10
Vol. 2, 1951-1959. December 1996 Clinical Cancer Research 1951 Activation of Human Effector Cells by a Tumor Reactive Recombinant Anti-Ganglioside GD2 Interleukin-2 Fusion Protein (chl4.18-1L2)’ Jacquelyn A. Hank,2 Jean E. Surfus, Jacek Gan, Peter Jaeger, Stephen D. Gillies, Ralph A. Reisfeld, and Paul M. Sondel Department of Human Oncology. University of Wisconsin-Madison, Madison. Wisconsin 53792 Ii. A. H.. J. E. S., J. G.. P. J., P. M. 5.1: Fuji ImmunoPharmaceuticals Corporation. Lexington. Massachusetts 02173 15. D. G.l: The Scripps Research Institute. La Jolla. Califrrnia 92037 iR. A. RI: and Departments of Pediatrics and Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin 53792 P. M. 5.1 ABSTRACT Cytotoxic effector cells interact with target cells through various mechanisms. CTLs use the antigen-specific T cell receptor, whereas Fe receptor-positive natural killer cells use this receptor to interact with antibody-coated target cells. We evaluated the tumor-binding and lymphocyte-ac- tivating capability of a recombinant fusion protein consist- ing of a tumor-selective human/mouse chimeric anti-gangli- oside GD2 antibody (chl4.18) and recombinant human interleukin-2 (1L2) (chl4.18-IL2). This fusion protein bound specifically to GD2-positive melanoma and neuroblastoma tumor cell lines, and its 1L2 component stimulated in vitro proliferation of an IL2-dependent cell line, as well as periph- eral blood mononuclear cells, in healthy control individuals and in cancer patients receiving continuous infusion of 1L2. The 1L2 presented by the fusion protein, when bound to tumor cells, induced proliferation of IL2-responsive cells as well as a comparable amount of soluble 1L2 did. This sug- gests that localization of 1L2 at the site of contact between tumor and effector cells is an effective way of presenting this cytokine to 1L2-responsive cells. The chl4.18-1L2 fusion protein also mediated antibody-dependent cellular cytotox- icity with Fc receptor-positive effector cells to an extent similar to chl4.18. These results, together with those of previous studies documenting antitumor efficacy against hu- man tumor xenografts in SCID mice and GD2-positive mu- rine tumors in immunocompetent syngeneic mice, suggest that the chl4.18-1L2 fusion protein should be tested in Phase I and II trials in patients with GD2-positive tumors. Received 10/24/95; revised 8/27/96: accepted 9/18/96. This research was supported by NIH grants and contracts CA-5344 I. CA-05436. CA-32685, CM-87290, CA- I 4520, CA- I 3539. CM-47669, HL-02I43. RR-03186, and American Cancer Society Grant CH-237. 2 To whom requests for reprints should be addressed, at K4/454 CSC. 6()0 Highland Avenue, Madison. WI 53792. Phone: (608) 263-7262: Fax: (608) 263-4226. INTRODUCTION Immunotherapy with IL23 is of benefit to some patients with renal cell cancer and melanoma (I ). Although multiple immune mechanisms are activated in patients receiving IL2, the immune components induced by lL2 necessary for antitumor activity have not been identified. Continuous infusion of IL2. at doses tolerated in the outpatient setting. induces systemic lymphoid activation in virtually all treated patients: however, only a minority of such patients achieve antitumor responses ( 1-4). Included in this lymphoid cell activation is an expansion and activation of the CDl6, FcR NK cells (5. 6). Currently. attempts are being made clinically to target these cells to tumor through the use of tumor-specific mAbs (7. 8). These include clinical evaluation of regimens combining effector cell activa- tion through 1L2 infusions with infusions of munine or chimeric tumor-selective mAb, such as the l4.G2a or chl4. 18 antibodies, which recognize the ganglioside GD2 expressed on neuroblas- toma. melanoma, and certain other tumors (7). In an effort to augment the stimulation of the FcR anti- body-directed” effector cells and activate FcR effector cells that express IL2 receptors (2, 9), a chl4.l8-IL2 fusion protein has been constructed by fusion of a synthetic sequence coding for human IL2 to the carboxyl end of the C-yl gene of the mAb ch I 4. 18 ( 10). When the antitumor variable region of this fusion protein binds to tumor, IL2 should be concentrated in the tumor microenvironment and provide activation of FcR effectors, such as NK cells, that did bind to the immunoglobulin Fe domain of the tumor-bound fusion protein. Furthermore, the IL2 component of this tumor-bound fusion protein may activate IL2-responsive cells. such as cytotoxic T cells and helper T cells, that do not necessarily have FcRs and the subpopulation of NK cells that express IL2 receptors but lack the FcR (9), thereby recruiting additional effector cells into the tumor microenviron- ment. The chI4.l8-1L2 fusion protein was previously shown to maintain antigen-binding activity and 1L2 activity ( 10) and has provided antitumor effects in SCID mice bearing human tumor xenografts of neuroblastoma ( II) and melanoma ( I 2), as well as in a syngeneic murine melanoma model ( 13). We have extended these findings by demonstrating that chl4. l8-IL2 bound to GD2 tumor cells can be visualized by flow cytornetry, detect- ing either the chl4.l8 or the 1L2 portion of this construct. The IL2 component of the fusion protein is able to stimulate lL2- dependent cells. The fusion protein bound to GD2 tumors 3 The abbreviations used are: lL2. interleukin 2: NK. natural killer: FcR, Fe receptor: mAb. monoclonal antibody: ADCC. antibody-dependent cellular cytotoxicity: HLR, Hoffmann LaRoche; PBMC, peripheral blood mononuclear cell: LAK. lymphokine-activated killer. Research. on June 26, 2021. © 1996 American Association for Cancer clincancerres.aacrjournals.org Downloaded from

Activation of Human Effector Cells by a Tumor Reactive … · Vol. 2, 1951-1959. December 1996 Clinical Cancer Research 1951 Activation of Human Effector Cells by a Tumor Reactive

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

  • Vol. 2, 1951-1959. December 1996 Clinical Cancer Research 1951

    Activation of Human Effector Cells by a Tumor Reactive

    Recombinant Anti-Ganglioside GD2 Interleukin-2

    Fusion Protein (chl4.18-1L2)’

    Jacquelyn A. Hank,2 Jean E. Surfus, Jacek Gan,

    Peter Jaeger, Stephen D. Gillies,

    Ralph A. Reisfeld, and Paul M. Sondel

    Department of Human Oncology. University of Wisconsin-Madison,

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

    Fuji ImmunoPharmaceuticals Corporation. Lexington. Massachusetts

    02173 15. D. G.l: The Scripps Research Institute. La Jolla. Califrrnia

    92037 iR. A. RI: and Departments of Pediatrics and Medical

    Genetics, University of Wisconsin-Madison, Madison, Wisconsin

    53792 P. M. 5.1

    ABSTRACT

    Cytotoxic effector cells interact with target cells

    through various mechanisms. CTLs use the antigen-specific

    T cell receptor, whereas Fe receptor-positive natural killer

    cells use this receptor to interact with antibody-coated target

    cells. We evaluated the tumor-binding and lymphocyte-ac-

    tivating capability of a recombinant fusion protein consist-

    ing of a tumor-selective human/mouse chimeric anti-gangli-

    oside GD2 antibody (chl4.18) and recombinant human

    interleukin-2 (1L2) (chl4.18-IL2). This fusion protein bound

    specifically to GD2-positive melanoma and neuroblastoma

    tumor cell lines, and its 1L2 component stimulated in vitro

    proliferation of an IL2-dependent cell line, as well as periph-

    eral blood mononuclear cells, in healthy control individuals

    and in cancer patients receiving continuous infusion of 1L2.

    The 1L2 presented by the fusion protein, when bound to

    tumor cells, induced proliferation of IL2-responsive cells as

    well as a comparable amount of soluble 1L2 did. This sug-

    gests that localization of 1L2 at the site of contact between

    tumor and effector cells is an effective way of presenting this

    cytokine to 1L2-responsive cells. The chl4.18-1L2 fusion

    protein also mediated antibody-dependent cellular cytotox-

    icity with Fc receptor-positive effector cells to an extent

    similar to chl4.18. These results, together with those of

    previous studies documenting antitumor efficacy against hu-

    man tumor xenografts in SCID mice and GD2-positive mu-

    rine tumors in immunocompetent syngeneic mice, suggest

    that the chl4.18-1L2 fusion protein should be tested in Phase

    I and II trials in patients with GD2-positive tumors.

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

    ‘ This research was supported by NIH grants and contracts CA-5344 I.

    CA-05436. CA-32685, CM-87290, CA- I 4520, CA- I 3539. CM-47669,HL-02I43. RR-03186, and American Cancer Society Grant CH-237.

    2 To whom requests for reprints should be addressed, at K4/454 CSC.6()0 Highland Avenue, Madison. WI 53792. Phone: (608) 263-7262:

    Fax: (608) 263-4226.

    INTRODUCTION

    Immunotherapy with IL23 is of benefit to some patients

    with renal cell cancer and melanoma ( I ). Although multiple

    immune mechanisms are activated in patients receiving IL2, the

    immune components induced by lL2 necessary for antitumor

    activity have not been identified. Continuous infusion of IL2. at

    doses tolerated in the outpatient setting. induces systemic

    lymphoid activation in virtually all treated patients: however,

    only a minority of such patients achieve antitumor responses

    ( 1-4). Included in this lymphoid cell activation is an expansion

    and activation of the CDl6�, FcR� NK cells (5. 6). Currently.

    attempts are being made clinically to target these cells to tumor

    through the use of tumor-specific mAbs (7. 8). These include

    clinical evaluation of regimens combining effector cell activa-

    tion through 1L2 infusions with infusions of munine or chimeric

    tumor-selective mAb, such as the l4.G2a or chl4. 18 antibodies,

    which recognize the ganglioside GD2 expressed on neuroblas-

    toma. melanoma, and certain other tumors (7).

    In an effort to augment the stimulation of the FcR� anti-

    body-directed” effector cells and activate FcR effector cells

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

    has been constructed by fusion of a synthetic sequence coding

    for human IL2 to the carboxyl end of the C-yl gene of the mAb

    ch I 4. 1 8 ( 10). When the antitumor variable region of this fusion

    protein binds to tumor, IL2 should be concentrated in the tumor

    microenvironment and provide activation of FcR� effectors,

    such as NK cells, that did bind to the immunoglobulin Fe

    domain of the tumor-bound fusion protein. Furthermore, the IL2

    component of this tumor-bound fusion protein may activate

    IL2-responsive cells. such as cytotoxic T cells and helper T

    cells, that do not necessarily have FcRs and the subpopulation of

    NK cells that express IL2 receptors but lack the FcR (9), thereby

    recruiting additional effector cells into the tumor microenviron-

    ment.

    The chI4.l8-1L2 fusion protein was previously shown to

    maintain antigen-binding activity and 1L2 activity ( 10) and has

    provided antitumor effects in SCID mice bearing human tumor

    xenografts of neuroblastoma ( I I ) and melanoma ( I 2), as well as

    in a syngeneic murine melanoma model ( 13). We have extended

    these findings by demonstrating that chl4. l8-IL2 bound to

    GD2� tumor cells can be visualized by flow cytornetry, detect-

    ing either the chl4.l8 or the 1L2 portion of this construct. The

    IL2 component of the fusion protein is able to stimulate lL2-

    dependent cells. The fusion protein bound to GD2� tumors

    3 The abbreviations used are: lL2. interleukin 2: NK. natural killer: FcR,Fe receptor: mAb. monoclonal antibody: ADCC. antibody-dependent

    cellular cytotoxicity: HLR, Hoffmann LaRoche; PBMC, peripheral

    blood mononuclear cell: LAK. lymphokine-activated killer.

    Research. on June 26, 2021. © 1996 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • 1952 Anti-GD2-IL2 Fusion Protein

    stimulates proliferation to the same extent as soluble fusion

    protein or soluble 1L2. The chl4.l8-1L2 fusion protein also

    stimulates cytolytic activity in vitro by cells able to mediate

    ADCC. FcR� NK cells obtained from melanoma patients in

    vivo following therapy with continuous infusion 1L2 are targeted

    to GD2� tumors that have bound chl4.l8-IL2 in vitro, and the

    lytic activity was comparable to that induced by the combination

    of free antibody and soluble 1L2.

    MATERIALS AND METHODS

    Recombinant IL2. HLR IL2 was provided through the

    Cancer Treatment and Evaluation Program of the National Can-

    cer Institute. The National Cancer Institute-Biological Response

    Modifiers program standard for unit dosage was used, and the

    specific activity of the IL2 was 15 X 106 units/mg. This unit

    corresponds closely with the international standard for IL2

    unitage (14).

    Chimenc Antibody and Fusion Protein. The mouse/

    human chimeric 14.18 antibody was constructed by combining

    the variable regions of the murine anti-GD2 14.18 antibody with

    the constant regions of human IgG1 heavy chain and K light

    chain (15, 16). The 14.l8-IL2 fusion protein was constructed by

    fusion of a synthetic sequence coding for human IL2 to the

    carboxyl end of the human C-yl gene of the mAb chl4. 18 (10).

    The fused gene was inserted into the vector pdHL2-l4. 18 as

    described previously (15). Transfection of the expression plas-

    mids in Sp2/0-Agl4 cells and propagation of the clones secret-

    ing chl4. 18-1L2, as well as its purification, have been described

    previously (10). To compare the 1L2 activity in the soluble IL2

    preparation and in the fusion protein, concentrations were based

    on weight/volume calculations of 1L2 in the two preparations.

    Because the fusion protein molecule consists of 80% chimeric

    14.18 immunoglobulin and 20% 1L2 by molecular weight, the

    fusion protein 1L2 concentration was based on IL2 comprising

    20% of the weight of the fusion protein. Thus, 50 ng/rnl of

    fusion protein would correspond to 10 ng/ml of IL2 in the fusion

    protein. Because 10 ng/ml of soluble IL2 corresponds to 150

    units/mi of soluble HLR 1L2, we have used this conversion to

    describe the units of 1L2 anticipated for the fusion protein

    preparation based on the molecular weight of IL2 and using the

    specific activity of 15 X 106 units/mg for the HLR IL2.

    Tumor Cell Lines. The GD2-positive LA-N-S neuro-blastoma target cell line, kindly provided by R. Seeger (Chil-

    dren’s Hospital of Los Angeles, Los Angeles, CA), was main-

    tamed as an adherent monolayer in Leibovitz’ s medium

    supplemented with 15% heat-inactivated fetal bovine serum. A

    trypsin-EDTA solution was used to harvest the cell monolayer.

    The M2l human melanoma line (GD2’) was described previ-

    ously (15), and the BT-20 human breast carcinoma cell line

    (GD2) was obtained from American Type Culture Collection.

    Both cell lines were maintained as adherent monolayers in

    RPM! 1640 supplemented with penicillin and streptomycin

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

    Flow Cytometry. Cell-bound fusion protein and anti-body were detected by standard indirect immunofluorescence

    methods (Becton Dickinson, San Jose, CA). Antibodies in-

    eluded a goat antihuman IgG (Caltag, San Francisco, CA) and a

    rabbit antihuman IL2 (Genzyme, Cambridge, MA).

    Proliferation Assays. Fresh PBMCs from healthy vol-

    unteer human donors or from patients who were treated with a

    96-h continuous infusion of IL2 (3. 4, 17) were cultured in

    0.2-ml round-bottom microplates at a concentration of 1 X l0�

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

    (Pel-Freez, Rogers, AR), 25 msi HEPES, 100 units/mI penicil-

    lin, and 100 p.g/ml streptomycin sulfate (RPMI-HS). Recombi-

    nant IL2 and fusion protein were added at concentrations as

    indicated in the “Results”. Concentrations of recombinant sol-

    uble 1L2 were also tested, which corresponded to the concen-

    tration of 1L2 in the fusion protein preparation based on the

    molecular weight and concentration of fusion protein: I �i.g of

    the fusion protein contains approximately 3000 units of IL2

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

    M21 and LA-N-S cells were used as a proliferative stimulus, the

    antibody or fusion protein at S �ig/ml was incubated with the

    tumor cells for 1 h on ice. Irradiation of these cells took place

    during this incubation, with LA-N-S and M2l receiving 10,000

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

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

    18 h, and harvested with a Filterrnate 196 Packard harvester, and

    [3H]thymidine incorporation was quantitated with a Matrix

    9600 direct 13 counter with a 5-mm counting period. Informed

    consent forms, approved by the University of Wisconsin Human

    Subjects Committee, were obtained prior to collection of all

    human blood specimens.

    For some proliferative studies, the 1L2-responsive cells

    used were the Tf-l myeloid leukemia cell line transfected with

    the gene for the 1L2 receptor �3 chain. This transfected line was

    designated Tf-l �, and the mock-transfected control line con-

    taming the LXSN vector but no IL2R�3 gene was designated

    Tf-IL (18). This Tf-l�3 cell line responded to 1L2 using inter-

    mediate affinity � receptor complexes ( 1 7. I 8) and thus isanalogous to the majority of NK cells in 1L2-treated patients,

    which also use intermediate affinity IL2 receptors (2). The Mik

    1�1 monoclonal antibody directed against the p70 IL-2 receptor1� chain was used in the blocking studies ( 19. 20).

    ADCC and Fusion Protein-mediated Cellular Cytotox-icity. All ADCC assays were performed in RPMI-HS. Effec-

    tor cells in a total volume of SO pi were plated in quadruplicate

    into 96-well U-bottomed microtiter plates at the indicated ef-

    fector/target ratios. Just prior to the addition of target cells, SO

    �il of antibody, antibody plus 1L2, or fusion protein were added

    to the effectors. While the effectors were being prepared, target

    cells were labeled for 2 h with 250 pCi of 5tCr in 0.2 ml of

    RPMI-HS. Target cells were mixed every 15-30 mm during

    labeling to keep the cells in suspension. After being washed

    twice with RPMI, S x l0� target cells in 50 �il of RPMI-HSwere added to effector cells and centrifuged at 200 X g for S

    mm. In the experiments using fusion protein-coated target cells.

    chl4.l8-1L2 was added to the targets following one wash and

    incubated on ice for 1 h. Two subsequent washes removed

    excess fusion protein and 51Cr. The effector cells were also

    plated in medium and in IL2 to determine their ability to

    mediate lysis of target cells in the absence of antibody or fusion

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

    and the supernatants were harvested using the Skatron Harvest-

    ing System (Skatron, McLean, VA). Maximum 5tCr release was

    measured by lysing target cells with the detergent cetrimide

    Research. on June 26, 2021. © 1996 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • M21

    chl4.18-1L2chl4.18

    BT-20

    JLRabbit Anti-1L2

    Rabbit IgG

    Rabbit lgGRabbit Anti-1L2

    . . . .�-

    Clinical Cancer Research 1953

    LAN-5

    1u�

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

    and GD2 BT-20 breast carcinoma cell lines were each coated with I p.g of either chl4.18-1L2 fusion protein or chl4.18 antibody. To detect theantibody component (top panels). a fluorescein-conjugated goat antihuman IgG secondary antibody was used. To detect the 1L2 component (bottom

    panels). a rabbit anti-IL2 antibody was used, followed by a phycoerythrin-conjugated goat antirabbit antibody.

    (Sigma). Spontaneous 5tCr release was measured by incubating

    target cells in RPMI-HS alone. Percent cytotoxicity values were

    calculated for each effector/target ratio as follows:

    C/� cytotoxicity

    experimental release - spontaneous release=l00X-�- .

    maximum release - spontaneous release

    Results are expressed as percent cytotoxicity or as lytic

    units, where 1 lytic unit is the number of effector cells necessary

    to achieve 20% lysis of S X I0� targets.

    RESULTS

    Fusion Protein chl4.18-1L2 Binds to GD2� Tumor

    Cells. The data shown in Fig. I demonstrate the binding of

    chl4.l8-IL2 to the GD2� M2l human melanoma and LA-N-S

    human neuroblastoma cell lines. The fusion protein binds to the

    tumor cells with a fluorescence intensity similar to that of the

    parental ch I 4. 18 antibody (Fig. 1, top panels). The specificity ofch I 4. 18-IL2 and ch I 4. 18 antibody is demonstrated by their lack

    of binding to the GD2 BT-20 human breast carcinoma cell

    line. The IL2 component of ch 14. 1 8-1L2 can be detected by

    rabbit anti-IL2 antibody when the fusion protein is bound to

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

    These data document that the IL2 component of the fusion

    protein remains associated with the tumor cells that bind

    chl4.l8-1L2 on their surfaces and is detectable by anti-IL2

    antibody. Furthermore, separate flow cytometric analyses have

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

    or �3 chains of the 1L2 receptor (data not shown). Thus, binding

    of the fusion protein to these cells is mediated through the

    chl4.18 rather than the IL2 component of the fusion protein.

    Soluble chl4.18-1L2 Stimulates IL2-induced Prolifera-

    tive Responses. Fig. 2 documents proliferative responses to

    1L2 attained with the Tf-I�3 cell line and PBMCs from a mel-

    anoma patient obtained 24 h after a 96 h continuous iv. infusion

    of IL2. Tf-l is a GM-CSF-dependent myeloid leukemia-derived

    cell line that constitutively expresses the common cytokine

    receptor -y chain. A variant cell line bearing functional interme-

    diate affinity IL2 receptors (�3-y dimers), designated Tf- I �, wasobtained by stable transfection of Tf- I with eDNA encoding the

    1� chain of the IL2 receptor ( 1 8). The Tf- 1 �3 cell line thatretained responsiveness to GM-CSF also responded to IL2 in a

    dose-dependent fashion. The mock-transfected Tf-lL cell line

    did not proliferate in response to 1L2, and the Tf-l�3 cell line did

    not respond to chl4.l8 antibody (data not shown). The results of

    proliferative assays shown in Fig. 2 were obtained by using

    dilutions of IL2 and chl4.l8-1L2 to achieve equivalent molar

    Research. on June 26, 2021. © 1996 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • 1200

    1000

    600

    400

    200

    0

    10000

    8000

    6000

    4000

    2000

    1954 Anti-GD2-IL2 Fusion Protein

    Cl)

    I-z

    0C�)

    0 -�L2 UImI 1.6 3.0 00 12 25 50 100 200 400FP ��JmI.B2O 1.20 2.0 5.0 10 20 40 80 ISO

    Fig. 2 Proliferation induced by soluble IL2 and IL2 within the solublefusion protein. The Tf-l�3 cell line (A) and PBMCs obtained from apatient following continuous infusion lL2 (B) were stimulated withincreasing concentrations of soluble IL2 (A) and soluble chl4.l8-1L2

    fusion protein (s). Dilutions of IL2 and fusion protein were made toachieve equivalent molar concentrations of IL2. The counts were deter-mined for [3H]TdR incorporation by proliferating cells.

    concentrations of IL2. Assuming the same specific activity of

    1L2 within the fusion protein as the recombinant HLR product,

    there are approximately 400 units/ml of 1L2 contained within

    the chl4.18-IL2 when it is applied at a concentration of 160

    ng/ml. When these data are expressed based on the concentra-

    tion of 1L2, dose-response curves indicate that at concentrations of

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

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

    proliferate. This finding was reproduced in two additional prolif-

    erative assays using the Tf- 1 �3 cell line as the responding cell.

    PBMCs obtained from six patients with melanoma who

    had just completed a continuous infusion of IL2 were also tested

    for their responsiveness to IL2 and chl4.l8-IL2. Results from

    one representative patient are shown at the bottom of Fig. 2.

    Previous studies have shown that NK cells obtained following in

    vivo IL2 treatment show rapid proliferative responses to re-

    stimulation with IL2 in vitro, using primarily the intermediate

    affinity 13’y receptor complex (2, 21). Similar proliferation by

    these cells was observed at most concentrations of IL2 and

    chl4.18-IL2 tested. These data indicate that the antibody com-

    ponent in the fusion protein did not adversely affect the ability

    of IL2 to interact with the IL2R complexes either on the Tf-l �3

    cells or on the PBMCs of a melanoma patients treated with IL2,

    nor did it affect the ability of the 1L2 component of the fusion

    protein to stimulate proliferation. chl4. I 8 antibody alone, at

    comparable concentrations, did not stimulate patient PBMCs to

    proliferate (data not shown). Table 1 presents data from two

    separate experiments indicating the in vitro proliferative re-

    sponse induced by 100 units of soluble IL2 and by a similar

    amount of IL2 contained within 40 ng of chl4.18-IL2. The

    responding cells were PBMCs obtained from five melanoma

    patients and PBMCs from one healthy control individual. The

    patient PBMCs were obtained 24 h after completion of a 96-h

    continuous infusion of IL2 in vivo. In these experiments, the

    fusion protein stimulated proliferation that was similar to that

    induced by the soluble 1L2 for patient I and for the control

    donor in experiment 1 and for the three patients in experiment 2.

    The chimeric chl4.l8 antibody itself was not stimulatory.

    Fusion Protein-induced Proliferation Mediated throughan Intermediate Affinity �#{176}y1L2 Receptor Is SpecificallyBlocked by Antibody to the � Chain of the IL2 Receptor.

    Fig. 3 demonstrates the proliferation induced by IL2 and fusion

    protein on cells with intermediate affinity receptors for 1L2,

    Tf-l �3, and PBMCs obtained from cancer patients following a

    4-day continuous infusion of 1L2. Cells with high-affinity a�j

    IL2 receptors (Kit 225 cells) were also assayed. Tf- 1 �3 and

    patient PBMCs responded to 1L2 and the fusion protein in a

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

    PBMCs from a healthy volunteer individual (control) and from two patients were assayed in experiment 1 . and PBMCs from three patients wereassayed in experiment 2. The patient PBMCs were obtained 24 h after a 96-h continuous infusion of IL2. They were stimulated with IL2 or

    chl4.l8-IL2 fusion protein at concentrations containing equivalent amounts of IL2 or with an excess of chl4.I8 antibody. The cells were culturedfor 4 days at 37#{176}Cin 5% CO,. pulsed with 3H-thymidine for 16 h, and harvested, and counts per 5 mm were obtained with a Packard 9600 Matrixcounter.

    Experiment

    3H-Thymidine incorporati on (count per 5 mm)

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

    I ControlPatient 1Patient 2

    197160184

    14,1949,741

    24,381

    13.3877,916

    7,413

    190161

    207

    II Patient 16Patient 17

    Patient 20

    16549

    31

    9,9592,321

    4,135

    11,2012,7006,317

    594643

    Research. on June 26, 2021. © 1996 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • 120000 120000

    90000 90000

    60000 60000

    30000 30000

    0 � �- 0

    200 50 12.5 66.5 16.6 4.1

    16000 16000

    L1�I-

    C-)

    0.I-a-

    I0

    C.”

    I-

    I 2000

    8000

    200

    12000

    8000

    4000

    0

    100000

    75000

    50000

    25000

    50

    100000

    75000

    50000

    25000

    0-200

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

    80000

    60000

    40000

    20000

    U)I-z

    0C)

    chl4.18

    LIt�chl4.18 FP

    LA-N-5FP

    M21Fig. 4 Proliferation induced by the lL2 component of the tumor-bound

    fusion protein. The M2l melanoma and LA-N-S neuroblastoma celllines were coated for I h at 4#{176}Cwith either chI4.18 antibody or the

    chl4.18-1L2 fusion protein (FP). Irradiation took place during thisincubation. These coated tumor target cells were washed twice and

    diluted to 500, 100, or 50 cells per well and used to stimulate l0� Tf-l�cells per well. Proliferation was quantitated after 72 h by I3HITdRincorporation.

    Clinical Cancer Research 1955

    Fig. 3 Antibody to the �3 chain of the IL2 receptor

    blocks fusion protein induced proliferation by cells

    expressing intermediate affinity, but not cells ex-

    pressing high-affinity lL2 receptors. Tf-l� cells.

    PBMCs, and KIT 225 cells were cultured with lL2(leftpa,iels) or the chI4.18-1L2 fusion protein (rightpanels). The humanized Mik �3l antibody at a finalconcentration of 3 p.g/ml (A) or human serum diluted

    66 5 16 6 4 1 1 1/50 as the control (�) was added at the initiation of

    12.5 � . . a 3-day I3HITdR incorporation assay. #{149},medium.

    ‘ . 0 � ‘50 12.5 66.5 16.6 4.1 1

    dose-dependent manner, and this proliferation was abrogated by

    inclusion of the Mik �3I antibody, which recognizes the �3 chain Coated

    of the IL2 receptor (20). Over the dose range examined, there Tumor Cells. . #{149}500

    was a strong proliferative response to both soluble IL2 and #{149}�fusion protein by the Kit 225 cell line, which expresses the � 50

    high-affinity afty IL2 receptor. The Mik �3 1 antibody did not

    block this proliferative response, as shown previously for IL2

    (19).

    chl4.18-1L2 Bound to GD2� Cells Stimulates Prolifer-ation. The flow cytometric studies (Fig. 1 ) document that the

    chl4.18-IL2 binds to GD2� tumor cells via the antibody van-

    able region and that its IL2 component is recognized by rabbit

    antihuman 1L2 antibody. Subsequent experiments tested the

    functional activity of the IL2 component of the fusion protein

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

    cell lines were coated with either chl4.18-IL2 or chl4.l8 and

    then washed free of any excess and cultured with the Tf-l�

    responding cell line or the Tf-lL control cell line. The data

    shown in Fig. 4 demonstrate that the ch 14. 18-IL2 fusion protein.

    when bound to a GD2� tumor cell line, effectively presents IL2

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

    M21 or LA-N-S cells it is able to efficiently present the bound

    Research. on June 26, 2021. © 1996 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • Patient 1

    400.

    300.

    200.

    L �oo

    LAN 5m4 18

    +112 FP

    cI,14.1O

    18

    Fp

    11.2

    Y

    Patient 2I

    C

    U

    N

    T

    S

    M2i

    ±14 10 +112

    ControlLAN-5

    200.

    150.

    100.

    50.

    c81418 +11.2� chI4.18Fr

    12

    M2�4.18+1U

    414.18

    112 Fr

    IL.2 (units) 2.5 25 ‘ 100FP (ng) I 10 40

    2.5 25 100I 10 40

    Fig. 5 Soluble fusion protein facilitated ADCC on GD2� tumor cells.

    PBMCs obtained from two patients following a 96-h continuous infusion of1L2 and from a volunteer control donor were the effector cells in a 4-h 51Crrelea.se assay. The GD2� LA-N-S and M21 cell lines were used as targets.The assay was performed in medium supplemented with 1L2 alone at 2.5.25, or 100 units/mI; with chl4.18-IL2 fusion protein (FP) alone at 1, 10, or40 ng/ml (corresponding to 2.5, 25, and 100 units of 1L2/ml): with thechI4.l8 antibody alone at 1, 10, or 40 ng/ml: or with a combination of 1L2

    and chl4.18 antibody at the same concentrations.

    1956 Anti-GD2-lL2 Fusion Protein

    two melanoma patients receiving IL2 were assayed for lytic

    Table 2 Stimulatory activity of IL2 in tumor-bound fusion protein

    The Tf-l�3 and control Tf-lL cell lines (experiment 1) or Tf-l�3 cells and PBMCs from IL2-treated patients (experiment 2) were stimulated ina 3-day proliferative assay with soluble IL2 or tumor cells coated with chI4.18 antibody or the chl4.18-IL2 fusion protein. Tumor cells (106) were

    incubated with S �i.g of chl4.18, washed twice, and diluted. Sixty % of the fusion protein used to coat the M2l cell remained bound to the cells:therefore, 100 cells have 0.3 ng of fusion protein, corresponding to approximately 1 unit of bound lL2. The total volume of the microwell is 0.2 ml,resulting in a total of 5 units of 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.pulsed with 3H-thymidine for 16 h, and then harvested, and counts per 5 mm were obtained with a Packard 9600 Matrix counter. The proliferationof Tf- 113 cells induced by 5 units/mI 1L2 as tumor-bound fusion protein was similar to the proliferation induced by 50 units/ml of soluble IL2. andthe proliferation of patient PBMCs induced by 25 units/mI 1L2 as tumor-bound fusion protein was similar to the proliferation induced by SO units/mI

    of soluble IL2.

    Stimulus

    3H-TdR incorporation (coun

    Experiment I

    Is X I0�), responding cell

    Experiment 2

    Tf- I �3 PatientTf-l �3 Tf- 1L

    Medium

    GM-CSF (5 ng/ml)1L2 (100 units/mI)IL2 (50 units/mI)1L2 (25 units/mI)100 M21 cells labeled with 0.5 ng of chl4.18-1L2 fusion protein (5.0 units of 1L2/ml)500 M2l cells labeled with 0.5 ng of chl4.l8-1L2 fusion protein (25 units of IL2/ml)

    6.7 10.8

    67.4 91.164.2 13.950.7 12.1

    41.9 12.8

    56.6 14.4

    70.8 13.4

    0.8 .03

    NT’ NT18.6 4.1

    9.3 1.7

    3.2 0.6

    1.2 0.4

    4.2 2.1

    ‘a NT, not tested.

    IL2. As few as SO such cells coated with the fusion protein can

    still produce a significant 1L2-specific proliferative response.

    The control Tf-lL cell line did not respond to tumor-bound

    fusion protein in these experiments (data not shown).

    Proliferative Response to “Equivalent” Doses of Tu-

    mor-bound Fusion Protein. In an attempt to determine

    whether there is a dose effect of the cell-bound fusion protein on

    the proliferative response, we compared the proliferation in-

    duced by soluble 1L2 to that induced by an equivalent amount of

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

    we determined the amount of fusion protein that remained

    bound to the coated GD2� tumor cells, based on the measure-

    ment of free fusion protein concentration in the supernatant

    following incubation. This indicated that approximately 60% of

    the chl4.l8-IL2 used to coat the cells actually remained bound

    to the cells. Data shown in Table 2 are from an experiment in

    which 106 M2l cells were coated with S �ag ofchl4.18-1L2 and

    washed twice. These cells were then diluted, and 100 or 500

    fusion protein-coated M2 1 cells per well were used to stimulate

    Tf- I 13 cells. With 60% of fusion protein bound to the M2 I cells,this corresponds to a total of 0.3 ng of fusion protein or approx-

    imately 1 unit of IL2 per 100 cells in the 0.2-ml microtiter well.

    This corresponds to 5.0 and 25 units of IL2/ml in the prolifer-

    ative assay with 100 and 500 fusion protein-coated M21 cells,

    respectively. Two experiments are presented in Table 2.

    chl4.18-IL2 fusion protein-coated M2l melanoma cells stimu-

    lated proliferative responses by the Tf-l �3 cell line (experiments

    1 and 2) and patient PBMCs obtained following an in vivo

    infusion of IL2 (experiment 2). These results indicate that the

    IL2 component of the fusion protein that remains coated to M2l

    cells is presented in a conformation able to stimulate the IL-2

    receptor and show that the tumor-bound IL-2 is as stimulatory as

    soluble 1L2.

    chl4.18-1L2 Fusion Protein Is Capable of Eliciting

    ADCC by PBMCs on GD2� Tumor Targets. PBMCs from

    Research. on June 26, 2021. © 1996 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • M21A300

    250

    200

    150

    100

    50

    0

    8 igG

    0 IgG+iL2

    iichl4.18

    Oichl4.18+1L2

    U FP

    600 �

    U)

    � 400z

    � 200

    800

    � 600

    #{163}(I)

    S

    :

    LA-N-5�600

    _I 5oo

    400

    300

    200

    100

    Clinical Cancer Research 1957

    0

    ,Ju1�,J

    Patient 6 Patient 7 PatIent 20 Patient 17

    Fig. 6 Tumor-bound fusion protein facilitated ADCC. Patient PBMCsobtained from four patients following a 96-h continuous infusion of 1L2

    were cryopreserved and thawed on the morning of the 4 h 51Cr release

    assay. The GD2� M2l (A) and LA-N-S (B) targets were coated with the

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

    during the 51Cr labeling. For the chl4.18 antibody-coated targets. sol-

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

    with the chl4.l8-IL2 fusion protein received no additional soluble 1L2.

    activity against the GD2� LA-N-S and M2l cell lines (Fig. 5).

    The fusion protein added in soluble form during the 4-h 51Cr

    release assay enhanced lysis mediated by both patients� PBMCs.

    The ch 14. 18-IL2 fusion protein mediated levels of ADCC sim-

    ilar to those mediated by ch I 4. 18 alone or antibody combined

    with soluble IL2 when tested on the NK-resistant LA-N-S target

    cells. Against the M2 I targets, the fusion protein also enhanced

    lysis over that achieved with 1L2 alone; however, the fusion

    protein was not as effective as antibody combined with soluble

    IL2. Similar effects were observed with PBMCs from a healthy

    volunteer donor that were not previously primed in vito with

    IL2.

    ADCC with Tumor Target Cells Coated with chl4.18-

    1L2 Fusion Protein. PBMCs obtained from four melanoma

    patients 24 h after a 96-h continuous infusion of IL2 were

    cryopreserved and thawed the day of the assay for use as

    effector cells. The GD2� tumor cell targets were coated with

    either chl4.l8-IL2, chI4.l8, or control lgG immediately fol-

    lowing the 5tCr labeling. Soluble IL2 (100 units/ml) was added

    ,� 400

    0 � 200

    chl4.18 FP chl4.18 FP

    M21 LA-N-5

    �. 0

    B Fig. 7 ADCC ofGD2� M21 and LA-N-S cells was facilitated by eitherchl4.l8 antibody or chl4.l8-IL2 fusion protein. Six separate 51Cr

    release cytotoxic assays using PBMCs obtained from nine patients

    receiving continuous infusion IL2 are included. Three ofthe assays used

    target cells coated with the chl4.18 antibody and fusion protein (-)

    and 3 assays used the antibody and fusion protein in soluble form

    ( ). For each experiment, the paired comparisons of ADCC by thechl4.18 or the fusion protein are connected by the line.

    to the medium during the 4-h assay with control IgG or mAb

    chl4.l8. Tumor-bound fusion protein elicited ADCC with both

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

    M2 1 target cells was comparable to that induced by mAb

    chl4.l8 alone. The level of cytotoxicity against the LA-N-S

    target cell was comparable to the augmented cytotoxicity ob-

    served when soluble IL2 was combined with chl4.l8. This

    similar pattern of lysis was also noted with the fusion protein

    added in soluble form (Fig. 4). These results suggest that for the

    LA-N-S neuroblastoma target, fusion protein is capable of aug-

    menting the level of ADCC observed with chl4.l8 alone.

    Fig. 7 shows the results from six separate experiments

    using PBMC samples from nine different cancer patients com-

    paring the cytotoxicity on both M2l melanoma and LA-N-S

    neuroblastoma targets. In three of the experiments, ch 14.18

    antibody or fusion protein was added in soluble form (as in Fig.

    5). and in three experiments the target cells were coated with the

    ch 14. 18 antibody or the fusion protein and washed free of any

    excess prior to the cytotoxic assay (as in Fig. 6). No reproduc-

    ible difference in ADCC was noted between coated targets or

    targets to which soluble fusion protein was added. Overall, the

    data presented in Fig. 7 show that the ADCC mediated by the

    ch I 4. 1 8 fusion protein was comparable to or better than that

    seen with an equivalent concentration of chl4.l8 antibody

    against both targets.

    DISCUSSION

    The fusion protein chl4. 18-1L2 was designed to create a

    molecule that would achieve enhanced in vivo effects over the

    combined use of the antibody and IL2 as separate molecules ( 10,

    1 1 ). The function of the antibody component of this fusion

    protein is to facilitate ADCC and to target the cytokine IL2 to

    the area of GD2� tumors. Thus, chl4.l8-IL2 is a single mole-

    cule containing both antitumor specificity and immunopotenti-

    Research. on June 26, 2021. © 1996 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • 1958 Anti-GD2-lL2 Fusion Protein

    ating capabilities. The results of in vitro experiments presented

    here confirm and extend previous reports demonstrating that the

    anti-GD2 cytokine fusion protein chl4.l8-IL2 retains the func-

    tional activity of both the antibody and IL2 (10). The fusion

    protein bound specifically to GD2� tumor cell lines, resulting in

    the same fluorescence intensity as the parent chl4. 1 8 antibody

    (Fig. 1 and Ref. 10). In addition, we demonstrated that tumor

    cell-bound fusion protein could be detected with antibody spe-

    cific for human IgG or IL2.

    A previous study using murine CTLL-2 cells as responders

    (10) demonstrated that the IL2 component of the chl4.l 8-1L2

    fusion protein was as active as native soluble 1L2 in inducing

    proliferative responses. In contrast, Fell et al. (22), using a

    fusion protein consisting of the Fab’ region of the human

    carcinoma-specific L6 antibody linked to IL2, found the specific

    activity of the IL2 component of that fusion protein to be

    200-fold less than native rIL2 when measured in a proliferative

    assay with the CTLL-2 cell line. We examined the ability of

    chl4.18-IL2 to stimulate proliferation of a human myeloid Ieu-

    kemia line, Tf-l�3, previously shown to respond to IL2 in a

    dose-dependent manner (18) and to stimulate PBMCs obtained

    from melanoma patients following 96-h continuous infusions of

    IL2. Both of these responding cell populations proliferate to IL2

    via interaction with the intermediate affinity (�3y�) IL2 receptor

    complex (2, 17, 21, 23). Both of these responding cell popula-

    tions demonstrated that the 1L2 component of the chl4. l8-IL2

    fusion protein was as stimulatory as soluble recombinant human

    1L2. In addition, the Mik �3l antibody specific for the �3 chain of

    the IL-2 receptor blocked these fusion protein-induced prolifer-

    ative responses, demonstrating that the activation was due to the

    IL-2 component. More importantly, we noted that tumor-bound

    fusion protein was capable of stimulating 1L2-induced prolifer-

    ative responses. Quantitative comparisons indicated that the 1L2

    component of the tumor cell-bound fusion protein was as stim-

    ulatory as soluble 1L2.

    Inclusion of 1L2 in vitro in the 51Cr release assay has been

    noted to augment 1L2-dependent LAK and ADCC activity me-

    diated by PBMCs obtained following in vivo therapy with IL2

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

    fusion protein also facilitates 1L2-dependent LAK killing of the

    Daudi target by patient PBMCs (data not shown). The current

    experiments demonstrate that the 1L2 component of the tumor-

    bound fusion protein also augments ADCC. When the fusion

    protein was used to coat tumor target cells prior to washing and

    inclusion in the cytotoxic assay, enhanced ADCC (over that

    induced by the chl4.l8 antibody) was noted with patient PB-

    MCs as effectors on the LAN-S target in over 50% of the assays.

    On the M2 I melanoma target, the level of ADCC was compa-

    rable to that seen with antibody alone. This ADCC activity

    extends the prior observation that chl4. l8-IL2 fusion protein

    can enhance T cell-mediated killing of an autologous tumor cell

    line (10). In the previously published case, using the human 660

    TIL line as a tumor-specific effector population, increased lysis

    of the autologous GD2� tumor was obtained by addition of the

    chl4. 18-IL2 fusion protein as compared to that mediated by

    addition of chl4.l8 antibody or IL2 alone (10).

    There is reason to hypothesize that in vivo localization of

    IL2 to the tumor via the tumor-selective chl4.18-1L2 fusion

    protein may induce more effective antitumor destruction than an

    equivalent amount of free soluble ]L2. Other studies have dem-

    onstrated that 1L2 produced in viva by tumor cells (following

    gene transfer) can result in enhanced rejection of the IL2-

    producing tumor cells (26). The fact that this rejection was

    immunologically mediated suggests that a higher local concen-

    tration of 1L2 may improve immune recognition of tumor.

    Previous findings with the 14.l8-IL2 fusion protein demon-

    strated suppression of human neuroblastoma tumor growth in an

    experimental hepatic metastases model in SCID mice (I I). In

    this model, human LAK cells and relatively low doses of

    chl4.18-IL2 fusion protein induced prolonged survival of ani-

    mals bearing micrometastases, comparable to the survival noted

    only with very high doses of recombinant human 1L2 (1 1).

    Mixtures of low doses of mAb chl4.18 plus IL2 do not effec-

    tively prolong the life span nor eradicate established metastases

    of neuroblastoma or melanoma in SCID mice, whereas the

    fusion protein chl4.l8-IL2 is able to accomplish both of these

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

    chl4.l8-IL2 fusion protein is effective against pulmonary and

    hepatic melanoma metastases in a syngeneic murine tumor

    model and that T cells are essential for achieving this antitumor

    effect. We note in the present in vitro study that in some

    instances, such as the lytic activity against M2l and LA-N-S

    targets, the fusion protein did not function any better than the

    combination of 1L2 and chl4.l8 as 2 independent reagents, yet

    four separate murine studies have noted a clear advantage of the

    fusion protein in vivo over the combination of soluble IL2 and

    chl4.l8 antibody (1 1-13, 27). This could be due to a number of

    factors. The human immunoglobulin component of the fusion

    protein actually lengthens the serum half life of IL2 (27) from a

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

    the chl4.l8-IL2 fusion protein. In addition, in vivo localization

    studies have shown targeting of anti-GD2 mAb to GD2� tumor

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

    can specifically localize to tumor sites in vivo and deliver IL2

    directly to tumor sites. This may result in augmented stimulation

    of IL2-responsive effector cells, including both NK and T cells.

    These preclinical in vitro data presented here and the

    previously obtained in vivo data from the munine xenograft and

    syngeneic models suggest that the chl4. 18-1L2 fusion protein

    prolongs the serum half-life of IL2, localizes to sites of GD2�

    tumor metastases, activates ADCC through FeR-bearing effec-

    tors, and activates IL2-responsive NK and T cells at tumor sites,

    potentially mediating a protective antitumor response. The cur-

    rent in vitro study documents that the 1L2 component of the

    tumor-bound fusion protein is as effective as soluble 1L2 in

    stimulating proliferation and cytotoxicity by effector cells ob-

    tamed from patients following in vivo therapy with IL2. The IL2

    component of the fusion protein facilitates ADCC of neuroblas-

    toma and melanoma targets by in vivo activated human effector

    cells. Toxicity testing of this fusion protein in experimental

    animals is now underway to determine how best to test ch 14. 18-

    IL2 in Phase I clinical trials for patients with GD2� tumors,

    including melanoma (29) and neuroblastoma.

    REFERENCES

    I. Rosenberg, S. A., Lotze. A. I., Yang, I. C., Aebersold, P. M.,Linehan, W. M., Seipp. C. A., and White, D. E. Experience with the use

    Research. on June 26, 2021. © 1996 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • Clinical Cancer Research 1959

    of high-dose interleukin-2 in the treatment of 652 cancer patients. Ann.

    Surg., 210: 474-484. 1989.

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

    Sosman, J. A.. Sugamura, K., and Sondel, P. M. Natural killer cells

    activated by Interleukin-2 treatment in rho respond to Interleukin-2

    primarily through the p75 receptor and maintain the p55 (TAC) negativephenotype. Cancer Res.. 50: 2683-2691 . I 990.3. Sosman, I. A., Kohler, P. C., Hank, J. A., Moore, K. H., Bechhofer,

    R.. Storer. B.. and Sondel, P. M. Repetitive weekly cycles of interleu-

    kin-2. II. Clinical and immunologic effects of dose, schedule. and

    indomethacin. I. Nail Cancer Inst., 80: 1451-1460, 1988.

    4. Sosman, J. A.. Hank. J. A., Moore, K. H., Borchert, A., Schell, K.,

    Kohler. P. C., Goldstein, D., Bechhofer, R., Storer, B., Albertini, M. R..Leung, R., Levitt, D., and Sondel, P. M. Prolonged interleukin-2 (IL-2)

    treatment can augment immune activation without enhancing antitumor

    activity in renal cell carcinoma. Cancer Invest., 9: 35-48, 1991.

    5. Phillips, J. H.. Gemlo, B. I.. Myers, W. W., Rayner, A. A., and

    Lanier, L. L. In vito and in vitro activation of natural killer cells in

    advanced cancer patients undergoing recombinant interleukin-2 and

    LAK cell therapy. J. Clin. Oncol.. 5: 1933-1941. 1987.

    6. McMannis, J. D., Fisher, R. I., Creekmore, S. P., Braun, D. P., Harris,

    J. E., and Ellis, T. M. In vito effects of recombinant IL-2. I. Isolation ofcirculating leul9+ lymphokine-activated killer effector cells from can-

    cer patients receiving recombinant IL-2. J. Immunol., 140: 1335-1340,1988.

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

    Reisfeld, R.. Seeger, R., Reynolds, C. P., Bauer, M., Wiersma, S.,

    Hammond, D., and Sondel, P. M. Treatment of neuroblastoma patientswith antiganglioside GD7 antibody plus interleukin-2 induces antibody

    dependent cellular cytotoxicity against neuroblastoma detected in vitro.

    J. Immunother., 15: 29-37. 1994.

    8. Ziegler, L. D., Palazzolo, P., Cunningham. J., Janus, M., Itoh, K.,

    Hayakawa, K., Helistrom, I., Hellstrom, K. E., Nicaise, C., and Dennin,R. Phase I trial of murine monoclonal antibody L6 in combination with

    subcutaneous interleukin-2 in patients with advanced carcinoma of the

    breast, colorectum, and lung. J. Clin. Oncol.. 10: 1470-1478, 1992.

    9. Weil-Hillman, G.. Fisch, P., Prieves, A. F., Sosman, I. A., Hank.

    J. A., and Sondel, P. M. Lymphokine-activated killer activity induced byin vito interleukin 2 therapy: predominant role for lymphocytes withincreased expression of CD2 and Leu I 9 antigens but negative expres-

    sion of CD16 antigens. Cancer Res., 49: 3680-3688, 1989.

    10. Gillies, S. D., Reilly, E. B., Lo, K. M.. and Reisfeld, R. A. Anti-

    body-targeted interleukin 2 stimulates I-cell killing of autologous tumor

    cells. Proc. Nail. Acad. Sci. USA, 89: 1428-1432, 1992.

    1 1 . Sabzevari, H., Gillies, S. D., Mueller, B. M., Pancook, I. D., and

    Reisfeld, R. A. A recombinant antibody-interleukin 2 fusion protein

    suppresses growth of hepatic human neuroblastoma metastases in severe

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

    9626-9630, 1994.

    12. Becker. J. C., Pancook, I. D., Gillies, S. D., Mendelsohn, I.,

    Reisfeld, R. A. Eradication of human hepatic and pulmonary melanoma

    metastases in SCID mice by antibody-interleukin 2 fusion proteins.

    Proc. NatI. Acad. Sci. USA, 93: 2702-2707, 1996.

    13. Becker. J. C.. Pancook, I. D., Gillies, S. D., Furukawa, K., andReisfeld, R. A. T cell mediated eradication of murine metastatic mela-

    noma Induced by targeted Interleukin 2 therapy. I. Exp. Med., 183:

    2361-2366. 1996.

    14. Gearing. A. J. H., and Thorpe, R. The international standard forhuman interleukin-2. Calibration by international collaborative study.J. Immunol. Methods, 114: 3-9, 1988.

    15. Mueller, B. M., Romberdahl, C. A., Gillies, S. D., and Reisfeld.

    R. A. Enhancement of antibody-dependent cytotoxicity with chimericanti-GD2 antibody. I. Immunol., 144: 1282-1286, 1990.

    16. Gillies, S. D.. Young, D., Lo, K-M., Foley, S. F.. and Reisfeld, R. A.Expression of genetically engineered immunoconjugates of lympho-

    toxin and a chimeric anti-ganglioside GD2 antibody. Hybridoma, /0:347-356, 1991.

    17. Sondel, P. M., Kohler, P. C., Hank. J. A.. Moore, K. H.. Rosenthal,N., Sosman, J., Bechhofer, R., and Storer, B. Clinical and immunolog-

    ical effects of recombinant Interleukin-2 given by repetitive weekly

    cycles to patients with cancer. Cancer Res.. 48: 2561-2567, 1988.

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

    G., Ju, G., and Sondel, P. M. Distinction between -y� detection andfunction in YT lymphoid cells and in the GM-CSF responsive human

    myeloid cell line. Tf-l. Blood, 86: 4568-4578. 1995.

    19. Tsudo, M., Kitamura, F., and Miyasaka, M. Characterization of theinterleukin 2 receptor B chain using 3 distinct monoclonal antibodies.

    Proc. NatI. Acad. Sci. USA, 86: 1982-1986. 1989.

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

    M., Payne, P. W., Waldman, T. A., Grant, A, J., and Tsien, W. H.Humanized Mik �3 I : a humanized antibody to the IL-2 receptor beta-chain that acts synergistically with humanized anti-TAC. J. Immunol.,

    15/: 1075-1085, 1993.

    21. Voss, S. D.. Sondel, P. M., and Robb, R. I. Characterization of the

    Interleukin 2 receptors (IL-2R) expressed on human natural killer cellsactivated in 100 by IL-2: association of the p64 IL-2R -y chain with theIL-2R y chain with the Il-2R 3 chain in functional intermediate-affinity

    IL-2R. I. Exp. Med., 176: 531-541, 1992.

    22. Fell. H. P.. Gayle, M. A., Grosmaire, L., and Ledbetter, J. A.

    Genetic construction and characterization of a fusion protein consisting

    of a chimeric F(ab’) with specificity for carcinomas and human IL-2.

    J. Immunol., 146: 2446-2452, 1991.

    23. Hank, J. A., Sosman, J. A., Kohler, P. C., Bechhofer, R., Storer, B.,and Sondel, P. M. Depressed in vitro T cell responses concomitant withaugmented Interleukin-2 responses by lymphocytes from cancer patients

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

    9. 5-14, 1990.

    24. Hank, J. A. Robinson, R. R., Surfus, I.. Mueller, B. M., Reisfeld,

    R. A., Cheung, N-K., and Sondel, P. M. Augmentation of antibodydependent cell mediated cytotoxicity following in vito therapy withrecombinant Interleukin-2. Cancer Res., 50: 5234-5239. 1990.

    25. Munn, D. H., and Cheung, N. K. V. Interleukin-2 enhancement of

    monoclonal antibody mediated cellular cytotoxicity against human mel-anoma. Cancer Res., 47: 6600-6605, 1987.

    26. Fearon, E., Pardoll, D., Itaya, T., Golumbeck. P., Levitsky. H. I..Simons, J. W., Karasuyama, H., Vogelstein, B., and Frost, P. Interleu-

    kin-2 production by tumor cells bypasses T helper function in thegeneration of an antitumor response. Cell, 60: 397-403, 1990.

    27. Gillies, S. D., Young, D., Lo, K-M., and Roberts, S. Biologicalactivity and in vito clearance of antitumor antibody/cytokine fusionproteins. Bioconjugate Chem., 4: 230-235, 1993.

    28. Reuland, P., Handgretinger, R., Smykowski, H., Dopfer, R.,Klingebiel. T., Miller, B. M., Reisfeld, R. A., Gallagher, S., Koscelniak,

    E., and Treuner, J. Application of the murine anti-GD-2 antibody 14.GD-2a for diagnosis and therapy of neuroblastoma. Intl. J. Radiat. AppI.Instrum. B, 18: 121-125, 1991.

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

    T.. Surfus, I., Reisfeld, R. A., Schiller, J. H.. and Sondel, P. M. SystemicInterleukin-2 treatment can inhibit the anti-idiotypic response to chi-

    meric anti-GD2 antibody in melanoma patients. I. Immunother.. inpress, 1996.

    Research. on June 26, 2021. © 1996 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • 1996;2:1951-1959. Clin Cancer Res J A Hank, J E Surfus, J Gan, et al. (ch14.18-IL2).recombinant anti-ganglioside GD2 interleukin-2 fusion protein Activation of human effector cells by a tumor reactive

    Updated version

    http://clincancerres.aacrjournals.org/content/2/12/1951

    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

    Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

    .http://clincancerres.aacrjournals.org/content/2/12/1951To request permission to re-use all or part of this article, use this link

    Research. on June 26, 2021. © 1996 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/content/2/12/1951http://clincancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://clincancerres.aacrjournals.org/content/2/12/1951http://clincancerres.aacrjournals.org/