a low-toxicity il-2 based immunocytokine retains anti-tumor activity … · 2011. 4. 29. ·...

39
A low-toxicity IL-2 based immunocytokine retains anti-tumor activity despite its high degree of IL-2 receptor selectivity Stephen D. Gillies * , Yan Lan**, Thore Hettmann # , Beatrice Brunkhorst, Yaping Sun, Stefan O. Mueller 1,2 and Kin-Ming Lo EMD Serono Research Institute, 45A Middlesex Turnpike, Billerica, MA 01821-3936, 1 Merck Serono, 64297 Darmstadt, Germany and 2 Karlsruhe Institute of Technology (KIT), Institut für Angewandte Biowissenschaften, 76128 Karlsruhe, Germany * Present address: Provenance Biopharmaceuticals, 830 Winter Street, Waltham, MA 02451 # Present address: U3 Pharma AG Bunsenstr. 1, 81243 Munchen, Germany ** Corresponding author: Yan Lan, e-mail, [email protected] Key Words: immunocytokines, interleukin-2, immune therapy, toxicity, bioactivity on July 9, 2021. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

Upload: others

Post on 19-Feb-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

  • A low-toxicity IL-2 based immunocytokine retains anti-tumor activity

    despite its high degree of IL-2 receptor selectivity

    Stephen D. Gillies*, Yan Lan**, Thore Hettmann#, Beatrice Brunkhorst, Yaping Sun,

    Stefan O. Mueller1,2 and Kin-Ming Lo

    EMD Serono Research Institute, 45A Middlesex Turnpike, Billerica, MA 01821-3936,

    1 Merck Serono, 64297 Darmstadt, Germany and 2 Karlsruhe Institute of Technology

    (KIT), Institut für Angewandte Biowissenschaften, 76128 Karlsruhe, Germany

    * Present address: Provenance Biopharmaceuticals, 830 Winter Street, Waltham, MA 02451 # Present address: U3 Pharma AG Bunsenstr. 1, 81243 Munchen, Germany

    ** Corresponding author: Yan Lan, e-mail, [email protected]

    Key Words: immunocytokines, interleukin-2, immune therapy, toxicity, bioactivity

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    2

    STATEMENT OF TRANSLATIONAL RELEVANCE

    The approach of selective IL-2 receptor activation such as the use of low-dose IL-

    2 in the clinic and the screening for IL-2 mutants that selectively activate the high affinity

    receptor is aimed at immune activation with less side effects. We have been using

    antibody targeting of cytokines to deliver immune stimulators to the tumor

    microenvironment in order to minimize systemic toxicity. In the current study we

    describe the characterization of such an immunocytokine called Selectikine, which

    contains an IL-2 (D20T) variant that is selective for both the human and mouse high

    affinity IL-2 receptors, and hence translational research can be performed in mouse

    models to evaluate anti-tumor efficacy and toxicity. Selectikine is currently in Phase I

    clinical trial, and this potent tumor-targeting immune stimulator with a low side effect

    profile is a good candidate for combination with traditional chemo and radiotherapies that

    by themselves have immune potentiating activity.

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    3

    ABSTRACT

    Purpose The goal of the study was to engineer a form of IL-2 that, when delivered as a

    tumor-specific antibody fusion protein, retains the ability to stimulate an anti-tumor

    immune response via interaction with the high affinity IL-2 receptor, but has lower

    toxicity due to the reduced activation of the intermediate affinity IL-2 receptor.

    Experimental Design We investigated changes in the proposed toxin motif of IL-2 by

    introducing a D20T mutation that has little effect on the activity of free IL-2. We

    expressed this IL-2 variant as a fusion protein with an antibody (NHS76) that targets the

    necrotic core of tumors, and characterized this molecule (NHS-IL2LT) in vitro and in

    vivo.

    Results NHS-IL2LT was shown to have near normal biological activity in vitro using T

    cell lines expressing the high affinity IL-2 receptor, but little or no activity using lines

    expressing only the intermediate IL-2 receptor. Relative to the control antibody fusion

    protein containing wild-type IL-2, NHS-IL2LT retained anti-tumor activity against

    established neuroblastoma and non-small cell lung cancer metastases in syngeneic mouse

    tumor models, but was much better tolerated in immune competent mice as well as in

    cynomolgus monkeys.

    Conclusions The demonstrated qualities of low toxicity and single agent efficacy

    suggest that NHS-IL2LT is a good candidate for therapeutic approaches combining

    standard cytotoxic and immune therapies. In fact, this molecule (also known as

    Selectikine or EMD 521873) is currently in Phase I clinical trial.

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    4

    INTRODUCTION

    Despite many years of clinical use, the mechanisms of IL-2 toxicity and

    therapeutic efficacy for cancer are not well understood. Clinical dose schedules showing

    the highest objective response rates in renal carcinoma and melanoma are associated with

    severe toxicities, especially those of the vascular compartment such as vascular leak

    syndrome (1). Many mechanisms have been proposed for this vascular toxicity. In one

    case, toxicity has been attributed to direct binding of IL-2 to endothelial cells via a motif

    resembling a component of bacterial toxins (2) and centered around aspartic acid residue

    20 (D20). Another group has reported a vasopermeability enhancing fragment of IL-2

    extending from residues 22-58 that increases vascular permeability independent of IL-2

    bioactivity (3). A third group of investigators has proposed that activation of cells bearing

    the intermediate affinity IL-2 receptor in the vascular compartment leads to inflammatory

    cytokine release by NK and other cells (4). In the latter case, it was proposed that a

    receptor-selective form of IL-2, i.e. one that effectively binds the high affinity and not the

    intermediate affinity IL-2 receptor, would avoid this activation in the vascular

    compartment, but provide IL-2 to activated T-cells expressing the high affinity receptor.

    This approach of selective receptor activation is not unlike the use of low-dose

    IL-2 in the clinic that has been shown to result in immune activation with far less side

    effects (5). Surprisingly, this treatment regimen appears to result in the expansion of a

    subset of CD56bright NK cells rather than T cells, at least in the circulation, so it is not

    clear whether this approach can effectively stimulate an anti-tumor T cell response. It is

    generally accepted that this treatment approach results in fewer long term clinical

    responses (1).

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    5

    One potential reason for this lack of efficacy with low-dose protocols may be due

    to the inability to deliver sufficient levels of IL-2 to the tumor microenvironment where it

    is needed to activate anti-tumor T cells. We and others have been using antibody

    targeting of cytokines (immunocytokines) such as IL-2 to deliver these immune

    stimulators to the tumor microenvironment with the hope of increasing efficacy without

    associated toxicity (reviewed in ref (6)). Numerous studies have documented that it is

    possible to generate (or enhance) potent T cell responses to tumors in mice and that these

    responses are mostly MHC class I restricted CD8 T cell responses, and are dependent on

    specific tumor targeting. Unlike many studies with IL-2, our treatment is based on short-

    term dosing (e.g. 3 to 5 successive days followed by at least two weeks without dosing)

    rather than repeated dosing over extended periods. The consequence is that we do not

    provide IL-2 to cells after their initial activation that includes up-regulation of high

    affinity IL-2 receptor and enhanced responsiveness. In this way toxicity is minimized, but

    apparently the targeting aspect results in efficacy that is not possible with short term

    dosing with non-targeted IL-2. In fact, many of our studies have included the use of IL-2

    (at equal or higher doses) that was found to be completely ineffective in situations where

    our targeted molecules were completely effective in eradicating tumor cells (7). In the

    current study, we used an antibody NHS76 that was selected from a phage display library

    using Raji Burkitt’s lymphoma nuclear extracts as the binding ligand (8), to target the

    DNA/histone complex in the necrotic core of tumors. Such Tumor Necrosis Treatment

    (TNT) antibodies have been shown to be effective in targeting tumors in animal models

    and cancer patients (9, 10).

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    6

    Despite the ability to target IL-2 to tumors, systemic administration of

    immunocytokines still exposes the cells in the vascular compartment to IL-2 prior to their

    delivery to the tumor. Therefore we have investigated ways of reducing their potential

    toxicity in order to increase the doses that can be administered. We first investigated

    changes in the proposed toxin motif (discussed above) by testing mutations of D20 of IL-

    2 that were reported to maintain normal IL-2 bioactivity. One such mutation to threonine

    (D20T) has little or no effect on activity of free IL-2, but would be predicted to eliminate

    the toxin motif responsible for endothelial cell binding (2). Surprisingly, when this form

    of IL-2 was expressed as a whole antibody immunocytokine, we found it to be highly

    specific for activating the high affinity IL-2 receptor. The resulting molecule would be

    expected to have two beneficial properties including selectivity for activated T cells, and

    reduced binding to endothelial cells. In the current study we describe the characterization

    of this molecule in vitro and in vivo, and demonstrated for the first time that a form of IL-

    2 with receptor specificity for the mouse high affinity IL-2 receptor can demonstrate anti-

    tumor responses without the normal levels of IL-2 toxicity. An immune stimulator with

    such a low side effect profile is a good candidate for combination with traditional chemo

    and radiotherapies that by themselves have immune potentiating activity (11).

    MATERIALS AND METHODS

    Cell lines and animals. The mouse myeloma NS/0 cell line was obtained from

    the European Collection of Animal Cell Cultures (Salisbury, UK). The murine T cell line

    CTLL-2 and the human astrocytoma U-87 MG were obtained from American Type

    Culture Collection (Rockville, MD). The murine neuroblastoma NXS2 was a gift from

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    7

    Dr. Ralph Reisfeld at Scripps Research Institute. The murine Lewis lung carcinoma

    (LLC) was a gift from the late Dr. Judah Folkman at Children’s Hospital, Boston. The

    human T cell line Kit-225 (K6) was provided by Dr. Angus Grant, EMD

    Pharmaceuticals, Durham, NC. The human TF-1β cell line was provided by Dr. Paul

    Sondel, University of Wisconsin at Madison, WI. All cell culture media were purchased

    from Invitrogen (Carlsbad, CA) and all cytokines from R&D Systems (Minneapolis,

    MN). C57Bl/6 (female), A/J (female), SCID CB17 mice (male), and BALB/c (female)

    mice (8-9 weeks old) were purchased from Taconic Farms (Germantown, NY) and

    Jackson Lab (Bar Harbor, ME).

    Expression and purification of NHS-IL2 immunocytokines. The pdHL vector

    used for the expression of NHS-IL2 immunocytokines was described previously (12).

    The genes encoding the V regions of the NHS antibody were based on the protein

    sequences of NHS76, a single-chain Fv derived from screening a human scFv phage

    library against Raji Burkitt’s lymphoma cell nuclear extracts (8). The VH and the VL

    were chemically synthesized, using optimized codons for mammalian expression. The

    VH was joined to the human IgG2 constant regions, which was in turn fused in frame to

    the sequence encoding the mature IL-2 in the vector. At the fusion junction, the C-

    terminal lysine residue of the CH3 was changed to alanine to increase serum half life

    (13). The sequence of the VL revealed that it was derived from a human lambda chain

    and contained the J3 region, hence it was joined to the constant region of human lambda

    3, which was used to replace the human kappa chain in the pdHL vector. The D20T

    mutation in the IL-2 moiety and the N297 mutation in the CH2 domain were introduced

    by overlapping polymerase chain reaction. The antibody-IL2 fusion proteins were

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    8

    expressed in NS/0 cells by transfection and selection of producer cell clones as described

    (13). Proteins were purified from conditioned cell culture media by binding to and

    elution from protein A Sepharose, followed by diafiltration into PBS.

    IL-2 bioactivity assays. The IL-2 activity of IL-2 containing immunocytokines

    were assayed in standard T-cell proliferation assays using the mouse CTLL-2 cell line

    (14), human T cell line Kit-225 (K6) (15) or human TF-1β cell line (16), which are all

    dependent on IL-2 for growth. The CTLL-2 cell line expresses the high affinity mouse

    IL-2 receptor, the Kit-225 (K6) cell line expresses the high affinity human IL-2 receptor

    (17), and the TF-1β cell line expresses only the intermediate affinity human IL-2

    receptor. For the Kit-225 assay proliferation was measured by the reduction of Alamar

    Blue instead of 3H thymidine uptake (18). Briefly, washed Kit-225 cells that had been

    starved for 4 days in AIM-V serum free media (12,500 cells/well) were incubated with

    IL-2 or IL-2 containing immunocytokines for 36 hours. Alamar Blue (Trek Diagnostics

    Cleveland, OH) was then added (30 μl/well) and the incubation continued for an

    additional 34 hours. The fluorescence was then read using a fluorescence plate reader

    (excitation 530 nm, emission 590 nm). The maximum response used for ED50 calculation

    was attained using an internal reference standard in each assay. The ED50 concentration

    was calculated using least squares analysis (TREND analysis from Excel).

    Mouse NK cell isolation and bioassay. Cells were harvested from spleens taken

    from C57Bl6 mice and NK cells enriched using a kit from Stem Cell Technologies

    (Vancouver Canada). NK cells were separated from the rest of the cells through negative

    selection using a depletion cocktail tailored to highly enrich NK cells from suspensions

    of murine spleen cells. NK cell purity was measured using cell staining for NK1.1 and

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    9

    DX5, known mouse NK cell markers. Cells were incubated with IL-2 or IL-2 containing

    immunocytokines for a total of 96 hours with 3H thymidine added during the last 16

    hours. The cells were harvested from the wells with water onto glass microfiber filter

    plates and radioactivity was measured by liquid scintillation counting.

    PBMC proliferation and FACS analyses. PBMC from normal donors were

    prepared by Ficoll gradient centrifugation and labeled with carboxyfluorescein diacetate

    succidimidyl ester (CFSE, Molecular Probes Inc., Eugene OR) for 15 min at 37o C. After

    washing with culture medium (RPMI containing 10% FBS), 2 x 106 cells were cultured

    per ml, alone or with added IL-2 or immunocytokine. In some cases, cultures included

    OKT3 anti-CD3 antibody at a final concentration of 0.1 μg/ml. Cell samples were

    collected at the indicated time points and stained with the following antibodies: anti-

    CD56PE, anti-CD4PE, anti-CD8PE and anti-CD25FITC according to the instructions of

    the manufacturer (BD Pharmingen, San Diego, CA). After incubation on ice for 30 min

    and washing, labeled cells were analyzed using a Coulter Epics XL-MCL flow cytometer.

    Pharmacokinetic analysis. BALB/c mice were injected with 25 μg of an

    immunocytokine in a volume of 0.2 ml in the tail vein using a slow push. At various time

    points, small blood samples were taken by retro-orbital bleeding and collected in tubes

    coated with heparin to prevent clotting. After centrifugation to remove the cells, the

    plasma was assayed by capture with anti-human IgG H&L antisera and detection with an

    anti-human IL2 antibody (12). Results were normalized to the initial concentration in the

    serum of each mouse taken immediately after injection.

    Pilot toxicity study in cynomolgus monkey. The animal experiment was

    approved by the local authorities and was conducted in compliance with the principles of

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    10

    Good Laboratory Practice (GLP) as well as the local animal welfare and health

    regulations. In this pilot monkey toxicity study (performed by MDS Pharma Services,

    France), a group of 2 male and 2 female cynomolgus monkeys (Macaca fascicularis) with

    an age of 25 to 27 months at the beginning of treatment were treated in an 3-week cyclic

    regimen that reflected the intended clinical dosing scheme of NHS-IL2LT. Animals were

    treated by a 1-hour intravenous (i.v.) infusion on 3 consecutive days (day 0 – day 2)

    followed by an 18-day treatment-free period (21-day cycle) for 3 cycles. The vehicle

    control was 0.9% (v/v) NaCl solution. NHS-IL2LT was administered at 1, 3 and 10

    mg/kg/d in 0.9% (v/v) NaCl solution. Animals were analyzed for standard toxicity

    parameters, i.e. clinical observations, body weight determinations, vital functions (e.g.,

    blood pressure measurements, electrocardiogram (ECG)), clinical pathology (hematology

    including lymphocyte subset analysis, serum chemistry, urinalysis), toxicokinetic and

    immunogenicity investigations, and histopathology examinations.

    Blood samples for measurement of the α-subunit of the soluble IL-2 receptor

    (sIL-2Rα) (2ml samples collected in tubes containing EDTA) and lymphocyte subtyping

    (1 ml samples collected without anticoagulant) were taken at day 0, 3, 5, 7 and 14 in each

    cycle. Blood samples were analysed by flow cytometry using a FACScan (Becton

    Dickinson, France) with anti-CD4 (FITC-labeled; #556615), anti-CD8 (PerCP - Cy5.5-

    labeled; #341050 and #341051) and anti-CD25 antibodies (PE-labeled; #557138, BD

    Pharmingen, France) according to the instruction provided by the manufacturer. SIL-2Rα

    was measured using a commercially available ELISA kit (#DR2A00, R&D Systems,

    France) according to the method described by the supplier.

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    11

    Tumor models. Experimental liver or lung metastases were induced by tail vein

    injection of viable single cells of NXS2 (0.4 x 106) or Lewis Lung Carcinoma (0.5 x 106)

    in 0.2 ml PBS into A/J mice or C57Bl/6 mice (n=7), respectively, on Day 0. Mice

    received 5 daily intravenous injections of PBS, fusion proteins, or free IL-2 with or

    without the naked NHS antibody, on days 4 to 8. Metastases were scored and organ

    weights were measured on day 28 as previously described (12). For depletion studies,

    mice received intraperitoneal injections of 50 μg of rat IgG2b anti-murine CD4 mAb

    (clone 2.43, TIB-210, ATCC), 100 μg of rat IgG2b anti-murine CD8 mAb (clone GK1.5,

    TIB-207, ATCC), or 20 μl of NK-cell-specific rabbit anti-asialo GM1 antiserum

    (WAKO, Richmond, VA) on day 3 and once weekly thereafter for 3 weeks, which had

    been shown previously to result in about 95% depletion of the respective T cell subsets or

    NK cells by indirect immunofluorescence staining and cytofluorometric analysis of

    lymph nodes and spleens.

    RESULTS

    Bioactivity of a mutant IL-2 immunocytokine. We originally tested the D20T

    mutation of IL-2 based on data suggesting that this change conferred no selectivity for

    activating either the intermediate or high affinity forms of the IL-2 receptor. Such a

    mutation would allow us to test the effect of removing this key residue in the proposed

    LDL toxin motif (2) and its possible role in the vascular toxicity of IL-2, independent of

    its receptor selectivity. While this mutation has little selectivity as free IL-2 (unpublished

    data), we found that when it is fused to the carboxyl terminus of the human IgG H chain,

    the selectivity for the high affinity receptor-mediated proliferation is increased

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    12

    dramatically. Biological activity as measured by cell proliferation was compared using

    two human cell lines: the Kit-225 T cell line expressing both intermediate βγ and high

    affinity αβγ, and TF-1β, an erythroleukemic line naturally expressing the common γ

    chain and transfected to express the human β chain. The resulting cell line responds to

    IL-2 through the heterodimeric βγ intermediate affinity receptor (16). Bioactivity of the

    final antibody-IL2 fusion molecule, NHS-IL2LT (LT stands for low toxicity), containing

    the D20T mutation, was compared by dose titrations to both free human rIL-2 as well as

    the same immunocytokine bearing wild-type IL2, NHS-IL2. While the ability of NHS-

    IL2LT to induce proliferation of Kit-225 cells was reduced 4-5 fold relative to free IL-2,

    the ability to induce proliferation of TF-1β was reduced more than 6000 fold (Fig. 1A

    and B). This resulted in selectivity for high affinity IL-2R of more than 1000-fold. Since

    we would be using mouse models for in vivo testing of toxicity and anti-tumor efficacy,

    we also examined the receptor selectivity on murine immune cells. In this case we used

    the standard IL-2 responsive T cell line, CTLL-2, for the high affinity measurement and

    purified mouse NK cells for measuring response through the intermediate IL-2R. We

    found that the NHS-IL2LT mutant immunocytokine had an even more profound

    selectivity for the high affinity IL-2R (Fig. 1 C and D). We figured that such biological

    activity assays are more sensitive and meaningful than just measuring binding affinity to

    the different receptors, because the selectivity for activating the high affinity receptor

    may not be completely accounted for by the difference in binding. In fact, we found that

    NHS-IL2LT retained binding affinities to both the high and intermediate affinity IL-2

    receptors (Supplementary Fig. 1A and B), and its differential activities on the

    proliferation of Kit225 and TF-1β cells seemed to be mainly due to impaired signaling, as

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    13

    measured by phosphorylated Stat5a (Supplementary Fig. 1C and D). NHS-IL2LT

    attained substantial levels of pStat5a signaling on TF-1β cells only at very high

    concentrations of about 100 nM to 1 μM (i.e., containing respectively 1800 and 18000

    ng/ml of IL-2 equivalents, Supplementary Fig 1D), consistent with the proliferation data

    of these cells in Fig. 1B.

    These results are in contrast to earlier studies with the N88R mutant form of IL-2

    that reported selectivity for high versus intermediate IL-2R, but was later found to

    demonstrate this effect only for human and monkey cells but not for mouse immune cells

    (19). Therefore, in the case of NHS-IL2LT, it is justified to use mouse models to

    determine relative toxicity and efficacy and thereby establish any potential improvements

    in the therapeutic index. Although not the original intention of our studies, we found it

    possible to test for the first time in relevant tumor models, the hypothesis that a form of

    IL-2 with selectivity for the high affinity IL-2R would be less toxic and still maintain

    anti-tumor efficacy. We also found in these studies that immunocytokines with wild type

    IL-2 also show a moderate degree of selectivity for the high affinity over the intermediate

    form of IL-2R, relative to free IL-2 (Fig. 1). We have also confirmed this to be the case

    with other immunocytokines utilizing different antibody V regions (data not shown).

    IL-2 receptor selectivity in cultured PBMC. Since both mouse and human

    immune cells responded with the same degree of IL-2 receptor selectivity, we used

    human PBMC to study differential proliferative responses in CD56+ NK cells as well as

    CD4+ and CD8+ T cell populations. We also monitored expression of the CD25

    component of the high affinity receptor in the same experiment. For proliferation

    analysis, resting PBMC were labeled with CFSE and cultured with rIL-2 or equimolar

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    14

    amounts of either NHS-IL2 or NHS-IL2LT immunocytokines. Significant proliferation of

    NK cells were observed in cultures containing rIL-2 and NHS-IL2 as visualized by CFSE

    dilution in the CD56 gated population on days 6 and 9 of culture (Fig. 2A). In contrast,

    little or no proliferation was observed in the culture containing NHS-IL2LT as measured

    by CFSE dilution. This result is consistent with the fact that little or no CD25 expression

    was observed in this CD56+ cell population and that only cells expressing intermediate

    receptor accounted for the observed proliferation. Despite this lack of proliferation, the

    number of CD56+ cells in the culture supplemented with NHS-IL2LT was increased on

    day 6 compared to the culture with no added IL-2, suggesting that NK cell survival was

    prolonged by this molecule.

    Analysis of proliferation and IL-2R expression in T cell populations was

    performed in the same manner, but in the presence and absence of anti-CD3 antibody

    stimulation. Again, PBMC were labeled with CFSE and cultured with rIL-2, ΝΗS-IL2 or

    NHS-IL2LT. In this case, CD4+ and CD8+ cells were gated and analyzed separately for

    proliferation and CD25 expression. In the absence of anti-CD3 stimulation, little or no

    CD4+ T cell proliferation was observed with rIL2 or either immunocytokine (Fig. 2B),

    since the IL-2Rβ chain is expressed constitutively in CD8 T cells, but not in un-

    stimulated CD4 T cells (20). In cultures activated by anti-CD3 antibody, strong

    proliferative responses were seen by day 6 in all cultures, including those with NHS-

    IL2LT. Response to this molecule correlated with a strong up-regulation of CD25 in the

    CD4 gated cell population as measured on day 6 of culture (Fig. 2B). Since anti-CD3

    activation is known to induce CD25 expression (21), we assume that the CD4+CD25+

    cells observed at this time have survived and proliferated in response to the selective

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    15

    NHS-IL2LT molecule, whereas the culture containing anti-CD3 antibody but no IL-2

    would not have maintained expression of CD25 until day 6.

    When CD8+ cells were examined in the same cultures, a somewhat different

    pattern of response was observed. In this case, cultures without anti-CD3 activation

    showed proliferative responses to the wild-type forms of IL-2, i.e. those capable of

    triggering both intermediate and high affinity receptors, but not to the selective NHS-

    IL2LT molecule (Fig. 2C). Also unlike CD4+ cells, some degree of CD8+ cell

    proliferation was observed in response to anti-CD3 antibody alone (no added IL-2), but

    that the addition of either the wild-type or mutant IL-2 molecules increased the number of

    cell divisions dramatically and to roughly the same extent. Again, the ability of NHS-

    IL2LT to stimulate cell proliferation was associated with strong induction of CD25

    expression in the CD8 gated population in the anti-CD3 activated culture.

    Effects on pharmacokinetic behavior in mice. Before comparing wild-type and

    mutant immunocytokines, it was essential to eliminate other parameters that could effect

    vascular toxicity and anti-tumor efficacy – especially changes in pharmacokinetics. We

    reported earlier that the fusion of IL-2 to an antibody molecule has unpredictable

    consequences to the pharmacokinetic behavior of immunocytokines in mice and man (22,

    23). Generally, the distribution α phase in the blood is far more rapid than that of a whole

    antibody and the t1/2 in the vascular compartment is shorter as well. These effects can be

    partially abrogated by reducing FcR binding or by modifying the linker between the

    antibody and IL-2 so that intracellular degradation is reduced (13). The NHS-IL2

    immunocytokines described in this study have been optimized for both reduced FcR

    binding (by using the γ2 isotype) and intracellular degradation (by mutating the C-

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    16

    terminal residue of the CH3 domain from K to A). Despite these changes, we found that

    the mutation of D20 to T had a dramatic effect on clearance from the circulation,

    primarily as a result of prolonging the distribution α phase (t1/2 α = 0.62 hr, t1/2 β = 4.6 hr,

    AUC (area under the curve) = 18.7 μg-hr/ml), as compared to that of NHS-IL2 (t1/2 α =

    0.93 hr, t1/2 β = 5.1 hr, AUC = 75.0 μg-hr/ml. Interestingly, this could be reversed by

    removing the N-linked glycosylation at N297 of the CH2 domain, either by enzymatic

    treatment (not shown) or through mutation to N297Q. This results in a longer exposure

    time for the aglycosylated form in the circulation (AUC = 59.4 μg-hr/ml), despite

    similarities in clearance rates (t1/2 α = 0.79 hr, t1/2 β = 4.9hr). The same phenomenon was

    observed with other IgG isotypes indicating that this was not particular to the use of the

    γ2 isotype (not shown). Since our antibody targets the necrotic portion of tumors through

    binding of DNA released from dying cells, use of such a non-FcR binding antibody

    should have little impact on anti-tumor efficacy.

    Tolerability of NHS-IL2LT in mice. Our initial tolerability studies utilized

    immune competent mice that make high-titered antibody responses to this human fusion

    protein beginning around day 5. For this reason we limited intravenous (i.v.) dosing to

    five consecutive days and increased dose amounts until significant body weight loss or

    death were observed. For NHS-IL2 containing wild-type IL-2, a dosing regimen of 50

    μg/mouse/day x 5 days resulted in animal deaths consistently in multiple experiments

    (Fig. 3A). This is a lower tolerated dose than we had seen in earlier studies with other

    immunocytokines, but was likely due to this molecule’s longer circulating t1/2 that led to

    accumulation with daily administration. In contrast, using NHS-IL2LT, daily i.v. doses of

    1 mg/mouse/day x 5 could be administered with no animal deaths, however at this point,

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    17

    mice were beginning to show signs of toxicity including transient weight loss (Fig. 3B).

    These data demonstrate that NHS-IL2LT is greater than 20-fold less toxic than the same

    molecule containing wild-type IL-2 when administered with this dosing schedule.

    Since treatment with IL-2 is expected to modulate IL-2R on immune cells, we

    also examined longer dosing schedules to see if there was IL-2R up-regulation or

    expansion of cells with high affinity IL-2R. Due to anti-immunocytokine antibody

    responses, we performed these studies in immune deficient SCID mice recognizing that

    the lack of functional B and T cells would likely make any results difficult to interpret in

    the context of immune competence. In the study shown in Fig. 3C, consecutive dosing for

    5 days repeated weekly resulted in significant toxicity and death with doses as low as 20

    μg per day, and with a delay of only about 5 days compared to the NHS-IL2 wild type

    control molecule. Additional pilot experiments in SCID mice identified the shortest time

    between 3-day repeated courses of treatment was 21 days (not shown), a schedule that is

    quite similar to those used for standard chemotherapies. These data show that the benefit

    of receptor selectivity is lost with continuous dosing, but that intermittent dosing

    protocols may be able to maintain the low toxicity benefit of this immunocytokine and its

    potential for use in combination therapies. In order to test this hypothesis in a more

    relevant model, where biological responses are more similar to humans, and where

    immunogenicity should be far less pronounced, we next studied repeated intermittent

    dosing in non-human primates.

    Toxicity and pharmacological activity of NHS-IL2LT in cynomolgus

    monkey. We assessed NHS-IL2LT in a GLP-compliant pilot toxicology study in

    cynomolgus monkeys, a relevant species for the assessment of IL-2 toxicities (24). The

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    18

    lowest dose administered (1 mg/kg) corresponded to the highest, but poorly tolerated,

    dose we have administered to cynomolgus monkeys using other immunocytokines

    containing un-mutated IL-2. The other doses were 3 and 10-fold higher. The maximum

    tolerated dose (MTD) of immunocytokines in humans have been determined to be 6.4

    and 7.5 mg/m2 for huKS-IL2 and hu14.18-IL2, respectively, when administersed as once-

    daily 4-hour infusions in a 3-day cycle repeated every 4 weeks (23, 25). This MTD range

    for humans corresponds to about 0.52 to 0.61 mg/kg for monkeys based on allometric

    scaling (26). However, the actual MTD of IL-2 containing immunocytokines in monkeys

    was determined empirically to be several-fold lower, about 0.1 mg/kg, and monkeys and

    importantly, humans dosed at their respective MTD’s had very similar Cmax and AUC

    (23, 25, and unpublished data).

    Monkeys were treated using the intended clinical regimen, i.e., a 21-day cycle

    consisting of treatment (1 h, i.v infusion) on 3 consecutive days (day 0-2) followed by an

    18-day treatment-free period, which is a more aggressive schedule with the anticipated

    lower toxicity of Selectikine. Toxicokinetic analysis showed a systemic and dose-

    proportional exposure of all NHS-IL2LT-treated animals. The peak plasma levels were in

    the range of 103 to 161 µg/ml in high dose treated male and female animals, which are

    many times higher than the peak plasma levels of 2 to 5 µg/ml obtained at the MTD of

    huKS-IL2 and hu14.18-IL2 in humans (23, 25), and in monkeys (unpublished data).

    Although most animals developed low levels of antibodies against NHS-IL2LT, this fact

    did not affect its kinetic properties and therefore drug exposure. NHS-IL2LT elicited no

    overt clinical signs or changes in body weight. In addition, no treatment-related effects at

    the injection sites or on cardiovascular parameters were observed. Treatment-related

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    19

    alterations with respect to hematology, biochemistry and pathology were mild to

    moderate; the major target organs being the lymphatic system, liver, kidney and intestine

    with lymphoid cell infiltrations. In summary, NHS-IL2LT induced moderate and typical

    IL-2-derived toxicity (27) and was clinically well tolerated using the intermittent dosing

    scheme, defined in our mouse models, up to the high-dose of 10 mg/kg tested.

    In addition to the comprehensive toxicity assessment, we also evaluated some

    pharmacological IL-2 response in the cynomolgus monkeys to prove the relevance of this

    species and to show NHS-IL2LT’s pharmacologic activity in vivo. We first analyzed the

    response of total lymphocytes, T-lymphocytes (CD3+) and NK (CD8+ CD16+) cells to

    NHS-IL2LT treatment. The total lymphocytes (Fig. 4A) and T-cells (Fig. 4B) first

    showed a very mild lymphopenia (considering these high doses) followed by a strong

    lymphocytosis around days 5 and 7 that were increased (2-3 fold) over baseline,

    compared to the vehicle control. Interestingly, all dose groups showed essentially the

    same degree of total lymphocyte and T cell expansions indicating saturation of receptors

    at the lowest dose level tested. NK cells showed a somewhat different pattern of response

    (Fig. 4C). In this case, the degree of lymphopenia was much stronger as a percentage of

    total NK cells, and lymphocytosis in the lowest dose group was clearly reduced compared

    to the two higher dose groups. In fact, the peak level after recovery in the low dose group

    was essentially a return to baseline. This level of response was not different from what

    was observed in the vehicle control group.

    We then looked at more specific IL-2 markers as the soluble IL-2 receptor α-

    subunit (sIL2Rα) and CD25 positive (activated) CD4+ and CD8+ lymphocytes in serum.

    The soluble form of the high affinity IL-2 receptor subunit α increases in serum

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    20

    concomitant with its cellular expression. Although the function of sIL2Rα is unclear, it

    correlates with increased T-cell and immune system activation (28). It has been shown

    previously that mutated IL-2 molecules are able to induce activated (CD25+) T-Helper

    (CD4+) as well as cytotoxic T-cells (CD8+) cells in non-human primate species (4).

    Cynomolgus monkeys showed a typical and strong IL-2 response with a sharp and dose-

    dependent increase in sIL2Rα one day after the last treatment in each cycle (day3, 24, 45)

    (Fig. 4D), whereas CD25 positive CD4+ (Fig. 4E) and CD8+ (Fig. 4F) cells increased on

    day 5 in each cycle. The peak expansion of activated CD4+ and CD8+ cells as well as the

    increase of sIL2Rα was 20-40-fold compared to the vehicle control. Overall the effects

    were reversible and returned to baseline values at the end of each cycle (day 20 and 41).

    These results confirmed our observations in vitro and show that NHS-IL2LT also has a

    selective pharmacological activity on CD4/CD8 positive cells in non-human primates.

    Anti-tumor activity in mouse models. Next we tested whether treatment with a

    low-toxicity form of IL-2 could show efficacy in mouse tumor models. We were also

    interested in testing whether tumor necrotic targeting could be effective in the setting of

    minimal residual disease. Previous studies of the uptake of radiolabeled antibodies in

    mouse tumor models had demonstrated that tumor necrosis can be used to target

    micrometastases in well oxygenated organs when lesions are greater than 20 to 30 cells in

    diameter (9). At this stage, cells undergoing apoptosis are known to express DNA or

    DNA containing complexes on their surface (29, 30). Furthermore, for tumor cells with

    high rates of mutation and daughter cell deaths, in vitro experiments using tumor

    spheroids to model micrometastases had shown that 20 to 30% of the cells in the

    proliferating rim are non-viable and necrotic (31).

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    21

    To test anti-tumor activity, we induced experimental lung metastases by

    intravenous injection of a lethal does of LLC cells in immune competent Bl/6 mice or

    experimental liver metastases by intravenous injection of NXS2 neuroblastoma cells in

    immune competent A/J mice. Treatment was initiated on day 4 when only small

    metastases have been established. For these studies we limited dosing to 5 consecutive

    days to avoid treatment after IL-2R up-regulation and thereby maintained receptor

    selectivity. Animals were sacrificed when control mice showed signs of toxicity from

    tumor burden (approximately day 28) and lungs or livers were removed for analysis of

    tumor burden by two criteria. Surface metastases were apparent after staining, but most

    surface lesions tended to fuse and could not be easily counted. Therefore, surface

    metastases were semi-quantitated by estimation of percent surface coverage. For the LLC

    model we first correlated this assessment method with tumor burden as measured by

    histological examination after H&E staining, and found a strong correlation between %

    surface coverage and overall tumor burden (Fig. 5A and B). In this experiment potent

    antitumor activity was demonstrated with a daily dose of 80 μg and an intermediate level

    of activity was observed using 20 μg per dose. The control groups treated with either the

    same amounts of IL-2 or combinations of IL-2 and the NHS antibody showed no activity

    at all in this model.

    In the NXS2 neuroblastoma model we measured both % surface coverage by

    metastases as well as overall organ weight as a measure of tumor burden. Again we

    confirmed that diseased livers consisted of tumor mass by H&E staining (not shown).

    Therefore, tumor-containing liver weight (approximately 5% of body weight) in excess of

    normal liver weight was taken as a measure of tumor mass. Under these conditions we

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    22

    found that equal doses of NHS-IL2 and NHS-IL2LT showed nearly the same level of

    activity as assessed by the percent of surface coverage by tumor (Fig. 6A), and both

    treatment groups were significantly different from the PBS control (P< 0.001). Treatment

    was somewhat better with NHS-IL2, but this was not significant (P = 0.18). When data

    were assessed by organ weights – a more objective measure of tumor burden - the

    similarity between the treatment groups (shown as individual animal organ weights) were

    even more striking. Recombinant IL-2 had no anti-tumor activity in the same model (not

    shown).

    In order to assess what effector cells were responsible for anti-tumor activity, we

    tested the effect of antibody depletion of CD4, CD8 or NK cells in the LLC experimental

    metastasis model. Depletion of the respective cell type was confirmed by FACS analysis

    of peripheral blood for each antibody. The in vivo efficacy results showed that depletion

    of CD4+ cells did not abrogate the anti-tumor effect of the two immunocytokines

    containing either wild-type or mutated IL-2 (Fig. 6B) and may have improved their

    activities, although the difference was not statistically significant (P = 0.16 for NHS-IL2

    and 0.096 for NHS-IL2LT). This improvement in anti-tumor activity suggests that

    elimination of CD4+CD25+ Treg cells might make this treatment approach more

    effective.

    In contrast to the results with CD4 depletion, treatment with anti-CD8 antibody

    resulted in a complete loss of anti-tumor activity for both NHS-IL2 and NHS-IL2LT (Fig.

    6B), indicating that CD8+ cells are the primary effectors in this model. This is in

    agreement with most of our earlier results in other syngeneic tumor models (6). The

    results with NK cell depletion were less dramatic and suggested some role for these cells

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    23

    in anti-tumor activity (Fig. 6B), possibly providing a supporting function to the more

    essential CD8+ effector cells. Interestingly, there was no difference between the groups

    treated with NHS-IL2 and NHS-IL2LT with respect to NK cell depletion, despite the

    dramatic selectivity differences in cytokine response in vitro and toxicity in vivo.

    DISCUSSION

    Several factors can play a role in the ability of a cytokine to effectively activate

    the immune system against a foreign agent or a tumor cell. One of the first cytokines to

    be approved for treatment of cancer is IL-2 despite the fact that its biology is one of the

    most complex and, in some cases, contradictory to its proposed role in immune

    stimulation. Not only does IL-2 potently stimulate NK and T cells to expand in numbers

    and increase their cytolytic activity, but in the case of T cells, it sensitizes them to

    activation-induced cell death (32) and is required for T regulatory (Treg) cells to suppress

    ongoing immune responses (33). In addition, activation of many cell types in the

    circulation through the intermediate IL-2R can lead to numerous side effects through the

    production of secondary cytokines and over-activation of NK cells – both of which can

    result in vascular injury (34).

    The initial goal of the present study was to engineer a form of IL-2 as an

    immunocytokine with reduced potential for causing side effects while retaining the

    ability to stimulate an anti-tumor immune response. An earlier report from another group

    reported such a study with free IL-2 containing an N88R mutation (4), however it was

    later found that the basis for the reduced toxicity (selectivity for high affinity over

    intermediate IL-2R) was not maintained in the mouse tumor model used to show efficacy

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    24

    (19). In our case, we tested the biological and anti-tumor activity of a different IL-2

    mutation (D20T) that was found by others to be non-receptor-selective in the context of

    free IL-2. This aspartic acid residue is the critical part of a toxin-like domain that is

    believed to be responsible, in part, for direct vascular toxicity of IL-2. Surprisingly, we

    found that the D20T mutation, in the context of a whole antibody immunocytokine, is

    highly selective for the high affinity IL-2R (as measured by IL-2 induced proliferation)

    for both human and mouse immune cells. Thus, for the first time, it was possible to test

    the hypothesis of whether such a selective molecule could be both less toxic and retain

    anti-tumor activity in a mouse model. Results indicate that the D20T form of IL-2,

    contained in the NHS-IL2LT molecule, retained the majority of its anti-tumor activity

    when tested in syngeneic mouse tumor models of experimental metastases. Even when it

    was dosed at higher levels than the control NHS-IL2 molecule containing wild-type IL-2,

    such doses represent a far greater therapeutic index. For example, we showed that for a

    single dosing cycle, the NHS-IL2LT molecule is roughly 20-fold less toxic than NHS-

    IL2 (containing normal IL2). In the tumor efficacy experiments we found no more than a

    four-fold increase in dose was required to achieve the same level of efficacy. Thus, the

    therapeutic index was improved a minimum of five-fold.

    The tolerability of NHS-IL2 LT was also tested in cynomolgus monkeys and

    shown to be dramatically improved over what we have observed with similar

    immunocytokines containing normal IL-2. The lowest dose in these studies was 1 mg/kg

    that resulted in sustained blood levels of NHS-IL2LT of several μg/ml - a level

    approaching the ED50 of this molecule for the intermediate IL-2R. Interestingly, this level

    showed a diminished ability to increase circulating levels of NK cells (following a brief

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    25

    lymphopenia), compared to the two higher doses. T cells, on the other hand, appeared to

    respond just as well at the low dose and proliferated to a much greater extent than NK

    cells. This expansion correlated to the up-regulation of CD25 on both CD4 and CD8

    positive T cells, as well as an increase in sIL-2 in the blood. We do not know yet whether

    the cells responding at the lower dose were already expressing CD25 or whether the

    initial response was stimulated through intermediate receptor-mediated induction of

    CD25 on resting cells. In any case, the overall effect was to selectively expand both T

    cell subsets, especially those expressing CD25, and to do this at increasingly high dose

    levels in the apparent absence of typical IL-2 side effects.

    How this selectivity for high affinity IL-2 receptor in NHS-IL2LT (Selectikine)

    may translate to anti-tumor activity and lower toxicity remains to be tested clinically. In

    fact, Selectikine (EMD 521873) is currently in Phase I clinical trial. The inherent

    problem with immune therapy is that it works less well as tumor burden increases and, in

    the case of NHS-IL2LT, we used a targeting approach (anti-DNA) that might be expected

    to require large tumors as a source of antigen. The fact that even small metastases could

    be treated with this approach, likely due to the appearance of DNA containing structures

    on apoptotic cells, allowed us to demonstrate anti-tumor activity in this minimal disease

    setting after a single dosing cycle and this activity was due mostly to CD8+ effector cells.

    Only low titers of anti-human antibodies were seen in monkeys after an 8-week treatment

    and these have not limited the number of cycles of NHS-IL2LT that can be administered.

    Therefore, it is likely that multiple cycles of treatment in the clinic could be administered

    safely and with the potential for improved efficacy. One challenge with IL-2 in general,

    and with forms specific for the high affinity receptor in particular, is the possibility of

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    26

    generating more T regulatory cells than anti-tumor effectors. In this regard it was

    interesting to note that our anti-tumor activity in the Lewis Lung model was improved

    when mice were treated with anti-CD4 antibody, although significant anti-tumor activity

    was seen without CD4 depletion. Nonetheless, many approaches to reducing T regulatory

    cells (11) could be combined with NHS-IL2LT, including many standard chemotherapies

    and local radiation. The fact that its administration, even at high doses, is associated with

    only mild side effects makes such an approach clinically attractive.

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    27

    Figure 1 Relative IL-2 bioactivity of normal and mutant IL-2 based

    immunocytokines (NHS-IL2 ( ), NHS-IL2LT( ), and control IL-2( )).

    Proliferation responses of (A) the high affinity IL2 receptor expressing human T cell line

    Kit-225 (K6), (B) the intermediate affinity IL2 receptor expressing human cell line TF-

    1β, (C) the high affinity IL2 receptor-expressing mouse T cell line CTLL-2, and (D) the

    intermediate IL2 receptor expressing mouse NK cells. Proliferation of Kit-225 (A) was

    measured by the reduction of Alamar Blue, while that of the other cells (B, C and D) was

    by 3H thymidine uptake. Mouse NK cells were freshly isolated from Bl/6 mice as

    described in the Materials and Methods. Data are representative of at least 3 experiments.

    Figure 2 CD45 positive immune cell proliferation in PBMC cultures containing

    wild-type and mutated IL-2 immunocytokine as assessed by CFSE dilution. A. CD56

    positive NK cells were gated and analyzed for CFSE staining intensity on days 6 and 9 of

    culture. CD25 expression on CD56+ cells was assessed on day 6. A decrease in the

    intensity of staining (shift to the left) was used as a measure of NK cell division. B.

    CD4+ cells were gated and analyzed for CD25 expression and CFSE dilution on day 6 of

    culture. One culture was activated with anti-CD3 antibody while a second was not. C.

    CD8+ cells were gated and analyzed for CD25 expression and CFSE dilution on day 6 of

    culture. One culture was activated with anti-CD3 antibody while a second was not.

    Figure 3 Tolerability of normal and mutant IL-2 based immunocytokines. Bl/6 or

    SCID mice (3 per group) were injected i.v. with either PBS () or the indicated

    immunocytokine, and survival (A and C) and body weight (B and D) were monitored. In

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    28

    Bl/6 mice (A and B), doses of NHS-IL2 were given for 5 consecutive days at 50

    μg/mouse () and for NHS-IL2LT, at 1000 μg/mouse (). Results shown were

    representative of 3 experiments. In SCID mice (C and D), doses of 20 μg/mouse of

    NHS-IL2 () or NHS-IL2LT () were given for 5 consecutive days, every week, until

    overt toxicity occurred.

    Figure 4 Pharmacological activity of NHS-IL2LT in cynomolgus monkey. Blood

    samples of animals treated with 0, 1, 3 and 10 mg/kg/day NHS-IL2LT were analyzed for

    total lymphocyte counts (A), CD3+ cells (B), CD8+ CD16+ cells (C), sIL2Rα (D), CD4+

    CD25+ cells (E), and CD8+ CD25+ cells (F) at the indicated days. Data (except D) were

    expressed as absolute counts (calculated based on total white blood cell count). Note that

    animals were treated in a cyclic regimen on day (D) 0, 1, 2 and 21, 22, 23 and 42, 43, 44,

    as indicated by arrows on the x-axis of (C) and (F). Female data as average are shown,

    males showed a comparable response.

    Figure 5 Efficacy in a syngeneic experimental metastasis model of non-small cell

    lung cancer as assessed by both lung surface coverage and histolological examination.

    Bl/6 mice were injected i.v. with LLC cells and then treated with the indicated dose of

    NHS-IL2LT, the combination of the corresponding amounts of antibody and free IL-2, or

    IL-2 alone for 5 consecutive days beginning 4 days later. Tumor outgrowth was assessed

    on day 28 by histological staining of lung sections (A) or by measuring surface coverage

    of lung metastases (B). The % surface coverage values of the lung sections shown in A

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    29

    are indicated below a representative section stained with H&E. The results shown were

    representative of three separate experiments.

    Figure 6 NHS-IL2LT shows efficacy in experimental models of both

    neuroblastoma and non-small cell lung cancer metastases. A. A/J mice were injected i.v.

    with NXS2 neuroblastoma and treated with the indicated immunocytokine for 5

    consecutive days beginning 4 days later. Tumor outgrowth in the liver was assessed by %

    surface coverage as well as by weighing the diseased organ. The results shown were

    representative of two separate experiments. B. Bl/6 mice were injected i.v. with LLC

    cells and treatment began 4 days thereafter with NHS-IL2 (20 μg/mouse), NHS-IL2LT

    (80 μg/mouse), or equimolar amounts of free IL-2 with or without the NHS antibody, for

    5 consecutive days. Individual groups of mice were treated with depleting antibodies as

    described in Materials and Methods. Tumor burden was assessed on day 28.

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    30

    REFERENCES

    1. Atkins MB, Lotze MT, Dutcher JP, et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. J Clin Oncol 1999;17: 2105-16.

    2. Baluna R, Rizo J, Gordon BE, Ghetie V, Vitetta ES. Evidence for a structural motif in toxins and interleukin-2 that may be responsible for binding to endothelial cells and initiating vascular leak syndrome. Proc Natl Acad Sci U S A 1999;96: 3957-62.

    3. Epstein AL, Mizokami MM, Li J, Hu P, Khawli LA. Identification of a protein fragment of interleukin 2 responsible for vasopermeability. J Natl Cancer Inst 2003;95: 741-9.

    4. Shanafelt AB, Lin Y, Shanafelt MC, et al. A T-cell-selective interleukin 2 mutein exhibits potent antitumor activity and is well tolerated in vivo. Nat Biotechnol 2000;18: 1197-202.

    5. Caligiuri MA, Zmuidzinas A, Manley TJ, Levine H, Smith KA, Ritz J. Functional consequences of interleukin 2 receptor expression on resting human lymphocytes. Identification of a novel natural killer cell subset with high affinity receptors. J Exp Med 1990;171: 1509-26.

    6. Davis CB, Gillies SD. Immunocytokines: amplification of anti-cancer immunity. Cancer Immunol Immunother 2003;52: 297-308.

    7. Gillies SD, Lan Y, Williams S, et al. An anti-CD20-IL-2 immunocytokine is highly efficacious in a SCID mouse model of established human B lymphoma. Blood 2005;105: 3972-8.

    8. Sharifi J, Khawli LA, Hu P, King S, Epstein AL. Characterization of a phage display-derived human monoclonal antibody (NHS76) counterpart to chimeric TNT-1 directed against necrotic regions of solid tumors. Hybrid Hybridomics 2001;20: 305-12.

    9. Epstein AL, Khawli LA, Chen F-M, Hu P, Glasky MS, Taylor CR. Tumor necrosis imaging and treatment of solid tumors. In: Torchilin VP, editor. Handbook of Targeted Delivery of Imaging Agents: CRC Press; 1995.

    10. Chen S, Yu L, et al. Pivotal study of Iodine-131-labeled chimeric Tumor Necrosis Treatment radioimmunotherpay in patients with advanced lung cancer. J Clin Oncol 2005;23: 1538-1547.

    11. Emens LA, Jaffee EM. Leveraging the activity of tumor vaccines with cytotoxic chemotherapy. Cancer Res 2005;65: 8059-64.

    12. Gillies SD, Lan Y, Wesolowski JS, et al. Antibody-IL-12 fusion proteins are effective in SCID mouse models of prostate and colon carcinoma metastases. J Immunol 1998;160: 6195-203.

    13. Gillies SD, Lo KM, Burger C, Lan Y, Dahl T, Wong WK. Improved circulating half-life and efficacy of an antibody-interleukin 2 immunocytokine based on reduced intracellular proteolysis. Clin Cancer Res 2002;8: 210-6.

    14. Gillis S, Ferm MM, Ou W, Smith KA. T cell growth factor: parameters of production and a quantitative microassay for activity. J Immunol 1978;120: 2027-32.

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    31

    15. Hori T, Uchiyama T, Tsudo M, et al. Establishment of an interleukin 2-dependent human T cell line from a patient with T cell chronic lymphocytic leukemia who is not infected with human T cell leukemia/lymphoma virus. Blood 1987;70: 1069-72.

    16. Farner NL, Voss SD, Leary TP, et al. Distinction between gamma c detection and function in YT lymphoid cells and in the granulocyte-macrophage colony-stimulating factor-responsive human myeloid cell line, Tf-1. Blood 1995;86: 4568-78.

    17. Damjanovich S, Bene L, Matko J, et al. Preassembly of interleukin 2 (IL-2) receptor subunits on resting Kit 225 K6 T cells and their modulation by IL-2, IL-7, and IL-15: a fluorescence resonance energy transfer study. Proc Natl Acad Sci U S A 1997;94: 13134-9.

    18. Johnson DR, Hauser IA, Voll RE, Emmrich F. Arterial and venular endothelial cell costimulation of cytokine secretion by human T cell clones. J Leukoc Biol 1998;63: 612-9.

    19. Hartmann G. Technology evaluation: BAY-50-4798, Bayer. Curr Opin Mol Ther 2004;6: 221-7.

    20. Taniguchi T, Minami Y. The IL-2/IL-2 receptor system: a current overview. Cell 1993;73: 5-8.

    21. Kabouridis PS, Tsoukas CD. Regulation of expression of interleukin 2 receptors upon triggering of the TCR-CD3 complex on human T lymphocytes. Immunol Invest 1990;19: 385-401.

    22. Gillies SD, Young D, Lo KM, Roberts S. Biological activity and in vivo clearance of antitumor antibody/cytokine fusion proteins. Bioconjug Chem 1993;4: 230-5.

    23. Ko YJ, Bubley GJ, Weber R, et al. Safety, pharmacokinetics, and biological pharmacodynamics of the immunocytokine EMD 273066 (huKS-IL2): results of a phase I trial in patients with prostate cancer. J Immunother 2004;27: 232-9.

    24. Harada Y, Yahara I. Pathogenesis of toxicity with human-derived interleukin-2 in experimental animals. Int Rev Exp Pathol 1993;34 Pt A: 37-55.

    25. King DM, Albertini MR, Schalch H, et al. Phase 1 clinical trial of the immunocytokine EMD273063 in melanoma patients. J Clin Oncol 2004;22 :4463- 4473. 26. Reagan-Shaw S, Nihal M, Ahmad N. Dose translation from animal to human studies revisited. FASEB J 2007;22:659-661. 27. Vial T, Descotes J. Clinical toxicity of interleukin-2. Drug Saf 1992;7: 417-33. 28. Waldmann, T. A. The IL-2/IL-2 receptor system: a target for rational immune intervention. Immunology Today 1993;14: 264-70 29. Casciola-Rosen LA, Anhalt G, Rosen A. Autoantigens targeted in systemic lupus erythematosus are clustered in two populations of surface structures on apoptotic keratinocytes. J Exp Med 1994;179: 1317-30. 30. Gorgani NN, Smith BA, Kono DH, Theofilopoulos AN. Histidine-rich glycoprotein binds to DNA and Fc gamma RI and potentiates the ingestion of apoptotic cells by macrophages. J Immunol 2002;169: 4745-51. 31. Cheng FM, Hansen EB, Taylor CR, Epstein AL. Diffusion and binding of monoclonal antibody TNT-1 in multicellular tumor spheroids. J Natl Cancer Inst 1991;83: 200-4.

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • NHS-IL2LT low toxicity immunocytokine

    32

    32. Lenardo MJ. Interleukin-2 programs mouse alpha beta T lymphocytes for apoptosis. Nature 1991;353: 858-61. 33. de la Rosa M, Rutz S, Dorninger H, Scheffold A. Interleukin-2 is essential for

    CD4+CD25+ regulatory T cell function. Eur J Immunol 2004;34: 2480-8. 34. Siegel JP, Puri RK. Interleukin-2 toxicity. J Clin Oncol 1991;9: 694-704.

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/

  • Published OnlineFirst April 29, 2011.Clin Cancer Res Stephen D. Gillies, Yan Lan, Thore Hettmann, et al. activity despite its high degree of IL-2 receptor selectivityA low-toxicity IL-2 based immunocytokine retains anti-tumor

    Updated version

    10.1158/1078-0432.CCR-10-2921doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://clincancerres.aacrjournals.org/content/suppl/2011/06/03/1078-0432.CCR-10-2921.DC1

    Access the most recent supplemental material at:

    Manuscript

    Authoredited. Author manuscripts have been peer reviewed and accepted for publication but have not yet been

    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/early/2011/04/29/1078-0432.CCR-10-2921To request permission to re-use all or part of this article, use this link

    on July 9, 2021. © 2011 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 29, 2011; DOI: 10.1158/1078-0432.CCR-10-2921

    http://clincancerres.aacrjournals.org/lookup/doi/10.1158/1078-0432.CCR-10-2921http://clincancerres.aacrjournals.org/content/suppl/2011/06/03/1078-0432.CCR-10-2921.DC1http://clincancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://clincancerres.aacrjournals.org/content/early/2011/04/29/1078-0432.CCR-10-2921http://clincancerres.aacrjournals.org/

    revisedNHS-IL2LT.CCRFigure 1Figure 2Figure 3Figure 4Figure 5Figure 6