12
Cancer Therapy: Preclinical NKTR-214, an Engineered Cytokine with Biased IL2 Receptor Binding, Increased Tumor Exposure, and Marked Efcacy in Mouse Tumor Models Deborah H. Charych, Ute Hoch, John L. Langowski, Steve R. Lee, Murali K. Addepalli, Peter B. Kirk, Dawei Sheng, Xiaofeng Liu, Paul W. Sims, Laurie A.VanderVeen, Cherie F. Ali, Thomas K. Chang, Marina Konakova, Rhoneil L. Pena, Rupesh S. Kanhere, Yolanda M. Kirksey, Chunmei Ji,Yujun Wang, Jicai Huang,Theresa D. Sweeney, Seema S. Kantak, and Stephen K. Doberstein Abstract Purpose: Aldesleukin, recombinant human IL2, is an effective immunotherapy for metastatic melanoma and renal cancer, with durable responses in approximately 10% of patients; however, severe side effects limit maximal dosing and thus the number of patients able to receive treatment and potential cure. NKTR-214 is a prodrug of conjugated IL2, retaining the same amino acid sequence as aldesleukin. The IL2 core is conjugated to 6 releasable polyethylene glycol (PEG) chains. In vivo, the PEG chains slowly release to generate active IL2 conjugates. Experimental Design: We evaluated the bioactivity and recep- tor binding of NKTR-214 and its active IL2 conjugates in vitro; the tumor immunology, tumor pharmacokinetics, and efcacy of NKTR-214 as a single agent and in combination with antiCTLA-4 antibody in murine tumor models. Tolerability was evaluated in non-human primates. Results: In a murine melanoma tumor model, the ratio of tumor-killing CD8 þ T cells to Foxp3 þ regulatory T cells was greater than 400 for NKTR-214 compared with 18 for aldesleukin, supporting preferential activation of the IL2 receptor beta over IL2 receptor alpha, due to the location of PEG molecules. NKTR- 214 provides a 500-fold greater exposure of the tumor to conju- gated IL2 compared with aldesleukin. NKTR-214 showed efcacy as a single agent and provided durable immunity that was resistant to tumor rechallenge in combination with antiCTLA- 4 antibody. NKTR-214 was well tolerated in non-human primates. Conclusions: These data support further evaluation of NKTR- 214 in humans for a variety of tumor types, adding to the repertoire of potent and potentially curative cancer immunothera- pies. Clin Cancer Res; 22(3); 68090. Ó2016 AACR. Introduction Immunotherapy offers potential for durable responses in a growing list of tumor types (1). Aldesleukin, recombinant human IL2, was the rst cancer immunotherapy, and one of the rst recombinant proteins, approved by the FDA in 1992. High-dose IL2 (HD-IL2) administration results in severe hypotension and vascular leak syndrome, which has relegated its use to specialized care centers. With use of appropriate supportive care, aldesleukin has demonstrated complete cancer regression in about 10% of patients treated for metastatic melanoma and renal cancer (24). Approximately 70% of patients with complete responses have been cured, maintaining complete regression for more than 25 years after initial treatment (26). Unfortunately, the side-effect prole restricts optimal IL2 dosing, limiting the number of patients who might achieve durable responses. IL2 stimulates immune cell proliferation and activation through a receptor-signaling complex containing alpha (IL2Ra, CD25), beta (IL2Rb, CD122), and common gamma chain receptors (g c , CD132; refs. 7, 8), but the effects are pleiotropic and contextual. At high doses, IL2 binds to heterodimeric IL2Rbg receptor leading to desired expansion of tumor killing CD8 þ memory effector T (CD8 T) cells (9). IL2 also binds to its heterotrimeric receptor IL2Rabg with greater afnity, which expands immunosuppressive CD4 þ CD25 þ regulatory T cells (Tregs) expressing high constitutive levels of IL2Ra. Expansion of Tregs represents an undesirable effect of IL2 for cancer immunotherapy (713). We hypothesized IL2 conjugation with polyethylene glycol (PEG) could be used to mask the region of IL2 that interacts with the IL2Ra subunit responsible for activating immunosuppressive Tregs, biasing activity towards tumor killing CD8 þ T cells. PEGy- lation could alter the immunostimulatory prole of aldesleukin without mutation of its amino acid sequence while creating an inactive prodrug, mitigating rapid systemic immune activation, and improving tolerability (14). We report the development of, NKTR-214, a biologic prodrug consisting of IL2 bound by 6 releasable polyethylene glycol (PEG) chains. We evaluated receptor bias, bioactivity, tumor immunol- ogy, pharmacokinetics, pharmacodynamics, and tolerability of NKTR-214. Antitumor effects of NKTR-214 were compared with aldesleukin as a single agent and in combination with an Nektar Therapeutics, San Francisco, California. Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Corresponding Author: Deborah H. Charych, Nektar Therapeutics, 455 Mission Bay Blvd South, San Francisco, CA 94158. Phone: 415-482-5558; Fax: 415-339- 5300; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-15-1631 Ó2016 American Association for Cancer Research. Clinical Cancer Research Clin Cancer Res; 22(3) February 1, 2016 680 on August 11, 2020. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

NKTR-214, an Engineered Cytokine with Biased IL2 Receptor ...Seema S. Kantak, and Stephen K. Doberstein Abstract Purpose: Aldesleukin, recombinant human IL2, is an effective immunotherapy

  • Upload
    others

  • View
    8

  • Download
    0

Embed Size (px)

Citation preview

Page 1: NKTR-214, an Engineered Cytokine with Biased IL2 Receptor ...Seema S. Kantak, and Stephen K. Doberstein Abstract Purpose: Aldesleukin, recombinant human IL2, is an effective immunotherapy

Cancer Therapy: Preclinical

NKTR-214, an Engineered Cytokine with BiasedIL2 Receptor Binding, Increased Tumor Exposure,and Marked Efficacy in Mouse Tumor ModelsDeborah H. Charych, Ute Hoch, John L. Langowski, Steve R. Lee, Murali K. Addepalli,Peter B. Kirk, Dawei Sheng, Xiaofeng Liu, Paul W. Sims, Laurie A. VanderVeen,Cherie F. Ali, Thomas K. Chang, Marina Konakova, Rhoneil L. Pena, Rupesh S. Kanhere,Yolanda M. Kirksey, Chunmei Ji, Yujun Wang, Jicai Huang, Theresa D. Sweeney,Seema S. Kantak, and Stephen K. Doberstein

Abstract

Purpose: Aldesleukin, recombinant human IL2, is an effectiveimmunotherapy for metastatic melanoma and renal cancer, withdurable responses in approximately 10% of patients; however,severe side effects limit maximal dosing and thus the number ofpatients able to receive treatment and potential cure. NKTR-214is a prodrug of conjugated IL2, retaining the same amino acidsequence as aldesleukin. The IL2 core is conjugated to 6 releasablepolyethylene glycol (PEG) chains. In vivo, the PEG chains slowlyrelease to generate active IL2 conjugates.

Experimental Design:We evaluated the bioactivity and recep-tor binding of NKTR-214 and its active IL2 conjugates in vitro; thetumor immunology, tumor pharmacokinetics, and efficacy ofNKTR-214 as a single agent and in combination with anti–CTLA-4 antibody in murine tumor models. Tolerability wasevaluated in non-human primates.

Results: In a murine melanoma tumor model, the ratio oftumor-killingCD8þT cells to Foxp3þ regulatory T cellswas greaterthan 400 for NKTR-214 compared with 18 for aldesleukin,supporting preferential activation of the IL2 receptor beta overIL2 receptor alpha, due to the location of PEG molecules. NKTR-214 provides a 500-fold greater exposure of the tumor to conju-gated IL2 compared with aldesleukin. NKTR-214 showed efficacyas a single agent and provided durable immunity that wasresistant to tumor rechallenge in combination with anti–CTLA-4 antibody. NKTR-214 was well tolerated in non-humanprimates.

Conclusions: These data support further evaluation of NKTR-214 in humans for a variety of tumor types, adding to therepertoire of potent andpotentially curative cancer immunothera-pies. Clin Cancer Res; 22(3); 680–90. �2016 AACR.

IntroductionImmunotherapy offers potential for durable responses in a

growing list of tumor types (1). Aldesleukin, recombinant humanIL2, was the first cancer immunotherapy, and one of the firstrecombinant proteins, approved by the FDA in 1992. High-doseIL2 (HD-IL2) administration results in severe hypotension andvascular leak syndrome, which has relegated its use to specializedcare centers. With use of appropriate supportive care, aldesleukinhas demonstrated complete cancer regression in about 10% ofpatients treated for metastatic melanoma and renal cancer (2–4).Approximately 70% of patients with complete responses havebeen cured, maintaining complete regression for more than 25years after initial treatment (2–6). Unfortunately, the side-effectprofile restricts optimal IL2 dosing, limiting the number ofpatients who might achieve durable responses.

IL2 stimulates immune cell proliferation and activationthrough a receptor-signaling complex containing alpha (IL2Ra,CD25), beta (IL2Rb, CD122), and common gamma chainreceptors (gc, CD132; refs. 7, 8), but the effects are pleiotropicand contextual. At high doses, IL2 binds to heterodimericIL2Rbg receptor leading to desired expansion of tumor killingCD8þ memory effector T (CD8 T) cells (9). IL2 also binds to itsheterotrimeric receptor IL2Rabg with greater affinity, whichexpands immunosuppressive CD4þ CD25þ regulatory T cells(Tregs) expressing high constitutive levels of IL2Ra. Expansionof Tregs represents an undesirable effect of IL2 for cancerimmunotherapy (7–13).

We hypothesized IL2 conjugation with polyethylene glycol(PEG) could be used to mask the region of IL2 that interacts withthe IL2Ra subunit responsible for activating immunosuppressiveTregs, biasing activity towards tumor killing CD8þ T cells. PEGy-lation could alter the immunostimulatory profile of aldesleukinwithout mutation of its amino acid sequence while creating aninactive prodrug, mitigating rapid systemic immune activation,and improving tolerability (14).

We report the development of, NKTR-214, a biologic prodrugconsisting of IL2 boundby 6 releasable polyethylene glycol (PEG)chains. We evaluated receptor bias, bioactivity, tumor immunol-ogy, pharmacokinetics, pharmacodynamics, and tolerabilityof NKTR-214. Antitumor effects of NKTR-214 were comparedwith aldesleukin as a single agent and in combination with an

Nektar Therapeutics, San Francisco, California.

Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

Corresponding Author: Deborah H. Charych, Nektar Therapeutics, 455 MissionBay Blvd South, San Francisco, CA 94158. Phone: 415-482-5558; Fax: 415-339-5300; E-mail: [email protected]

doi: 10.1158/1078-0432.CCR-15-1631

�2016 American Association for Cancer Research.

ClinicalCancerResearch

Clin Cancer Res; 22(3) February 1, 2016680

on August 11, 2020. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 2: NKTR-214, an Engineered Cytokine with Biased IL2 Receptor ...Seema S. Kantak, and Stephen K. Doberstein Abstract Purpose: Aldesleukin, recombinant human IL2, is an effective immunotherapy

anti-CTLA-4 antibody, using murine tumor models. The resultssupport the future development of NKTR-214 for treatment ofcancer.

Materials and MethodsRecombinant protein and antibodies

Aldesleukin was purchased from Myoderm and diluted in therecommended product formulation buffer for in vivo studies.NKTR-214 was manufactured by Nektar Therapeutics. ELISAstudy materials are described in Supplementary Materials andMethods.

For in vivo murine studies, aldesleukin and anti–CTLA-4 anti-bodywere administered intraperitoneally; NKTR-214was admin-istered intravenously by tail-vein injection. For each animal, theconcentration was adjusted to achieve the specified dose at aninjection volumeof 0.2mL for aldesleukin, 0.1mL for anti–CTLA-4 antibody, and 8 to 10 mL/kg for NKTR-214. NKTR-214 dosingwas based on the IL2 equivalent content of the conjugate.

AnimalsAnimal studies were conducted under accreditation by the

Association for Assessment and Accreditation of Laboratory Ani-malCare or theCanadianCouncil onAnimalCare. All studiesmetthe ethical and humane criteria for transportation, housing, andcare established by United States NIH guidelines or Canadiananimal care regulators. Female C57BL/6 NCrl mice aged 6 to 8weeks weighing 15 to 20 g and female Balb/CNCrlmice aged 6 to8 weeks weighing 16 to 20 g were purchased from Charles RiverLaboratories. Male and female cynomolgus monkeys, aged 2 to 3years weighing 2.3 to 2.8 kg at treatment initiation, were obtainedfrom Primus Bio-Resources Inc. Male na€�ve adult cynomolgusmonkeys, aged 3 to 4 years weighing 3.7 to 4.3 kg at treatmentinitiation, were obtained from Worldwide Primates Inc.

Cell linesIL2-dependent CTLL-2 mouse T-lymphocyte cell line, B16F10

melanoma, EMT6 mammary, and CT26 colon carcinoma celllines were obtained from ATCC. All cells were verified pathogen

andmycoplasma (IDEXX BioResearch) free and authenticated byATCC using Cytochrome C subunit I PCR assay.

Production and purification of anti–CTLA-4UC10-4F10-11 hybridoma (15), which secretes hamster anti-

mouse CTLA-4 antibody with mouse IgG2a isotype, was pur-chased from ATCC (HB-304). PFHM-II medium (Invitrogen) wasused to culture the cell line under 5% CO2. The cell line wasmaintained between 1 � 105 and 1 � 106 cells/mL and anti–CTLA-4 was expressed and purified (16).

Degree of PEGylation for NKTR-214A releasable conjugate of glycine was used as a reference

standard. Reference standard orNKTR-214was incubated at roomtemperature for 45 minutes with 37.5 mmol/L NaOH to releasePEG from IL2 or glycine. The reaction was neutralized with anequal volume of 100 mmol/L HCl. Samples were analyzed byreverse phase high-performance liquid chromatography (HPLC)to quantify released protein, glycine, and PEG. PEG molesreleased fromNKTR-214were calculated using the standard curvefrom releasable PEG-glycine standard. Degree of PEGylation wascalculated as ratio of PEGmoles to protein moles, or ratio of areaunder the curve (AUC) of released PEG to AUC of IL2. Proceduresfor mapping PEGylation sites are included in the SupplementaryMaterials and Methods.

In vitro hydrolysis kinetics of NKTR-214Hydrolysis ofNKTR-214wasmonitored in vitro inPBS at pH7.4

at 37�C. At indicated time points, levels of released IL2 conjugateswere measured using reverse phase HPLCwith corresponding IL2conjugate reference standards. Data were fit to a kinetic modelusing the Berkeley-Madonna software. The study was performedlong enough to obtain the half-life of release.

Identification of the active released IL2 conjugatesActivity of NKTR-214 and its released IL2 conjugates was

measured by activation of pSTAT5 in CTLL-2 cells using SectorImager 2400 plate reader and assay reagents from Meso ScaleDiscovery, LLC as described in Supplementary Materials andMethods. Total and phosphorylation of STAT5 (pSTAT5) levelswere detected using the MSD Phospho(Tyr694)/Total STAT5a,bWhole Cell Lysate Kit. Methods for biophysical characterizationand affinity of NKTR-214 binding to IL2 receptor subunits aredescribed in Supplementary Materials and Methods.

Tumor exposure of NKTR-214 compared with aldesleukinC57BL/6 mice were implanted subcutaneously into the right

flank with B16F10 melanoma cells (1 � 106 per animal). Sevendays after implantation, when tumors measured 200 mm3, ani-mals were administered NKTR-214 (2 mg/kg � 1) or aldesleukin(3 mg/kg daily � 5). Tumors were harvested (n ¼ 4 per obser-vation time), homogenized in ice-cold PBS containing proteaseinhibitor (Roche) and 0.25% acetic acid, and centrifuged toobtain supernatant. To quantify NKTR-214 levels in tumor tissue,PEG was released from IL2 by incubating the supernatant in a pH9 buffer at 37�C overnight. IL2 was measured by sandwich ELISAspecific for human IL2. To calculate AUC, data were fit withPheonix WinNonLin using a noncompartmental model. AUCafter aldesleukin was estimated on the basis of day 1 AUCmultiplied by 5.

Translational Relevance

Recent attention in immunotherapy for treatment of cancerhas focused on the successes of checkpoint inhibitor antibo-dies. However, administration of the recombinant cytokineIL2 aldesleukin produces durable responses lasting up to twodecades in approximately 10% of patients treated for meta-static melanoma and renal cancer. IL2 is an activator andsuppressor of the immune system and is associatedwith severeside effects such as hypotension and vascular leak syndrome,limiting widespread use. Recent studies have shown thatdurable responses may increase with combined immu-notherapies. We evaluated NKTR-214, a polymer conjugateof IL2 that tips the balance in favor of immune activation oversuppression in the tumor, with improved efficacy and toler-ability compared with aldesleukin. Our data provide therationale for further evaluation ofNKTR-214 in clinical studiesas both a single agent and in combination with other immu-notherapies including checkpoint inhibitor antibodies.

NKTR-214, an Engineered Cytokine for Cancer Immunotherapy

www.aacrjournals.org Clin Cancer Res; 22(3) February 1, 2016 681

on August 11, 2020. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 3: NKTR-214, an Engineered Cytokine with Biased IL2 Receptor ...Seema S. Kantak, and Stephen K. Doberstein Abstract Purpose: Aldesleukin, recombinant human IL2, is an effective immunotherapy

Immune cell phenotypingMice bearing B16F10 melanoma tumors (as described

above) were administered NKTR-214 (2 mg/kg � 1), aldesleukin(3mg/kg daily� 5), or vehicle (daily� 5). Tumors were collected5, 7, and 10 days following initial dose andwere within one SD ofmean tumor volume for each group. Following mechanical andenzymatic digestion, phenotypic assessment of immune cellsubtypes in tumor microenvironment was determined by flowcytometry. Immunohistochemical analysis of T-cell density wasperformed on frozen tumor sections 7 days after administration(Supplemental Materials and Methods).

Activity of NKTR-214 in murine tumor modelsAll studies were conducted in therapeutic mode with dosing

initiated in the presence of established tumors with volume inexcess of 100mm3. Tumormeasurements and body weights wererecorded 3 times perweek. For all studies,mean tumor volume foreach groupwas graphically represented until >20%of the originalcohort was removed due to tumor volume constraints. For thesingle-agent study, mice bearing B16F10 melanoma tumors wererandomized 7 days after implantation and administered NKTR-214 2 mg/kg every 9 days for 3 doses (n ¼ 9 mice), aldesleukin 3mg/kg twice daily for 5 days (2 cycles, n¼ 9mice), or vehicle for 5doses (n¼ 12mice). For EMT6mammary (n¼ 10 per group) andCT26 colon carcinoma (n ¼ 12 per group) combination efficacystudies, 2�106 cellswere implanted into the rightflankof BALB/cmice. Seven days after implantation, mice were randomized andadministered NKTR-214 0.8mg/kg every 9 days for 3 doses, anti–CTLA-4 100 mg twice weekly through day 18, vehicle control, oranti–CTLA-4 and NKTR-214 given in combination. Two groupsimplanted with EMT6 were administered either aldesleukin 0.5mg/kg daily for 5 days (2 cycles) or aldesleukin combination withanti–CTLA-4.

For the tumor rechallenge study, Balb/c mice were implantedsubcutaneously in the right flank with 2 � 106 EMT6 mammarycarcinoma cells. Seven days after inoculation, mice were random-ized and administered antibody control (n ¼ 10 mice) or acombination of anti–CTLA-4 and NKTR-214 (n ¼ 80 mice).Forty-nine days after dosing initiation, tumor-free animals exhi-biting complete response to combined immunotherapy wereenrolled into two groups receiving either rechallenge of 2 �106 EMT6or unrelated CT26 colon carcinoma cells. An additionalcohort of age-appropriate na€�ve animals was challenged withEMT6 cells (n ¼ 10 mice). After 109 days, animals that rejectedthe EMT6 rechallenge were inoculated with 2 � 106 EMT6 cells(n ¼ 14 mice). Age-appropriate na€�ve animals (n ¼ 5 mice) weresimilarly challenged; all groups were followed with no additionaltreatment.

Evaluation of immune markers and potential hypotension andvascular leak syndrome in non-human primates

For evaluation of immune markers, cynomolgus monkeys(n ¼ 3/sex/dose) received a single intravenous bolus dose intothe saphenous veinofNKTR-2140.01, 0.03, and0.1mg/kg. Bloodsamples were collected to evaluate effects of NKTR-214 on lym-phocyte and soluble CD25 counts as described in SupplementaryMaterials and Methods.

Arterial blood pressure was used to monitor potential hypo-tension and vascular leak syndrome. Male monkeys (n¼ 4) weresurgically implanted with telemetry transmitters (TL11M2-D70-PCT; Data Sciences International). Each received NKTR-214 as a

slow intravenous bolus every 14 days for 4 doses (0, 0.01, 0.03,and 0.1 mg/kg) according to Latin-square study design. Contin-uous telemetry recordings were collected from each consciousunrestricted monkey for 0 to 24 hours (time for peak NKTR-214concentration), 30 to 54 hours (time for peak active 2-PEG-IL2and 1-PEG-IL2 concentration), and 80 to 104 hours (peak immu-nological and clinical signs observed) after each dose. Arterialblood pressure was collected every 15 minutes over each telem-etry-recording period.

Inflammatory cytokines TNFa and IFNg were analyzed inmonkeys at 1, 3, 6, 12, 24, 48, 72, and 168 hours after a single0.1 mg/kg dose of NKTR-214 by flow cytometry using a Cyto-metric Bead Array for Non-Human Primate (NHP) Th1/Th2 kit(BD Biosciences). Onemilliliter of blood was collected via saphe-nous vein and resulting serum samples were diluted 1 in 10 intubes containing a mixture of beads coated with capture anti-bodies specific to IFNg and TNFa. Bound cytokines were detectedusing phycoerythrin-conjugated detection antibodies. The IFNgand TNFa concentrations were interpolated using a 5-parameterlogistic nonlinear regression equation, established from thehuman Th1/Th2 cytokine standard curves ranging from 20 to5,000 pg/mL for IFNg and of 10 to 5,000 pg/mL for TNFa.

Statistical analysesFor identification of active released IL2 conjugates, EC50 values

for each compound were determined from sigmoidal dose–response curves using GraphPad Prism 5.01. In mouse efficacystudies, results are shown as mean � SEM. Tumor growth inhibi-tion was calculated as percent reduction in volume for treatedgroups relative to vehicle. The significance between experimentaland vehicle groups was compared using ANOVA, with the Tukeypost test. Differences between NKTR-214 and aldesleukin wereanalyzed using unpaired Student t test. P values < 0.05 were consi-dered significant. Kaplan–Meier survival curves represent percentof tumors in each group with a volume less than their 4-fold ofinitial day 0 value (tumor volume quadrupling time, TVQT). Stati-stical significance was calculated utilizing GraphPad Prism v6.04.

ResultsDesign and characterization of NKTR-214

NKTR-214 is a prodrug consisting of IL2 conjugated to 6releasable PEG chains. The amino acid sequence of the IL2 is thesame as aldesleukin. The PEG reagent and conjugation reactionwere optimized to facilitate localization on IL2 at lysine residuesclustered at the IL2/IL2Ra interface (Fig. 1). Enzymatic digestionof NKTR-214 followed by hydrolytic removal of PEG on thepeptides confirmed on average 6 PEG chains conjugated anddistributed at K31, K34, K42, K47, K48, and K75 (cleavagepeptides 20–51 and 68–94). Amino acids within these cleavagepeptides form the hydrophobic center of the IL2/IL2Ra interface,surrounded by a polar periphery containing key ion-pairingresidues (17). These key hydrophobic and charged IL2/IL2Rainteraction sites are at or near sites of PEGylation. When NKTR-214 is hydrolyzed to its most active 1-PEG-IL2 state (describedbelow), the last remaining PEG is distributed at the IL2/IL2Rainterface.

Because of the high degree of PEGylation initially, NKTR-214is an inactive prodrug and serves as "reservoir" revealing activeconjugated IL2 after slow release of PEG chains in vivo (Fig. 2A).The prodrug effect is designed tomitigate rapid systemic immune

Charych et al.

Clin Cancer Res; 22(3) February 1, 2016 Clinical Cancer Research682

on August 11, 2020. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 4: NKTR-214, an Engineered Cytokine with Biased IL2 Receptor ...Seema S. Kantak, and Stephen K. Doberstein Abstract Purpose: Aldesleukin, recombinant human IL2, is an effective immunotherapy

activation such as what might occur with HD-IL2 regimen, whileallowing sustained concentrations of active conjugated IL2 in thetumor. In vitro hydrolysis studies demonstrate PEG release occurssequentially, with a half-life of 20 hours for each step in therelease, supporting the lack of a "burst" IL2 release effect (Sup-plementary Fig. S1). Unconjugated IL2 is cleared rapidly in vivowith a half-life of 13 minutes in mice and 1.4 hours in humans(18, 19). Therefore, slow release of PEG relative to fast clearance ofIL2 provides minimal exposure to free IL2 in vivo.

Identification of the active IL2 conjugates released fromNKTR-214

To assess bioactivity of NKTR-214 and identify active conju-gated IL2 species formed after PEG release, we evaluated levels ofpSTAT5 in murine T cells (Fig. 2B) after treatment in culture. Asanticipated, NKTR-214 was not active with an EC50 > 4,000 ng/mL, consistent with the prodrug design. Unconjugated aldesleu-kin had an EC50 of 0.4 ng/mL, similar to reported values (18). Invitro, PEG chains were released hydrolytically from NKTR-214,forming a mixture of conjugated IL2 species ranging from 5-PEG-IL2 to 1-PEG-IL2, which were each purified. 1-PEG-IL2 and 2-PEG-IL2 species were identified as the most active IL2 conjugateswith EC50 values of 2 and 20 ng/mL, respectively. Positionalisomer analysis of synthesized 1-PEG-IL2 indicated two position-al isomers (of the six potential K sites described above), whichwere identically bioactive in the pSTAT assay. The 3-PEG-IL2 wasfound to be inactive with EC50 > 1,300 ng/mL. Therefore, speciescontaining � 3-PEGs are inactive; those with < 3-PEGs are active.

Biophysical affinity characterization of NKTR-214 binding to IL2receptor subunits indicates PEG chains impact affinity to IL2Ra toa greater extent than IL2Rb compared with IL2 (SupplementaryTable S1; Supplementary Fig. S2).

NKTR-214 provides 500-fold increased tumor exposure relativeto aldesleukin

To assess tumor exposure, NKTR-214 or aldesleukin levels weremeasured in mice bearing B16F10 melanoma tumors. As shownin Fig. 3, tumor aldesleukin levels rapidly reached Cmax and thenrapidly declined, leading to <4 ng/g concentrations 24 hours aftereach dose and a daily AUC of 0.09� 0.02 mg/hour/g. In contrast,NKTR-214 was detectable in tumors for up to 8 days after a singledose achieving an AUC of 30 � 6.9 mg/hour/g. On the basis ofAUC, a single dose of NKTR-214 led to a 67-fold higher exposurecompared with 5 daily doses of aldesleukin, even though 7.5fewer IL2 equivalents were dosed using NKTR-214 (3mg/kg daily� 5¼ 15 mg/kg vs. 2 mg/kg). Thus, normalizing exposure on thebasis of IL2 equivalents, NKTR-214 achieved a 500-fold increasedexposure relative to aldesleukin. The active conjugated IL2 formofNKTR-214 (2-PEG-IL2 and 1-PEG-IL2 together) was also quan-tified and remained detectable in tumor for up to 5 days yieldinganAUCof 23�4.4mg/g.Hence, exposure to active conjugated IL2was 50-times higher compared with aldesleukin, translating to380 times increased exposure relative to an equivalent dose ofaldesleukin. The tumor exposure ofNKTR-214 allowsdosing onceevery 9 days in mice compared with twice daily for two 5-daycycles for aldesleukin.

Immune cell phenotyping: NKTR-214 favors proliferation andinfiltration of memory CD8 T cells over Tregs in the tumor

The in vitro binding and activation profiles described abovesuggest PEGylation interfered with the interaction between IL2and IL2Ra relative to aldesleukin. We therefore tested whetherthese effects bias the profile of immune cell subtypes in vivo. Thenumber of CD8 T and Treg cells in the tumor following admin-istration of either NKTR-214 or IL2 is a key measure of whetherpleiotropic effects of IL2 have been shifted due to conjugation ofIL2 to PEG at the IL2/IL2Ra interface. To address the question,mice bearing subcutaneous B16F10 mouse melanoma tumorswere treated with a single dose of NKTR-214 or 5 doses ofaldesleukin, and immune cells in the tumor microenvironmentwere quantified by flow cytometry. In tumors of aldesleukin-treated mice, total and memory CD8 cells were increased as apercentage of tumor-infiltrating lymphocytes; however, theseeffects were transient, reaching significance relative to vehicle onday 5 (Fig. 4A). In contrast, significant (P < 0.05) and sustainedtotal andmemory CD8 T-cell stimulation was achieved followinga single NKTR-214 administration, with superior percentages ofmemory CD8 (day 7) and total CD8 (days 7 and 10) relative toaldesleukin. Both NKTR-214 and aldesleukin treatment resultedin increased activated natural killer (NK) cells 5 and 7 days aftertreatment initiation, though this effect was diminished by day 10.CD4 cell percentages of tumor-infiltrating lymphocytes werediminished following NKTR-214 treatment relative to vehicle onday 5. On day 10, NKTR-214 resulted in fewer CD4 cell percen-tages compared with vehicle and aldesleukin. The CD4 cellpopulation was further analyzed for the FoxP3þ subset, whichdefines the Treg population. NKTR-214 administration reducedpercentage of Tregs at every time point, consistent with reducedaccess to the IL2Ra subunit arising from the PEG chains. In

Figure 1.Model demonstrating region of PEGylation sites at IL2 interface with IL2Ra.NKTR-214 is IL2 at its core (green) bound by lysines to 6 PEG chains permolecule as determined by reverse phase HPLC quantification of completelyreleased PEG and protein (8 replicates for 4 lots of NKTR-214). Sites ofPEGylation are depicted by red circles. The gc receptor is depicted in pink,IL2Rb in aqua blue, and IL2Ra in dark blue. Only peptide 68–94(VLNLAQSK75NFHLRPRDLISNINVIVLE) and peptide 20–51(LQMILNGINNYK31NPK34LTRMLTFK42FYMPK47K48ATE) were bound by PEG.The pegylation results represent 6 separate experiments, each with 2replicates, using 3 batches of NKTR-214.

NKTR-214, an Engineered Cytokine for Cancer Immunotherapy

www.aacrjournals.org Clin Cancer Res; 22(3) February 1, 2016 683

on August 11, 2020. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 5: NKTR-214, an Engineered Cytokine with Biased IL2 Receptor ...Seema S. Kantak, and Stephen K. Doberstein Abstract Purpose: Aldesleukin, recombinant human IL2, is an effective immunotherapy

contrast, Treg reduction with aldesleukin was modest achievingsignificance on day 5. The increase of CD8 T cells and reduction ofTregs led to amarked elevation of the CD8/Treg ratio in the tumorby day 7. The ratio of CD8/Treg for NKTR-214, aldesleukin, andvehicle was 449, 18, and 4, respectively.

While these results provide quantification of immune cells inthe tumor microenvironment, the spatial relationship between Tcells and tumor cells cannot be visualized. Therefore, immuno-histochemical staining was performed (Fig. 4B), with represen-tative images showing CD8 T cells are not only increased innumber but are interspersed with tumor cells.

These results indicate NKTR-214 is capable of inducing morerobust in vivomemory effectorCD8T-cell response, than seenwithaldesleukin, without a commensurate stimulation of Tregs intumor, consistent with in vitro IL2Rb-biased binding profile ofactive IL2 conjugates derived fromNKTR-214. These in vivo effectsarise although 7.5-fold fewer IL2 equivalents were administeredfor NKTR-214 compared with aldesleukin.

NKTR-214 provides superior single-agent efficacy comparedwith aldesleukin in an aggressive mouse melanoma model

Given the markedly increased ratio of CD8 T cells/Tregs inthe tumormicroenvironment following NKTR-214 treatment, wecompared the efficacy of NKTR-214 to aldesleukin in the B16F10

murine melanoma model. NKTR-214 and aldesleukin led tosignificant tumor growth inhibition at day 9 compared withvehicle, 80% and 91%, respectively (Fig. 5A). In animals whoreceived vehicle, the tumors rapidly grew to the endpoint of 1,500mm3 8 days after treatment, leading to median tumor growthdelay, defined as time to reach 4 times the initial volume or TVQTof 5 days. Aldesleukin significantly increased the TVQT to 10 dayscompared with vehicle. For animals treated with NKTR-214, theTVQTwas increased 16.7 days providing significant tumor growthinhibition compared with aldesleukin, despite 10-fold fewer IL2equivalents and reduced dosing frequency (Fig. 5B). Although nosignificant body weight loss was recorded,mice in the aldesleukingroup became hypothermic and exhibited shivering behavior,unlike the NKTR-214–treated group where mild weakness wasobserved.

NKTR-214 is synergistic with an anti–CTLA-4 antibodyCheckpoint inhibitor antibody anti–CTLA-4 antagonizes the

inhibitory interaction between CTLA-4 and its ligand B7 onantigen-presenting cells (20). To test the concept of T-cell activa-tion using dual mechanisms of checkpoint blockade and directcytokine stimulation, NKTR-214 and murine anti–CTLA-4 wereevaluated in two syngeneic tumormodels. No significant effect ontumor growth was seen when the EMT6 murine breast tumor

Figure 2.A, schematic of the inactive NKTR-214 prodrug releasing to IL2 conjugates bound by fewer PEG chains with increasing bioactivity. Red depicts the IL2 cytokine core,and orange depicts the polymer chains located at the IL2/IL2Ra interface. B, identification of the most active IL2 conjugates created by hydrolysis ofNKTR-214 as measured by phosphorylation of STAT5 in murine CTLL-2 T cells. Purified forms of the released conjugated IL2 formswere assayed. On the basis of theEC50, the 2-PEG-IL2 and 1-PEG-IL2 derived from NKTR-214 are identified as the most active conjugates released from NKTR-214. EC50 values for STAT5phosphorylation were determined from two or more independent experiments, each performed in triplicate. The IL2 control is a purified reference standard.

Charych et al.

Clin Cancer Res; 22(3) February 1, 2016 Clinical Cancer Research684

on August 11, 2020. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 6: NKTR-214, an Engineered Cytokine with Biased IL2 Receptor ...Seema S. Kantak, and Stephen K. Doberstein Abstract Purpose: Aldesleukin, recombinant human IL2, is an effective immunotherapy

model was treated with single-agent NKTR-214, aldesleukin, oranti–CTLA-4, Fig. 6A.When aldesleukinwas combinedwith anti–CTLA-4, tumor growth inhibition of 83%was achieved on day 18;however, the combinationofNKTR-214 andCTLA-4provedmoreefficacious leading to tumor regression of�3.8%. After treatmentended on day 21, both groups were followed for long-term tumorgrowth delay. At the end of study (day 100), the aldesleukincombination group maintained 40% tumor-free animals, with amean TVQT of 32.5 days, which was significant compared withvehicle (TVQT 10.8 days). On the other hand, the TVQT of theNKTR-214 and anti–CTLA-4 group could not be determined as70% of the animals were tumor-free at day 100; 78 days after thelast dose of NKTR-214 was given. Although the CT26 murinecolon tumor model also exhibits aggressive growth, single-agentNKTR-214 and anti–CTLA-4 each produced significant, if mod-erate, tumor growth inhibition of 51% and 60%, respectively, atday 11 comparedwith vehicle (Fig. 6B).Neither single agent led tosignificant mean TVQT changes, with 10% of animals remainingtumor free at day 100. However, the combination of these agentsproved synergistic, with 89% tumor growth inhibition at day 11and 67% tumor-free animals at day 100. For all treatment groups,the combinations were well tolerated with no body weight lossand no other obvious clinical signs. These data indicate synergybetween NKTR-214 and checkpoint blockade, with each using acomplementary mechanism of immune activation. Data demon-strating the antitumor immunity stimulated by this combinationrequires innate and adaptive immune responses are shown inSupplementary Fig. S3.

NKTR-214 combined with anti–CTLA-4 induces durable andspecific antitumor immunity

The combination studies described above suggest durabilityof response; animals remain tumor-free for an extended periodafter their last therapeutic dose. To more fully explore thedurability and specificity of antitumor activity induced by

NKTR-214 combined with anti–CTLA-4 in the EMT6 model,mice with complete responses to therapy (n ¼ 30) were rechal-lenged with a second EMT6 tumor implant or nonrelated CT26implant (n ¼ 10) and monitored without additional therapy(Fig. 6C and D). Tumor-free animals proved resistant to EMT6rechallenge with 70% of the mice remaining tumor-free, whileCT26 tumor implants proceeded to grow. The tumor-free micefrom the EMT6 tumor rechallenge were challenged a third timewith EMT6, and followed without additional treatment. After60 days, 100% of these animals remained tumor-free. Theresistance to tumor growth strongly suggests antitumor immu-nity induced by this combination therapy is tumor-specific andlong-lasting. Antibody depletion experiments (SupplementaryMaterials and Methods and Supplementary Fig. S3) demon-strated CD8 T and NK cells were required for protection fromrechallenge.

NKTR-214 does not produce hypotension or vascular leaksyndrome in non-human primates at the maximumtolerated dose

To proceed with clinical testing of NKTR-214 in humans,safety was evaluated in cynomolgus monkeys. To establish thecynomolgus monkey as a suitable toxicology species, humanand monkey peripheral blood mononuclear cells from threedonors each were exposed to human IL2 and active 1-PEG-IL2from NKTR-214. The resulting IL2R activation measured bypSTAT5 signaling was similar between monkey and humanprimary T cells (Supplementary Table S2). The maximumtolerated dose (MTD) was 0.1 mg/kg. Soluble CD25 and totallymphocyte counts were used as pharmacodynamic markers ofimmune stimulation (21). Both biomarkers increased in adose-responsive manner above baseline producing 24-fold and4-fold increases in soluble CD25 and lymphocytes respectivelyat MTD (Supplementary Fig. S4A). As vascular leak syndrome isa known toxicity of HD-IL2 that is detected through hypoten-sion, cardiovascular safety (ECG and blood pressure) wasmonitored in cynomolgus monkeys using telemetry. No hypo-tension or ECG abnormalities were observed at MTD for 104hours after dose administration (Supplementary Fig. S4B). Lackof pulmonary edema was further confirmed by histopathology(data not shown) with no evidence of lung infiltrates. Studies ofinflammatory cytokines TNFa and IFNg showed no detectablelevels, suggesting lack of toxicity-associated cytokine release,despite robust immune activation (22). Together with themurine antitumor efficacy, these results support the improvedsafety and tolerability of NKTR-214.

DiscussionThis report describes the development of NKTR-214 for

cancer therapy. To our knowledge, this is the first use of aprodrug design for biologic therapy using PEG, releasable PEGlinkers, and site-directed PEG to alter the immunologic, phar-macokinetic, and biodistribution profile of the molecule (Figs.3 and 4). The agent shows marked efficacy in aggressive murinetumors as a single agent and in combination with anti–CTLA-4(Figs. 5 and 6). NKTR-214 is a highly "combinable cytokine"dosed more like an antibody than a cytokine; anticipateddosing schedule in humans is once every 21 days. Yet NKTR-214 provides a mechanism of direct immune stimulation

Figure 3.Tumor pharmacokinetics of NKTR-214 and its released active conjugated-IL2 forms. B16F10 mouse melanoma tumor tissue was analyzed after asingle dose of NKTR-214 or 5 daily doses of aldesleukin. NKTR-214 (allconjugated IL2 species, black squares) and active conjugated IL2 fromNKTR-214 (1-PEG-IL2 and 2-PEG-IL2 blue circles) are shown. Aldesleukin isindicated by red triangles. When adjusted for dose equivalency, tumorexposure for NKTR-214 was approximately 500 times higher as 7.5-foldfewer total IL2 equivalents were administered for NKTR-214. Activeconjugated IL2 forms from NKTR-214 are 380 times higher thanaldesleukin. For aldesleukin, solid red lines are measured; dotted red linesrepresent extrapolated values based on measured values after day 1 and 5.Graph shows mean � SD, n ¼ 4 mice per time point.

NKTR-214, an Engineered Cytokine for Cancer Immunotherapy

www.aacrjournals.org Clin Cancer Res; 22(3) February 1, 2016 685

on August 11, 2020. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 7: NKTR-214, an Engineered Cytokine with Biased IL2 Receptor ...Seema S. Kantak, and Stephen K. Doberstein Abstract Purpose: Aldesleukin, recombinant human IL2, is an effective immunotherapy

characteristic of cytokines. The combination of NKTR-214 withmurine anti–CTLA-4 results in tumor-free animals remainingtumor-free for up to 100 days and resistant to tumor rechal-lenge (Fig. 6). NKTR-214 is well tolerated in non-humanprimates, with no evidence of hypotension and vascular leaksyndrome at its MTD (Supplementary Fig. S4B).

PEGylation technology has existed for decades leading toavailability of multiple protein conjugates across diverse ther-apeutic areas including oncology (pegfilgrastim), infectiousdisease (peginterferon alpha-2b) and autoimmune disease(certolizumab pegol; ref. 23). In the past, the primary functionof PEG was to improve half-life using standard stable conju-gation chemistry (24–26). In this report, PEG is used to bias

receptor selectivity and alter pharmacokinetics. No mutationswere introduced into the molecule; therefore, the core of NKTR-214 is identical to the well-understood aldesleukin amino acidsequence. The presence of multiple PEG chains creates aninactive prodrug, which prevents rapid systemic immune acti-vation upon administration. Use of releasable linkers allowsPEG chains to slowly hydrolyze, continuously forming activeconjugated IL2 bound by 2-PEGs or 1-PEG. PEGylation dra-matically alters the pharmacokinetics of NKTR-214 comparedwith IL2, providing a 500-fold increase in AUC in the tumorcompared with an IL2 equivalent dose. The location of PEGchains at the IL2/IL2Ra interface interferes with binding tohigh-affinity IL2Ra, while leaving binding to low-affinity IL2Rb

Figure 4.Immune cell alterations in B16F10 mouse melanoma models following treatment with a single dose of NKTR-214 or 5 daily doses of aldesleukin. Tumor-infiltrating lymphocytes were isolated from animals at the time points indicated and immune cell populations were assessed by flow cytometry. Each datapoint represents an individual mouse tumor and the line represents the mean. Data are combined from 2 to 4 independent studies with 3 to 4 replicates ateach time point. A, in the tumor, NKTR-214 administration led to marked increases in total and memory CD8 T cells, greater in magnitude and durationthan with aldesleukin. Both treatments led to increased NK cells. NKTR-214 also led to significant decreases in Treg percentages, leading to higherCD8/Treg ratios of >400 compared with aldesleukin (ratio of 18) and vehicle (ratio of 4). B, immunohistochemical staining of B16F10 tumors showingCD8 T cells (brown) in close proximity to tumor cells (purple). Representative fields from 30 to 60 images obtained per group are shown, n¼ 3 mice per group.Data shown are for day 7 after dose administration.

Charych et al.

Clin Cancer Res; 22(3) February 1, 2016 Clinical Cancer Research686

on August 11, 2020. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 8: NKTR-214, an Engineered Cytokine with Biased IL2 Receptor ...Seema S. Kantak, and Stephen K. Doberstein Abstract Purpose: Aldesleukin, recombinant human IL2, is an effective immunotherapy

unperturbed (Supplementary Fig. S2). The bias observed in vitrochanges the immune cell profile in vivo in the tumor microen-vironment, leading to a maximal CD8/Tregs ratio of >400 forNKTR-214 compared with 18 for aldesleukin treatment (Fig.4A). Overcoming immune tolerance in the tumor microenvi-ronment may require tipping the balance in favor of CD8 T overTregs in the tumor (10–13). Furthermore, CD8 T cells areextensively interspersed with tumor cells (Fig. 4B). Infiltrationof CD8 T cells is relevant because previous immunotherapiessuch as tumor antigen–specific vaccines may have failed due topoor trafficking of these cells into the tumor (11). NK cellresponses are also mobilized by NKTR-214 treatment; bothinnate and adaptive immune responses appear to play a role inantitumor immunity. Together, these results suggest NKTR-214favors immune activation over suppression in the tumor, with a

mechanism different from the original cytokine and comple-mentary to checkpoint blockade antibodies.

Numerous approaches to improve the safety profile ofaldesleukin have been reported; however, these were chal-lenged by the absence of a defined mechanism for IL2 toxicity.One hypothesis implicated NK cells as the mechanism oftoxicity; a mutein biased towards IL2Ra and against activationof NK cells was developed. However, this proved ineffective inthe clinic with a lower MTD than aldesleukin (27, 28). Sub-sequently, other reports described modified forms of IL2 thathave attempted to favor ILRbg binding, prolong exposure, ormitigate toxicity. These include mutation of the IL2 amino acidsequence, fusion to a targeting antibody, or use of IL2/anti-IL2antibody complexes (29–31). These alterations, like every newagent, need to be evaluated clinically. Notably, NKTR-214provides increased tumor exposure without assuming tumortarget selectivity and without the complications of fusing anantibody to a cytokine. Unnatural protein fusion junctionspose greater risk for immunogenicity. Receptor bias shownwith NKTR-214 is achieved using the clinically validated alde-sleukin amino acid sequence and PEG, which is regarded assafe and nonimmunogenic.

To assess the impact of receptor bias, prodrug design, and hightumor concentrations on in vivo efficacy, we chose the subcuta-neous B16F10 mouse melanoma model, a tumor known to beaggressive and difficult to treat (32). NKTR-214 showed signifi-cantly improved efficacy in this model as a single agent comparedwith aldesleukin. The improvement was achieved despite using10-fold fewer IL2 equivalents. While 20 total doses of aldesleukinwere needed to achievemodest efficacy, only 3doses ofNKTR-214were administered (Fig. 5). Interestingly anti–CTLA-4 was notactive in the B16F10 melanoma model as a single agent (32, 33).The combination of NKTR-214 with anti–CTLA-4 proved syner-gistic in mouse efficacy models of breast and colon tumors, with83% and 67% of animals remaining tumor-free up to 100 days,respectively (Fig. 6A and B). Tumor-free animals could be chal-lenged twice with the same tumors, and up to 100% of theseanimals resisted tumor regrowth without further therapy (Fig.6C). Recent clinical and preclinical studies combining immu-notherapies such anti–CTLA-4 and anti–PD-1 indicate morepatients may benefit when multiple mechanisms of immuneactivation are targeted (34–37). Our results suggest NKTR-214 ispositioned to provide a complementary mechanism of immuneactivation in combination, using dosing schedules concurrentwith antibody therapy.

NKTR-214 is administered in vivo as an inactive prodrug,potentially mitigating toxicities associated with rapid off-tar-get systemic immune activation, which would occur uponadministration of HD-IL2 or other biologically active cyto-kines. Ability to dose fewer IL2 equivalents at a significantlyreduced frequency may also contribute to the improved tol-erability observed with NKTR-214. In cynomolgus monkeys,NKTR-214 could be dosed at MTD without cardiovascularadverse effects, reflected in the absence of hypotension andvascular leak syndrome. Total lymphocyte and soluble CD25levels were used as pharmacodynamic markers and indicateclear dose-dependent immune stimulation in monkeys, withno evidence of cytokine release syndrome. These results sug-gest unlike HD-IL2, NKTR-214 may be dosed safely inhumans, potentially increasing the number of patients achiev-ing durable responses.

Figure 5.Single-agent efficacy of NKTR-214 compared with aldesleukin in well-established subcutaneous B16F10 mouse melanoma tumors. Mice weretreated with NKTR-214, aldesleukin, or vehicle. Changes in mean tumorvolume over time (A) and the percentage of tumors below 4 times the initialvolume over time (B) are shown. Symbols are mean � SEM. � , statisticalsignificance (P < 0.05) compared with vehicle (ANOVA, Tukey post test);z, P < 0.05 compared with aldesleukin (unpaired Student t test). Theexperiment was repeated a total of 7 times with similar results.

NKTR-214, an Engineered Cytokine for Cancer Immunotherapy

www.aacrjournals.org Clin Cancer Res; 22(3) February 1, 2016 687

on August 11, 2020. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 9: NKTR-214, an Engineered Cytokine with Biased IL2 Receptor ...Seema S. Kantak, and Stephen K. Doberstein Abstract Purpose: Aldesleukin, recombinant human IL2, is an effective immunotherapy

Although it is difficult to directly translate efficacy in mousetumor models to human cancers, it is encouraging that the IL2pathway is already a validated cancer immunotherapy target witha history of durable benefit for some patients. The efficacy ofcombination therapy using checkpoint blockade antibodiesappears to translate between mice and humans, increasing theprobability that NKTR-214 will produce robust efficacy in pati-ents with cancer (34, 37). Although much of the current focus

in cancer immunotherapy is on immune checkpoint blockadeusing antibodies, cytokines will play an important role in pro-viding complementary mechanisms of action, as single agentsand in combination. The data presented in this report indicatethe immunologic, pharmacokinetic, and pharmacologic proper-ties of the endogenous cytokine IL2 were significantly alteredby strategic polymer conjugation. As such, NKTR-214 is currentlybeing evaluated in a Phase 1 clinical study in solid tumors.

Figure 6.Combination of NKTR-214 withcheckpoint inhibition and tumorrechallenge. A, EMT6 breast model:aldesleukin or NKTR-214 incombination with anti–CTLA-4generated significant tumor growthinhibition (left; � , day 18) comparedwith vehicle. A higher percentage oftumor-free animals was achievedwhen NKTR-214 was combined withanti–CTLA-4 (right; z, 70% day 100);n ¼ 12 mice per group. B, CT-26 colonmodel: anti–CTLA-4 and NKTR-214 assingle agents generated significantgrowth inhibition (�), but thecombination of both agents wassynergistic (left; z, day 11) and lead tosignificant percentage of tumor-freeanimals (right; � , 67% day 100);n ¼ 12 mice per group. Tumorrechallenge data: mean tumorvolumes (C) and individual animalmeasurements (D). Combinationtherapy induced complete responsesin 44% of treated animals. Tumor-freeresponders were resistant to firstEMT6 rechallenge, with 70%remaining tumor-free, but weresusceptible to CT26 tumor growth.Complete responders from the firstrechallenge group were rechallengeda second timewith EMT6 implants, and100% remained tumor-free withoutadditional treatment. Symbolsrepresent mean � SEM. � , P < 0.05compared with vehicle (ANOVA,Tukey post test); z, P < 0.05 comparedwith aldesleukin (unpairedStudent t test).

Charych et al.

Clin Cancer Res; 22(3) February 1, 2016 Clinical Cancer Research688

on August 11, 2020. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 10: NKTR-214, an Engineered Cytokine with Biased IL2 Receptor ...Seema S. Kantak, and Stephen K. Doberstein Abstract Purpose: Aldesleukin, recombinant human IL2, is an effective immunotherapy

Disclosure of Potential Conflicts of InterestD. Sheng, T.K. Chang, and J. Huang have ownership interest (including

patents) inNektar Therapeutics. No potential conflicts of interest were disclosedby the other authors.

Authors' ContributionsConception and design: D.H. Charych, U. Hoch, J.L. Langowski, S.R. Lee,P.B. Kirk, X. Liu, C.F. Ali, J. Huang, S.S. Kantak, S.K. DobersteinDevelopment of methodology: D.H. Charych, U. Hoch, J.L. Langowski,S.R. Lee, M.K. Addepalli, P.B. Kirk, D. Sheng, X. Liu, L.A. VanderVeen, C.F. Ali,T.K. Chang, M. Konakova, R.S. Kanhere, Y. Wang, J. Huang, S.S. Kantak,S.K. DobersteinAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): D.H. Charych, S.R. Lee, M.K. Addepalli, P.B. Kirk,X. Liu, P.W. Sims, L.A. VanderVeen, C.F. Ali, T.K. Chang, Y.M. Kirksey, C. Ji,Y. Wang, T.D. Sweeney, S.S. KantakAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): D.H. Charych, U. Hoch, J.L. Langowski, S.R. Lee,M.K. Addepalli, P.B. Kirk, X. Liu, L.A. VanderVeen,C.F. Ali, T.K.Chang, R.L. Pena,R.S. Kanhere, Y.M. Kirksey, C. Ji, Y. Wang, T.D. Sweeney, S.S. KantakWriting, review, and/or revision of the manuscript: D.H. Charych, J.L. Lan-gowski, S.R. Lee, P.B. Kirk, D. Sheng, P.W. Sims, L.A. VanderVeen, C.F. Ali, T.K.Chang, M. Konakova, C. Ji, J. Huang, T.D. Sweeney, S.S. Kantak, S.K. Doberstein

Administrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): J.L. Langowski, S.R. Lee, M.K. Addepalli,Y.M. Kirksey, C. Ji, S.S. KantakStudy supervision: D.H. Charych, U. Hoch, J.L. Langowski, S.R. Lee,M.K. Addepalli, R.L. Pena, C. Ji, S.S. Kantak, S.K. DobersteinOther (initiated and developed the concept of the program and moleculewith help from the team): D.H. Charych

AcknowledgmentsThe authors thank Dr. Mario Sznol, Yale University, for his careful reading

and critique of this manuscript. The authors also thank Pai Charaweeyawong,Nektar Therapeutics, for his technical contributions and Phillips GilmoreOncology Communications, Inc. for technical support with manuscriptpreparation.

Grant SupportThis study was financially supported by Nektar Therapeutics.The costs of publication of this articlewere defrayed inpart by the payment of

page charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received July 14, 2015; revisedOctober 21, 2015; acceptedOctober 23, 2015;published online February 1, 2016.

References1. Pennock G, Chow L. The evolving role of immune checkpointe inhibitors

in cancer treatment. Oncologist 2015;20:1–11.2. Rosenberg SA. Raising the bar: the curative potential of human cancer

immunotherapy. Sci Transl Med 2012;4:127ps8.3. Smith FO, Downey SG, Klapper JA, Yang JC, Sherry RM, Royal RE, et al.

Treatment of metastatic melanoma using interleukin-2 alone or in con-junction with vaccines. Clin Cancer Res 2008;14:5610–8.

4. Klapper JA, Downey SG, Smith FO, Yang JC, Hughes MS, Kammula US,et al. High-dose interleukin-2 for the treatment of metastatic renal cellcarcinoma: a retrospective analysis of response and survival in patientstreated in the Surgery Branch at theNational Cancer Institute between 1986and 2006. Cancer 2008;113:293–301.

5. AtkinsMB, LotzeMT,Dutcher JP, Fisher RI,WeissG,MargolinK, et al.High-dose recombinant interleukin 2 therapy for patients with metastatic mel-anoma: analysis of 270 patients treated between 1985 and 1993. J ClinOncol 1999;17:2105–16.

6. Rosenberg SA, Yang JC, White DE, Steinberg SM. Durability of completeresponses in patients with metastatic cancer treated with high-dose inter-leukin-2 - Identification of the antigens mediating response. Ann Surg1998;228:307–19.

7. Wang X, Rickert M, Garcia KC. Structure of the quaternary complex ofinterleukin-2 with its alpha, beta, and gammac receptors. Science 2005;310:1159–63.

8. Margolin K. Cytokine therapy in cancer. Expert Opin Biol Ther 2008;8:1495–505.

9. Boyman O, Sprent J. The role of interleukin-2 during homeostasis andactivation of the immune system. Nat Rev Immunol 2012;12:180–90.

10. Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 combi-nation blockade expands infiltrating T cells and reduces regulatory T andmyeloid cells within B16 melanoma tumors. Proc Natl Acad Sci U S A2010;107:4275–80.

11. Gajewski TF, Woo SR, Zha Y, Spaapen R, Zheng Y, Corrales L, et al. Cancerimmunotherapy strategies based on overcoming barriers within the tumormicroenvironment. Curr Opin Immunol 2013;25:268–76.

12. Zhou P, Zheng X, Zhang H, Liu Y, Zheng P. B7 blockade alters the balancebetween regulatory T cells and tumor-reactive T cells for immunotherapy ofcancer. Clin Cancer Res 2009;15:960–70.

13. Wang HY, Lee DA, Peng G, Guo Z, Li Y, Kiniwa Y, et al. Tumor-specifichuman CD4þ regulatory T cells and their ligands: implications for immu-notherapy. Immunity 2004;20:107–18.

14. Rickert M, Wang X, Boulanger MJ, Goriatcheva N, Garcia KC. The structureof interleukin-2 complexed with its alpha receptor. Science 2005;308:1477–80.

15. Walunas TL, Lenschow DJ, Bakker CY, Linsley PS, Freeman GJ, Green JM,et al. CTLA-4 can function as a negative regulator of T cell activation.Immunity 1994;1:405–13.

16. UC10-4F10-11 (ATCC� HB-304�) culture method; 2015 [accessed2015 Jun 29]. Available from http://www.atcc.org/Products/All/HB-304.aspx#culturemethod.

17. Stauber DJ, Debler EW, Horton PA, Smith KA, Wilson IA. Crystal structureof the IL-2 signaling complex: paradigm for a heterotrimeric cytokinereceptor. Proc Natl Acad Sci U S A 2006;103:2788–93.

18. Gibbons JA, Luo ZP, Hannon ER, Braeckman RA, Young JD. Quantitationof the renal clearance of interleukin-2 using nephrectomized and ureter-ligated rats. J Pharmacol Exp Ther 1995;272:119–25.

19. Konrad MW, Hemstreet G, Hersh EM, Mansell PW, Mertelsmann R, KolitzJE, et al. Pharmacokinetics of recombinant interleukin 2 inhumans. CancerRes 1990;50:2009–17.

20. Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunityby CTLA-4 blockade. Science 1996;271:1734–6.

21. Thompson JA, Curti BD, Redman BG, Bhatia S,Weber JS, Agarwala SS, et al.Phase I study of recombinant interleukin-21 in patients with metastaticmelanoma and renal cell carcinoma. J Clin Oncol 2008;26:2034–9.

22. Anderson JA, Lentsch AB,Hadjiminas DJ,Miller FN,Martin AW,NakagawaK, et al. The role of cytokines, adhesion molecules, and chemokines ininterleukin-2-induced lymphocytic infiltration in C57BL/6 mice. J ClinInvest 1996;97:1952–9.

23. Patel JN,Walko CM. Sylatron: a pegylated interferon for use inmelanoma.Ann Pharmacother 2012;46:830–8.

24. Parveen S, Sahoo SK. Nanomedicine: clinical applications of polyethyleneglycol conjugated proteins and drugs. Clin Pharmacokinet 2006;45:965–88.

25. Fishburn CS. The pharmacology of PEGylation: balancing PD with PK togenerate novel therapeutics. J Pharm Sci 2008;97:4167–83.

26. Kunugi K, Yamaoka T. Polymers in nanomedicine. Springer-Verlag, Hei-delberg; New York, NY; 2012. p.xi, 281p.

27. Shanafelt AB, Lin Y, Shanafelt MC, Forte CP, Dubois-Stringfellow N,Carter C, et al. A T-cell-selective interleukin 2 mutein exhibits potentantitumor activity and is well tolerated in vivo. Nat Biotechnol2000;18:1197–202.

28. Margolin K, Atkins MB, Dutcher JP, Ernstoff MS, Smith JW, Clark JI, et al.Phase I trial of BAY 50-4798, an interleukin-2-specific agonist in advancedmelanoma and renal cancer. Clin Cancer Res 2007;13:3312–9.

29. Krieg C, L�etourneau S, Pantaleo G, Boyman O. Improved IL-2 immuno-therapy by selective stimulation of IL-2 receptors on lymphocytes andendothelial cells. Proc Natl Acad Sci U S A 2010;107:11906–11.

www.aacrjournals.org Clin Cancer Res; 22(3) February 1, 2016 689

NKTR-214, an Engineered Cytokine for Cancer Immunotherapy

on August 11, 2020. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 11: NKTR-214, an Engineered Cytokine with Biased IL2 Receptor ...Seema S. Kantak, and Stephen K. Doberstein Abstract Purpose: Aldesleukin, recombinant human IL2, is an effective immunotherapy

30. Gillies SD, Lan Y, Hettmann T, Brunkhorst B, Sun Y, Mueller SO, et al. Alow-toxicity IL-2-based immunocytokine retains antitumor activitydespite its high degree of IL-2 receptor selectivity. Clin Cancer Res2011;17:3673–85.

31. LevinAM, BatesDL, RingAM,Krieg C, Lin JT, Su L, et al. Exploiting a naturalconformational switch to engineer an interleukin-2 `superkine'. Nature2012;484:529–33.

32. Sundstedt A, Celander M, Eriksson E, Torngren M, Hedlund G. Mono-therapeutically nonactive CTLA-4 blockade results in greatly enhancedantitumor effects when combined with tumor-targeted superantigens in aB16 melanoma model. J Immunother 2012;35:344–53.

33. Quezada SA, Peggs KS, Curran MA, Allison JP. CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effectorand regulatory T cells. J Clin Invest 2006;116:1935–45.

34. Ott PA, Hodi FS, Robert C. CTLA-4 and PD-1/PD-L1 blockade: newimmunotherapeutic modalities with durable clinical benefit in melanomapatients. Clin Cancer Res 2013;19:5300–9.

35. Wolchok JD, Kluger H, CallahanMK, PostowMA, Rizvi NA, Lesokhin AM,et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med2013;369:122–33.

36. Hafalla JC, Claser C, Couper KN, Grau GE, Renia L, de Souza JB, et al. TheCTLA-4 and PD-1/PD-L1 inhibitory pathways independently regulate hostresistance to Plasmodium-induced acute immune pathology. PLoS Patho-log 2012;8:e1002504.

37. Duraiswamy J, Kaluza KM, Freeman GJ, Coukos G. Dual blockadeof PD-1 and CTLA-4 combined with tumor vaccine effectivelyrestores T-cell rejection function in tumors. Cancer Res 2013;73:3591–603.

Clin Cancer Res; 22(3) February 1, 2016 Clinical Cancer Research690

Charych et al.

on August 11, 2020. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 12: NKTR-214, an Engineered Cytokine with Biased IL2 Receptor ...Seema S. Kantak, and Stephen K. Doberstein Abstract Purpose: Aldesleukin, recombinant human IL2, is an effective immunotherapy

2016;22:680-690. Clin Cancer Res   Deborah H. Charych, Ute Hoch, John L. Langowski, et al.   Tumor Models

MouseBinding, Increased Tumor Exposure, and Marked Efficacy in NKTR-214, an Engineered Cytokine with Biased IL2 Receptor

  Updated version

  http://clincancerres.aacrjournals.org/content/22/3/680

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2016/01/29/22.3.680.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/22/3/680.full#ref-list-1

This article cites 35 articles, 18 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/22/3/680.full#related-urls

This article has been cited by 24 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected]

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

  Permissions

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

.http://clincancerres.aacrjournals.org/content/22/3/680To request permission to re-use all or part of this article, use this link

on August 11, 2020. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from