,Ju1,J<br />

chl4.18 FP chl4.18 FP<br />

M21<br />

LA-N-5<br />

600<br />

_I<br />

5oo<br />

400<br />

LA-N-5<br />

B Fig. 7 ADCC <strong>of</strong>GD2 M21 and LA-N-S cells was facilitated <strong>by</strong> either<br />

chl4.l8 antibody or chl4.l8-IL2 fusion protein. Six separate 51Cr<br />

release cytotoxic assays using PBMCs obtained from nine patients<br />

receiving continuous infusion IL2 are included. Three <strong>of</strong>the assays used<br />

target cells coated with the chl4.18 antibody and fusion protein (-)<br />

and 3 assays used the antibody and fusion protein in soluble form<br />

( ). For each experiment, the paired comparisons <strong>of</strong> ADCC <strong>by</strong> the<br />

chl4.18 or the fusion protein are connected <strong>by</strong> the line.<br />

300<br />

200<br />

100<br />

0<br />

Patient 6 Patient 7 PatIent 20 Patient 17<br />

Fig. 6 <strong>Tumor</strong>-bound fusion protein facilitated ADCC. Patient PBMCs<br />

obtained from four patients following a 96-h continuous infusion <strong>of</strong> 1L2<br />

were cryopreserved and thawed on the morning <strong>of</strong> the 4 h 51Cr release<br />

assay. The GD2 M2l (A) and LA-N-S (B) targets were coated with the<br />

chl4.18, control antibody (IgG), or the chl4.l8-IL2 fusion protein (FP)<br />

during the 51Cr labeling. For the chl4.18 antibody-coated targets. soluble<br />

1L2 was added at l()O units/mI during the 4-h assay. Targets coated<br />

with the chl4.l8-IL2 fusion protein received no additional soluble 1L2.<br />

activity against the GD2 LA-N-S and M2l cell lines (Fig. 5).<br />

The fusion protein added in soluble form during the 4-h 51Cr<br />

release assay enhanced lysis mediated <strong>by</strong> both patients PBMCs.<br />

The ch 14. 18-IL2 fusion protein mediated levels <strong>of</strong> ADCC similar<br />

to those mediated <strong>by</strong> ch I 4. 18 alone or antibody combined<br />

with soluble IL2 when tested on the NK-resistant LA-N-S target<br />

cells. Against the M2 I targets, the fusion protein also enhanced<br />

lysis over that achieved with 1L2 alone; however, the fusion<br />

protein was not as effective as antibody combined with soluble<br />

IL2. Similar effects were observed with PBMCs from a healthy<br />

volunteer donor that were not previously primed in vito with<br />

IL2.<br />

ADCC with <strong>Tumor</strong> Target <strong>Cells</strong> Coated with chl4.18-<br />

1L2 Fusion Protein. PBMCs obtained from four melanoma<br />

patients 24 h after a 96-h continuous infusion <strong>of</strong> IL2 were<br />

cryopreserved and thawed the day <strong>of</strong> the assay for use as<br />

effector cells. The GD2 tumor cell targets were coated with<br />

either chl4.l8-IL2, chI4.l8, or control lgG immediately following<br />

the 5tCr labeling. Soluble IL2 (100 units/ml) was added<br />

to the medium during the 4-h assay with control IgG or mAb<br />

chl4.l8. <strong>Tumor</strong>-bound fusion protein elicited ADCC with both<br />

M21 and LA-N-S targets (Fig. 6, A and B). The ADCC against<br />

M2 1 target cells was comparable to that induced <strong>by</strong> mAb<br />

chl4.l8 alone. The level <strong>of</strong> cytotoxicity against the LA-N-S<br />

target cell was comparable to the augmented cytotoxicity observed<br />

when soluble IL2 was combined with chl4.l8. This<br />

similar pattern <strong>of</strong> lysis was also noted with the fusion protein<br />

added in soluble form (Fig. 4). These results suggest that for the<br />

LA-N-S neuroblastoma target, fusion protein is capable <strong>of</strong> augmenting<br />

the level <strong>of</strong> ADCC observed with chl4.l8 alone.<br />

Fig. 7 shows the results from six separate experiments<br />

using PBMC samples from nine different cancer patients comparing<br />

the cytotoxicity on both M2l melanoma and LA-N-S<br />

neuroblastoma targets. In three <strong>of</strong> the experiments, ch 14.18<br />

antibody or fusion protein was added in soluble form (as in Fig.<br />

5). and in three experiments the target cells were coated with the<br />

ch 14. 18 antibody or the fusion protein and washed free <strong>of</strong> any<br />

excess prior to the cytotoxic assay (as in Fig. 6). No reproducible<br />

difference in ADCC was noted between coated targets or<br />

targets to which soluble fusion protein was added. Overall, the<br />

data presented in Fig. 7 show that the ADCC mediated <strong>by</strong> the<br />

ch I 4. 1 8 fusion protein was comparable to or better than that<br />

seen with an equivalent concentration <strong>of</strong> chl4.l8 antibody<br />

against both targets.<br />

DISCUSSION<br />

The fusion protein chl4. 18-1L2 was designed to create a<br />

molecule that would achieve enhanced in vivo effects over the<br />

combined use <strong>of</strong> the antibody and IL2 as separate molecules ( 10,<br />

1 1 ). The function <strong>of</strong> the antibody component <strong>of</strong> this fusion<br />

protein is to facilitate ADCC and to target the cytokine IL2 to<br />

the area <strong>of</strong> GD2 tumors. Thus, chl4.l8-IL2 is a single molecule<br />

containing both antitumor specificity and immunopotenti-


1958 Anti-GD2-lL2 Fusion Protein<br />

ating capabilities. The results <strong>of</strong> in vitro experiments presented<br />

here confirm and extend previous reports demonstrating that the<br />

anti-GD2 cytokine fusion protein chl4.l8-IL2 retains the functional<br />

activity <strong>of</strong> both the antibody and IL2 (10). The fusion<br />

protein bound specifically to GD2 tumor cell lines, resulting in<br />

the same fluorescence intensity as the parent chl4. 1 8 antibody<br />

(Fig. 1 and Ref. 10). In addition, we demonstrated that tumor<br />

cell-bound fusion protein could be detected with antibody specific<br />

for human IgG or IL2.<br />

A previous study using murine CTLL-2 cells as responders<br />

(10) demonstrated that the IL2 component <strong>of</strong> the chl4.l 8-1L2<br />

fusion protein was as active as native soluble 1L2 in inducing<br />

proliferative responses. In contrast, Fell et al. (22), using a<br />

fusion protein consisting <strong>of</strong> the Fab’ region <strong>of</strong> the human<br />

carcinoma-specific L6 antibody linked to IL2, found the specific<br />

activity <strong>of</strong> the IL2 component <strong>of</strong> that fusion protein to be<br />

200-fold less than native rIL2 when measured in a proliferative<br />

assay with the CTLL-2 cell line. We examined the ability <strong>of</strong><br />

chl4.18-IL2 to stimulate proliferation <strong>of</strong> a human myeloid Ieukemia<br />

line, Tf-l3, previously shown to respond to IL2 in a<br />

dose-dependent manner (18) and to stimulate PBMCs obtained<br />

from melanoma patients following 96-h continuous infusions <strong>of</strong><br />

IL2. Both <strong>of</strong> these responding cell populations proliferate to IL2<br />

via interaction with the intermediate affinity (3y) IL2 receptor<br />

complex (2, 17, 21, 23). Both <strong>of</strong> these responding cell populations<br />

demonstrated that the 1L2 component <strong>of</strong> the chl4. l8-IL2<br />

fusion protein was as stimulatory as soluble recombinant human<br />

1L2. In addition, the Mik 3l antibody specific for the 3 chain <strong>of</strong><br />

the IL-2 receptor blocked these fusion protein-induced proliferative<br />

responses, demonstrating that the activation was due to the<br />

IL-2 component. More importantly, we noted that tumor-bound<br />

fusion protein was capable <strong>of</strong> stimulating 1L2-induced proliferative<br />

responses. Quantitative comparisons indicated that the 1L2<br />

component <strong>of</strong> the tumor cell-bound fusion protein was as stimulatory<br />

as soluble 1L2.<br />

Inclusion <strong>of</strong> 1L2 in vitro in the 51Cr release assay has been<br />

noted to augment 1L2-dependent LAK and ADCC activity mediated<br />

<strong>by</strong> PBMCs obtained following in vivo therapy with IL2<br />

(24, 25). We have noted that the IL2 within the chl4.18-IL2<br />

fusion protein also facilitates 1L2-dependent LAK killing <strong>of</strong> the<br />

Daudi target <strong>by</strong> patient PBMCs (data not shown). The current<br />

experiments demonstrate that the 1L2 component <strong>of</strong> the tumorbound<br />

fusion protein also augments ADCC. When the fusion<br />

protein was used to coat tumor target cells prior to washing and<br />

inclusion in the cytotoxic assay, enhanced ADCC (over that<br />

induced <strong>by</strong> the chl4.l8 antibody) was noted with patient PB-<br />

MCs as effectors on the LAN-S target in over 50% <strong>of</strong> the assays.<br />

On the M2 I melanoma target, the level <strong>of</strong> ADCC was comparable<br />

to that seen with antibody alone. This ADCC activity<br />

extends the prior observation that chl4. l8-IL2 fusion protein<br />

can enhance T cell-mediated killing <strong>of</strong> an autologous tumor cell<br />

line (10). In the previously published case, using the human 660<br />

TIL line as a tumor-specific effector population, increased lysis<br />

<strong>of</strong> the autologous GD2 tumor was obtained <strong>by</strong> addition <strong>of</strong> the<br />

chl4. 18-IL2 fusion protein as compared to that mediated <strong>by</strong><br />

addition <strong>of</strong> chl4.l8 antibody or IL2 alone (10).<br />

There is reason to hypothesize that in vivo localization <strong>of</strong><br />

IL2 to the tumor via the tumor-selective chl4.18-1L2 fusion<br />

protein may induce more effective antitumor destruction than an<br />

equivalent amount <strong>of</strong> free soluble ]L2. Other studies have demonstrated<br />

that 1L2 produced in viva <strong>by</strong> tumor cells (following<br />

gene transfer) can result in enhanced rejection <strong>of</strong> the IL2-<br />

producing tumor cells (26). The fact that this rejection was<br />

immunologically mediated suggests that a higher local concentration<br />

<strong>of</strong> 1L2 may improve immune recognition <strong>of</strong> tumor.<br />

Previous findings with the 14.l8-IL2 fusion protein demonstrated<br />

suppression <strong>of</strong> human neuroblastoma tumor growth in an<br />

experimental hepatic metastases model in SCID mice (I I). In<br />

this model, human LAK cells and relatively low doses <strong>of</strong><br />

chl4.18-IL2 fusion protein induced prolonged survival <strong>of</strong> animals<br />

bearing micrometastases, comparable to the survival noted<br />

only with very high doses <strong>of</strong> recombinant human 1L2 (1 1).<br />

Mixtures <strong>of</strong> low doses <strong>of</strong> mAb chl4.18 plus IL2 do not effectively<br />

prolong the life span nor eradicate established metastases<br />

<strong>of</strong> neuroblastoma or melanoma in SCID mice, whereas the<br />

fusion protein chl4.l8-IL2 is able to accomplish both <strong>of</strong> these<br />

tasks (11, 12). Becker et a/. (13) also demonstrated that this<br />

chl4.l8-IL2 fusion protein is effective against pulmonary and<br />

hepatic melanoma metastases in a syngeneic murine tumor<br />

model and that T cells are essential for achieving this antitumor<br />

effect. We note in the present in vitro study that in some<br />

instances, such as the lytic activity against M2l and LA-N-S<br />

targets, the fusion protein did not function any better than the<br />

combination <strong>of</strong> 1L2 and chl4.l8 as 2 independent reagents, yet<br />

four separate murine studies have noted a clear advantage <strong>of</strong> the<br />

fusion protein in vivo over the combination <strong>of</strong> soluble IL2 and<br />

chl4.l8 antibody (1 1-13, 27). This could be due to a number <strong>of</strong><br />

factors. The human immunoglobulin component <strong>of</strong> the fusion<br />

protein actually lengthens the serum half life <strong>of</strong> IL2 (27) from a<br />

li/2f3 <strong>of</strong> 6 mm for recombinant human IL2 to a ‘1/2(3 <strong>of</strong> 30 h for<br />

the chl4.l8-IL2 fusion protein. In addition, in vivo localization<br />

studies have shown targeting <strong>of</strong> anti-GD2 mAb to GD2 tumor<br />

in viva (28). Thus, it is likely that the ch 14. 1 8-IL2 fusion protein<br />

can specifically localize to tumor sites in vivo and deliver IL2<br />

directly to tumor sites. This may result in augmented stimulation<br />

<strong>of</strong> IL2-responsive effector cells, including both NK and T cells.<br />

These preclinical in vitro data presented here and the<br />

previously obtained in vivo data from the munine xenograft and<br />

syngeneic models suggest that the chl4. 18-1L2 fusion protein<br />

prolongs the serum half-life <strong>of</strong> IL2, localizes to sites <strong>of</strong> GD2<br />

tumor metastases, activates ADCC through FeR-bearing effectors,<br />

and activates IL2-responsive NK and T cells at tumor sites,<br />

potentially mediating a protective antitumor response. The current<br />

in vitro study documents that the 1L2 component <strong>of</strong> the<br />

tumor-bound fusion protein is as effective as soluble 1L2 in<br />

stimulating proliferation and cytotoxicity <strong>by</strong> effector cells obtamed<br />

from patients following in vivo therapy with IL2. The IL2<br />

component <strong>of</strong> the fusion protein facilitates ADCC <strong>of</strong> neuroblastoma<br />

and melanoma targets <strong>by</strong> in vivo activated human effector<br />

cells. Toxicity testing <strong>of</strong> this fusion protein in experimental<br />

animals is now underway to determine how best to test ch 14. 18-<br />

IL2 in Phase I clinical trials for patients with GD2 tumors,<br />

including melanoma (29) and neuroblastoma.<br />

REFERENCES<br />

I. Rosenberg, S. A., Lotze. A. I., Yang, I. C., Aebersold, P. M.,<br />

Linehan, W. M., Seipp. C. A., and White, D. E. Experience with the use


Clinical Cancer Research 1959<br />

<strong>of</strong> high-dose interleukin-2 in the treatment <strong>of</strong> 652 cancer patients. Ann.<br />

Surg., 210: 474-484. 1989.<br />

2. Weil-Hillman, G., Voss, S. D., Finch, P., SchelI, K., Hank, J. A.,<br />

Sosman, J. A.. Sugamura, K., and Sondel, P. M. Natural killer cells<br />

activated <strong>by</strong> Interleukin-2 treatment in rho respond to Interleukin-2<br />

primarily through the p75 receptor and maintain the p55 (TAC) negative<br />

phenotype. Cancer Res.. 50: 2683-2691 . I 990.<br />

3. Sosman, I. A., Kohler, P. C., Hank, J. A., Moore, K. H., Bechh<strong>of</strong>er,<br />

R.. Storer. B.. and Sondel, P. M. Repetitive weekly cycles <strong>of</strong> interleukin-2.<br />

II. Clinical and immunologic effects <strong>of</strong> dose, schedule. and<br />

indomethacin. I. Nail Cancer Inst., 80: 1451-1460, 1988.<br />

4. Sosman, J. A.. Hank. J. A., Moore, K. H., Borchert, A., Schell, K.,<br />

Kohler. P. C., Goldstein, D., Bechh<strong>of</strong>er, R., Storer, B., Albertini, M. R..<br />

Leung, R., Levitt, D., and Sondel, P. M. Prolonged interleukin-2 (IL-2)<br />

treatment can augment immune activation without enhancing antitumor<br />

activity in renal cell carcinoma. Cancer Invest., 9: 35-48, 1991.<br />

5. Phillips, J. H.. Gemlo, B. I.. Myers, W. W., Rayner, A. A., and<br />

Lanier, L. L. In vito and in vitro activation <strong>of</strong> natural killer cells in<br />

advanced cancer patients undergoing recombinant interleukin-2 and<br />

LAK cell therapy. J. Clin. Oncol.. 5: 1933-1941. 1987.<br />

6. McMannis, J. D., Fisher, R. I., Creekmore, S. P., Braun, D. P., Harris,<br />

J. E., and Ellis, T. M. In vito effects <strong>of</strong> recombinant IL-2. I. Isolation <strong>of</strong><br />

circulating leul9+ lymphokine-activated killer effector cells from cancer<br />

patients receiving recombinant IL-2. J. Immunol., 140: 1335-1340,<br />

1988.<br />

7. Hank. J., Surfus, J., Gan, I., Chew, T-L.. Hong, R., Tans. K..<br />

Reisfeld, R.. Seeger, R., Reynolds, C. P., Bauer, M., Wiersma, S.,<br />

Hammond, D., and Sondel, P. M. Treatment <strong>of</strong> neuroblastoma patients<br />

with antiganglioside GD7 antibody plus interleukin-2 induces antibody<br />

dependent cellular cytotoxicity against neuroblastoma detected in vitro.<br />

J. Immunother., 15: 29-37. 1994.<br />

8. Ziegler, L. D., Palazzolo, P., Cunningham. J., Janus, M., Itoh, K.,<br />

Hayakawa, K., Helistrom, I., Hellstrom, K. E., Nicaise, C., and Dennin,<br />

R. Phase I trial <strong>of</strong> murine monoclonal antibody L6 in combination with<br />

subcutaneous interleukin-2 in patients with advanced carcinoma <strong>of</strong> the<br />

breast, colorectum, and lung. J. Clin. Oncol.. 10: 1470-1478, 1992.<br />

9. Weil-Hillman, G.. Fisch, P., Prieves, A. F., Sosman, I. A., Hank.<br />

J. A., and Sondel, P. M. Lymphokine-activated killer activity induced <strong>by</strong><br />

in vito interleukin 2 therapy: predominant role for lymphocytes with<br />

increased expression <strong>of</strong> CD2 and Leu I 9 antigens but negative expression<br />

<strong>of</strong> CD16 antigens. Cancer Res., 49: 3680-3688, 1989.<br />

10. Gillies, S. D., Reilly, E. B., Lo, K. M.. and Reisfeld, R. A. Antibody-targeted<br />

interleukin 2 stimulates I-cell killing <strong>of</strong> autologous tumor<br />

cells. Proc. Nail. Acad. Sci. USA, 89: 1428-1432, 1992.<br />

1 1 . Sabzevari, H., Gillies, S. D., Mueller, B. M., Pancook, I. D., and<br />

Reisfeld, R. A. A recombinant antibody-interleukin 2 fusion protein<br />

suppresses growth <strong>of</strong> hepatic human neuroblastoma metastases in severe<br />

combined immunodeficiency mice. Proc. NatI. Acad. Sci. USA, 9/:<br />

9626-9630, 1994.<br />

12. Becker. J. C., Pancook, I. D., Gillies, S. D., Mendelsohn, I.,<br />

Reisfeld, R. A. Eradication <strong>of</strong> human hepatic and pulmonary melanoma<br />

metastases in SCID mice <strong>by</strong> antibody-interleukin 2 fusion proteins.<br />

Proc. NatI. Acad. Sci. USA, 93: 2702-2707, 1996.<br />

13. Becker. J. C.. Pancook, I. D., Gillies, S. D., Furukawa, K., and<br />

Reisfeld, R. A. T cell mediated eradication <strong>of</strong> murine metastatic melanoma<br />

Induced <strong>by</strong> targeted Interleukin 2 therapy. I. Exp. Med., 183:<br />

2361-2366. 1996.<br />

14. Gearing. A. J. H., and Thorpe, R. The international standard for<br />

human interleukin-2. Calibration <strong>by</strong> international collaborative study.<br />

J. Immunol. Methods, 114: 3-9, 1988.<br />

15. Mueller, B. M., Romberdahl, C. A., Gillies, S. D., and Reisfeld.<br />

R. A. Enhancement <strong>of</strong> antibody-dependent cytotoxicity with chimeric<br />

anti-GD2 antibody. I. Immunol., 144: 1282-1286, 1990.<br />

16. Gillies, S. D.. Young, D., Lo, K-M., Foley, S. F.. and Reisfeld, R. A.<br />

Expression <strong>of</strong> genetically engineered immunoconjugates <strong>of</strong> lymphotoxin<br />

and a chimeric anti-ganglioside GD2 antibody. Hybridoma, /0:<br />

347-356, 1991.<br />

17. Sondel, P. M., Kohler, P. C., Hank. J. A.. Moore, K. H.. Rosenthal,<br />

N., Sosman, J., Bechh<strong>of</strong>er, R., and Storer, B. Clinical and immunological<br />

effects <strong>of</strong> recombinant Interleukin-2 given <strong>by</strong> repetitive weekly<br />

cycles to patients with cancer. Cancer Res.. 48: 2561-2567, 1988.<br />

18. Farner, N. L., Voss, S. D., Leary, T. P.. Gan, I., Hakimi, J., Evans,<br />

G., Ju, G., and Sondel, P. M. Distinction between -y detection and<br />

function in YT lymphoid cells and in the GM-CSF responsive human<br />

myeloid cell line. Tf-l. Blood, 86: 4568-4578. 1995.<br />

19. Tsudo, M., Kitamura, F., and Miyasaka, M. Characterization <strong>of</strong> the<br />

interleukin 2 receptor B chain using 3 distinct monoclonal antibodies.<br />

Proc. NatI. Acad. Sci. USA, 86: 1982-1986. 1989.<br />

20. Hakimi, J., Ha, V. C., Lin, P., Campbell, E., Gately, M. K., Tsudo,<br />

M., Payne, P. W., Waldman, T. A., Grant, A, J., and Tsien, W. H.<br />

<strong>Human</strong>ized Mik 3 I : a humanized antibody to the IL-2 receptor betachain<br />

that acts synergistically with humanized anti-TAC. J. Immunol.,<br />

15/: 1075-1085, 1993.<br />

21. Voss, S. D.. Sondel, P. M., and Robb, R. I. Characterization <strong>of</strong> the<br />

Interleukin 2 receptors (IL-2R) expressed on human natural killer cells<br />

activated in 100 <strong>by</strong> IL-2: association <strong>of</strong> the p64 IL-2R -y chain with the<br />

IL-2R y chain with the Il-2R 3 chain in functional intermediate-affinity<br />

IL-2R. I. Exp. Med., 176: 531-541, 1992.<br />

22. Fell. H. P.. Gayle, M. A., Grosmaire, L., and Ledbetter, J. A.<br />

Genetic construction and characterization <strong>of</strong> a fusion protein consisting<br />

<strong>of</strong> a chimeric F(ab’) with specificity for carcinomas and human IL-2.<br />

J. Immunol., 146: 2446-2452, 1991.<br />

23. Hank, J. A., Sosman, J. A., Kohler, P. C., Bechh<strong>of</strong>er, R., Storer, B.,<br />

and Sondel, P. M. Depressed in vitro T cell responses concomitant with<br />

augmented Interleukin-2 responses <strong>by</strong> lymphocytes from cancer patients<br />

following in on’o treatment with interleukin-2. J. Biol. Response Modif.,<br />

9. 5-14, 1990.<br />

24. Hank, J. A. Robinson, R. R., Surfus, I.. Mueller, B. M., Reisfeld,<br />

R. A., Cheung, N-K., and Sondel, P. M. Augmentation <strong>of</strong> antibody<br />

dependent cell mediated cytotoxicity following in vito therapy with<br />

recombinant Interleukin-2. Cancer Res., 50: 5234-5239. 1990.<br />

25. Munn, D. H., and Cheung, N. K. V. Interleukin-2 enhancement <strong>of</strong><br />

monoclonal antibody mediated cellular cytotoxicity against human melanoma.<br />

Cancer Res., 47: 6600-6605, 1987.<br />

26. Fearon, E., Pardoll, D., Itaya, T., Golumbeck. P., Levitsky. H. I..<br />

Simons, J. W., Karasuyama, H., Vogelstein, B., and Frost, P. Interleukin-2<br />

production <strong>by</strong> tumor cells <strong>by</strong>passes T helper function in the<br />

generation <strong>of</strong> an antitumor response. Cell, 60: 397-403, 1990.<br />

27. Gillies, S. D., Young, D., Lo, K-M., and Roberts, S. Biological<br />

activity and in vito clearance <strong>of</strong> antitumor antibody/cytokine fusion<br />

proteins. Bioconjugate Chem., 4: 230-235, 1993.<br />

28. Reuland, P., Handgretinger, R., Smykowski, H., Dopfer, R.,<br />

Klingebiel. T., Miller, B. M., Reisfeld, R. A., Gallagher, S., Koscelniak,<br />

E., and Treuner, J. Application <strong>of</strong> the murine anti-GD-2 antibody 14.<br />

GD-2a for diagnosis and therapy <strong>of</strong> neuroblastoma. Intl. J. Radiat. AppI.<br />

Instrum. B, 18: 121-125, 1991.<br />

29. Albertini. M. R., Gan, J., Jaeger, P., Hank, I. A., Storer. B.. Rivest,<br />

T.. Surfus, I., Reisfeld, R. A., Schiller, J. H.. and Sondel, P. M. Systemic<br />

Interleukin-2 treatment can inhibit the anti-idiotypic response to chimeric<br />

anti-GD2 antibody in melanoma patients. I. Immunother.. in<br />

press, 1996.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!