30
1 Single-dose anti-PD-L1/IL-15 fusion protein KD033 generates synergistic anti- tumor immunity with robust tumor-immune gene signatures and memory responses Stella A. Martomo 1 *, Dan Lu 1 , Zhanna Polonskaya 1 , Xenia Luna 1 , Zhikai Zhang 1 , Sam Feldstein 1 , Radovan Lumban-Tobing 1 , Danielle K. Almstead 1 , Faical Miyara 1 , Jeegar Patel 1 1 Kadmon Corporation, 450 East 29th Street, New York, NY 10016 *Corresponding author: [email protected] Address: 450 East 29 th Street, New York, NY 10016 Running Title: Anti-PD-L1/IL-15 induces cytotoxic gene signature in tumors Keywords: cytokine fusions, bi-functional antibodies, tumor-microenvironment, immuno-stimulatory Conflict of Interest: All authors are current or former employees/interns of Kadmon and hold stock and/or stock options or interests in Kadmon. on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

Single-dose anti-PD-L1/IL-15 fusion protein KD033 generates ......2020/12/08  · 14, Bio Xcell), blocking antibodies, or combinations thereof. Anti-mouse CTLA-4 (clone 9D9, BioXcell)

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

  • 1

    Single-dose anti-PD-L1/IL-15 fusion protein KD033 generates synergistic anti-

    tumor immunity with robust tumor-immune gene signatures and memory

    responses

    Stella A. Martomo1*, Dan Lu

    1, Zhanna Polonskaya

    1, Xenia Luna

    1, Zhikai Zhang

    1, Sam Feldstein

    1,

    Radovan Lumban-Tobing1, Danielle K. Almstead

    1, Faical Miyara

    1, Jeegar Patel

    1

    1Kadmon Corporation, 450 East 29th Street, New York, NY 10016

    *Corresponding author: [email protected]

    Address: 450 East 29th

    Street, New York, NY 10016

    Running Title: Anti-PD-L1/IL-15 induces cytotoxic gene signature in tumors

    Keywords: cytokine fusions, bi-functional antibodies, tumor-microenvironment, immuno-stimulatory

    Conflict of Interest: All authors are current or former employees/interns of Kadmon and hold stock

    and/or stock options or interests in Kadmon.

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    mailto:[email protected]://mct.aacrjournals.org/

  • 2

    Abstract

    Immunocytokines hold great potential as anti-cancer agents as they utilize a specific anti-tumor

    antibody to deliver an immune activating cytokine directly to the immunosuppressive tumor-

    microenvironment (TME). We have developed a novel immunocytokine (KD033) comprised of a fully

    human, high affinity anti-programmed death-ligand 1 (PD-L1) linked to the sushi domain of the human

    IL-15/IL-15 receptor alpha (IL-15/IL-15Rα) complex. A murine PD-L1 cross-reactive KD033

    surrogate (srKD033) and a non-targeting antibody (ntKD033) were also developed to investigate

    mechanism of action in murine tumor models. Efficacy analyses showed a robust anti-tumor effect of

    single-dose srKD033 in several diverse syngeneic murine tumor models. In a CT26 murine colon

    tumor model, single dose srKD033 produced durable anti-tumor immunity as evidenced by resistance

    to subsequent tumor re-challenges. Mice responding to srKD033 treatment showed increased retention

    of PD-L1/IL-15 in the TME which likely facilitated prolonged IL-15-induced expansion of cytotoxic

    cells. Importantly, target-based PD-L1/IL-15 delivery via srKD033 was well-tolerated and induced

    significant anti-tumor activity in murine carcinoma models that are non- or minimally responsive to

    IL-15 or anti-PD-L1/PD-1 monotherapy.

    Introduction

    Programmed death-ligand-1 (PD-L1) and programmed death-1 (PD-1) interaction have been shown to

    play non-redundant roles in reducing tumor-specific T-cell response in the TME (1-3). Blocking the

    PD-1/PD-L1 interaction with anti-PD-L1 or anti-PD-1 agents increases cytotoxic T-cell activation

    leading to long-lasting anti-tumor responses in some patients (4-6). Despite the demonstrated efficacy

    of PD-1/PD-L1 immune checkpoint inhibitors (ICIs) in a variety of cancers, a large percentage of

    patients do not respond or rapidly become refractory to these therapies (7). A rational strategy to

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 3

    increase clinical responses and also reduce resistance to ICIs, is to combine ICIs with cytokines that

    enhance T-cell activation while avoiding T-cell exhaustion/inactivation (anergy) (7,8). The selectivity

    of some cytokines in expanding activated T cells make them ideal candidates for combination therapies

    with ICIs (9).

    Immunostimulatory cytokines such as IL-15 have demonstrated clinical activity in the treatment of

    certain cancers (10,11). In patients with metastatic renal cell carcinoma (RCC), recombinant IL-15 (rIl-

    15) produced a dramatic efflux of NK and memory CD8 T cells from circulating blood and produced

    clearance of lung lesions in two patients without evidence of regulatory T cells (Treg) stimulation or

    significant adverse events (12). IL-15 acts through its specific receptor, IL-15Rα, which is expressed

    on antigen-presenting dendritic cells, monocytes and macrophages. The membrane-bound IL-15/IL-

    15Rα complex is then trans-presented to neighboring NK or CD8 T cells that express the IL-15RβƔ

    receptor to induce anti-tumor activity (13).

    Unfortunately, the short half-life of rIL-15 and the limited availability of IL-15Rα in vivo have limited

    its usefulness. Therefore, efforts to increase rIL-15 half-life, reduce systemic toxicity, and direct

    activity to the TME are expanding. IL-15 shares two components of its receptor (the common gamma-

    chain and the IL-2/15RβƔ complex) with IL-2 (14), which is effective in the treatment of RCC and

    metastatic melanoma. Thus, IL-15 and IL-2 can mediate overlapping, differing or even opposing

    functions, particularly in the early phases of T-cell activation when T cells enter their differentiation

    program (15). Importantly, administration of rIL-2 requires careful management to limit severe

    toxicities, such as cardiopulmonary toxicity and end-organ dysfunction (16). Unlike rIL-15, rIL-2 also

    expands regulatory T cells (Tregs) which contributes to an immunosuppressive TME and prevents

    generation of tumor immunity (10,11). It has been shown that complexation of the IL-15Rα sushi-

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 4

    domain with IL-15 increases IL-15 half-life and bioavailability in vivo (17) and is effective in

    mimicking endogenous IL-15/IL-15Rα complex binding to IL2/15RβƔ-expressing cells (18).

    In this report we describe biological activity and efficacy of a novel human anti-PD-L1/ IL-15Rα-IL-

    15 immunocytokine (KD033) which binds to human and non-human primate (NHP) cells expressing

    PD-L1. A KD033 surrogate (srKD033) which binds to human, NHP, and murine cells expressing PD-

    L1 was also developed to evaluate treatment efficacy in syngeneic murine tumor models. KD033 and

    srKD033 are engineered to minimize antibody-dependent cellular cytotoxicity (ADCC) and also

    facilitate IL-15 trans-presentation from PD-L1-expressing cells. KD033 targeted anti-PD-L1 delivery

    is expected to activate/expand cytotoxic T-cell memory as well as antigen presentation when PD-L1-

    expressing immune and tumor cells are present at the tumor site. We expect this effect will lead to

    robust anti-tumor and memory responses and reduced systemic toxicities.

    Materials and Methods

    IL-15 Fusion Molecules

    Production of KD033, a high affinity anti-human/monkey-PD-L1; srKD033, a high affinity anti-

    human/monkey/murine-PD-L1; and ntKD033, a non-targeting antibody is described in Supplementary

    Methods 1. Anti-PD-L1 IgG1 sequences were ID 246, 250 and 267 (US Patent 10,407,502 B2). For

    ntKD033, aa31-35 (ID267) were WYGMD instead of AYRMF. Validation of activity is included in

    Supplementary Figure 1. All antibodies were of IgG1 class and were fused to the human IL-15/IL-

    15Rα complex (Figure 1A). A LALA modification, in which leucine residues at positions 234 and 235

    of human IgG1 Fc domain are substituted with alanine residues, was incorporated into the Fc region in

    order to reduce FcƔ receptor binding thereby minimizing ADCC (19,20).

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 5

    Cell Proliferation Assay

    Mouse splenocytes (Cellero) were used to measure IL-15-induced proliferation. Increasing

    concentrations of fusion proteins were added to 50,000 thawed splenocytes/well in 96-well U-bottom

    plates in IMDM, 10% FBS with 55 µM 2-Mercaptoethanol (Invitrogen). Proliferation was measured at

    day four using CellTitreGlo (Promega). Data points represented average of duplicate wells and were

    repeated three times.

    Cell Lines for Murine in vivo Studies

    Cell lines were routinely tested for mycoplasma before inoculation. Cell lines, sources, and culture

    conditions used are described in Table 1 and Supplementary Methods 2.

    In vivo Syngeneic Murine Tumor Studies

    The maximal tolerated dose (MTD, antibody dose level that does not cause untoward toxicity or death)

    of srKD033 and ntKD033 was determined using single escalating intravenous (i.v.) doses in naive or

    CT26-bearing Balb/c mice. The MTD of srKD033 and ntKD033 was determined to be 3 mg/kg and 1

    mg/kg, respectively. These doses, or lower, were used for all subsequent in vivo studies. The efficacy

    of srKD033 was evaluated in eleven subcutaneously implanted syngeneic mouse models (n=10 per

    arm). Tumor cells and mouse strains used are listed in the Supplementary Methods 2. Efficacy was

    determined by comparing measured tumor volumes in srKD033-treated versus vehicle-treated mice.

    Assessment of srKD033 efficacy in CT26-bearing Balb/c mice was repeated and compared to efficacy

    observed with ntKD033, anti-mouse PD-L1 (developed by Kadmon), anti-mouse PD-1 (RMP1-14, Bio

    Xcell), blocking antibodies, or combinations thereof. Anti-mouse CTLA-4 (clone 9D9, BioXcell) was

    used with srKD033 in the 4T1 breast cancer model. All fusion proteins were administered as a single

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 6

    i.v. dose, unless otherwise noted. Anti-PD-L1 mouse anti-PD-1 and anti-CTLA-4 were administered by

    intraperitoneal (i.p.) injection twice per week over a 3-week period.

    To determine which immune cell populations contribute to srKD033 efficacy, antibodies that deplete

    either CD8 T (YTS 169.4, Bio Xcell) or NK cells (Asialo GM1, ThermoFisher Scientific) were used as

    described in Supplementary Figure 2A. Depletions were confirmed by flow cytometry of peripheral

    blood as described in Supplementary Methods 2.

    MOA studies were also performed using CT26 colon carcinoma bearing Balb/c mice. Mice were

    treated with either a single i.v. dose of srKD033 or ntKD033, or two i.v. doses of anti-PD-L1 when

    tumors reached approximately 130 mm3. At 7 days post-treatment, peripheral blood, spleens and

    tumors from each group were collected for flow cytometry and immunohistochemistry (IHC). Details

    of IHC methods and detection of srKD033/ntKD033 in serum are provided in Supplementary Methods

    2.

    All in vivo mouse studies were conducted for Kadmon by Crown Bioscience Inc. Animal care and use

    protocols and any amendment(s) were reviewed and approved by the Institutional Animal Care and

    Use Committee (IACUC) of Crown Biosciences. All animal studies were conducted in accordance

    with the regulations of the Association for Assessment and Accreditation of Laboratory Animal Care

    (AAALAC).

    Flow Cytometry Analysis

    Serially diluted KD033, srKD033, ntKD033 and, where indicated, the anti-PD-L1 antibody were added

    to 50,000 cells/well. Binding was detected using Allophycocyanin (APC)-conjugated goat anti-human

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 7

    IgG (Jackson ImmunoResearch, 1:200 dilution) and evaluated using the Guava EasyCyte™ HT Flow

    Cytometer with GuavaSoft 2.2.2. (EMD Millipore) for data analysis. Data points are the means ± SD

    of duplicate measurements, and were repeated three times. Additional description for in vitro flow

    cytometry analyses is provided in Supplementary Methods 1. Analysis of PD-L1 expression on tumor

    cells and immune cell populations following treatment with srKD033 in vivo can be found in

    Supplementary Methods 2. Antibodies used are listed in Table 2.

    Immunohistochemistry

    FFPE or OCT blocks of tumor samples were sectioned (4 µm thickness/section) with a manual rotary

    microtome and processed using a Leica Bond RX autostainer. Antibodies used are listed in Table 2. All

    stained sections were scanned with a NanoZoomer-HT 2.0 Image system (40x magnification;

    Hamamatsu photonics). IHC images were analyzed using the HALOTM

    Image Analysis platform.

    Whole sections were analyzed and areas of necrosis were excluded. Total cell numbers (or area) and

    IHC positive cells (or area) were scored. The IHC score represents the positive to total cell count/area

    ratio in the examined section..

    Gene Transcription Analysis

    Tumors were isolated on day 7 after treatment, snapped frozen and used for RNA transcription using

    the NanoString mouse PanCancer IO 360 immune-oncology panel. NanoString gene expression

    analyses and tumor RNA extractions were conducted by Canopy Biosciences as described in

    Supplementary Figure 3.

    Statistics

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 8

    Group comparisons were performed using one-way ANOVA with Tukey’s multiple comparisons test.

    Comparison of tumor growth for treated and control groups were performed using RM ANOVA. The

    log-rank (Mantel-Cox) test was used for survival calculations. Tests were done on GraphPad Prism

    software (version 8.1.2). Significance is indicated as follows: ns (not significant), p

  • 9

    murine tumor models with varying percentage of PD-L1 positive tumor cells (Figure 1E). srKD033

    was not effective in EMT-6 breast or Renca kidney tumor models. However, anti-tumor activity as

    measured by tumor growth inhibition (TGI) was observed in the other models (Figure 1F-L). In the

    murine CT26 colon carcinoma model, single-dose srKD033 produced a curative effect as evidenced by

    the absence of tumor mass in some mice (Figure 1F). A similar effect was observed in the H22 liver

    tumor model (Figure 1H). These data show that srKD033 is efficacious in the treatment of tumor types

    that express varied levels of PD-L1, consistent with anti-PD-L1 activity on tumor and/or immune cells

    in TME (21,22). srKD033 also showed significant efficacy in the B16F10 and B16BL6 melanoma

    models, which are known not to respond to ICIs (Figure 1J, K).

    srKD033 is better tolerated than ntKD033

    To compare the mode of anti-PD-L1/IL-15 (srKD033) action to IL-15 alone (ntKD033) and to

    ntKD033-anti-PD-L1 combination, CT26 tumor-bearing mice were treated with single-dose srKD033

    or ntKD033, repeat dose anti-PD-L1, or combination ntKD033+anti-PD-L1. Analysis of body weight

    in mice treated with single-dose ntKD033 or srKD033 showed that ntKD033 was well tolerated up to 1

    mg/kg; srKD033 was tolerated up to 3 mg/kg. Body weight loss observed in srKD033-treated mice (3

    mg/kg) on Day 4 after treatment was largely recovered by Day 6 (Figure 2A). These findings are

    consistent with data from srKD033-treated naïve Balb/c mice (Figure 1D). Notably, mice treated with

    ntKD033 at 1 mg/kg showed a significant reduction in body weight at Day 4 that did not recover by

    Day 6.

    At the highest tolerated dose levels, srKD033 was more effective than ntKD033 in the CT26 colon

    carcinoma tumor model (Figure 2B and C). Interestingly, to achieve an efficacy comparable to that

    observed with single-dose srKD033 (3mg/kg), a combination of single-dose ntKD033 (1 mg/kg) and

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 10

    repeat dose anti-PD-L1 (10 mg/kg) was required (Figure 2D, E). However, mice treated with this

    combination experienced significant increases in spleen weights, compared to srKD033-treated mice

    (Figure 2G). In addition, at Day 7, human IL-15 serum levels were higher in ntKD033-treated mice,

    compared to srKD033-treated mice, although this difference did not reach statistical significance

    (Figure 2H). Both srKD033 and ntKD033 treatments resulted in comparable increases in CD8 T cells

    in the spleen (Figure 2H). In contrast, levels of splenic NK cells were significantly higher in ntKD033-

    treated mice (Figure 2I).

    In a comparison of one srKD033 “best-responder” to one ntKD033 “best-responder” (indicated by a

    “+” in the tumor volume plots, Figure 2, panels J and K, respectively), the srKD033 responder showed

    an increase in CD8 T lymphocyte infiltration into and surrounding the tumor which was not observed

    in the tumor from the ntKD033 responder (Figure 2J, K). Because the response associated with single

    treatments of srKD033 or ntKD033 were highly variable, the extent of NK cell tumor infiltration

    between srKD033 and ntKD033 responders did not reach statistical significance.

    srKD033 increased both adaptive and innate immune cell activation in tumors

    To further evaluate the srKD033 MOA, eight responders (%TGI>50) and five non-responders

    (%TGI

  • 11

    in srKD033 responders when compared to vehicle-treated mice. While an increase of CD8 T cells in

    peripheral blood was observed in all treatment groups, the highest levels were observed in srKD033

    responders. In contrast, the highest increase in NK cells was observed in both ntKD033 responder- and

    non-responder mice. Immunohistochemistry showed CD8 T and NK cell infiltration into tumors of

    both srKD033- and ntKD033-responders, but statistical significance was only achieved in the srKD033

    responders (Figure 3C). Moreover, in srKD033-responders, more anti-PD-L1/IL-15 was retained and a

    significant increase of B220+ cell tumor infiltration was observed (Figure 3C, D). Notably, changes in

    the peripheral blood lymphocyte population of srKD033-treated mice was also observed in

    cynomolgus monkeys treated with single dose KD033 by i.v. administration. In these studies, KD033

    treatment produced dose-dependent increases in CD8 T, NK and NKT-like cells in the periphery at 7

    days post-dose (Supplementary Table 1).

    The NanoString gene expression platform was used to evaluate treatment effect on gene expression in

    tumors. Overall changes in the gene expression profiles were relatively comparable between srKD033-

    and ntKD033-treated mice (Supplementary Figure 3). Because these fusion proteins both contain the

    IL-15/IL-15Rα complex, it is likely that the majority of the observed changes were mediated by the

    activity of IL-15. A notable difference between srKD033- and ntKD033-treated mice was a

    significantly higher increase in cytotoxic genes (Gzma, Gzmb, Gzme and Prf1) in both srKD033

    responders and non-responders (Figure 3E). There was a higher expression of exhausted T-cell related

    genes as well as of genes associated with innate immune response in srKD033-responders compared to

    ntKD033-reponders (Figure 3F, Supplementary Figure 3). Expression of T-cell related genes was

    significantly reduced in srKD033 non-responders compared to srKD033 responders (Figure 3F).

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 12

    To evaluate further the cytotoxic immune cell population critically involved in mediating efficacy of

    srKD033, select immune cell populations in CT26 colon carcinoma bearing Balb/c mice were depleted

    prior to srKD033 treatment. Efficacy following single-dose srKD033 treatment was then assessed in

    survival analyses. Administration of a CD8 T cell depleting antibody decreased CD8 T cells, but also

    increased NK cells in peripheral blood (Figure 3F). Antibody depletion of NK cells produced the

    expected decrease of NK cells in the periphery without increasing the population of other immune cells

    that were evaluated. Importantly, depletion of NK cells did not decrease the efficacy of srKD033 in the

    CT26 colon carcinoma model as evidenced by unchanged survival. Meanwhile, depletion of CD8 T

    cells produced a significant reduction in srKD033-mediated tumor growth inhibition and survival, but

    not immediately or completely (Figure 3G), suggesting a complex involvement of immune cell

    populations, in addition to CD8 T cells, in mediating srKD033 efficacy.

    srKD033 produced durable memory responses

    Both srKD033-responder and non-responder mice showed greater increases in CTLA-4 expression,

    compared to ntKD033-treated mice (Figure 3E). The axis of PD-1/PD-L1/CD80/CTLA-4 interactions

    in TME was recently described (23,24). The administration of anti-PD-L1 was shown to decrease

    CD80 on antigen presenting cells (APCs) in the CT26 and 4T1 tumor models (24). As CTLA-4 gene

    transcription was also increased in srKD033 non-responders, we sought to determine if administration

    of anti-CTLA-4 in combination with srKD033 would produce an anti-tumor effect in the 4T1 model

    where srKD033 monotherapy was ineffective. The regimen of srKD033 in combination with repeat

    dose anti-CTLA-4 was indeed observed to significantly increase TGI in the 4T1 model (Figure 4A).

    The combination of anti-PD-1 and anti-PD-L1 can be synergistic as they mediate overlapping and non-

    overlapping interactions between tumors and immune cells (PD-1/PD-L1), or between immune cells in

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 13

    TME (PD-L1/CD80) (22). PD-1 expression is increased significantly in srKD033 responders (Figure

    3F). To determine if continuous anti-PD-1 blocking would be synergistic with srKD033, the

    combination of single or repeat doses of srKD033 with repeat dose anti-PD-1 was evaluated. The

    combination of single-dose srKD033 (3 mg/kg) with repeat doses of anti-PD-1 showed synergistic

    activity when compared to either treatment alone (Figure 4B). Repeat dosing of srKD033 (1 mg/kg)

    with repeat doses of anti-PD-1 was effective to eliminate tumors in 30% of treated mice; whereas the

    combination of single-dose srKD033 (3 mg/kg) with repeat dose of anti-PD-1 cured 60% of the mice

    in this treatment group (Figure 4C, D). Mice demonstrating a curative effect following srKD033

    monotherapy or srKD033/anti-PD-1 combination therapy were re-challenged with the same tumor

    (CT26) without additional antibody administration. All mice rejected the CT26 re-challenge (Figure

    4E).

    To determine if a CT26-specific T-cell response was generated in the cured (tumor-free) mice, either a

    CT26-specific gp70 tetramer or a non-specific β-galactosidase tetramer was used to stain CD8 T

    splenocytes for flow cytometry (Figure 4F). A significant increase in gp70 tetramer CD8 T cells, but

    not non-specific β-galactosidase CD8 T cells, was observed in mice treated with srKD033 or the

    srKD033/anti-PD-1 combination, confirming the generation of a sustained CT26-specific CD8 T cell

    memory response.

    Discussion

    In this report, we have evaluated biological activity and treatment efficacy of a novel PD-L1/ IL-15/IL-

    15Rα immunocytokine (KD033). One of the features of this bi-functional antibody is its ability to

    guide IL-15 to the TME, thereby stimulating intratumoral cytolytic immune responses. Data from our

    studies has shown that treatment with the KD033 murine analog (srKD033) increases survival and

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 14

    tumor growth inhibition in multiple murine tumor models. In the TME, srKD033 enhances activation

    of cytotoxic cells and induces a synergistic effect on innate and adaptive immune cell. By fusing IL-15

    to anti-PD-L1, the effects of IL-15 in vivo shifted from changing the ratio of cytotoxic/Treg cells to

    eliciting a robust activation of cytotoxic cells without significantly affecting Treg, resulting in the

    generation of a potent memory response after single-dose srKD033 treatment.

    KD033 and srKD033 differ from the recently described anti-PD-L1/IL-15 fusion protein, N-809 (25)

    in that the mode of KD033/srKD033 action was not intended to involve ADCC or complement-

    dependent cytotoxicity. In KD033/srKD033, IL-15 was linked to the C-terminal Fc-domain, rather than

    the N-terminal. The C-terminal Fc-fusion likely preserves high affinity binding to PD-L1 (due to

    opposite sites of binding) on either tumor or immune cells in the TME. The increased retention of

    srKD033 in the TME is consistent with this MOA (Figure 3C). Accordingly, single-dose srKD033

    treatment produced a varied level of efficacy in a variety of tumors, including tumors which are known

    to be non-responsive to ICIs, such as the B16F10 melanoma model.

    Previous studies have shown that IL-15 treatment, or combination treatment of IL-15 or IL-2 with PD-

    1/PD-L1 blockade, increases expression of PD-L1 and produces robust anti-tumor responses in murine

    cancer models (26,27). In the present study, single-dose srKD033 treatment in a CT26 colon carcinoma

    model showed increased efficacy when compared to repeat doses of anti-PD-L1 alone, or the highest

    tolerated dose of ntKD033. Durability of the srKD033 treatment response was superior to that

    observed with ntKD033. Importantly, 3 mg/kg srKD033 was better tolerated than 1 mg/kg of

    ntKD033, or combination ntKD033/anti-PD-L1, as assessed by changes in body and spleen weights.

    These changes were coincident with significantly higher levels of IL-15 in serum (Figure 2G) and

    increased percentage of splenic NK cells (Figure 2I), which was not observed following srKD033

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 15

    treatment, despite the lower dose of ntKD033 (1 mg/kg ntKD033 vs. 3 mg/kg srKD033). By targeting

    IL-15 to PD-L1-expressing cells, greater localized immune stimulation was achieved with reduced

    systemic toxicity (Figure 3E). In contrast, ntKD033 treatment resulted in prolonged presence of IL-15

    in the periphery resulting in increased systemic NK cell stimulation in spleen and PBMCs (Figure 3B).

    Depletion of CD8 T cells resulted in a significant increase of NK cells in the periphery following

    srKD033 treatment (Figure 3F). This finding suggests that srKD033 changes the balance of cytotoxic

    cell activation in the periphery. The ability of srKD033 to redirect IL-15 toward the TME differentiate

    its mode of action from that of ICIs, free IL-15, modified free IL-2, or IL-15/IL-2 combination with

    ICIs, which are currently being evaluated in various cancer indications.

    Our data also suggest that srKD033 has a different mode of action in the TME compared to free rIL-

    15, rIL-2 or ICIs. Recently it has been shown that anti-PD-L1 treatment disrupts the balance of PD-

    L1/CD80 cis interaction (23,24) resulting in increased availability of B7.1 to bind to CTLA-4 on Tregs.

    We hypothesize that when srKD033 is bound to PD-L1 expressing APCs in the TME, srKD033 can act

    both as a PD-L1 blocker and a co-stimulatory molecule for CD8 T cells (Supplementary Figure 1H).

    With co-stimulatory activity of srKD033 on APCs, even if PD-L1 blocking resulted in increased

    CTLA-4/CD80 interaction (24), activation/proliferation of CD8 T cells could still occur. In the case of

    srKD033 non-responders, srKD033 could be bound to PD-L1 expressing cells other than APCs. The

    Tregs/CTLA-4 axis could play a significant role, resulting in less CD8 T cell activation/proliferation.

    srKD033 could also directly increase activation of cytotoxic cells resulting in increased exhaustion. In

    agreement with this hypothesis, co-administration of srKD033 with ICIs such as anti-CTLA-4 and anti-

    PD-1 increased the anti-tumor efficacy of srKD033 (Figure 4A, B). In contrast to srKD033, the relative

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 16

    non-specificity of ntKD033 is consistent with the observed increase of cytotoxic cells in the periphery,

    especially NK cells, which in turn non-specifically increase trafficking of these cells into tumors.

    Data from our gene expression analysis revealed elevated levels of cytotoxic genes (Gzma, Gzmb,

    Gzme and Prf1) in both srKD033-responsive and non-responsive mice, compared to ntKD033

    responsive mice (Figure 3E). The absence of anti-tumor activity in srKD033 non-responders despite

    the increased expression of cytotoxic genes suggests that increased levels of cytotoxic proteins in

    tumors alone may not be sufficient to elicit an anti-tumor response. In addition to the efficient co-

    stimulatory effect of srKD033 on CD8 T cells, the increased retention of anti-PD-L1/IL-15 in

    srKD033-responsive tumors (Figure 3C) could facilitate prolonged and increased infiltration of other

    immune cells such as B220+ cells in the TME (Figure 3C). The increase of B220+ cells observed in

    the srKD033-responsive tumors correlated with the increase of cytotoxic genes identified in the gene

    transcription analysis. This is consistent with these cells being pre-mNK cells possessing NK-like

    killing with functional APCs capabilities (28,29). Furthermore, increased expression of genes such as

    Itgax and Nos2 (Figure 3F) suggests involvement of both innate and adaptive immune response in

    srKD033-responders.

    The lower levels of NK cell proliferation in the periphery of srKD033-responsive mice, compared to

    ntKD033-treated mice, did not decrease NK cell infiltration into tumors. The ability of some CD8 T

    cells to acquire NK cell markers after exposure to IL-15 or IL-2 has been previously demonstrated

    (30,31). It is possible that these cell types were induced by srKD033 treatment and could have

    contributed to the observed efficacy. srKD033-responsive mice showed higher levels of NKT-like cells

    in peripheral blood, compared to non-responsive and ntKD033-treated mice, and increased in NKT-

    like cells was also seen in the periphery of KD033-treated cynomolgus monkeys (Supplementary Table

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 17

    1). The production of invariant NKT (iNKT) cells may account for this observation. The finding that

    intratumoral iNKT-cell infiltration is a positive prognostic indicator in colorectal cancer (32,33) is

    consistent with our findings in the CT26 colon carcinoma model and support the use of

    immunophenotyping to predict and/or evaluate response to treatment. Additional studies are ongoing to

    determine the exact nature of these NKT-like cell populations and their potential involvement in

    KD033/srKD033-mediated tumor immunity.

    Our PD-L1 targeting immunocytokine has been shown to increase T-cell activity via inhibition of PD-

    1/PD-L1 interaction, deliver IL-15 to the TME leading to activation of T- and NK-cells locally,

    achieve a robust and durable anti-tumor efficacy, and reduce toxicity associated with systemic IL-15

    exposure. Additionally, clinical development of a single bi-functional, versus combination, therapy

    would likely reduce cost and burden for patients and health care professionals. Because PD-L1 is

    expressed in various cell types, the potential for off-target effects of KD033 in patients is of concern.

    In this regard, it is important to note that repeat administration of srKD033 in mice, and repeat

    administration of KD033 at therapeutic doses in monkeys, did not produce long-term adverse effects.

    We believe that the biological activity, efficacy, and safety of KD033 support its continued

    development toward clinical evaluation.

    Acknowledgments

    The study was supported by Kadmon. Editorial support was provided by Nathan Mata of Halloran

    Consulting Group working on behalf of Kadmon.

    References

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 18

    1. Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, et al. Engagement of the

    PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of

    lymphocyte activation. J Exp Med 2000;192:1027-34

    2. Sheppard KA, Fitz LJ, Lee JM, Benander C, George JA, Wooters J, et al. PD-1 inhibits T-cell

    receptor induced phosphorylation of the ZAP70/CD3zeta signalosome and downstream

    signaling to PKCtheta. FEBS Lett 2004;574:37-41

    3. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity

    2013;39:1-10

    4. Ribas A, Hamid O, Daud A, Hodi FS, Wolchok JD, Kefford R, et al. Association of

    Pembrolizumab With Tumor Response and Survival Among Patients With Advanced

    Melanoma. JAMA 2016;315:1600-9

    5. von Pawel J, Bordoni R, Satouchi M, Fehrenbacher L, Cobo M, Han JY, et al. Long-term

    survival in patients with advanced non-small-cell lung cancer treated with atezolizumab versus

    docetaxel: Results from the randomised phase III OAK study. Eur J Cancer 2019;107:124-32

    6. Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, et al. Five-year survival outcomes

    for patients with advanced melanoma treated with pembrolizumab in KEYNOTE-001. Ann

    Oncol 2019;30:582-8

    7. Zou W, Wolchok JD, Chen L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy:

    Mechanisms, response biomarkers, and combinations. Sci Transl Med 2016;8:328rv4

    8. Sznol M. Blockade of the B7-H1/PD-1 pathway as a basis for combination anticancer therapy.

    Cancer J 2014;20:290-5

    9. Raeber ME, Zurbuchen Y, Impellizzieri D, Boyman O. The role of cytokines in T-cell memory

    in health and disease. Immunol Rev 2018;283:176-93

    10. Lee S, Margolin K. Cytokines in cancer immunotherapy. Cancers (Basel) 2011;3:3856-93

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 19

    11. Waldmann TA. Cytokines in Cancer Immunotherapy. Cold Spring Harb Perspect Biol 2018;10

    12. Steel JC, Waldmann TA, Morris JC. Interleukin-15 biology and its therapeutic implications in

    cancer. Trends Pharmacol Sci 2012;33:35-41

    13. Huntington ND, Legrand N, Alves NL, Jaron B, Weijer K, Plet A, et al. IL-15 trans-

    presentation promotes human NK cell development and differentiation in vivo. J Exp Med

    2009;206:25-34

    14. Giri JG, Anderson DM, Kumaki S, Park LS, Grabstein KH, Cosman D. IL-15, a novel T cell

    growth factor that shares activities and receptor components with IL-2. J Leukoc Biol

    1995;57:763-6

    15. Mitchell DM, Ravkov EV, Williams MA. Distinct roles for IL-2 and IL-15 in the

    differentiation and survival of CD8+ effector and memory T cells. J Immunol 2010;184:6719-

    30

    16. Dutcher JP, Schwartzentruber DJ, Kaufman HL, Agarwala SS, Tarhini AA, Lowder JN, et al.

    High dose interleukin-2 (Aldesleukin) - expert consensus on best management practices-2014. J

    Immunother Cancer 2014;2:26

    17. Bergamaschi C, Rosati M, Jalah R, Valentin A, Kulkarni V, Alicea C, et al. Intracellular

    interaction of interleukin-15 with its receptor alpha during production leads to mutual

    stabilization and increased bioactivity. J Biol Chem 2008;283:4189-99

    18. Mortier E, Quemener A, Vusio P, Lorenzen I, Boublik Y, Grotzinger J, et al. Soluble

    interleukin-15 receptor alpha (IL-15R alpha)-sushi as a selective and potent agonist of IL-15

    action through IL-15R beta/gamma. Hyperagonist IL-15 x IL-15R alpha fusion proteins. J Biol

    Chem 2006;281:1612-9

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 20

    19. Canfield SM, Morrison SL. The binding affinity of human IgG for its high affinity Fc receptor

    is determined by multiple amino acids in the CH2 domain and is modulated by the hinge

    region. J Exp Med 1991;173:1483-91

    20. Chappel MS, Isenman DE, Everett M, Xu YY, Dorrington KJ, Klein MH. Identification of the

    Fc gamma receptor class I binding site in human IgG through the use of recombinant

    IgG1/IgG2 hybrid and point-mutated antibodies. Proc Natl Acad Sci U S A 1991;88:9036-40

    21. Diskin B, Adam S, Cassini MF, Sanchez G, Liria M, Aykut B, et al. PD-L1 engagement on T

    cells promotes self-tolerance and suppression of neighboring macrophages and effector T cells

    in cancer. Nat Immunol 2020;21:442-54

    22. Oh SA, Wu D-C, Cheung J, Navarro A, Xiong H, Cubas R, et al. PD-L1 expression by

    dendritic cells is a key regulator of T-cell immunity in cancer. Nature Cancer 2020;1:681-91

    23. Sugiura D, Maruhashi T, Okazaki IM, Shimizu K, Maeda TK, Takemoto T, et al. Restriction of

    PD-1 function by cis-PD-L1/CD80 interactions is required for optimal T cell responses.

    Science 2019;364:558-66

    24. Zhao Y, Lee CK, Lin CH, Gassen RB, Xu X, Huang Z, et al. PD-L1:CD80 Cis-Heterodimer

    Triggers the Co-stimulatory Receptor CD28 While Repressing the Inhibitory PD-1 and CTLA-

    4 Pathways. Immunity 2019;51:1059-73.e9

    25. Jochems C, Tritsch SR, Knudson KM, Gameiro SR, Rumfield CS, Pellom ST, et al. The multi-

    functionality of N-809, a novel fusion protein encompassing anti-PD-L1 and the IL-15

    superagonist fusion complex. Oncoimmunology 2019;8:e1532764

    26. Knudson KM, Hicks KC, Alter S, Schlom J, Gameiro SR. Mechanisms involved in IL-15

    superagonist enhancement of anti-PD-L1 therapy. J Immunother Cancer 2019;7:82

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 21

    27. West EE, Jin HT, Rasheed AU, Penaloza-Macmaster P, Ha SJ, Tan WG, et al. PD-L1 blockade

    synergizes with IL-2 therapy in reinvigorating exhausted T cells. J Clin Invest 2013;123:2604-

    15

    28. Chan CW, Crafton E, Fan HN, Flook J, Yoshimura K, Skarica M, et al. Interferon-producing

    killer dendritic cells provide a link between innate and adaptive immunity. Nat Med

    2006;12:207-13

    29. Guimont-Desrochers F, Lesage S. Revisiting the Prominent Anti-Tumoral Potential of Pre-

    mNK Cells. Frontiers in Immunology 2013;4

    30. Bakker AB, Phillips JH, Figdor CG, Lanier LL. Killer cell inhibitory receptors for MHC class I

    molecules regulate lysis of melanoma cells mediated by NK cells, gamma delta T cells, and

    antigen-specific CTL. J Immunol 1998;160:5239-45

    31. McMahon CW, Raulet DH. Expression and function of NK cell receptors in CD8+ T cells.

    Curr Opin Immunol 2001;13:465-70

    32. Krijgsman D, de Vries NL, Skovbo A, Andersen MN, Swets M, Bastiaannet E, et al.

    Characterization of circulating T-, NK-, and NKT cell subsets in patients with colorectal

    cancer: the peripheral blood immune cell profile. Cancer Immunol Immunother 2019;68:1011-

    24

    33. Tachibana T, Onodera H, Tsuruyama T, Mori A, Nagayama S, Hiai H, et al. Increased

    intratumor Valpha24-positive natural killer T cells: a prognostic factor for primary colorectal

    carcinomas. Clin Cancer Res 2005;11:7322-7

    Table 1. Cell lines and mouse strains used for in vivo studies

    Cell line Cancer type Cell source Media* Mouse

    Strain Supplier

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 22

    EMT-6 Breast ATCC (American Type Culture

    Collection) DMEM BALB/C LC

    CT26 Colon SIBS (Shanghai Institutes for Biological

    Sciences) RPMI1640 BALB/C LC

    H22 Liver CCTCC (China Center for Type Culture

    Collection) RPMI1640 BALB/C LC

    Pan02 Pancreatic NIH (National Institutes of Health) RPMI1640 C57BL/6 LC B16BL6 Melanoma Nanjing KeyGen Biotech Co., Ltd. RPMI1640 C57BL/6 LC

    RM-1 Prostate SIBS (Shanghai Institutes for Biological

    Sciences) RPMI1640 C57BL/6 LC

    Renca Kidney ATCC (American Type Culture

    Collection) DMEM BALB/C LC

    B16F10 Melanoma SIBS (Shanghai Institutes for Biological

    Sciences) DMEM C57BL/6 LC

    MC38 Colon FDCC (FuDan IBS Cell Center ) DMEM C57BL/6 LC MBT2 Bladder RIKEN (RIkagaku KENkyusho) RPMI1640 C3H VR

    LL/2 Lung SIBS (Shanghai Institutes for Biological

    Sciences) DMEM C57BL/6 LC

    4T1 Breast SIBS (Shanghai Institutes for Biological

    Sciences) RPMI1640 BALB/C LC

    * all with 10% FBS LC: Shanghai Lingchang Bio-Technology Co. Ltd (LC, Shanghai, China)

    VR: Vital River Laboratory Animal Technology Co., Ltd. (VR, Beijing, China)

    Table 2. Antibodies used for flow cytometry and immunohistochemistry

    Markers Fluorochrome Clone Cat. Isotypes Vendor Note CD45 Percp-cy5.5 30-F11 103132 Rat IgG2b, κ Biolegend FC CD3 BUV395 17A2 740268 Rat IgG2b, κ BD FC CD4 FITC GK1.5 100406 Rat IgG2b, κ Biolegend FC CD8 APC-H7 53-6.7 560182 Rat IgG2a, κ BD FC FoxP3 PE FJK-16s 12-5773-82 Rat IgG2a, κ eBioscience FC CD25 APC PC61 102012 Rat IgG1, λ Biolegend FC CD335 BV421 29A1.4 137612 Rat IgG2a, κ Biolegend FC Live/Dead eFluor®506 NA 65-0866-14 N/A eBioscience FC Anti-PD-L1 BV711 M1H5 563369 Rat IgG2a, λ BD FC

    gp70 tetramer APC H-2Ld

    MuLV gp70 TB-M521-2 N/A MBL FC

    β-gal tetramer PE H-2Ld β-gal TS-M511-1 N/A MBL FC

    CD8 N/A 4SM15 14-0808-82 Rat IgG2a

    mAb eBioscience

    IHC,

    1:400

    CD335 N/A N/A AF2225 Goat pAb R&D IHC,

    1:200

    B220 N/A RA3-6B2 550286 Rat IgG2a

    mAb BD

    IHC,

    1:500 Human-Fc N/A N/A ab98616 Goat pAb Abcam IHC, 1:50 ImmPRESS

    HPR Anti-Rat N/A N/A MP-7444-15 N/A Vector IHC, 2ry

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 23

    Ig ImmPRESS

    HRP Anti-Goat

    Ig N/A N/A MP-7405 N/A Vector IHC, 2ry

    FC: Flow Cytometry IHC: Immunohistochemistry

    Figure Legends

    Figure 1. PD-L1 binding and IL-15 function of srKD033 in vitro and in vivo

    A. Schematic representation of fusion proteins: KD033 and srKD033 (anti-human and -human/mouse

    PD-L1 IgG1, respectively), and ntKD033 (an IgG1 antibody that does not bind to any known targets in

    mouse), all fused to human IL-15/IL-15Rα. B. Analysis of fusion protein binding to PD-L1-expressing

    mouse CT26 colon carcinoma cells. C. Human IL-15/IL-15Rα proliferation activity in murine

    splenocytes. D. Analysis of body weight in Balb/c mice following a single i.v. dose of srKD033 (0.3, 1

    or 3 mg/kg). E. Efficacy analysis in eleven different mouse models with varied tumor PD-L1

    expression (measured when tumors reached 100 mm3) following a single i.v. dose srKD033 (3 mg/kg).

    Tumor Growth Inhibition (TGI, %), was measured against vehicle treated animals on the day the first

    animal in the control arm reached 3000 mm3 in tumor volume. F-L. Analysis of tumor growth

    following a single i.v. dose srKD033 (3 mg/kg).

    Figure 2. Tolerability and efficacy of single-dose srKD033 and ntKD033 in CT26 tumor-bearing

    Balb/c mice

    A. Body weight change in CT26 tumor-bearing Balb/c mice treated with single-dose ntKD033,

    srKD033, anti-PD-L1, or combination treatment as indicated in the legend. B-E. Anti-tumor activity of

    single-dose srKD033, ntKD033 (evaluated at their respective MTDs), or other treatments as indicated

    in the legend. F. IL-15 levels in sera, and G. spleen weights in mice treated with single-dose srKD033,

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 24

    ntKD033, or other treatments as indicated in the legend (see panel B). H, I. Measurement of splenic

    CD8 T and NK cells in mice treated with single-dose srKD033, ntKD033, or other treatments as

    indicated in the legend (see panel B). J, K. Tumor volumes and corresponding IHC analysis (panels to

    right of E and F) of tumors from CT26 Balb/c mice treated with srKD033 or ntKD033.

    Figure 3. Analysis of tumor histochemistry, peripheral immune cell markers and gene expression

    in mice treated with srKD033 and ntKD033

    A. Tumor growth of mice designated responders and non-responders after srKD033 or ntKD033

    treatment. B. Flow cytometry of peripheral blood cells from srKD033 and ntKD033 responders and

    non-responders. C, D. IHC analysis of CD8, NK, and B220+ cells, and fusion protein levels in tumors

    from CT26 tumor-bearing Balb/c mice treated with single-dose srKD033 or ntKD033. E. Tumor

    growth and NanoString analysis of gene expression changes in CT26 tumors. F. Volcano plots

    showing changes in gene expression between srKD033 and ntKD033 responders (top) and srKD033

    responders and non-responders (bottom). G. Flow cytometry of peripheral blood after CD8 T or NK

    cell depletion prior to srKD033 treatment in CT26 model. G. Effect of CD8 T and NK cell depletion

    on srKD033-mediated efficacy and survival in the CT26 model.

    Figure 4. Efficacy and durability of srKD033 treatment in the murine colon carcinoma model

    A. Analysis of tumor growth in orthotopically-implanted 4T1 tumor-bearing Balb/c mice treated with

    srKD033 and anti-CTLA-4 combination. Analysis of tumor growth (panel B, spider plots in panel D)

    and survival (panel C) in CT26 tumor-bearing Balb/c mice treated with different treatment regimens of

    srKD033 or anti-PD-1. D. Analysis of tumor growth in CT26 mice cured by treatment (tumor-free

    mice) following re-challenge with CT26 (1st), and H22 (2

    nd) tumors. E. Measurement of Gp70+ CD8 T

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • 25

    cells specific for CT26, compared to levels of non-specific B-galactosidase+ CD8 T cells in

    splenocytes of mice treated with either srKD033 or srKD033+anti-PD-1.

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/

  • Published OnlineFirst December 8, 2020.Mol Cancer Ther Stella A Martomo, Dan Lu, Zhanna Polonskaya, et al. gene signatures and memory responsessynergistic anti-tumor immunity with robust tumor-immune Single-dose anti-PD-L1/IL-15 fusion protein KD033 generates

    Updated version

    10.1158/1535-7163.MCT-20-0457doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://mct.aacrjournals.org/content/suppl/2020/12/08/1535-7163.MCT-20-0457.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://mct.aacrjournals.org/content/early/2020/12/08/1535-7163.MCT-20-0457To request permission to re-use all or part of this article, use this link

    on July 1, 2021. © 2020 American Association for Cancer Research. mct.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 December 8, 2020; DOI: 10.1158/1535-7163.MCT-20-0457

    http://mct.aacrjournals.org/lookup/doi/10.1158/1535-7163.MCT-20-0457http://mct.aacrjournals.org/content/suppl/2020/12/08/1535-7163.MCT-20-0457.DC1http://mct.aacrjournals.org/cgi/alertsmailto:[email protected]://mct.aacrjournals.org/content/early/2020/12/08/1535-7163.MCT-20-0457http://mct.aacrjournals.org/

    Article FileFigure 1Figure 2Figure 3Figure 4