cancer nk cell mediated cytotoxicity contributes to tumor ... · cancer nk cell–mediated...

8
CANCER NK cellmediated cytotoxicity contributes to tumor control by a cytostatic drug combination Marcus Ruscetti 1 *, Josef Leibold 1 *, Matthew J. Bott 1 *, Myles Fennell 1 , Amanda Kulick 2 , Nelson R. Salgado 1 , Chi-Chao Chen 1 , Yu-jui Ho 1 , Francisco J. Sanchez-Rivera 1 , Judith Feucht 3 , Timour Baslan 1 , Sha Tian 1 , Hsuan-An Chen 1 , Paul B. Romesser 1 , John T. Poirier 2,4 , Charles M. Rudin 2,4 , Elisa de Stanchina 2 , Eusebio Manchado 1 , Charles J. Sherr 5,6 , Scott W. Lowe 1,6 Molecularly targeted therapies aim to obstruct cell autonomous programs required for tumor growth. We show that mitogen-activated protein kinase (MAPK) and cyclin-dependent kinase 4/6 inhibitors act in combination to suppress the proliferation of KRAS-mutant lung cancer cells while simultaneously provoking a natural killer (NK) cell surveillance program leading to tumor cell death. The drug combination, but neither agent alone, promotes retinoblastoma (RB) protein-mediated cellular senescence and activation of the immunomodulatory senescence-associated secretory phenotype (SASP). SASP components tumor necrosis factora and intercellular adhesion molecule1 are required for NK cell surveillance of drug-treated tumor cells, which contributes to tumor regressions and prolonged survival in a KRAS-mutant lung cancer mouse model. Therefore, molecularly targeted agents capable of inducing senescence can produce tumor control through noncell autonomous mechanisms involving NK cell surveillance. T he KRAS oncogene is frequently mutated in several human cancers. It drives tumor- igenesis by constitutively activating growth factor signaling pathways that promote un- controlled proliferation, namely the mitogen- activated protein kinase (MAPK) or phosphoinositide 3-kinase pathways. Although much effort has been placed on targeting KRAS or its downstream effectors, to date, most therapeutic agents have failed, owing to an inability to sustain inhibition of RAS-driven signaling (1, 2). Combinatorial strategies are being developed to circumvent these effects, for example, by combining MAPK kinase (MEK) inhibitors with upstream recep- tor tyrosine kinase inhibitors to thwart adaptive resistance mechanisms (3, 4). Another approach involves combining MEK inhibitors with down- stream cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors that, in principle, could more potent- ly block the proliferation of KRAS-mutant cells by simultaneously reducing MAPK-regulated cyclin D levels and directly targeting CDK4 kinase activity (5). In addition to the intrinsic effects on tumor cell proliferation, both MEK and CDK4/6 inhibitors can modulate T cell function as single agents or in combination with T cell checkpoint blockade (68). We explored the cell autonomous and noncell autonomous effects of combining MEK and CDK4/6 inhibitors using KRAS-mutant tumor models. We first tested a number of highly selec- tive CDK4/6 inhibitors (palbociclib, abemaciclib, ribociclib) in combination with the U.S. Food and Drug Administrationapproved MEK in- hibitor trametinib in human KRAS-mutant lung and pancreatic cancer cell lines. Compared with treatment with either single agent, the two-drug combination was substantially more effective at inhibiting proliferation as well as phospho- rylation of the retinoblastoma (RB) protein, a direct CDK4 and 6 target (Fig. 1A and fig. S1). Accordingly, the combination of trametinib and palbociclib was more effective at impairing tu- mor growth and inducing tumor stasis in mice harboring a KRAS-mutant lung cancer patient- derived xenograft (PDX), when treated at the maximally tolerated dose for each agent (Fig. 1B) (9, 10). Similar results were also observed in other KRAS-mutant PDX models treated at lower doses (Fig. 1C and fig. S2, A and B), confirming that the combination produces biological effects that neither drug can achieve alone. These human xenograft studies require the use of immunodeficient NOD-scid IL2Rg null (NSG) mice. To assess whether and to what extent tumor cell responses are altered by the immune system, we made use of an established syngeneic transplant mouse model of lung cancer. Mouse tumor cells derived from a Kras G12D/+ ;Trp53 -/- (KP) lung tumor (11) were engineered to express luciferase and green fluorescent protein (GFP) and tested for drug sensitivity in vitro or after intravenous injection into C57BL/6 immuno- competent mice (Fig. 1D), where they produced aggressive lung adenocarcinomas by 1-week post transplantation. As in the human models, the trametinib plus palbociclib combination syner- gistically suppressed the growth of cultured KP tumor cells (fig. S2C) and significantly increased the survival of lung tumorbearing animals (Fig. 1E). This effect was substantially impaired when the same cells were transplanted into immuno- deficient NSG mice (fig. S2D and E), suggest- ing that the immune system might contribute to treatment efficacy. Immune profiling of lung tumors after 1 week of combination treatment revealed a general influx of CD45 + immune cells compared with single-agent treatment (fig. S3A). Whereas there was no change in B cell or macrophage num- bers in tumors after treatment, there was an increased infiltration of CD4 + and CD8 + T cells in the lungs and spleens of both tumor-bearing and naïve (tumor-free) mice after either single- agent or combination therapy. However, changes in CD4 + or CD8 + T cell activation were not ob- served, as assessed by staining for CD69, KLRG1, and CD107a, a marker of lymphocyte degranula- tion (figs. S3, B to D; S4, A and B; and S5, A to D). Thus, the drug combination does not appear to produce selective activation of T cells in this lung cancer model. By contrast, combined treatment with trame- tinib and palbociclib triggered the selective ac- cumulation of natural killer (NK) cells and a decrease in Gr-1 hi CD11b + myeloid-derived sup- pressor cells (MDSCs) in the lungs of tumor- bearing mice compared with treatment with either agent alone (Fig. 1, F and G; and figs. S3, E and F; S4, C and D; and S5F). NK cells de- rived from combination-treated tumors appeared activated and more mature, as indicated by cell surface expression of CD107a and other activat- ing receptors and altered transcriptional profiles showing a reduction in proliferation-associated genes and an increase in expression of genes linked to NK cell maturation and cytotoxicity (Fig. 1G and figs. S4C, S5E, and S6). Combined treatment also led to NK cell accumulation (but not activation) and a decrease in MDSCs in the spleens of tumor-bearing mice but not in the lungs or spleens of naïve mice (figs. S3, F to H, and S5, E to G), suggesting that this im- mune effect is tumor dependent. To determine whether specific immune cells contribute to the outcome of combination ther- apy, we assessed the impact of perturbing immune cell function on the survival of tumor- bearing (immunocompetent) mice after vehicle or trametinib and palbociclib therapy. Neither depletion of macrophages, Gr-1 + granulocytes/ MDSCs, and CD4 + and CD8 + T cells using block- ing antibodies nor antiPD-1 immunotherapy (to stimulate exhausted T cells) had any im- pact on the survival of vehicle or combination- treated mice (figs. S7, A to D, and S8). By contrast, RESEARCH Ruscetti et al., Science 362, 14161422 (2018) 21 December 2018 1 of 7 1 Department of Cancer Biology and Genetics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. 2 Department of Molecular Pharmacology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. 3 Center for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. 4 Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. 5 Department of Tumor Cell Biology, St. Jude Childrens Research Hospital, Memphis, TN 38105, USA. 6 Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA. *These authors contributed equally to this work. Corresponding Author. Email: [email protected] on June 13, 2020 http://science.sciencemag.org/ Downloaded from

Upload: others

Post on 06-Jun-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: CANCER NK cell mediated cytotoxicity contributes to tumor ... · CANCER NK cell–mediated cytotoxicity contributes to tumor control bya cytostatic drug combination Marcus Ruscetti

CANCER

NK cell–mediated cytotoxicitycontributes to tumor control by acytostatic drug combinationMarcus Ruscetti1*, Josef Leibold1*, Matthew J. Bott1*, Myles Fennell1, Amanda Kulick2,Nelson R. Salgado1, Chi-Chao Chen1, Yu-jui Ho1, Francisco J. Sanchez-Rivera1,Judith Feucht3, Timour Baslan1, Sha Tian1, Hsuan-An Chen1, Paul B. Romesser1,John T. Poirier2,4, Charles M. Rudin2,4, Elisa de Stanchina2, Eusebio Manchado1,Charles J. Sherr5,6, Scott W. Lowe1,6†

Molecularly targeted therapies aim to obstruct cell autonomous programs requiredfor tumor growth. We show that mitogen-activated protein kinase (MAPK) andcyclin-dependent kinase 4/6 inhibitors act in combination to suppress theproliferation of KRAS-mutant lung cancer cells while simultaneously provoking anatural killer (NK) cell surveillance program leading to tumor cell death. The drugcombination, but neither agent alone, promotes retinoblastoma (RB) protein-mediatedcellular senescence and activation of the immunomodulatory senescence-associatedsecretory phenotype (SASP). SASP components tumor necrosis factor–a andintercellular adhesion molecule–1 are required for NK cell surveillance of drug-treatedtumor cells, which contributes to tumor regressions and prolonged survival in aKRAS-mutant lung cancer mouse model. Therefore, molecularly targeted agents capableof inducing senescence can produce tumor control through non–cell autonomousmechanisms involving NK cell surveillance.

The KRAS oncogene is frequently mutatedin several human cancers. It drives tumor-igenesis by constitutively activating growthfactor signaling pathways that promote un-controlled proliferation, namely themitogen-

activatedproteinkinase (MAPK)orphosphoinositide3-kinase pathways. Although much effort hasbeen placed on targeting KRAS or its downstreameffectors, to date, most therapeutic agents havefailed, owing to an inability to sustain inhibitionof RAS-driven signaling (1, 2). Combinatorialstrategies are being developed to circumventthese effects, for example, by combining MAPKkinase (MEK) inhibitors with upstream recep-tor tyrosine kinase inhibitors to thwart adaptiveresistance mechanisms (3, 4). Another approachinvolves combining MEK inhibitors with down-stream cyclin-dependent kinase 4 and 6 (CDK4/6)inhibitors that, in principle, could more potent-ly block the proliferation of KRAS-mutant cellsby simultaneously reducing MAPK-regulatedcyclin D levels and directly targeting CDK4 kinaseactivity (5). In addition to the intrinsic effects on

tumor cell proliferation, both MEK and CDK4/6inhibitors canmodulate T cell function as singleagents or in combination with T cell checkpointblockade (6–8).We explored the cell autonomous and non–

cell autonomous effects of combining MEK andCDK4/6 inhibitors using KRAS-mutant tumormodels. We first tested a number of highly selec-tive CDK4/6 inhibitors (palbociclib, abemaciclib,ribociclib) in combination with the U.S. Foodand Drug Administration–approved MEK in-hibitor trametinib in human KRAS-mutant lungand pancreatic cancer cell lines. Compared withtreatment with either single agent, the two-drugcombination was substantially more effectiveat inhibiting proliferation as well as phospho-rylation of the retinoblastoma (RB) protein, adirect CDK4 and 6 target (Fig. 1A and fig. S1).Accordingly, the combination of trametinib andpalbociclib was more effective at impairing tu-mor growth and inducing tumor stasis in miceharboring a KRAS-mutant lung cancer patient-derived xenograft (PDX), when treated at themaximally tolerated dose for each agent (Fig.1B) (9, 10). Similar results were also observed inother KRAS-mutant PDX models treated at lowerdoses (Fig. 1C and fig. S2, A and B), confirmingthat the combination produces biological effectsthat neither drug can achieve alone.These human xenograft studies require the

use of immunodeficient NOD-scid IL2Rgnull

(NSG) mice. To assess whether and to what extenttumor cell responses are altered by the immunesystem, we made use of an established syngeneictransplant mouse model of lung cancer. Mousetumor cells derived from a KrasG12D/+;Trp53−/−

(KP) lung tumor (11) were engineered to expressluciferase and green fluorescent protein (GFP)and tested for drug sensitivity in vitro or afterintravenous injection into C57BL/6 immuno-competent mice (Fig. 1D), where they producedaggressive lung adenocarcinomas by 1-week posttransplantation. As in the human models, thetrametinib plus palbociclib combination syner-gistically suppressed the growth of cultured KPtumor cells (fig. S2C) and significantly increasedthe survival of lung tumor–bearing animals (Fig.1E). This effect was substantially impaired whenthe same cells were transplanted into immuno-deficient NSG mice (fig. S2D and E), suggest-ing that the immune system might contributeto treatment efficacy.Immune profiling of lung tumors after 1 week

of combination treatment revealed a generalinflux of CD45+ immune cells compared withsingle-agent treatment (fig. S3A). Whereas therewas no change in B cell or macrophage num-bers in tumors after treatment, there was anincreased infiltration of CD4+ and CD8+ T cellsin the lungs and spleens of both tumor-bearingand naïve (tumor-free) mice after either single-agent or combination therapy. However, changesin CD4+ or CD8+ T cell activation were not ob-served, as assessed by staining for CD69, KLRG1,and CD107a, a marker of lymphocyte degranula-tion (figs. S3, B to D; S4, A and B; and S5, A toD). Thus, the drug combination does not appearto produce selective activation of T cells in thislung cancer model.By contrast, combined treatment with trame-

tinib and palbociclib triggered the selective ac-cumulation of natural killer (NK) cells and adecrease in Gr-1hiCD11b+ myeloid-derived sup-pressor cells (MDSCs) in the lungs of tumor-bearing mice compared with treatment witheither agent alone (Fig. 1, F and G; and figs. S3,E and F; S4, C and D; and S5F). NK cells de-rived from combination-treated tumors appearedactivated and more mature, as indicated by cellsurface expression of CD107a and other activat-ing receptors and altered transcriptional profilesshowing a reduction in proliferation-associatedgenes and an increase in expression of geneslinked to NK cell maturation and cytotoxicity(Fig. 1G and figs. S4C, S5E, and S6). Combinedtreatment also led to NK cell accumulation(but not activation) and a decrease in MDSCsin the spleens of tumor-bearing mice but notin the lungs or spleens of naïve mice (figs. S3, Fto H, and S5, E to G), suggesting that this im-mune effect is tumor dependent.To determine whether specific immune cells

contribute to the outcome of combination ther-apy, we assessed the impact of perturbingimmune cell function on the survival of tumor-bearing (immunocompetent) mice after vehicleor trametinib and palbociclib therapy. Neitherdepletion of macrophages, Gr-1+ granulocytes/MDSCs, and CD4+ and CD8+ T cells using block-ing antibodies nor anti–PD-1 immunotherapy(to stimulate exhausted T cells) had any im-pact on the survival of vehicle or combination-treated mice (figs. S7, A to D, and S8). By contrast,

RESEARCH

Ruscetti et al., Science 362, 1416–1422 (2018) 21 December 2018 1 of 7

1Department of Cancer Biology and Genetics, Sloan KetteringInstitute, Memorial Sloan Kettering Cancer Center, New York,NY 10065, USA. 2Department of Molecular Pharmacology,Sloan Kettering Institute, Memorial Sloan Kettering CancerCenter, New York, NY 10065, USA. 3Center for CellEngineering and Immunology Program, Memorial SloanKettering Cancer Center, New York, NY 10065, USA.4Department of Medicine, Memorial Sloan Kettering CancerCenter, New York, NY 10065, USA. 5Department of TumorCell Biology, St. Jude Children’s Research Hospital, Memphis,TN 38105, USA. 6Howard Hughes Medical Institute,Chevy Chase, MD 20815, USA.*These authors contributed equally to this work.†Corresponding Author. Email: [email protected]

on June 13, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 2: CANCER NK cell mediated cytotoxicity contributes to tumor ... · CANCER NK cell–mediated cytotoxicity contributes to tumor control bya cytostatic drug combination Marcus Ruscetti

Ruscetti et al., Science 362, 1416–1422 (2018) 21 December 2018 2 of 7

G

E

D

A

B

Pal

boci

clib

(µM

)

0.1

0.3

0.5

1

A549, trametinib (nM)

1 5 10 25 100

Rib

ocic

lib (

µM)

A549, trametinib (nM)

0.1

0.3

0.5

1

1 5 10 25 100

Abe

mac

iclib

(µM

)

A549, trametinib (nM)

0.1

0.3

0.5

1

1 5 10 25 100

F

H

0

50

100

0

0

0

P <0.0001

Vehicle

Trametinib

Palbociclib

Combo

Sur

viva

l (%

)

10 15 20 25 30 Time (d)

5.325.143.99

Vehicle Trametinib Palbociclib Combo

3.98 5.14 5.32

NK1.1-BV605

10 10 0 10 10 2

CD

3-B

V65

0

0

10

10

10

10

2

9.44

3

3 4 5

5

4

**** ****

**

**** ****

****

**

* *

C

D10

7a+

+ cel

ls)

(%)

5

10

15

20

25

(per

10

3 GF

P+ tu

mor

cel

ls)

(%)

100

200

300

400

2

4

6

8

10

N

K1.

1+ (

of C

D45

+ cel

ls)

(%)

0 0 0

N

K1.

1+

(of

NK

1.1

Vehicle Tramet Palbo ComboVehicle Tramet Palbo Combo Vehicle Tramet Palbo ComboTime (d)

10 20 30 0

50

100

Sur

viva

l (%

)

Isotype

Combo + NK1.1

Combo + Isotype

NK1.1

D

C57BL/6 Mouse KP (KrasG12D/+;p53-/-)

Trametinib Palbociclib

MSCV-Luc-GFP

P = 0.0001

9.44

Time (d)

Vehicle

Trametinib (lo)

Combo (lo)

0 10 20 30 40 50 0

500

1000

1500 Trametinib (hi)

Combo (hi) n.s.

*

**** ****

Palbociclib ****

C

Vol

ume

(m

m3)

Time (d)

Vol

ume

(m

m3

0

500

1000

1500

2000

2500

0 7 14 21 28 35 42 49

)

****

Vehicle

Trametinib

Combo

Palbociclib

*

***

Fig. 1. NK cell immunity is required for the efficacy of combinationMEK and CDK4/6 inhibitor therapy. (A) Clonogenic assay ofA549 lung cancer cells treated with MEK (trametinib) and/or variousCDK4/6 inhibitors (palbociclib, ribociclib, abemaciclib); representativeof three biological replicates. (B) Tumor volumes of mice bearingKRAS-mutant MSK-LX27 PDX lung tumors treated with vehicle, trametinib(3 mg/kg body weight), palbociclib (150 mg/kg body weight), or bothin combination (Combo) for indicated times (n = 5 mice per group).(C) Tumor volumes of mice bearing KRAS-mutant MSK-LX68 PDX lungtumors treated with vehicle, trametinib [1 mg/kg (lo) or 3 mg/kg (hi)body weight], palbociclib (150 mg/kg body weight), or both incombination for indicated times (n = 8 mice per group). n.s., notsignificant. (D) Syngeneic KP transplant lung cancer model. (E) Kaplan-Meier survival curve of KP transplant mice treated with vehicle,

trametinib (1 mg/kg body weight), palbociclib (100 mg/kg bodyweight), or both in combination (n ≥ 8 per group) (log-rank test).(F) Representative flow cytometry plots of NK cell populationsin lung tumors from KP transplant mice treated for 1 week as in (E).(G) Percentage of NK cells within the CD45+ population (left), total NKcells relative to tumor cell number (middle), and percentage of CD107a+

degranulating NK cells (right) (n ≥ 4 mice per group). Palbo, palbociclib;Tramet, trametinib. (H) Kaplan-Meier survival curve of KP transplantmice treated with vehicle or combined trametinib (1 mg/kg bodyweight) and palbociclib (100 mg/kg body weight) and either an isotypecontrol antibody (C1.18.4) or NK1.1-depleting antibody (PK136) (n ≥ 8per group) (log-rank test). (B and C) Two-way ANOVA. (G) One-wayANOVA. Error bars, mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001,****P < 0.0001.

RESEARCH | REPORTon June 13, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 3: CANCER NK cell mediated cytotoxicity contributes to tumor ... · CANCER NK cell–mediated cytotoxicity contributes to tumor control bya cytostatic drug combination Marcus Ruscetti

depletion of NK cells using an anti-NK1.1 anti-body (PK136) significantly reduced the survivaladvantage produced by combination therapywhile having no effect on cohorts treated withvehicle or single agents (Fig. 1H and figs. S7Eand F, and S8). This effect was independent ofthe MDSC reduction after treatment (fig. S3F),as Gr-1 depletion did not influence the sur-vival of combination-treated mice in the pres-ence or absence of NK cells (fig. S7B). Hence,in this model, the trametinib and palbociclibcombination triggers a potent and selective NKcell–mediated response that contributes to treat-ment efficacy.We found the contribution of immune sur-

veillance to the action of an apparently cyto-static drug combination intriguing and set outto study the underlying mechanisms in moredetail. CDK4 and 6–mediated phosphorylationof RB (the intended target of trametinib and pal-bociclib treatment) cancels its growth-suppressiveaction to facilitate E2F-mediated G1-S progres-sion (5). We confirmed in all our models thatcombination therapy produced a more potentreduction in RB phosphorylation and prolifer-ation without inducing apoptosis (fig. S9). Besidesits role in modulating cell cycle progression, RBplays a crucial role in mediating cellular se-nescence, a tumor-suppressive program that in-volves a stable (if not permanent) cell cycle arrestprogram coupled to an immune modulatorycomponent (12–14). Specifically, senescent cellsdisplay an RB-dependent down-regulation ofproliferation genes (15, 16) and a concomitantup-regulation of genes encoding a wide rangeof secretory proteins and other factors that mod-ulate the microenvironment (17). Of note, thissecretory program, often referred to as thesenescence-associated secretory phenotype (SASP),can have either tumor-suppressive or tumor-promoting functions, depending on context (18–23).In its tumor-suppressive role, RB often collab-orates with the p53 tumor suppressor to limitthe proliferation of premalignant cells (24),and disruption of both genes is linked to se-nescence escape during tumorigenesis (25).To test whether trametinib and/or palboci-

clib treatment induced senescence in humanor mouse KRAS-mutant lung cancer cells, weevaluated a series of markers and functional prop-erties associated with the senescent state (14, 26).Only combination treatment resulted in signif-icant senescence-associated beta-galactosidase(SA-b-gal) activity, the accumulation of senescence-associated heterochromatin foci, loss of the nu-clear envelope protein Lamin B1, and increasedexpression of DEC1, DCR2, and CDKN2B (Fig. 2Aand fig. S10).Colony-forming assays revealed that many

KRAS-mutant cell lines treated with eitherpalbociclib or trametinib alone resumed prolifera-tion upon drug washout, whereas combination-treated cells did not (Fig. 2B and fig. S11, A to B).This durable, senescent-like arrest was RB de-pendent and did not occur in tumor cells harbor-ing RB genomic loss (Fig. 2B and figs. S11 andS12). Both human and murine KRAS-mutant

tumor cells lacking p53 also displayed senes-cence markers in response to the trametiniband palbociclib combination (Fig. 2B and figs.S11 and S12). Therefore, this treatment can re-store senescence to tumor cells that have es-caped p53 tumor suppressive programs duringtumor evolution provided that RB function isretained.The senescence program provoked by com-

bined trametinib and palbociclib treatment wasalso associated with a potent SASP induction.RNA sequencing (RNA-seq) analysis of humanKRAS-mutant tumor cell lines after drug treat-ment revealed that the drug combination pro-duced a greater reduction in proliferation genesand increase in SASP factor expression com-pared with either treatment alone (Fig. 2C, fig.S13B, and tables S1 and S2). Among the immunemodulatory genes that were induced and se-creted were chemokines involved in NK cell re-cruitment (CCL2, CCL4, CCL5, CXCL10, CX3CL1),as well as cytokines that promote NK cell pro-liferation and activation [interleukin-15 (IL-15),IL-18, tumor necrosis factor–a (TNF-a)] (Fig. 2Dand fig. S13, A and C). Many of these same SASPfactors were also induced in murine KP lungcancer cells and a PDX lung cancer model in vivo(fig. S13, D and E). Gene Set Enrichment Analysisrevealed that signatures linked to oncogene-induced and replicative senescence (22, 27), nu-clear factor kB (NF-kB) and TNF-a signaling,and NK cell–mediated cytotoxicity were alsoselectively enriched in the transcriptional pro-files of combination treated cells (fig. S13, F toH). Although not secretory per se, NK cell ligands(which are required for activation of NK cellcytotoxicity and tumor cell targeting) are part ofthe transcriptional module linked to the SASP(28, 29). We found that intercellular adhesionmolecule–1 (ICAM-1) and the NKG2D ligandsULBP2 and MICA were induced after combina-tion treatment in human KRAS-mutant tumorcells, PDXs, and murine KP lung tumor cells (Fig.2E and fig. S14). Overall, these data suggest that,in addition to a more stable cell cycle arrest con-ferred by RB-mediated senescence, combinedMEK and CDK4/6 inhibition may promote tu-mor cell immune surveillance through inductionof the SASP program.To explore this possibility, we tested whether

NK cells could functionally target tumor cellsafter combined trametinib and palbociclib treat-ment. Using an in vitro NK cell coculture assaythat quantitatively measures both NK cell–tumorcell interactions and cytotoxicity, we observedthat treatment with the drug combination, butnot with single agents, triggered the rapid asso-ciation and eventual killing of KRAS-mutanttumor cells by the human YT NK cell line evenin the presence of the drugs (Fig. 2F; fig. S15,A and B; and movies S1 to S4). These resultswere corroborated using freshly isolated primaryhuman NK cells (fig. S15, C and D) and using themurine system, in which senescent KP cellstreated with the drug combination were suf-ficient to induce degranulation and cytotoxicityin drug-treated splenic NK cells upon coculture

(fig. S15, E and F). In vivo, NK cells also ap-peared to preferentially target senescent cells,as tumors in NK cell–depleted mice that re-ceived the trametinib and palbociclib combi-nation were significantly larger and retainedmore SA-b-gal+ cells compared with those incontrol mice with NK cells intact (Fig. 2, G andH, and fig. S15G). NK cells are therefore capableof eliminating senescent tumor cells after com-bined trametinib and palbociclib treatment.We performed a genetic experiment to dis-

able the SASP program in KP tumor cells. Thetranscription factor NF-kB is a master regulatorof the SASP program but plays only a limitedrole in senescence-induced cell cycle arrest (30–32).KP tumor cells expressing a well-characterizedshort hairpin RNA (shRNA) targeting the p65subunit of NF-kB underwent growth arrest inresponse to the drug combination but displayeda reduction in many SASP factors and were nottargeted by spleen-derived murine NK cells in vitro(Fig. 3, A and B, and fig. S16, A to D). In vivo,tumors derived from p65-suppressed KP cellsshowed similar levels of NK cell accumulationas tumors expressing a control shRNA targetingthe nonexpressed gene Renilla luciferase aftercombination therapy; however, these infiltratingNK cells were not activated, and the treatmentwas not as effective (Fig. 3C and fig. 16E). Thus,the SASP appears necessary for therapy-inducedNK cell surveillance and the efficacy of the drugcombination in vivo.To pinpoint SASP factors needed for NK cell

attack, we tested a range of neutralizing anti-bodies against NF-kB-regulated SASP factors(IL-15, IL-18, TNF-a, CCL2, CCL5) for theirability to alter survival in KP transplant micetreated with the trametinib and palbociclib com-bination. Only TNF-a depletion reduced animalsurvival, which was similar in magnitude tothat produced by NK cell depletion (Fig. 3D andfig. S16, F and G) and was associated with areduction in activated NK cells present in thetreated tumors (Fig. 3E). This effect was inpart due to tumor cell–derived TNF-a, as shRNAscapable of suppressing Tnfa in combination-treated KP cells markedly inhibited NK cellcytotoxicity in vitro (Fig. 3F and fig. S17, A to D).Still, Tnfa suppression (fig. S17E) was not aseffective as p65 depletion (fig. S16E) at impairingthe survival of tumor-bearing mice after combi-nation treatment, indicating that an interplaybetween multiple SASP factors is required forNK cell surveillance in vivo.We also explored potential mechanisms by

which NK cells target tumor cells undergoingtherapy-induced senescence. Cell surface NK cellligands and adhesion molecules are up-regulatedalong with the SASP during many forms ofsenescence (29). We found that both ICAM-1 andmembers of the MICA/B, ULBP, Rae-1, and H60family of NKG2D ligands were increased aftercombination therapy in an NF-kB-dependent orindependent manner, respectively (Figs. 2E and3A and figs. S14 and 16D). Furthermore, blockingICAM-1 and its receptor LFA-1 (and to a lesserextent NKG2D) blunted NK cell cytotoxicity in

Ruscetti et al., Science 362, 1416–1422 (2018) 21 December 2018 3 of 7

RESEARCH | REPORTon June 13, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 4: CANCER NK cell mediated cytotoxicity contributes to tumor ... · CANCER NK cell–mediated cytotoxicity contributes to tumor control bya cytostatic drug combination Marcus Ruscetti

Ruscetti et al., Science 362, 1416–1422 (2018) 21 December 2018 4 of 7

CA

B

Vehicle Trametinib

Palbociclib

Combo

*** **

** *

** **

Cha

nge

in tu

mor

cel

ls (

%)

- + - -

- + +

+ Palbociclib Trametinib - +

- - - + +

+

A549 H460

row

max

ro

w m

in

IL-15 IL-18

IFN- 2 IFN

CCL5

CCL2

CCL3

VEGF

PDGF

G-CSF

GM-CSF

IL-7

IL-4

IL-2

IL-5

sCD40L

TNF-

IL-12p70

TGF-

FGF2 EGF

FLT-3L

CCL22

CCL11

CX3CL1

IL-8

IL-1

IL-10

IL-6 IL-12p40

CCL7

CCL4

CXCL10

PDGF

CXCL1

IL-17AIL-1RA

IL-1

IL-3

TNF-

IL-9

IL-13

H2030

HCC15

A549 +

+

-

RB p53

+

-

-

D

SA

SP

Cell C

ycle

E

G *

SA-

-gal

(%)

0

5

10

15

20

25

Isotype NK1.1

Trametinib

*** **

** **

Vehicle

Palbo

Combo + Isotyp

e

Combo + NK1.1

50

0

-50

-100

-150

-200 Cha

nge

in tu

mor

are

a (%

)

HICAM-1-FITC

Vehicle Tramet Palbo Combo

Isotype NK1.1

H2030 A549-75

-50

-25

0

25

rametinib

oa

row

min

IL-15

IL-12p40

IFN

IL-6

CCL5

CCL2

CCL4

PDGFFGF2

GM-CSF

IL-13

EGF

CXCL10

CCL11

IL-4

IL-9

IL-2

IL-5

TGF-

CCL3

VEGF

G-CSF FLT-3L

CXCL1

IL-7

IL-1

CCL22

CX3CL1

CCL7

PDGF

IL-1RA

TNF-

IL-17A

IL-8

IFN- 2

IL-10

sCD40L

IL-1

IL-3

IL-12p70

IL-18

TNF-

M

FI

Vehicle Trametinib Palbociclib Combination

0

10

20

30

40

50

60

70

80

A549

H2030

H460

SA-

-gal

+ (%

)

F

ICAM-1 ULBP2/5/6 MICA0

500

1000

1500

2000

5000

10000

15000

20000 VehicleTrametinibPalbociclibCombo

*********

********

****

*****n.s.

0 102 103 104 105

CPTVCPTV

deniatsnuα

1-M

ACI

S100A11TNFAIP2CYP1B1MAP1LC3BSAT1OPTNTNFAIP3CDKN1AEPS8SQSTM1NPC1CCND1RNF149CCNE1RAB27BORC6WEE1IQGAP3NEURL1BID1PIRCOMMD4IMPA2RAD51AP1NUCKS1RHNO1H2AFVNUSAP1DUTFAM111BCENPMRNASEH2APOLE2HMGB3DEPDC1BNUF2FAM83DCCNB1AURKAPLK1SGOL2UBE2CCDC20CHEK1CTDSP1ASF1BFBXO5MCM6HELLSBUB1BTK1CDK2DEKUSP1CDK1CDCA3CCNB2NCAPD2KIF2CFOXM1BIRC5AURKBH2AFXCENPACCNA2NASPKIF11TOP2ATMPOHMMRMKI67PRC1LMNB1SMC4DLGAP5TYMSHMGB2SPC24CDKN3KIF20ASPC25STMN1MYBL2H1FXHMGN2EZH2CDK4MCM3EXO1DNMT1HMGA1MTND2L1CAMSTXBP1GM2ACSTBRHOBKRT8MUC1DDR1CLUGAS6IL6NFKB2CX3CL1CCL2NFE2L2SERPINE1CXCL11ACVRL1PVRCD82PLAUIL15SGMS2ATP2C1CD40BST2ITGA5HIF1AATP2A2CSF1IGFBP7KLF6IFNAR1PSEN1TAPBPIRF1IL1R1NAMPTCXCL10IL15RAIFNGR1TNFRSF9HLACHLABHLAATAP1ATF3IFIT2GADD45ARCAN1FOSL2BIRC2NINJ1PNRC1KDM6BDNAJB4STAT1ISG15XAF1MX1IFIT3B2MSTAT3

VTPC

2

1

0

1

2

H460 A549H2030

Fig. 2. Senescence and SASP induction after combination MEK andCDK4/6 inhibitor therapy induces NK cell immune surveillance.(A) Quantification of SA-b-gal+ cells in human KRAS-mutant lung tumorcell lines after 8-day treatment with trametinib (25 nM) and/orpalbociclib (500 nM). Mean of three biological replicates is plotted.(B) Clonogenic assay of human KRAS-mutant lung cancer cells replatedin the absence of drugs after 8-day pretreatment as in (A); representa-tive of three biological replicates. RB and p53 genomic status is indicatedon right. (C) Heat map of senescence-associated cell cycle and SASPgene expression in human KRAS-mutant lung cancer cell lines aftertreatment as in (A), as assessed by RNA-seq. Two biological replicatesper cell line are shown. C, combination; P, palbociclib; T, trametinib;V, vehicle. (D) Heat map of cytokine array results from humanKRAS-mutant lung tumor cells treated with trametinib (25 nM) and/orpalbociclib (500 nM) for 8 days. Data are presented as mean of threebiological replicates. (E) Flow cytometry analysis of ICAM-1 expressionand levels of other NK cell–activating ligands after 8-day treatment of A549

lung tumor cells as described in (A). Quantification of mean fluorescenceintensity (MFI) from three biological replicates is shown on the right.(F) Quantification of NK cell cytotoxicity (by live cell imaging) afterpretreatment of A549 and H2030 lung tumor cell lines with indicateddrugs for 8 days and coculturing with the YT NK cell line at a 10:1 effectorto target cell (E:T) ratio for 20 hours in the presence or absence ofindicated drugs. Change in tumor cells is normalized to controlwells lacking NK cells. Mean of three biological replicates is plotted.(G) Representative SA-b-gal staining of KP transplant lung tumors after1-week treatment with combined trametinib (1 mg/kg body weight)and palbociclib (100 mg/kg body weight) and either an isotype controlantibody (C1.18.4) or NK1.1 depleting antibody (PK136) (scale bar,50 mm). Quantification shown on right (n = 3 mice per group). (H) Effectof 1-week treatment as in (G) on lung tumor burden (relative to vehicle)(n = 5 mice per group). (E, F, and H) One-way ANOVA. (G) Unpairedtwo-tailed t test. Error bars, mean ± SEM. *P < 0.05, **P < 0.01, ***P <0.001, ****P < 0.0001.

RESEARCH | REPORTon June 13, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 5: CANCER NK cell mediated cytotoxicity contributes to tumor ... · CANCER NK cell–mediated cytotoxicity contributes to tumor control bya cytostatic drug combination Marcus Ruscetti

Ruscetti et al., Science 362, 1416–1422 (2018) 21 December 2018 5 of 7

D

B

C

E

Vehicle (Ren) Combo (Ren) Vehicle (p65) Combo (p65)

Vehicle le l Combo Vehicle Combo

p65Ren

***

n.s.

Cha

nge

in tu

mor

cel

ls (

%)

0

-10

-20

-30

-40

-50

10

e e

0

0

0 n.s. ***

NK

1.1

(%

)+

0

5

10

15

0

5

10

15

20

eeVehicle e Combo eeo Vehicle le Combo

p65Ren

Vehicle e Combo o Vehicle le Combo

p65 Ren

Sur

viva

l (%

)

0

50

100

10 15 20 30 25 Time (d)

Isotype

Combo + Isotype

Combo + NK1.1

Combo + TNF-P = 0.0057

P = 0.0173

R

elat

ive

Exp

ress

ion

(AU

)

**

0 1

2 3 4 5

0 5

10 15 20 25

0

1

2

3

4

***

0

1

2

3

4 **

0

5

10

15

20

0 5

10 15 20 25 *** ****

0

2

4

6

0

1

2

3 *** **** **

p65 Combo

Ren Vehicle

Vehicle

Combo

A

C

D10

7a+

+ cel

ls)

(%)

(of

NK

1.1

F

Cha

nge

in tu

mor

cel

ls (

%)

***

Vehicle Combo Combo

TnfaScramble

n.s.

-30

-20

-10

0

10

o CVehicle Vehicle e Combo o Vehicle le Combo

Isotype

5

0

10

15

NK

1.1+

(%

)

TNF-

n.s.

Vehicle e Combo o Vehicle le Combo

Isotype TNF-

0

5

10

15

25

C

D10

7a+

+ cel

ls)

(%)

(of

NK

1.1

20

**

p65 Tnfa Il15 Ccl2

Il6 Ccl5 Icam1 Cxcl1

α α

α

Fig. 3. NF-kB-mediated SASP and TNF-a secretion is required fortreatment-induced NK cell activity. (A) Quantitative reverse tran-scription polymerase chain reaction analysis of SASP gene expression inKP tumor cells transduced with indicated shRNAs after treatmentwith vehicle or trametinib (25 nM) and palbociclib (500 nM) for 8 days.Mean of two biological replicates associated with three technical replicatesis plotted. AU, arbitrary units. (B) Representative images of KP tumorcells with indicated shRNAs (green) pretreated as in (A) and coculturedwith primary murine splenic NK cells (red) at a 20:1 E:T ratio for 20 hoursin the presence or absence of indicated drugs (scale bar, 25 mm).Quantification of NK cell cytotoxicity (by live cell imaging) is shown onthe right. Change in tumor cells is normalized to control wells lackingNK cells. Mean of three biological replicates is plotted. (C) Flowcytometry analysis of total and CD107a+ degranulating NK cells withinthe CD45+ population in the lungs after 1-week treatment of micetransplanted with KP tumor cells containing control Renilla (Ren) or p65

shRNAs with vehicle or combined trametinib (1 mg/kg body weight)and palbociclib (100 mg/kg body weight) (n ≥ 3 mice per group).(D) Kaplan-Meier survival curve of KP transplant mice treated withvehicle or combined trametinib (1 mg/kg body weight) and palbociclib(100 mg/kg body weight) and either an isotype control (HRPN), NK1.1(PK136), or TNF-a (XT3.11) targeting antibody (n ≥ 6 per group) (log-ranktest). (E) Flow cytometry analysis as in (C) after 1-week treatment ofKP transplant mice with vehicle or combined trametinib (1 mg/kg bodyweight) and palbociclib (100 mg/kg body weight) and either an isotypecontrol (HRPN) or TNF-a (XT3.11) targeting antibody (n = 5 per group).(F) Quantification of NK cell cytotoxicity (by live cell imaging) towardKP tumor cells with indicated shRNAs pretreated as in (B) and coculturedwith primary murine splenic NK cells at a 20:1 E:T ratio for 20 hoursin the presence or absence of indicated drugs. (A to C and E and F)One-way ANOVA. Error bars, mean ± SEM. **P < 0.01, ***P < 0.001,****P < 0.0001.

RESEARCH | REPORTon June 13, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 6: CANCER NK cell mediated cytotoxicity contributes to tumor ... · CANCER NK cell–mediated cytotoxicity contributes to tumor control bya cytostatic drug combination Marcus Ruscetti

combination-treated tumor cells in live cell imagingand chromium release assays and significantlyreduced the survival of combination-treated KPtransplant mice in vivo (figs. S15, C and D, andS17, F to H). Therefore, the SASP transcriptionalmodule contributes both secreted (i.e., TNF-a)and cell surface (i.e., ICAM-1) factors to facilitateNK cell surveillance.The orthotopic KP model represents a flex-

ible syngeneic experimental system but is extra-ordinarily aggressive with tumors disseminatedthroughout the lungs, making assessment of tu-mor response to therapy challenging. To examinethe impact of this senescence-inducing therapyon NK cell immune surveillance and tumor pro-gression in a more physiologically relevant set-ting, we utilized an autochthonous model ofKRAS-mutant lung cancer. Genetically engineered

KrasLSL-G12D/wt;Trp53flox/flox (KP GEMM) micewere infected intratracheally with an adenovirusexpressing Cre recombinase to induce endoge-nous lung tumor formation (33), leading to thedevelopment of focal KRAS-driven tumors thatcould be monitored using microcomputed tomo-graphy (mCT). Consistent with our findings in thesyngeneic transplant model, combined trame-tinib and palbociclib treatment after endogenoustumor formation led to a significant reduction inRB phosphorylation and tumor cell proliferation,accumulation of SA-b-gal+ senescent tumor cells,induction of SASP factors including TNF-a andICAM-1, and subsequent recruitment of NK cellswithin lung adenocarcinomas of KP GEMMmice(Fig. 4A and fig. S18, A to D).The contribution of NK cells to the activity

of the combination therapy was marked. Two

weeks post treatment, we observed tumor re-gressions in mice treated with the combinationtherapy and an isotype control antibody but notin those mice receiving an NK1.1-depleting anti-body or single-agent trametinib or palbociclibwhose tumors continued to progress (Fig. 4, Band C). Moreover, the overall survival of micetreated with trametinib plus palbociclib, whichincluded a notable number of long-term survi-vors, was significantly longer than mice receiv-ing single-agent treatment or the combinationtreatment in the context of NK cell depletion(Fig. 4D). Antibody-mediated blockade of SASPfactors TNF-a or ICAM-1 also prevented tumorregressions and blunted the survival benefit ofthe drug combination (Fig. 4, C and D). There-fore, senescence-inducing targeted therapiescan lead to tumor control in KRAS-mutant lung

Ruscetti et al., Science 362, 1416–1422 (2018) 21 December 2018 6 of 7

C

Vehicle Combo Vehicle Combo

Kras G12D; Trp53 -/- GEMM mice

-50

0

50

100

150

Tum

or v

olum

e (%

cha

nge)

Pre-treatment 2 weeks

D -/-Kras G12D; Trp53 GEMM mice

Sur

viva

l (%

)

0 50 100 150

Time (d)

50

100

250 200

Combo Isotype

Combo NK1.1

Vehicle

Palbociclib

Trametinib

0

Combo TNF-αCombo ICAM1

****

**** ***

**

**** Vehicle

Trametinib

Palbociclib

Combo Isotype

Combo TNF-αCombo NK1.1

Combo ICAM1

********** ***

**

pERK

CC3

NKp46

pRB

Ki67

SA-β -gal

A B

V

Combo + NK1.1

ehicle

Combo + Isotype

Fig. 4. Combination trametinib and palbociclib treatment drives NKcell–mediated lung tumor regressions in genetically engineeredmice. (A) Immunohistochemical staining of KP GEMM tumors treated withvehicle or combined trametinib (1 mg/kg body weight) and palbociclib(100 mg/kg body weight) for 2 weeks (scale bar, 50 mm). CC3, cleavedcaspase-3; pERK, phosphorylated extracellular signal–regulated kinase.(B) Representative mCT images of KP GEMM lung tumors prior totreatment and after 2 weeks of treatment with vehicle or combinedtrametinib (1 mg/kg body weight) and palbociclib (100 mg/kg body

weight) and either an isotype control antibody (C1.18.4) or NK1.1 depletingantibody (PK136). Yellow boxes indicate lung tumors. (C) A waterfallrepresentation of the response of each tumor after 2 weeks of treatmentwith vehicle, trametinib (1 mg/kg body weight), palbociclib (100 mg/kgbody weight), or both, and either an isotype control (C1.18.4), NK1.1(PK136), TNF-a (XT3.11), or ICAM-1 (YN1/1.7.4) blocking antibody (n ≥ 6per group). (D) Kaplan-Meier survival curve of KP GEMM mice treated asin (C) (n ≥ 6 per group) (log-rank test). (C) One-way ANOVA. *P < 0.05,**P < 0.01, ***P < 0.001, ****P < 0.0001.

RESEARCH | REPORTon June 13, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 7: CANCER NK cell mediated cytotoxicity contributes to tumor ... · CANCER NK cell–mediated cytotoxicity contributes to tumor control bya cytostatic drug combination Marcus Ruscetti

tumors, with SASP-provoked andNK cell–mediatedimmune surveillance facilitating tumor regres-sion and prolonged survival.Leveraging immunocompetent mouse models

of KRAS-mutant lung cancer, we explored thenon–cell autonomous effects of combined MEKand CDK4/6 inhibitor therapy, revealing an im-mune modulatory component to the antitu-mor response. Complementary to previous workin other systems demonstrating activation ofT cell responses using similar monotherapies(6, 7), our studies highlight a distinct mech-anism of innate immune attack by NK cells,which act as a natural senolytic to eliminatetumor cells in combination drug–treated miceharboring autochthonous tumors. The ability ofNK cells to target drug-treated tumor cells re-quires the induction of RB-mediated cellularsenescence and acquisition of an NF-kB–dependentSASP program that culminates in the secretionof proinflammatory cytokines (e.g., TNF-a) andsurface expression of NK cell–activating mol-ecules (e.g., ICAM-1) (fig. S18E). Thus, whereasdeveloping tumors evade both senescence andimmune surveillance to become fully malignant,these processes can be reestablished in tumorcells by certain targeted cancer therapies. Al-though chronic SASP induction can have delete-rious consequences in some settings (17, 34), thetimely clearance of senescent cells by NK cells inthis model establishes senescence induction asa beneficial outcome of targeted therapy, andexplains how some cytostatic agents can be cyto-toxic in vivo. Because NK cells do not requireneoantigen recognition to target tumor cells (28),strategies to exploit and enhance this form ofimmune surveillance may complement exist-ing efforts to harness adaptive immune surveil-lance by means of T cell checkpoint blockadeimmunotherapies.

REFERENCES AND NOTES

1. A. A. Samatar, P. I. Poulikakos, Nat. Rev. Drug Discov. 13,928–942 (2014).

2. P. Lito et al., Cancer Cell 25, 697–710 (2014).

3. E. Manchado et al., Nature 534, 647–651 (2016).4. A. Prahallad et al., Nature 483, 100–103 (2012).5. C. J. Sherr, D. Beach, G. I. Shapiro, Cancer Discov. 6, 353–367

(2016).6. J. Deng et al., Cancer Discov. 8, 216–233 (2018).7. S. Goel et al., Nature 548, 471–475 (2017).8. P. J. R. Ebert et al., Immunity 44, 609–621 (2016).9. D. W. Fry et al., Mol. Cancer Ther. 3, 1427–1438 (2004).10. A. G. Gilmartin et al., Clin. Cancer Res. 17, 989–1000

(2011).11. N. Dimitrova et al., Cancer Discov. 6, 188–201 (2016).12. J. W. Shay, O. M. Pereira-Smith, W. E. Wright, Exp. Cell Res.

196, 33–39 (1991).13. T. Kuilman, C. Michaloglou, W. J. Mooi, D. S. Peeper, Genes

Dev. 24, 2463–2479 (2010).14. N. E. Sharpless, C. J. Sherr, Nat. Rev. Cancer 15, 397–408

(2015).15. A. Chicas et al., Cancer Cell 17, 376–387 (2010).16. M. Narita et al., Cell 113, 703–716 (2003).17. J. P. Coppé et al., PLOS Biol. 6, e301 (2008).18. T. Eggert et al., Cancer Cell 30, 533–547 (2016).19. T. W. Kang et al., Nature 479, 547–551 (2011).20. V. Krizhanovsky et al., Cell 134, 657–667 (2008).21. W. Xue et al., Nature 445, 656–660 (2007).22. N. Tasdemir et al., Cancer Discov. 6, 612–629 (2016).23. J. P. Coppé, P. Y. Desprez, A. Krtolica, J. Campisi, Annu. Rev.

Pathol. 5, 99–118 (2010).24. M. Serrano, A. W. Lin, M. E. McCurrach, D. Beach, S. W. Lowe,

Cell 88, 593–602 (1997).25. S. W. Lowe, C. J. Sherr, Curr. Opin. Genet. Dev. 13, 77–83

(2003).26. M. Collado, M. Serrano, Nat. Rev. Cancer 6, 472–476

(2006).27. A. L. Fridman, M. A. Tainsky, Oncogene 27, 5975–5987

(2008).28. M. G. Morvan, L. L. Lanier, Nat. Rev. Cancer 16, 7–19

(2016).29. A. Sagiv et al., Aging (Albany NY) 8, 328–344 (2016).30. J. C. Acosta et al., Cell 133, 1006–1018 (2008).31. A. V. Orjalo, D. Bhaumik, B. K. Gengler, G. K. Scott,

J. Campisi, Proc. Natl. Acad. Sci. U.S.A. 106, 17031–17036(2009).

32. Y. Chien et al., Genes Dev. 25, 2125–2136 (2011).33. E. L. Jackson et al., Genes Dev. 15, 3243–3248 (2001).34. B. G. Childs, M. Durik, D. J. Baker, J. M. van Deursen, Nat. Med.

21, 1424–1435 (2015).

ACKNOWLEDGMENTS

We thank H. Varmus, T. Jacks, and J. P. Morris IV for sharingcell lines; A. Banito, X. Li, D. Alonso-Curbelo, V. Low, H. C. Chen,L. Zamechek, W. Luan, F. Luna, and M. Rowicki for technicalassistance and advice; N. Adams and J. Sun for graciouslyproviding murine NKG2D-Fc antibodies and helpful commentson the manuscript; J. Boudreau and K. Hsu for insightsconcerning in vitro NK cell assays; and other members of the

Lowe laboratory for insightful discussions. Funding: Wethank W. H. Goodwin and A. Goodwin and the CommonwealthFoundation for Cancer Research for research support. This workwas supported by three grants from the National CancerInstitute (PO1 CA129243-06, P30 CA008748-S5, and U54OD020355-01), a grant from the Center of ExperimentalTherapeutics of Memorial Sloan Kettering Cancer Center(MSKCC), and an MSKCC-Parker Institute for CancerImmunotherapy pilot grant. M.R. was supported by an MSKCCTranslational Research Oncology Training Fellowship (NIH T32-CA160001) and an American Cancer Society PostdoctoralFellowship (129040-PF-16-115-01-TBG). J.L. was supported by afellowship of the German Research Foundation (DFG). M.J.B.was supported by an American Association for ThoracicSurgery-Andrew Morrow Research Scholarship. T.B. wassupported by the William C. and Joyce C. O'Neil Charitable Trustand Memorial Sloan Kettering Single Cell Sequencing Initiative.F.J.S.-R. is an HHMI Hanna Gray Fellow and was partiallysupported by an MSKCC Translational Research OncologyTraining Fellowship (NIH T32-CA160001). P.B.R. was supportedin part by a K12 Paul Calebresi Career Development Award forClinical Oncology (K12 CA184746). E.d.S. received supportthrough a U54 OD020355-01 NIH grant and the Geoffrey BeeneCancer Research Center. C.M.R. was supported by a Stand UpTo Cancer grant from the American Association for CancerResearch. S.W.L. is the Geoffrey Beene Chair of Cancer Biology.S.W.L. and C.J.S. are Howard Hughes Medical Instituteinvestigators. Author contributions: M.R. and J.L. conceivedthe project, performed and analyzed experiments, and wrote thepaper with assistance from all authors. M.J.B., M.F., A.K., N.R.S.,C.-C.C., Y.-j.H., F.J.S.-R., J.F., T.B., S.T., H.A.C., and E.M.performed and analyzed experiments. P.B.R., J.T.P., C.M.R., andE.d.S. provided patient-derived tumor specimens. C.J.S. andS.W.L. conceived the project, supervised experiments, and wrotethe paper. Competing interests: P.B.R. is a shareholder ofPfizer and a consultant for EMD Serono and AstraZeneca andhas received an honorarium from Corning. E.M. is an employeeand share holder of Novartis. S.W.L. is a founder and scientificadvisory board member of Blueprint Medicines and ORICPharmaceuticals and received an award and honorarium fromEli Lilly and Company. Data and materials availability:RNA-seq data presented in this study are deposited in the GeneExpression Omnibus database under accession number GSE110397.

SUPPLEMENTARY MATERIALS

www.sciencemag.org/content/362/6421/1416/suppl/DC1Materials and MethodsFigs. S1 to S18Movies S1 to S4Tables S1 and S2References (35–41)

4 January 2018; resubmitted 10 June 2018Accepted 19 November 201810.1126/science.aas9090

Ruscetti et al., Science 362, 1416–1422 (2018) 21 December 2018 7 of 7

RESEARCH | REPORTon June 13, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 8: CANCER NK cell mediated cytotoxicity contributes to tumor ... · CANCER NK cell–mediated cytotoxicity contributes to tumor control bya cytostatic drug combination Marcus Ruscetti

mediated cytotoxicity contributes to tumor control by a cytostatic drug combination−NK cell

Poirier, Charles M. Rudin, Elisa de Stanchina, Eusebio Manchado, Charles J. Sherr and Scott W. LoweHo, Francisco J. Sanchez-Rivera, Judith Feucht, Timour Baslan, Sha Tian, Hsuan-An Chen, Paul B. Romesser, John T. Marcus Ruscetti, Josef Leibold, Matthew J. Bott, Myles Fennell, Amanda Kulick, Nelson R. Salgado, Chi-Chao Chen, Yu-jui

DOI: 10.1126/science.aas9090 (6421), 1416-1422.362Science 

, this issue p. 1416; see also p. 1355Sciencerecruitment and elimination of senescent lung cancer cells, which did not occur when either agent was used alone.

induced natural killer (NK) cell−−mitogen-activated protein kinase inhibitor and a cyclin-dependent kinase 4/6 inhibitor a−−KRAS-mutant lung tumors in mice (see the Perspective by Cornen and Vivier). The two small molecules

report that combining two clinically approved cancer drugs promoted immune surveillance and killing of et al.Ruscetti simultaneous strategies that mobilize the immune system and directly target malignant cells may be more effective.

Immunotherapy is a powerful treatment for certain cancers. Yet for those patients that do not respond,Small molecules spark NK cell response

ARTICLE TOOLS http://science.sciencemag.org/content/362/6421/1416

MATERIALSSUPPLEMENTARY http://science.sciencemag.org/content/suppl/2018/12/19/362.6421.1416.DC1

CONTENTRELATED

http://stm.sciencemag.org/content/scitransmed/7/283/283ra55.fullhttp://stm.sciencemag.org/content/scitransmed/8/357/357ra123.fullhttp://stm.sciencemag.org/content/scitransmed/6/243/243ra87.fullhttp://science.sciencemag.org/content/sci/362/6421/1355.full

REFERENCES

http://science.sciencemag.org/content/362/6421/1416#BIBLThis article cites 41 articles, 12 of which you can access for free

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Terms of ServiceUse of this article is subject to the

is a registered trademark of AAAS.ScienceScience, 1200 New York Avenue NW, Washington, DC 20005. The title (print ISSN 0036-8075; online ISSN 1095-9203) is published by the American Association for the Advancement ofScience

Science. No claim to original U.S. Government WorksCopyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of

on June 13, 2020

http://science.sciencemag.org/

Dow

nloaded from