molecularly targeted drug combinations …...2017/08/02  · molecularly targeted drug combinations...

10
Molecularly targeted drug combinations demonstrate selective effectiveness for myeloid- and lymphoid- derived hematologic malignancies Stephen E. Kurtz a , Christopher A. Eide a,b , Andy Kaempf c , Vishesh Khanna a,b , Samantha L. Savage a , Angela Rofelty a , Isabel English a , Hibery Ho a , Ravi Pandya d , William J. Bolosky d , Hoifung Poon d , Michael W. Deininger e , Robert Collins f , Ronan T. Swords g , Justin Watts g , Daniel A. Pollyea h , Bruno C. Medeiros i , Elie Traer a , Cristina E. Tognon a , Motomi Mori c,j , Brian J. Druker a,b,1 , and Jeffrey W. Tyner k,1 a Division of Hematology & Medical Oncology, Oregon Health & Science University, Portland, OR 97239; b Howard Hughes Medical Institute, Oregon Health & Science University, Portland, OR 97239; c Biostatistics Shared Resource, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239; d Microsoft Research, Redmond, WA 98052; e Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112; f UT Southwestern Medical Center, Dallas, TX 75390; g University of Miami School of Medicine, Miami, FL 33136; h Division of Hematology, University of Colorado School of Medicine, Aurora, CO 80045; i Stanford University School of Medicine, Stanford, CA 94305; j Oregon Health & Science UniversityPortland State University School of Public Health, Portland, OR 97239; and k Department of Cell, Development & Cancer Biology, Oregon Health & Science University, Portland, OR 97239 Contributed by Brian J. Druker, July 6, 2017 (sent for review March 9, 2017; reviewed by Keith Flaherty and A. Y. Leung) Translating the genetic and epigenetic heterogeneity underlying human cancers into therapeutic strategies is an ongoing challenge. Large-scale sequencing efforts have uncovered a spectrum of mutations in many hematologic malignancies, including acute myeloid leukemia (AML), suggesting that combinations of agents will be required to treat these diseases effectively. Combinatorial approaches will also be critical for combating the emergence of genetically heterogeneous subclones, rescue signals in the micro- environment, and tumor-intrinsic feedback pathways that all contribute to disease relapse. To identify novel and effective drug combinations, we performed ex vivo sensitivity profiling of 122 pri- mary patient samples from a variety of hematologic malignancies against a panel of 48 drug combinations. The combinations were designed as drug pairs that target nonoverlapping biological pathways and comprise drugs from different classes, preferably with Food and Drug Administration approval. A combination ratio (CR) was derived for each drug pair, and CRs were evaluated with respect to diagnostic categories as well as against genetic, cytoge- netic, and cellular phenotypes of specimens from the two largest disease categories: AML and chronic lymphocytic leukemia (CLL). Nearly all tested combinations involving a BCL2 inhibitor showed additional benefit in patients with myeloid malignancies, whereas select combinations involving PI3K, CSF1R, or bromodomain inhib- itors showed preferential benefit in lymphoid malignancies. Ex- panded analyses of patients with AML and CLL revealed specific patterns of ex vivo drug combination efficacy that were associated with select genetic, cytogenetic, and phenotypic disease subsets, warranting further evaluation. These findings highlight the heuristic value of an integrated functional genomic approach to the identi- fication of novel treatment strategies for hematologic malignancies. targeted therapies | drug combinations | ex vivo assay | hematologic malignancies T he promise of precision medicine is the ability to align medical interventions with individual patients at the time of diagnosis and to alter treatment regimens as new mutations arise and responses diminish. Although technical developments in next- generation sequencing and computational biology have accelerated progress in precision medicine, fundamental challenges remain. Whole-genome and whole-exome sequencing technologies can identify many target mutations, but these techniques are analyti- cally intensive and may not reliably detect translocations, zygosity changes, and low-allele burden mutations with clinical significance. A further hindrance to the clinical utility of mutation status is a lack of drug therapies that selectively target cancer-associated mutations, with effective Food and Drug Administration (FDA)-approved drugs existing for only a subset of the genes currently known to underlie tumorigenesis (1). These limitations invite a complementary strategy that assesses drug sensitivities obtained with targeted agents designed to inhibit discrete cellular processes as a means of identi- fying phenotypic indications for specific cancers (2). Associating phenotypic responses with particular genetic alterations may then beget precision-based therapies. For blood cancers, the success with targeted therapies de- veloped to inhibit BCR-ABL fusions that drive chronic myelog- enous leukemia has spurred efforts to identify similar therapies for other hematopoietic malignancy types. Acute myeloid leu- kemia (AML), which results from the enhanced proliferation and impaired differentiation of hematopoietic stem and progenitor cells, has substantial underlying heterogeneity, a feature that Significance Mononuclear cells obtained from freshly isolated patient sam- ples with various hematologic malignancies were evaluated for sensitivities to combinations of drugs that target specific cell- signaling pathways. The diagnostic, genetic/cytogenetic, and cellular features of the patient samples were correlated with effective drug combinations. For myeloid-derived tumors, such as acute myeloid leukemia, several combinations of targeted agents that include a kinase inhibitor and venetoclax, a selective inhibitor of BCL2, are effective. Author contributions: S.E.K., C.A.E., R.P., W.J.B., H.P., E.T., C.E.T., B.J.D., and J.W.T. de- signed research; S.E.K., C.A.E., A.K., S.L.S., A.R., I.E., and H.H. performed research; V.K., S.L.S., A.R., I.E., H.H., M.W.D., R.C., R.T.S., J.W., D.A.P., B.C.M., E.T., and B.J.D. contributed new reagents/analytic tools; S.E.K., C.A.E., A.K., M.M., B.J.D., and J.W.T. analyzed data; and S.E.K., C.A.E., A.K., D.A.P., B.C.M., C.E.T., M.M., and J.W.T. wrote the paper. Reviewers: K.F., Dana-Farber/Harvard Cancer Center; and A.Y.L., University of Hong Kong. Conflict of interest statement: D.A.P. serves on the advisory boards for Pharmacyclics and Gilead. J.W.T. receives research support from Agios, Array Biopharma, Aptose, AstraZeneca, Constellation, Genentech, Gilead, Incyte, Janssen R&D, Seattle Genetics, Syros, and Takeda and is a consultant for Leap Oncology. B.J.D. serves on the advisory boards for Gilead and Roche TCRC. B.J.D. is principal investigator or coinvestigator on Novartis and BMS clinical trials. His institution, Oregon Health & Science University, has contracts with these companies to pay for patient costs, nurse and data manager salaries, and institutional overhead. He does not derive salary, nor does his laboratory receive funds from these contracts. M.W.D. serves on the advisory boards and/or as a consultant for Novartis, Incyte, and BMS and receives research funding from BMS and Gilead. The authors certify that all compounds and combinations tested in this study were chosen without input from any of our industry partners. Freely available online through the PNAS open access option. 1 To whom correspondence may be addressed. Email: [email protected] or [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1703094114/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1703094114 PNAS Early Edition | 1 of 10 MEDICAL SCIENCES PNAS PLUS Downloaded by guest on August 3, 2020

Upload: others

Post on 07-Jul-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Molecularly targeted drug combinations …...2017/08/02  · Molecularly targeted drug combinations demonstrate selective effectiveness for myeloid- and lymphoid-derived hematologic

Molecularly targeted drug combinations demonstrateselective effectiveness for myeloid- and lymphoid-derived hematologic malignanciesStephen E. Kurtza, Christopher A. Eidea,b, Andy Kaempfc, Vishesh Khannaa,b, Samantha L. Savagea, Angela Rofeltya,Isabel Englisha, Hibery Hoa, Ravi Pandyad, William J. Boloskyd, Hoifung Poond, Michael W. Deiningere, Robert Collinsf,Ronan T. Swordsg, Justin Wattsg, Daniel A. Pollyeah, Bruno C. Medeirosi, Elie Traera, Cristina E. Tognona, Motomi Moric,j,Brian J. Drukera,b,1, and Jeffrey W. Tynerk,1

aDivision of Hematology & Medical Oncology, Oregon Health & Science University, Portland, OR 97239; bHoward Hughes Medical Institute, Oregon Health &Science University, Portland, OR 97239; cBiostatistics Shared Resource, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239;dMicrosoft Research, Redmond, WA 98052; eHuntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112; fUT Southwestern Medical Center,Dallas, TX 75390; gUniversity of Miami School of Medicine, Miami, FL 33136; hDivision of Hematology, University of Colorado School of Medicine, Aurora,CO 80045; iStanford University School of Medicine, Stanford, CA 94305; jOregon Health & Science University–Portland State University School of PublicHealth, Portland, OR 97239; and kDepartment of Cell, Development & Cancer Biology, Oregon Health & Science University, Portland, OR 97239

Contributed by Brian J. Druker, July 6, 2017 (sent for review March 9, 2017; reviewed by Keith Flaherty and A. Y. Leung)

Translating the genetic and epigenetic heterogeneity underlyinghuman cancers into therapeutic strategies is an ongoing challenge.Large-scale sequencing efforts have uncovered a spectrum ofmutations in many hematologic malignancies, including acutemyeloid leukemia (AML), suggesting that combinations of agentswill be required to treat these diseases effectively. Combinatorialapproaches will also be critical for combating the emergence ofgenetically heterogeneous subclones, rescue signals in the micro-environment, and tumor-intrinsic feedback pathways that allcontribute to disease relapse. To identify novel and effective drugcombinations, we performed ex vivo sensitivity profiling of 122 pri-mary patient samples from a variety of hematologic malignanciesagainst a panel of 48 drug combinations. The combinations weredesigned as drug pairs that target nonoverlapping biologicalpathways and comprise drugs from different classes, preferablywith Food and Drug Administration approval. A combination ratio(CR) was derived for each drug pair, and CRs were evaluated withrespect to diagnostic categories as well as against genetic, cytoge-netic, and cellular phenotypes of specimens from the two largestdisease categories: AML and chronic lymphocytic leukemia (CLL).Nearly all tested combinations involving a BCL2 inhibitor showedadditional benefit in patients with myeloid malignancies, whereasselect combinations involving PI3K, CSF1R, or bromodomain inhib-itors showed preferential benefit in lymphoid malignancies. Ex-panded analyses of patients with AML and CLL revealed specificpatterns of ex vivo drug combination efficacy that were associatedwith select genetic, cytogenetic, and phenotypic disease subsets,warranting further evaluation. These findings highlight the heuristicvalue of an integrated functional genomic approach to the identi-fication of novel treatment strategies for hematologic malignancies.

targeted therapies | drug combinations | ex vivo assay |hematologic malignancies

The promise of precision medicine is the ability to alignmedical interventions with individual patients at the time of

diagnosis and to alter treatment regimens as new mutations ariseand responses diminish. Although technical developments in next-generation sequencing and computational biology have acceleratedprogress in precision medicine, fundamental challenges remain.Whole-genome and whole-exome sequencing technologies canidentify many target mutations, but these techniques are analyti-cally intensive and may not reliably detect translocations, zygositychanges, and low-allele burden mutations with clinical significance.A further hindrance to the clinical utility of mutation status is a lackof drug therapies that selectively target cancer-associated mutations,with effective Food and Drug Administration (FDA)-approved

drugs existing for only a subset of the genes currently known tounderlie tumorigenesis (1). These limitations invite a complementarystrategy that assesses drug sensitivities obtained with targeted agentsdesigned to inhibit discrete cellular processes as a means of identi-fying phenotypic indications for specific cancers (2). Associatingphenotypic responses with particular genetic alterations may thenbeget precision-based therapies.For blood cancers, the success with targeted therapies de-

veloped to inhibit BCR-ABL fusions that drive chronic myelog-enous leukemia has spurred efforts to identify similar therapiesfor other hematopoietic malignancy types. Acute myeloid leu-kemia (AML), which results from the enhanced proliferation andimpaired differentiation of hematopoietic stem and progenitorcells, has substantial underlying heterogeneity, a feature that

Significance

Mononuclear cells obtained from freshly isolated patient sam-ples with various hematologic malignancies were evaluated forsensitivities to combinations of drugs that target specific cell-signaling pathways. The diagnostic, genetic/cytogenetic, andcellular features of the patient samples were correlated witheffective drug combinations. For myeloid-derived tumors, suchas acute myeloid leukemia, several combinations of targetedagents that include a kinase inhibitor and venetoclax, a selectiveinhibitor of BCL2, are effective.

Author contributions: S.E.K., C.A.E., R.P., W.J.B., H.P., E.T., C.E.T., B.J.D., and J.W.T. de-signed research; S.E.K., C.A.E., A.K., S.L.S., A.R., I.E., and H.H. performed research; V.K.,S.L.S., A.R., I.E., H.H., M.W.D., R.C., R.T.S., J.W., D.A.P., B.C.M., E.T., and B.J.D. contributednew reagents/analytic tools; S.E.K., C.A.E., A.K., M.M., B.J.D., and J.W.T. analyzed data;and S.E.K., C.A.E., A.K., D.A.P., B.C.M., C.E.T., M.M., and J.W.T. wrote the paper.

Reviewers: K.F., Dana-Farber/Harvard Cancer Center; and A.Y.L., University of Hong Kong.

Conflict of interest statement: D.A.P. serves on the advisory boards for Pharmacyclics andGilead. J.W.T. receives research support from Agios, Array Biopharma, Aptose, AstraZeneca,Constellation, Genentech, Gilead, Incyte, Janssen R&D, Seattle Genetics, Syros, and Takedaand is a consultant for Leap Oncology. B.J.D. serves on the advisory boards for Gilead andRoche TCRC. B.J.D. is principal investigator or coinvestigator on Novartis and BMS clinicaltrials. His institution, Oregon Health & Science University, has contracts with these companiesto pay for patient costs, nurse and data manager salaries, and institutional overhead. Hedoes not derive salary, nor does his laboratory receive funds from these contracts. M.W.D.serves on the advisory boards and/or as a consultant for Novartis, Incyte, and BMS andreceives research funding from BMS and Gilead. The authors certify that all compoundsand combinations tested in this study were chosen without input from any of ourindustry partners.

Freely available online through the PNAS open access option.1Towhom correspondence may be addressed. Email: [email protected] or [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1703094114/-/DCSupplemental.

www.pnas.org/cgi/doi/10.1073/pnas.1703094114 PNAS Early Edition | 1 of 10

MED

ICALSC

IENCE

SPN

ASPL

US

Dow

nloa

ded

by g

uest

on

Aug

ust 3

, 202

0

Page 2: Molecularly targeted drug combinations …...2017/08/02  · Molecularly targeted drug combinations demonstrate selective effectiveness for myeloid- and lymphoid-derived hematologic

indicates the need for multiple targeted therapies or the use ofcombinations.AML diagnosis relies on cytogenetic analysis, as recurrent

chromosomal variations represent established prognostic markers,even though nearly half of patients with AML have a normalkaryotype.DNA sequencing of 200 patients with AML uncovered an

average of 13 somatic mutations in each genome, of which5 mutations were recurrent (3). Common mutations in AML thatare also driver mutations represent potential therapeutic targets.Recurrent mutations in transcription factors and epigeneticregulators identified in AML suggest that aberrant transcrip-tional circuits are a common feature underlying leukemogenesis(3, 4). These circuits may drive oncogenic gene expression pro-grams to alter differentiation, activate self-renewal, and generateleukemia stem cells responsible for the initiation and propaga-tion of disease (5–7). Of the many genes commonly mutated inAML, targeted therapies in clinical trials or clinical use havebeen developed for only five: PML-RARA, FLT3, KIT, IDH1,and IDH2.The standard treatment for AML is chemotherapy consisting

of cytarabine and anthracyclines, which is more effective inadults younger than 60 y of age; however, the overall survivalrate 5 y after diagnosis is 25% (8). The outcome in older pa-tients, who represent the majority of patients with this diseaseand are unable to receive intensive chemotherapy, is poor, with amedian survival of 5–10 mo (9). It is also noteworthy that asignificant proportion of older patients with AML do not receiveany antileukemic therapy (10). A key exception is the subset ofpatients with AML with acute promyelocytic leukemia, for whichthe use of all-trans retinoic acid therapy results in excellent anddurable responses, suggesting the potential value of targetedtherapies for other AML subgroups (11, 12). Recent advance-ments in understanding of the molecular pathogenesis of AMLhave resulted in a growing number of molecularly targeted drugcandidates. However, several factors hinder the development ofeffective single-agent targeted treatments, including the intra-tumoral heterogeneity of hematologic malignancies, the emer-gence of genetically heterogeneous subclones leading to relapse,and rescue signals from the tumor microenvironment. Attemptsto develop small-molecule inhibitors of the tyrosine kinase FLT3,in which activating mutations are detected in approximately 30%of adult AML cases (13, 14), illuminate the difficulty for effectivesingle-agent targeted therapies. The short duration of response toFLT3 inhibitors is largely attributable to the rapid selection forand expansion of drug-resistant subclones (15–17). Targeted drugsmay yet improve treatment outcomes. However, it may be difficultfor these compounds, if used as single agents, to produce durableremissions necessary for long-term disease management or“bridging” the patient to successful bone marrow transplantationtherapy, the only current potential for cure. Combinations thatmodulate distinct pathways may provide an opportunity for im-proved responses (18). For example, the combination of anMEK inhibitor (trametinib) with an RAF inhibitor (dabrafenib)is now an approved therapy for BRAF mutation-positive meta-static melanoma (19). A similarly attractive alternative strategyfor AML, supported by emerging data, is the use of molecularlyguided drug combinations, such as quizartinib and azacitidine,which inhibit FLT3 and DNA methyltransferase activities, re-spectively (20).In the absence of a comprehensive portfolio of therapeutic

drugs targeting specific mutations, we used ex vivo functionalscreening to identify drug sensitivities in primary samples frompatients with various hematologic malignancies. Based on dataaccumulated from this assay to date, many instances of ex vivosensitivity to small-molecule kinase inhibitors have been vali-dated against known genetic targets (e.g., BCR-ABL, FLT3-ITD,RAS), and many novel drug/mutation associations have been

discovered (21–24). These data suggest that a similar screeningplatform may identify combinations of targeted agents that aremore effective than either of their respective single agents, thusdefining and enabling a rational program for selecting clinicallyrelevant combinatorial therapies. Thus, to identify new therapeuticcombinations for AML and other hematologic malignancies, weassessed the sensitivity of primary patient samples to various drugcombinations by using this ex vivo functional platform.

ResultsFreshly isolated primary mononuclear cells from patients withvarious hematologic malignancies (N = 122) were cultured in thepresence of a panel of 48 drug combinations, each in a fixedmolar dose series. The drug combinations were designed as pairsof inhibitors that target nonoverlapping biological pathways,comprising different classes of compounds, including kinase in-hibitors, bromodomain inhibitors, BH3 mimetics, and histonedeacetylase (HDAC) inhibitors. To maximize the translationalimpact of any findings, combinations used FDA-approved drugsif possible. For comparison, cells were also tested against gradedconcentrations of each inhibitor alone, and sensitivity wasassessed by a methanethiosulfonate (MTS)-based viability assayafter 3 d. The efficacy of each combination relative to its re-spective single agents was quantified with combination ratio(CR) values, defined as the IC50 or area under the fitted dose–response curve for the combination divided by the lowest IC50 orarea under the curve (AUC) value for either single agent. By thismetric, a CR value of less than 1 indicates the drug combinationis more effective than either single agent. We derived these CRvalues because of known limitations of applying conventionalsynergy calculations when one or more of the single agents iscompletely ineffective on particular samples (25).Patients were classified according to four general diagnostic

groups: AML, chronic lymphocytic leukemia (CLL), acute lym-phoblastic leukemia (ALL), and myeloproliferative neoplasms(MPNs) or myelodysplastic syndromes (MPNs; Table 1 andDataset S1). Unsupervised hierarchical clustering of CR valuesfor each drug combination revealed several distinct patterns ofefficacy (Fig. 1A). Myeloid leukemia patient samples wereenriched within a cluster of sensitivity to combinations pairingthe BCL2 inhibitor venetoclax with select kinase inhibitors[dasatinib (multikinase), doramapimod (p38), sorafenib (multi-kinase), or idelalisib (PI3KCD)]. This clustering pattern segre-gated the majority of myeloid vs. lymphoid samples. Within eachof these larger myeloid and lymphoid clusters, subsets of samplesshowed sensitivity to combinations involving the MEK inhibitortrametinib and a second kinase inhibitor [idelalisib, palbociclib(CDK4/6), or quizartinib (FLT3/CSF1R)]. In addition, a smallcluster of patients with leukemia showed sensitivity to combi-nations of the HDAC inhibitor panobinostat in tandem with theJAK inhibitor ruxolitinib and/or the multikinase inhibitor sor-afenib. To confirm that these clusters result from the effective-ness of the combination rather than that of a single agent, IC50values for each single agent were mapped according to thecombination cluster pattern. Importantly, apart from venetoclax,which, as a single agent, demonstrated potent and selective ef-ficacy in lymphoid (predominantly CLL) patient samples, single-agent efficacies did not align uniquely to a combination efficacy-derived myeloid or lymphoid cluster (Fig. S1). This pattern ofvenetoclax sensitivity is consistent with its recent approval fortreatment of patients with CLL with 17p deletions (26).To enable comparisons among different measures of combi-

nation efficacy, AUC values were also calculated for each sam-ple/drug pair, and IC50 and AUC values are highly correlated(Spearman ρ = 0.806; Fig. 1B). Clustering of AUC CR valuesyielded similar sensitivity clusters as those seen with IC50 CRresults (Fig. S2). In addition, there was a broad distribution inthe frequency and type of samples demonstrating combination

2 of 10 | www.pnas.org/cgi/doi/10.1073/pnas.1703094114 Kurtz et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 3

, 202

0

Page 3: Molecularly targeted drug combinations …...2017/08/02  · Molecularly targeted drug combinations demonstrate selective effectiveness for myeloid- and lymphoid-derived hematologic

efficacy. For instance, the palbociclib/ruxolitinib, alisertib/crizo-tinib, vandetanib/vemurafenib, and palbociclib/quizartinib com-binations were effective, according to both CR measures, inmore than 100 of 122 patient samples surveyed, encompassing allfour diagnosis categories. Furthermore, the HDAC inhibitorpanobinostat as a single agent showed potency across most of thepatient samples tested (Fig. S3). To relate these findings withother definitions of synergy, IC50 and AUC CR effect measuresfor a subset of combinations and samples were compared withexcess over Bliss (EOB) determinations (27). A high level ofagreement was observed between the two methods (Spearman rvalues for IC50 CR or AUC CR vs. EOB, 0.953 and 0.928, re-spectively; P < 0.0001).To identify drug combinations that were more frequently ef-

fective within specific diagnostic categories, the median CRvalues for IC50 and AUC for each combination within each of thefour diagnostic subgroups were compared with a CR referencevalue of 1 (Datasets S2 and S3). Combinations in which medianIC50 CR and AUC CR were significantly <1 within a specificdiagnostic subgroup were mapped to regions of a four-way Venndiagram depending on subgroup efficacy observed (Fig. 1D).Consistent with findings from clustering of CR values, severalcombinations exhibited median CR values significantly less than1 selectively among patient samples with myeloid malignancies,including combinations of venetoclax and the CSF1R inhibitorARRY-382, p38 MAPK inhibitor doramapimod, or bromodo-main inhibitor JQ1 (Fig. 1E). In contrast, six combinations wereselectively effective in CLL patient samples only, including qui-zartinib/ibrutinib and JQ1/sorafenib (Fig. 1E). Representativesingle-agent and combination dose–response curves for thesecombinations are provided in Fig. S3. Several combinations weresignificantly effective beyond either single agent in myeloid andlymphoid samples, with the most common broad efficacy at-tributable to the palbociclib/ruxolitinib combination. Althoughno unique effective combinations were identified for ALL pa-tient samples, this was likely attributable to its smaller cohort size(n = 12) among the diagnostic categories surveyed. For a morecomplete evaluation of combination benefit, median IC50 andAUC values for each single agent were also compared acrossdiagnosis subgroups (Dataset S4). In particular, venetoclax sen-sitivity was significantly different among disease types, with thelowest median IC50 and AUC values observed for CLL samples(median IC50, 51.4 nM vs. 2,105 nM for AML samples). Con-trastingly, sensitivities to JQ1, quizartinib, sorafenib, and cabo-zantinib were also different across subgroups, with selectivepotency for AML samples by both CR effect measures. Notably,none of the single agents except venetoclax and quizartinib dem-

onstrated significant selectivity for the same diagnosis subgroupalone and in combination. Furthermore, CR values across thepatient cohort were not significantly associated with the generalpatient characteristics of sex, age (either as a categorical orcontinuous variable), or type of specimen analyzed (DatasetS4). Viewed comprehensively, these findings suggest patternsof disease-selective enhanced efficacy of targeted therapycombinations that highlight potential opportunities for thetreatment of heterogeneous diagnostic subtypes of myeloid andlymphoid malignancies.For the two largest diagnostic groups, AML and CLL, ex-

panded panels of clinical, prognostic, mutational, cytogenetic,and surface-antigen data were compiled for comparisons to CRvalues for each drug combination. For AML samples (n = 58),beyond general clinical characteristics such as age, sex, and whiteblood cell (WBC) count, additional annotations examined in-cluded mutational profiles from a focused panel of genes com-monly mutated in AML, cytogenetic features from standardchromosome analysis, and cell-surface antigen expressionaccording to flow cytometry (Fig. 2 and Datasets S5 and S6).Notably, the most prevalent mutation in this cohort was NPM1(33%), and approximately 50% of the cohort featured normalkaryotype. Patients harboring mutations in NPM1 demonstratedsignificantly enhanced sensitivity to the JQ1/sorafenib combina-tion [median IC50 CR, 0.437; false discovery rate (FDR)-adjusted P = 0.010]. Patients harboring mutations in DNMT3Aexhibited significantly enhanced sensitivity to the JQ1/palbociclibcombination (median IC50 CR, 0.119; FDR-adjusted P = 0.017;Fig. 3, Left). Patients with normal cytogenetics were significantlysensitive to combinations of ruxolitinib/cabozantinib and JQ1/sorafenib, whereas those harboring complex karyotypes weresignificantly sensitive to the combination of idelalisib/quizartinib(Fig. 3, Middle). Surface expression of several specific myeloidmarkers was also associated with significant sensitivity to com-binations involving venetoclax, including CD11b (integrin αM) tovenetoclax/JQ1 (median IC50 CR, 0.121; FDR-adjusted P =0.001) and CD58 (LFA-3) venetoclax/doramapimod (medianIC50 CR, 0.040; FDR-adjusted P = 0.003; Fig. 3, Right).CLL samples were characterized for the mutational status of

IgVH and TP53. Cytogenetic features for chromosomal deletionsand trisomy, as well as prognostic cell surface antigens such asCD38 and ZAP70, were determined by standard chromosomeanalysis and flow cytometry, respectively (Fig. 4 and Datasets S7and S8). Among the tested combinations, the most significantassociations with respect to disease characteristics were observedin patients harboring deletion of 13q, who showed significantsensitivity to combinations of palbociclib with venetoclax (median

Table 1. Summary of select patient characteristics for surveyed sample cohort

Characteristic All patients (n = 122) ALL (n = 12) AML (n = 58) CLL (n = 42) MPN or MDS/MPN (n = 10)

Sex (n evaluable) n = 116 n = 12 n = 56 n = 42 n = 6Female 59 8 30 20 1Male 57 4 26 22 5

Age, y (n evaluable) n = 106 n = 9 n = 54 n = 37 n = 6Median 62.1 29.7 53.7 65.9 60.7Range 5.5–83.8 5.5–70.2 8.2–83.8 44.7–79.4 35.2–74.9

Sample type (n evaluable) n = 122 n = 12 n = 58 n = 42 n = 10Peripheral blood 86 7 31 41 7Bone marrow aspirate 34 5 25 1 3Leukapheresis 2 0 2 0 0

WBC count (n evaluable) n = 115 n = 9 n = 55 n = 42 n = 9Median, ×1,000/μL 42.9 60.0 34.6 45.8 92.7Range, ×1,000/μL 0.5–309 10.3–169 0.5–240 5.8–309 26.3–240

In total, primary samples from 122 patients with a variety of hematologic malignancies were screened for ex vivo sensitivity toinhibitor combinations.

Kurtz et al. PNAS Early Edition | 3 of 10

MED

ICALSC

IENCE

SPN

ASPL

US

Dow

nloa

ded

by g

uest

on

Aug

ust 3

, 202

0

Page 4: Molecularly targeted drug combinations …...2017/08/02  · Molecularly targeted drug combinations demonstrate selective effectiveness for myeloid- and lymphoid-derived hematologic

A

E

C

D

B

Venetoclax - DasatinibVenetoclax - Doramapimod

Venetoclax - JQ1Venetoclax - ARRY-382Venetoclax - PalbociclibVenetoclax - RuxolitinibVenetoclax - SorafenibVenetoclax - Idelalisib

Venetoclax - QuizartinibPanobinostat - Venetoclax

Panobinostat - JQ1Panobinostat - Doramapimod

Panobinostat - IdelalisibPanobinostat - ARRY-382Panobinostat - QuizartinibPanobinostat - PalbociclibPanobinostat - RuxolitinibPanobinostat - Sorafenib

JQ1 - IdelalisibJQ1 - ARRY-382JQ1 - Sorafenib

JQ1 - DoramapimodJQ1 - PalbociclibJQ1 - Ruxolitinib

Palbociclib - ARRY-382Panobinostat - Trametinib

Quizartinib - ARRY-382Sorafenib - Entosplentinib

JQ1 - QuizartinibJQ1 - Trametinib

Cytarabine - Nutlin 3aRuxolitinib - Cabozantinib

Quizartinib - IbrutinibPalbociclib - IdelalisibIdelalisib - ARRY-382Idelalisib - RuxolitinibIdelalisib - Quizartinib

Alisertib - CrizotinibVandetanib - Vemurafenib

Palbociclib - DoramapimodPalbociclib - RuxolitinibPalbociclib - QuizartinibPalbociclib - SorafenibTrametinib - Idelalisib

Trametinib - ARRY-382Trametinib - PalbociclibTrametinib - QuizartinibTrametinib - Venetoclax

15

-00

69

91

5-0

06

92

15

-00

71

21

5-0

08

10

15

-00

66

11

5-0

08

04

15

-00

65

71

5-0

09

26

15

-00

67

21

5-0

08

13

15

-00

67

91

5-0

06

65

16

-00

05

21

5-0

07

42

16

-00

07

01

5-0

08

53

15

-00

90

41

5-0

08

09

15

-00

87

41

5-0

08

83

15

-00

69

81

5-0

07

111

5-0

09

71

15

-00

66

21

5-0

06

63

15

-00

67

71

5-0

06

95

15

-00

67

61

5-0

08

23

15

-00

83

31

5-0

07

55

16

-00

05

61

6-0

00

73

16

-00

05

41

5-0

07

32

15

-00

88

21

5-0

08

69

15

-00

67

11

5-0

07

49

15

-00

96

11

5-0

07

77

15

-00

73

41

5-0

07

67

15

-00

83

51

5-0

08

51

15

-00

85

51

5-0

07

23

15

-00

76

61

5-0

09

76

15

-00

88

11

5-0

09

12

15

-00

98

71

6-0

00

75

15

-00

86

81

5-0

09

35

15

-00

70

21

5-0

08

72

15

-00

75

61

5-0

09

90

15

-00

73

51

5-0

07

17

15

-00

811

15

-00

76

41

5-0

06

73

16

-00

07

71

5-0

08

92

15

-00

80

11

5-0

09

74

15

-00

87

01

5-0

07

90

16

-00

07

81

6-0

00

41

16

-00

04

81

6-0

00

94

15

-00

68

01

5-0

07

80

16

-00

09

01

5-0

08

15

15

-00

80

71

5-0

07

86

16

-00

00

11

5-0

08

93

16

-00

110

16

-00

10

61

5-0

06

64

15

-00

97

91

6-0

00

93

16

-00

09

91

5-0

09

30

16

-00

06

71

6-0

01

28

15

-00

97

51

5-0

09

42

15

-00

83

71

5-0

09

09

16

-00

115

15

-00

91

51

5-0

09

17

15

-00

94

91

5-0

09

46

15

-00

94

11

5-0

09

86

16

-00

02

31

5-0

09

00

15

-00

66

61

5-0

06

70

15

-00

81

61

5-0

09

51

15

-00

95

61

6-0

00

03

15

-00

90

31

5-0

08

29

15

-00

70

11

6-0

01

09

15

-00

74

31

6-0

00

88

15

-00

71

41

6-0

00

98

16

-00

12

51

5-0

09

44

16

-00

10

21

6-0

01

20

ALL AML

CLL MPN or MDS/MPN

-2.86 -0.71 ≥0

log(IC50 CR)

(95% CI:0.7973-0.8157)

-5-4-3-2-10123

-4 -3 -2 -1 0 1 2 3 4

log(

AU

C C

R)

log(IC50 CR)

Idelalisib - Quizartinib

Venetoclax - DoramapimodVenetoclax - JQ1

Venetoclax - ARRY-382

Venetoclax - PalbociclibVenetoclax - SorafenibVenetoclax - Ruxolitinib

Palbociclib - Idelalisib

Idelalisib - ARRY-382Idelalisib - Ruxolitinib

Palbociclib - Doramapimod

Palbociclib - Ruxolitinib

Cytarabine - Nutlin 3a

Trametinib - Idelalisib

Trametinib - ARRY-382

Trametinib - PalbociclibTrametinib - Venetoclax

Panobinostat - RuxolitinibJQ1 - IdelalisibJQ1 - Sorafenib

JQ1 - PalbociclibJQ1 - Ruxolitinib

Alisertib - Crizotinib

Vandetanib - Vemurafenib

Ruxolitinib - CabozantinibQuizartinib - IbrutinibPalbociclib - Quizartinib

Palbociclib - Sorafenib

ALL

AML

CLL

MPN or MDS/MPN

Venetoclax - Dasatinib

-4

-2

0

2

4

-4

-2

0

2

4

Venetoclax - ARRY-382 Venetoclax - Doramapimod

Quizartinib - Ibrutinib JQ1 - Sorafenib

ALL(n=12)

AML(n=58)

CLL(n=42)

MPN or MDS/MPN

(n=10)

ALL(n=12)

AML(n=58)

CLL(n=42)

MPN or MDS/MPN

(n=10)

ALL(n=12)

AML(n=58)

CLL(n=42)

MPN or MDS/MPN

(n=10)

ALL(n=12)

AML(n=58)

CLL(n=42)

MPN or MDS/MPN

(n=10)

log(

IC50

CR

)lo

g(IC

50 C

R)

***

***** *

******

020406080

100120140

All sam

ples

Palboc

iclib

- Rux

olitin

ib

Alisertib

- Criz

otinib

Vande

tanib

- Vem

urafen

ib

Palboc

iclib

- Quiz

artini

b

Cytarab

ine - N

utlin

3a

Idelal

isib - R

uxoli

tinib

JQ1 -

Rux

olitin

ib

Palboc

iclib

- Dora

mapim

od

Palboc

iclib

- Sora

fenib

Veneto

clax -

Palboc

iclib

Ruxoli

tinib

- Cab

ozan

tinib

Veneto

clax -

JQ1

Veneto

clax -

Rux

olitin

ib

Idelal

isib - A

RRY-382

Veneto

clax -

Sorafen

ib

JQ1 -

Palboc

iclib

Palboc

iclib

- Idela

lisib

JQ1 -

Sorafen

ib

Idelal

isib - Q

uizart

inib

Veneto

clax -

Das

atinib

Veneto

clax -

ARRY-382

Veneto

clax -

Dora

mapim

od

Trameti

nib - P

alboc

iclib

Quizart

inib -

Ibrut

inib

Veneto

clax -

Idela

lisib

Trameti

nib - I

delal

isib

Trameti

nib - A

RRY-382

JQ1 -

Idela

lisib

Trameti

nib - V

eneto

clax

Trameti

nib - Q

uizart

inib

Sorafen

ib - E

ntosp

lentin

ib

Palboc

iclib

- ARRY-38

2

Quizart

inib -

ARRY-382

Panob

inosta

t - Ven

etocla

x

Panob

inosta

t - Rux

olitin

ib

Veneto

clax -

Quiz

artini

b

JQ1 -

Dora

mapim

od

JQ1 -

ARRY-382

Panob

inosta

t - Palb

ocicli

b

Panob

inosta

t - ARRY-38

2

Panob

inosta

t - Dora

mapim

od

JQ1 -

Trameti

nib

Panob

inosta

t - Quiz

artini

b

Panob

inosta

t - Sora

fenib

Panob

inosta

t - Ide

lalisib

JQ1 -

Quiz

artini

b

Panob

inosta

t - JQ

1

Panob

inosta

t - Tram

etinib

Num

ber o

f sam

ples

ALL AML CLL MPN or MDS/MPN

Samples with IC50 CR and AUC CR < 1

Spearmanr=0.8067

Fig. 1. Differential patterns of selective efficacy of small-molecule combinations relative to single agents. (A) Unsupervised hierarchical clustering of IC50 CRvalues for 122 patients with leukemia across 48 tested combinations. IC50 CR values were log-transformed and row- and column-clustered using Pearsoncorrelation pairwise average linkage method. Darker red color (lower CR values) indicates drug combinations exhibiting higher efficacy than either singleagent alone. Diagnostic category annotation of each sample is also shown. (B) Correlation of IC50 CR and AUC CR effect measure values. Shaded regionindicates sample/drug pairs for which the combination was more effective than either single agent (CR < 1) by both effect measures. (C) Distribution ofeffective drug combinations based on frequency and diagnostic category. Bar labeled “all samples” indicates diagnosis group breakdown for all 122 surveyedpatient samples for comparison. (D) Overlap of significantly effective inhibitor combinations among four general diagnosis subgroups. Combinations forwhich median IC50 CR and AUC CR were significantly <1 within each diagnostic subgroup are shown; combinations listed in overlapping regions were sig-nificantly effective in each subgroup. (E) Select examples of combinations by diagnostic subgroup. Scatter plots of log-transformed IC50 CR values for selectcombinations. Black horizontal bars represent median CR. (*Median CR significantly <1 in that subgroup.)

4 of 10 | www.pnas.org/cgi/doi/10.1073/pnas.1703094114 Kurtz et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 3

, 202

0

Page 5: Molecularly targeted drug combinations …...2017/08/02  · Molecularly targeted drug combinations demonstrate selective effectiveness for myeloid- and lymphoid-derived hematologic

IC50 CR, 0.040; FDR-adjusted P = 0.003) or trametinib (medianIC50 CR, 0.116; P = 0.006; Fig. 5).Although strict association of a particular combination with a

patient feature may require a more complete understanding ofthe inter- and intrasubgroup heterogeneity and expanded samplesize, we found statistically significant associations for selectprevalent biomarkers (Table 2). Furthermore, in the broadercontext of the two largest diagnosis categories (AML and CLL),certain inhibitor combinations demonstrated disease-specificselective efficacy irrespective of genetic and clinical features.For example, several combinations of venetoclax with a kinaseinhibitor have enhanced efficacy in AML (e.g., venetoclax/dor-amapimod), whereas combinations of JQ1 with multiple kinaseinhibitors exhibit an enhanced efficacy preference for CLL (JQ1/sorafenib; Fig. 6). Importantly, these results, obtained by directlycomparing the median CR values of the two largest diseasesubsets, are consistent with our earlier findings from un-supervised clustering. Thus, this approach uncovers multiplepotential opportunities for application of combination therapiesin distinct diagnostic, genetic/cytogenetic, and phenotypic sub-sets of patients with AML and CLL.To address the reproducibility of these results, we compiled an

independent validation dataset (N = 151) of a similar distribu-tion of prospectively collected primary leukemia patient samples.There is a high level of correlation for the median IC50 and AUCCRs between the discovery and validation datasets for all com-binations and samples tested (Spearman r = 0.975 and 0.949,respectively; P < 0.0001; Fig. 6B). Given the particularly notableefficacy of combinations involving venetoclax in AML patientspecimens from our discovery cohort, a subset of the most ef-fective of these combinations was compared with those of thevalidation-set AML samples, revealing similar sensitivity distri-

butions in both groups (Fig. 6C). Additionally, expanded synergyanalysis of this same subset of combinations was performed foreach drug alone over a seven-point dose series and all 49 possiblecombinations of the two agents on a panel of human leukemiacell lines (MOLM-13, MOLM-14, HL-60, OCI-AML2, OCI-AML3). Overall, cell-line IC50 and AUC CR measures (de-termined by equimolar series) correlated well with the Blisssynergy score across the full matrix of combinations (Spearmanr = 0.649 and 0.787, respectively; P < 0.0001).

DiscussionAs the vast molecular heterogeneity of cancer continues to beunraveled, the necessity of defining actionable targeted thera-peutic strategies for patients remains paramount to improvementin outcomes. Given difficulties involving the often nondurableresponses and rapid development of resistance observed withmany single-agent targeted therapies, we sought to identify ef-fective combination strategies for patients with hematologicmalignancies. Before this study, we screened more than 1,000 pri-mary patient specimens against a panel of single-agent small-molecule inhibitors. Using these historical drug sensitivity data, weranked drugs by their IC50 and used these rankings to assemble aninitial panel of drug combinations consisting primarily of kinaseinhibitors that targeted nonoverlapping pathways. Primary patientsamples with various hematologic malignancies were screened, and,based on data from this initial panel, we generated a second iter-ation including new combinations of kinase inhibitors as well ascombinations of inhibitors from select additional drug classes.However, for both panels, broad cytotoxicity was problematic formany combinations, and unsupervised hierarchical clustering ofCR values revealed no distinct clusters tracking with diagnosiscategory. This preliminary work informed the design of the panel of

PositiveNegative

N/A

PositiveNegative

N/A

PositiveNegative

N/A

Sex --- 56Type of sample --- 58Age, grouped (yrs) --- 55Age, continuous (yrs) --- 55WBC (K/uL) --- 55Blasts in BM (%) --- 47Blasts in PB (%) --- 50Prognostic risk --- 49NPM1 19 58FLT3-ITD 15 58DNMT3A 9 48NRAS 8 48TET2 8 48RUNX1 7 48IDH2 6 48CEBPA 5 49IDH1 4 48PTPN11 4 48TP53 4 48U2AF1 4 47FLT3 D835 3 47WT1 3 48FLT3 other 2 47KIT 2 48KRAS 2 47EZH2 1 45Normal 25 51Complex karyotype 10 51-7 3 51-5 or del(5q) 3 51inv(16) or t(16;16)(p13.1;q22) 3 51t(15;17) 3 51t(8;21) 2 51inv(3) 1 51t(v;11)(v;q23) 1 51abnl(17p) 0 51t(6;9) 0 51t(9;11) 0 51CD33 45 49CD13 44 50CD117 42 48CD34 31 46CD64 24 43CD56 16 38CD11b 15 39CD58 14 31CD15 11 38CD14 9 36CD71 3 31

Clin

ical

Mut

atio

nsC

ytog

enet

ics

Sur

face

antig

ens

Posit

ive (n

)Ev

aluab

le (n

)

Adverse Intermediate-IFavorable N/A

Female Male

N/A

BM PB Leukapheresis≥65 40-64 18-39 <18Yrs (0-100)WBC (0-250)% (0-100)

AML

15-0

0811

15-0

0872

15-0

0755

15-0

0979

16-0

0109

15-0

0723

15-0

0883

15-0

0680

16-0

0041

15-0

0717

15-0

0892

16-0

0078

15-0

0990

15-0

0855

16-0

0056

16-0

0003

16-0

0102

15-0

0734

16-0

0128

15-0

0670

16-0

0120

15-0

0909

15-0

0975

16-0

0088

15-0

0764

16-0

0048

15-0

0813

15-0

0976

15-0

0702

15-0

0900

15-0

0807

15-0

0767

15-0

0903

15-0

0766

15-0

0673

15-0

0756

15-0

0874

16-0

0067

15-0

0692

15-0

0786

15-0

0829

15-0

0777

16-0

0077

15-0

0837

16-0

0075

16-0

0073

16-0

0094

16-0

0001

15-0

0742

15-0

0974

16-0

0070

15-0

0961

15-0

0930

15-0

0912

15-0

0942

16-0

0115

15-0

0701

15-0

0870

Fig. 2. Clinical and genetic features of patients with AML surveyed. Panels of the indicated disease-specific clinical, prognostic, mutation, cytogenetic, andsurface antigen features were compared among all 58 patients with AML in the study. The number of patients evaluable for each feature is given, along withthe number of positive samples for that feature (when relevant for categorical variables). Gray boxes indicate unavailable information. Each patient is shownin a unique column, and samples are sorted from left to right according to frequency of genetic mutations.

Kurtz et al. PNAS Early Edition | 5 of 10

MED

ICALSC

IENCE

SPN

ASPL

US

Dow

nloa

ded

by g

uest

on

Aug

ust 3

, 202

0

Page 6: Molecularly targeted drug combinations …...2017/08/02  · Molecularly targeted drug combinations demonstrate selective effectiveness for myeloid- and lymphoid-derived hematologic

combinations used herein, which added several additional classes ofinhibitors as well as an intentional inclusion of FDA-approved drugsor drugs in clinical development wherever possible. To this end,defining drug combinations for ex vivo screening is a highly iterativeprocess of refinement. Although logistical considerations precludecomprehensive evaluation of all possible pairwise combinations ofinhibitors, these cumulative data may also prove amenable to ap-plied machine learning-based computational models to predictnovel drug target pairings in specific malignancies (28, 29), and may

be adaptable as a testing scheme for other tumor types given recentdevelopments to isolate circulating tumor cells (30, 31).A critical finding in this study is the effectiveness of several

combinations of targeted agents that include a kinase inhibitorand venetoclax, a selective inhibitor of BCL2, for myeloid-derived tumors (Fig. 6). The relative sensitivity to these combi-nations in the ex vivo assay supports the notion that coupling akinase-derived proliferative signal inhibitor with an antiapoptoticagent improves efficacy. Furthermore, we confirmed the capacity

-4

-2

0

2

4

Positive Negative Positive Negative

DNMT3A NPM1

JQ1 - Palbociclib JQ1 - Sorafenib

Positive Negative Positive Negative

Normal karyotype Complex karyotype

Ruxolitinib -Cabozantinib

Idelalisib -Quizartinib

Positive Negative Positive Negative

CD11b CD58

Venetoclax - JQ1 Venetoclax -Doramapimod

*** *

* ** **

-4

-2

0

2

4

-4

-2

0

2

4

log(

IC50

CR

)AML mutations AML cytogenetic abnormalities AML surface antigen expression

-4

-3.5

-3

-2.5

-2

-1.5

-1

-0.5

0

log(

FDR

-adj

uste

d p-

valu

efo

r pos

itive

sam

ples

)

-2-1.5-1-0.500.511.52[median CR for positive samples][median CR for negative samples]( )log

-4

-3.5

-3

-2.5

-2

-1.5

-1

-0.5

0-2-1.5-1-0.500.511.52

[median CR for positive samples][median CR for negative samples]( )log

-4

-3.5

-3

-2.5

-2

-1.5

-1

-0.5

0-2-1.5-1-0.500.511.52

[median CR for positive samples][median CR for negative samples]( )log

NPM1(JQ1 - Palbociclib)

NPM1(JQ1 - Sorafenib)

DNMT3A(JQ1 - Palbociclib)

Complex karyotype(Idelalisib - Quizartinib)

Normal karyotype(Ruxolitinib - Cabozantinib)

Normal karyotype(JQ1 - Sorafenib)

CD11b(Venetoclax- JQ1)

CD58(Venetoclax - Ruxoltinib)

CD58(Venetoclax- Doramapimod)

CD11b(Venetoclax- Doramapimod)CD15

(Venetoclax- Idelalisib)

CD58(Venetoclax- ARRY-382)

CD58(Venetoclax - Dasatinib)

CD14(Venetoclax- JQ1)

A

B

Fig. 3. Associations of selective inhibitor combination benefit with mutation, cytogenetic, and surface antigen expression features in AML. (A) Scatter plotsof combination/feature pairing test significance vs. difference in median CR between subgroups. Summaries for mutation (Left), cytogenetic (Middle), andsurface antigens (Right). All plotted points correspond to combination/feature pairings in which (i) median IC50 and AUC CR of the negative samples were notsignificantly <1 and (ii) positive and negative subgroups contained at least 15% of total evaluable samples each. Points above the horizontal dashed gray linedemonstrated median IC50 CR and AUC CR values for positive subgroup that were significantly <1 (i.e., FDR-adjusted P < 0.05). Points to the right of thevertical dashed gray line represent those in which the median IC50 CR value of the positive subgroup was at least twofold lower than that of the negativesubgroup. (B) Scatter plots of log-transformed IC50 CR values for select combinations by AML mutation (Left), cytogenetic (Middle), and surface antigenexpression subgroups (Right). Black horizontal bars represent median CR value. (*Median CR significantly <1 in that subgroup.)

15-0

0677

15-0

0695

15-0

0868

15-0

0823

15-0

0657

15-0

0661

15-0

0671

15-0

0926

16-0

0093

16-0

0052

15-0

0666

15-0

0676

15-0

0698

15-0

0699

15-0

0749

15-0

0809

15-0

0816

15-0

0833

15-0

0853

15-0

0893

15-0

0951

15-0

0956

16-0

0098

15-0

0662

15-0

0904

15-0

0946

15-0

0714

15-0

0835

15-0

0712

16-0

0090

15-0

0664

15-0

0665

15-0

0711

15-0

0815

15-0

0869

15-0

0679

16-0

0110

15-0

0663

15-0

0882

15-0

0804

15-0

0917

16-0

0099

24---xeSType of sample 42Age, quartiles (yrs) --- 37Age, continuous (yrs) 37WBC (K/uL) --- 42Monocytes in PB (K/uL) --- 32Monocytes in PB (%) --- 42

2291HVGI11535PT9362)q31(led638)p71(led147lamroN734)q11(led

Trisomy 12 4 35532)q6(led22783DC71207PAZ

Clin

ical

Mutations

Cyt

ogen

etic

s

Surfaceantigens

CLL

PositiveNegativeN/A

Positive Negative N/A

Positive Negative N/A

Female Male

N/A

BM PB≥72 65-71 60-64 <60Yrs (0-100)WBC (0-350)

% (0-10)

Pos

itive

(n)

Eva

luab

le (n

)

Monocytes (0-10)

Fig. 4. Clinical and genetic features of patients with CLL surveyed. Panels of the indicated disease-specific clinical, mutation, cytogenetic, and surface antigenfeatures were compared among all 42 patients with CLL in the study. The number of patients evaluable for each feature is given, along with the number ofpositive samples for a given feature (when relevant for categorical variables). Gray boxes indicate unavailable information. Each patient is shown in a uniquecolumn, and samples are sorted from left to right according to frequency of cytogenetic abnormalities.

6 of 10 | www.pnas.org/cgi/doi/10.1073/pnas.1703094114 Kurtz et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 3

, 202

0

Page 7: Molecularly targeted drug combinations …...2017/08/02  · Molecularly targeted drug combinations demonstrate selective effectiveness for myeloid- and lymphoid-derived hematologic

of select venetoclax combinations to augment apoptotic celldeath in a panel of human AML cell lines (Fig. S4). Combina-tions such as dasatinib, doramapimod, sorafenib, or idelalisibwith venetoclax are broadly effective on myeloid-derived tumorsamples and may be useful for treatment of AML in particular.These combinations appear to be effective across a broad per-centage of AML patient samples, irrespective of cohort subtypeheterogeneity. Consistent with these observations, recent reportsindicate the combined inhibition of BCR-ABL1 and BCL2 is apromising strategy for targeting Philadelphia chromosome-positive ALL as well as the stem cell population in chronic my-eloid leukemia (32, 33). We also observed certain combinationswith venetoclax to be effective on CLL samples with 13q dele-tions. Although venetoclax has recently achieved FDA approvalfor patients with CLL with 17p deletions, our data suggest thatvenetoclax, even as a single agent, may be more broadly effectivein patients with CLL with diverse cytogenetic profiles, and

combinations may offer options particularly for disease states inwhich venetoclax as a single agent is not effective. It is note-worthy that venetoclax is effective for a variety of hematologicmalignancy subsets, including CLL, multiple myeloma, and AML(34–37). Additionally, we note that genetic features that areconsiderably less frequent in AML or CLL will require expandedsample sizes to more robustly interrogate possible associationswith inhibitor combination efficacy. Similarly, small sample sizesof other diagnosis groups (ALL, MPN, or MDS/MPN) are alimitation of the present study. We may potentially miss impor-tant drug combinations because of small sample sizes and lowerpower, and, as such, these diagnostic groups will require largersample sizes to permit the identification of relationships betweencombination efficacy and disease-specific features.Developments in functional screening technology have pro-

duced several assay platforms for the evaluation of responses oftumor cells to exogenous perturbations. Functional screening

Table 2. Selective inhibitor combination sensitivities by feature among AML and CLL patient samples surveyed

Feature type Diagnosis category Combination

IC50 AUC

Median CR P value* Median CR P value*

MutationDNMT3A AML JQ1/palbociclib 0.1187 0.0171 0.5693 0.0189NPM1 AML JQ1/palbociclib 0.1138 0.0045 0.6830 0.0120NPM1 AML JQ1/sorafenib 0.4375 0.0108 0.6952 0.0091

CytogeneticsComplex karyotype AML Idelalisib/quizartinib 0.1753 0.0227 0.6872 0.0078Normal karyotype AML Ruxolitinib/cabozantinib 0.3289 0.0126 0.8040 0.0015Normal karyotype AML JQ1/sorafenib 0.4375 0.0378 0.8495 0.0197del(13q) CLL Trametinib/palbociclib 0.1161 0.0057 0.4121 <0.0001del(13q) CLL Venetoclax/palbociclib 0.2673 0.0013 0.5269 <0.0001

Surface antigenCD11b AML Venetoclax/doramapimod 0.0441 0.0136 0.4503 0.0185CD11b AML Venetoclax/JQ1 0.1205 0.0014 0.2539 0.0070CD14 AML Venetoclax/JQ1 0.1004 0.0201 0.2539 0.0061CD15 AML Venetoclax/idelalisib 0.0308 0.0189 0.6769 0.0309CD58 AML Venetoclax / ARRY-382 0.0381 0.0012 0.2796 0.0006CD58 AML Venetoclax/dasatinib 0.0229 0.0007 0.2594 0.0021CD58 AML Venetoclax/doramapimod 0.0402 0.0031 0.4396 0.0035CD58 AML Venetoclax/ruxolitinib 0.0027 0.0036 0.3359 0.0053

*P values are FDR-adjusted.

-4

-2

0

2

4

Positive Negative Positive Negative

del(13q) del(13q)

Venetoclax -Palbociclib

Trametinib -Palbociclib

****

log(

IC50

CR

)

CLL cytogenetic abnormalities-4

-3.5

-3

-2.5

-2

-1.5

-1

-0.5

0-2-1.5-1-0.500.511.52

[median CR for positive samples][median CR for negative samples]( )log

log(

FDR

-adj

uste

d p-

valu

efo

r pos

itive

sam

ples

) del(13q)(Venetoclax - Palbociclib)

del(13q)(Trametinib - Palbociclib)

A B

Fig. 5. Sensitivity of CLL patient samples harboring del(13q)to combinations with the CDK4/6 inhibitor palbociclib. (A) Scatter plot of combination-featurepairing test significance vs. difference in median CR between subgroups. All plotted points correspond to combination/feature pairings in which (i) themedian IC50 and AUC CR of the negative samples were not significantly <1 and (ii) the positive and negative subgroups contained at least 15% of the totalevaluable samples each. Points above the horizontal dashed gray line demonstrate median IC50 CR and AUC CR values for the positive subgroup that weresignificantly <1 (i.e., FDR-adjusted P < 0.05). Points to the right of the vertical dashed gray line represent those for which the median IC50 CR value of thepositive subgroup was at least twofold lower than that of the negative subgroup. (B) Scatter plot of log-transformed IC50 CR values for select combinationseffective in del(13q)-positive CLL samples. Black horizontal bars represent the median CR value. (*Median CR is significantly <1 in that subgroup.)

Kurtz et al. PNAS Early Edition | 7 of 10

MED

ICALSC

IENCE

SPN

ASPL

US

Dow

nloa

ded

by g

uest

on

Aug

ust 3

, 202

0

Page 8: Molecularly targeted drug combinations …...2017/08/02  · Molecularly targeted drug combinations demonstrate selective effectiveness for myeloid- and lymphoid-derived hematologic

efforts in hematologic malignancies have often involved the cultureof patient cells in conventional 2D tissue culture platforms, some-times with conventional culture conditions (38–40) and other timesusing additives or feeder cell coculture that promote certain phe-notypic aspects of the cells, such as preservation of primitive celldifferentiation state (41) and cell proliferation (42). A key feature ofour ex vivo assay is that it provides drug sensitivity data within 4 d, atime frame that can support and influence clinical decision-making.Furthermore, this approach addresses some of the challenges indeploying effective therapies in which there is a substantial gap be-tween clinical, diagnostic, or genetic markers and available drugs.Ideally, the integration of functional and genomic data types mayfacilitate more precise insight into the molecular mechanisms con-tributing to disease. For example, our data suggest that combinedtargeting of the BTK inhibitor ibrutinib and the multikinase inhibitorquizartinib may represent a promising strategy for patients with CLL.The efficacy of ibrutinib is well established for CLL (reviewed in

ref. 43). Although quizartinib is primarily considered to be aFLT3 inhibitor, it is also a potent inhibitor of CSF1R, a targetrecently implicated for CLL treatment because of its effects onsupportive nurse-like monocyte/macrophage cells that expressCSF1R (44–46), highlighting the importance of teasing outtumor-intrinsic mechanisms from tumor-extrinsic microenviron-ment contributions to sensitivity/resistance. Accordingly, simulta-neous inhibition of BTK- and CSF1R-mediated signaling pathwaysmay result in further improvement of responses (Fig. 6).Although we have identified several links between actionable

diagnostic and genetic features and effective combinations oftargeted agents, we also acknowledge certain limitations to ouranalyses. For example, the inhibitor screening method requiresthe use of prospectively collected, freshly isolated patient sam-ples as a consequence of a clinical visit. Variability in the numberof mononuclear cells recovered from a specimen limits the scopeand number of inhibitors that may be tested. Furthermore, the

-2.5 -2.0 -1.5 -1.0 -0.5 0 0.5 1.0 2.0

Venetoclax - ARRY-382Venetoclax - DoramapimodVenetoclax - IdelalisibVenetoclax - JQ1Palbociclib - ARRY-382Venetoclax - DasatinibPalbociclib - IdelalisibVenetoclax - PalbociclibVenetoclax - RuxolitinibVenetoclax - SorafenibIdelalisib - RuxolitinibTrametinib - ARRY-382Cytarabine - Nutlin 3aIdelalisib - ARRY-382Panobinostat - VenetoclaxPanobinostat - RuxolitinibTrametinib - VenetoclaxPalbociclib - RuxolitinibIdelalisib - TrametinibTrametinib - PalbociclibPalbociclib - DoramapimodAlisertib - CrizotinibIdelalisib - QuizartinibVandetanib - VemurafenibPalbociclib - QuizartinibJQ1 - PalbociclibJQ1 - RuxolitinibPalbociclib - SorafenibRuxolitinib - CabozantinibJQ1 - SorafenibJQ1 - IdelalisibQuizartinib - Ibrutinib

Difference of median IC50 CR

2.51.5

AML selective CLL selective

Discovery set log(median CR)

Discovery set log(median CR)

Val

idat

ion

set l

og(m

edia

n C

R)

Val

idat

ion

set l

og(m

edia

n C

R)

A B

-3 -2 -1 0 2 31

-1 -0.5 0 10.5-1

-0.5

0

1

0.5

-3

-2

-1

0

2

3

1

IC50 CR

AUC CR

C Venetoclax - Doramapimod

AMLDiscovery set

AMLValidation set

-3

-2

-1

0

1

2

3

log(

IC50

CR

)

Venetoclax - JQ1

-3

-2

-1

0

1

2

3

log(

IC50

CR

)

Quizartinib - Ibrutinib

-3

-2

-1

0

1

2

3

log(

IC50

CR

)

AMLDiscovery set

AMLValidation set

CLLDiscovery set

CLLValidation set

FDR-adjustedp-value: <0.0001 0.0103 1000.0<1000.0<1000.0< 0.0048

Spearman r:0.975

Spearman r:0.949

Fig. 6. Validation of combination selectivity between AML and CLL. (A) The difference of median IC50 CR values (AML – CLL) was computed for each of48 indicated combinations by using the Hodges-Lehmann method. The median difference is represented by a closed circle, and the 95% CI is shown as thecolored bar. AML-selective and CLL-selective combinations are colored orange or green, respectively. (B) Spearman correlation of log-transformed median IC50

CR and AUC CR values between the discovery sample cohort (N = 122) and independent validation cohort (N = 151). (C) Validation of select effectivecombinations within AML or CLL diagnostic subgroups. Scatter plots of log-transformed IC50 CR values for the indicated combinations. Black horizontal barsrepresent median CR; FDR-adjusted P values (Wilcoxon signed-rank test of median) are shown.

8 of 10 | www.pnas.org/cgi/doi/10.1073/pnas.1703094114 Kurtz et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 3

, 202

0

Page 9: Molecularly targeted drug combinations …...2017/08/02  · Molecularly targeted drug combinations demonstrate selective effectiveness for myeloid- and lymphoid-derived hematologic

challenge of overcoming noise in the drug sensitivity data causedby variations in biological response and technique limit the utilityof applying conventional methods of determining synergy (25).Additionally, as a result of the prospective nature of samplecollection, there are differences in sample size and frequency ofgenetic parameters surveyed within a given diagnostic group. Toaddress these issues, we used an integrated and robust approach,including the development of a CR to measure relative efficacyof inhibitor combinations, requiring that both effect measures(IC50 and AUC) achieve significance, performing rank-basedtests of the median within and between relevant subgroups,and applying multiplicity adjustments to the P values from thesetests. We also note that developments in new oncology drugswithin various drug classes will present opportunities to increasetarget/pathway representation in combination screening. Finally,our assay makes use of aggregate readings of whole mononuclearcell population responses to drug combinations and singleagents, whereby a readout that offers granularity of responses atthe single-cell level would provide additional data for parsing ofdrug combination efficacy. Recent approaches that make use ofsingle-cell imaging or flow cytometry (47–51), or CRISPR/Cas9 gene silencing (52), will be useful in enhancing our initialview of combination efficacy and in identifying new targets.In summary, our data identify promising drug combinations

that were previously unrecognized but may promote the testingof a select number of these drug combinations in clinical trials.Validation of these combinations will require testing in clinicaltrials, which may be suitable initially for patients with AML withthe relapsed/refractory disease. Accordingly, we formed combi-nations with FDA-approved drugs to facilitate their translationaleffect. With respect to the BCL2 inhibitor venetoclax, there isnow compelling clinical evidence supporting its evaluation incombination with other agents for AML (53). Independent ofthe findings reported here, three clinical trials currently in therecruitment phase will provide opportunities to evaluate targetedcombinations of the BCL2 inhibitor (venetoclax) with an MEKinhibitor (cobimetinib; ClinicalTrials.gov ID code NCT02670044),an MDM2 inhibitor (idasanutlin; ClinicalTrials.gov ID codeNCT02670044), or a BET inhibitor (ABBV-075; ClinicalTrials.gov ID code NCT02391480) on patients with relapsed/refractoryAML who are not eligible for induction therapy. Collectively,these findings may yield new therapeutic options for patients whileadvancing the use of ex vivo functional testing as a valid assay inthe clinical decision-making process.

Materials and MethodsPatient Samples. All patients gave consent to participate in this study, whichhad the approval and guidance of the institutional review boards at OregonHealth & Science University (OHSU), University of Utah, University of TexasMedical Center (UT Southwestern), Stanford University, University of Miami,and University of Colorado. Mononuclear cells were isolated by Ficoll-gradient centrifugation from freshly obtained bone marrow aspirates orperipheral blood draws and plated into assays within 24 h. All samples wereanalyzed for clinical characteristics and drug sensitivity. AML and CLL patientsamples were additionally analyzed with respect to expanded, disease-specific panels of clinical, prognostic, genetic, cytogenetic, and surface an-tigen characteristics obtained from patient electronic medical records. Geneticcharacterization of AML samples included results of a clinical deep-sequencingpanel of genes commonly mutated in hematologic malignancies (GeneTrailspanel from Knight Diagnostic Laboratories, OHSU; Foundation Medicine re-ports from UT Southwestern).

Ex Vivo Functional Screen. Small-molecule inhibitors, purchased from LCLaboratories and Selleck Chemicals, were reconstituted in DMSO and storedat −80 °C. The CSF1R inhibitor ARRY-382 was obtained from Array Bio-pharma. Inhibitors were distributed into 384-well plates prepared with asingle agent per well in a seven-point concentration series ranging from10 μM to 0.0137 μM for each drug (except dasatinib, which was plated at aconcentration range of 1 μM to 0.00137 μM). Similar plates were preparedwith the 48 indicated pairwise inhibitor combinations in seven-point fixed

molar concentration series identical to those used for single agents. The finalconcentration of DMSO was ≤0.1% in all wells, and all sets of single-agentand combination destination plates were stored at −20 °C and thawed im-mediately before use. Primary mononuclear cells were plated across single-agent and combination inhibitor panels within 24 h of collection. Cells wereseeded into 384-well assay plates at 10,000 cells per well in RPMI 1640 mediasupplemented with FBS (10%), L-glutamine, penicillin/streptomycin, andβ-mercaptoethanol (10−4 M). After 3 d of culture at 37 °C in 5% CO2, MTSreagent (CellTiter96 AQueous One; Promega) was added, optical density wasmeasured at 490 nm, and raw absorbance values were adjusted to a refer-ence blank value and then used to determine cell viability (normalized tountreated control wells).

Inhibitor Dose–Response Curve Analysis and Effect-Measure Calculations.Normalized viability values (40) at each dose of a seven-point dilution se-ries for 21 small-molecule inhibitors and 48 pairwise combinations of two ofthese single agents were analyzed for each of 122 primary leukemia sam-ples. Dose concentrations were log10-transformed, and a probit regressioncurve was fit to each seven-point drug sensitivity profile by using maximum-likelihood estimation for the intercept and slope. This parametric model waschosen over a polynomial because the probit’s monotonic shape reflects adose–response curve typically seen in samples incubated with cytotoxic orinhibitory agents (54). Normalized viability values greater than 100%, in-dicating higher cell viability than the average viability across control wells ona given plate, were truncated to 100% to produce a percentage responsevariable amenable to probit modeling. From the fitted probit curve for eachsample/drug pairing, the IC50 was defined as the lowest concentration toachieve 50% predicted viability and the AUC was computed by integrationof the curve height across the tested dose range. If the predicted cell viability(i.e., probit curve height) was ≤50% at the lowest tested dose or >50%across the entire dose range, the IC50 was designated as the lowest dose orhighest dose, respectively. For sensitivity profiles with 100% normalized vi-ability at all seven dose points, the IC50 and AUC were designated as thehighest tested dose and the maximum possible AUC, respectively. For sen-sitivity profiles with 0% viability at all seven dose points, the IC50 and AUCwere designated as the lowest tested dose and a value (0.5) just below theminimum probit-derived AUC, respectively.

To quantify the efficacy of an equimolar drug combination in comparisonwith its constituent single agents, a CR effect measure was generated basedon the specific IC50 and AUC values for each inhibitor triad (the drug com-bination and the two single agents). The IC50 CR and AUC CR values weredefined as the ratio of the combination’s IC50 or AUC to the minimum IC50 orAUC for the two single agents, respectively. Each sensitivity profile modeledby probit regression was assigned a fit statistic based on the P value for thetest of whether the fitted curve’s slope was horizontal. Generally, a smallerfit statistic produced by a decreasing slope indicates a better fit and, byextension, provides a measure of confidence in the curve-derived IC50 andAUC for a particular sample/drug pair. A CR effect measure value less than1 indicates that a sample is more sensitive to the drug combination than it isto either of the single agents that constitute the combination.

Statistical Analysis. Unsupervised hierarchical clustering and heat-map dis-plays of inhibitor sensitivity were generated by using GenePattern software(Broad Institute). Inhibitor combination efficacy was compared for all samples(N = 122) across a panel of general clinical variables: diagnostic category(AML, ALL, CLL, MPN or MDS/MPN), age (categorized as <18, 18–39, 40–64,or ≥65 y), sex, and type of specimen (peripheral blood, bone marrow aspi-rate, leukapheresis). For each CR effect measure, a one-sample Wilcoxonsigned-rank test was used to assess whether the median CR value was sig-nificantly less than 1 to identify effective drug combinations for each sub-group. Furthermore, differences in median CR across the subgroups of thesevariables were evaluated with the Kruskal–Wallis test. Additional diagnosis-specific clinical and genetic variables were examined for AML (n = 58) andCLL (n = 42) patient samples. Similar tests were performed on a subgroup’smedian CR and on differences across subgroups for each of these clinical orgenetic variables. Subgroups for all mutations, cytogenetic abnormalities,and cell surface antigens were defined as positive or negative. Correlationsbetween continuous clinical variables (e.g., WBC count) and CR values wereappraised with Spearman correlation coefficients. For all within-subgroupand between-subgroup nonparametric tests, FDR adjustments (55) wereapplied to P values with an adaptive linear step-up method to account fortests being performed on each of the 48 inhibitor triads.

ACKNOWLEDGMENTS. We thank the 122 patients who contributed samplesto enable this study; David Patterson, David Haussler, and Pierrette Lo for

Kurtz et al. PNAS Early Edition | 9 of 10

MED

ICALSC

IENCE

SPN

ASPL

US

Dow

nloa

ded

by g

uest

on

Aug

ust 3

, 202

0

Page 10: Molecularly targeted drug combinations …...2017/08/02  · Molecularly targeted drug combinations demonstrate selective effectiveness for myeloid- and lymphoid-derived hematologic

helpful discussions; and Brian Junio for sample coordination. This work wassupported by The Leukemia & Lymphoma Society (B.J.D.), National Human Ge-nome Research Institute Grant U54HG007990 (to S.E.K.), the V Foundation forCancer Research (J.W.T.), the Gabrielle’s Angel Foundation for Cancer Research(J.W.T.), National Cancer Institute Grant 1R01CA183947-01 (to J.W.T.), University

ofMiami Clinical and Translational Science Institute Award KL2TR000461 (to J.W.),a Howard Hughes Medical Institute Medical Research Fellows Program (to V.K.),and the Biostatistics Shared Resource of the Oregon Health & Science UniversityKnight Cancer Institute via National Cancer Institute Grant 5P30CA69533 (to A.K.and M.M.). B.J.D. is a Howard Hughes Medical Institute investigator.

1. Dienstmann R, Jang IS, Bot B, Friend S, Guinney J (2015) Database of genomic bio-markers for cancer drugs and clinical targetability in solid tumors. Cancer Discov 5:118–123.

2. Friedman AA, Letai A, Fisher DE, Flaherty KT (2015) Precision medicine for cancer withnext-generation functional diagnostics. Nat Rev Cancer 15:747–756.

3. Ley TJ, et al.; Cancer Genome Atlas Research Network (2013) Genomic and epi-genomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med 368:2059–2074.

4. Döhner H, Weisdorf DJ, Bloomfield CD (2015) Acute myeloid leukemia. N Engl J Med373:1136–1152.

5. Chao MP, Seita J, Weissman IL (2008) Establishment of a normal hematopoietic andleukemia stem cell hierarchy. Cold Spring Harb Symp Quant Biol 73:439–449.

6. Reya T, Morrison SJ, Clarke MF, Weissman IL (2001) Stem cells, cancer, and cancer stemcells. Nature 414:105–111.

7. Somervaille TC, Cleary ML (2006) Identification and characterization of leukemia stemcells in murine MLL-AF9 acute myeloid leukemia. Cancer Cell 10:257–268.

8. Döhner H, et al.; European LeukemiaNet (2010) Diagnosis and management of acutemyeloid leukemia in adults: Recommendations from an international expert panel, onbehalf of the European LeukemiaNet. Blood 115:453–474.

9. Oran B, Weisdorf DJ (2012) Survival for older patients with acute myeloid leukemia: Apopulation-based study. Haematologica 97:1916–1924.

10. Medeiros BC, et al. (2015) Big data analysis of treatment patterns and outcomesamong elderly acute myeloid leukemia patients in the United States. Ann Hematol94:1127–1138.

11. Ravandi F, et al. (2009) Effective treatment of acute promyelocytic leukemia with all-trans-retinoic acid, arsenic trioxide, and gemtuzumab ozogamicin. J Clin Oncol 27:504–510.

12. Lo-Coco F, Orlando SM, Platzbecker U (2013) Treatment of acute promyelocytic leu-kemia. N Engl J Med 369:1472.

13. Nakao M, et al. (1996) Internal tandem duplication of the flt3 gene found in acutemyeloid leukemia. Leukemia 10:1911–1918.

14. Yamamoto Y, et al. (2001) Activating mutation of D835 within the activation loop ofFLT3 in human hematologic malignancies. Blood 97:2434–2439.

15. Man CH, et al. (2012) Sorafenib treatment of FLT3-ITD(+) acute myeloid leukemia:Favorable initial outcome and mechanisms of subsequent nonresponsiveness associ-ated with the emergence of a D835 mutation. Blood 119:5133–5143.

16. Shah NP, et al. (2013) Ponatinib in patients with refractory acute myeloid leukaemia:findings from a phase 1 study. Br J Haematol 162:548–552.

17. Baker SD, et al. (2013) Emergence of polyclonal FLT3 tyrosine kinase domain muta-tions during sequential therapy with sorafenib and sunitinib in FLT3-ITD-positiveacute myeloid leukemia. Clin Cancer Res 19:5758–5768.

18. Shafer D, Grant S (2016) Update on rational targeted therapy in AML. Blood Rev 30:275–283.

19. Spain L, Julve M, Larkin J (2016) Combination dabrafenib and trametinib in themanagement of advanced melanoma with BRAFV600 mutations. Expert OpinPharmacother 17:1031–1038.

20. Chang E, et al. (2016) The combination of FLT3 and DNA methyltransferase inhibitionis synergistically cytotoxic to FLT3/ITD acute myeloid leukemia cells. Leukemia 30:1025–1032.

21. Leonard JT, et al. (2016) Functional and genetic screening of acute myeloid leukemiaassociated with mediastinal germ cell tumor identifies MEK inhibitor as an activeclinical agent. J Hematol Oncol 9:31.

22. Maxson JE, et al. (2016) Identification and characterization of tyrosine kinase non-receptor 2 mutations in leukemia through integration of kinase inhibitor screeningand genomic analysis. Cancer Res 76:127–138.

23. Maxson JE, et al. (2015) Therapeutically targetable ALK mutations in leukemia.Cancer Res 75:2146–2150.

24. Maxson JE, et al. (2013) Oncogenic CSF3R mutations in chronic neutrophilic leukemiaand atypical CML. N Engl J Med 368:1781–1790.

25. Chou TC (2006) Theoretical basis, experimental design, and computerized simulationof synergism and antagonism in drug combination studies. Pharmacol Rev 58:621–681.

26. Croce CM, Reed JC (2016) Finally, an apoptosis-targeting therapeutic for cancer.Cancer Res 76:5914–5920.

27. Foucquier J, Guedj M (2015) Analysis of drug combinations: current methodologicallandscape. Pharmacol Res Perspect 3:e00149.

28. Tang J, et al. (2013) Target inhibition networks: Predicting selective combinations ofdruggable targets to block cancer survival pathways. PLOS Comput Biol 9:e1003226.

29. Behinaein B, Rudie K, Sangrar W (October 3, 2016) Petri net siphon analysis and graphtheoretic measures for identifying combination therapies in cancer. IEEE/ACM TransComput Biol Bioinformatics, 10.1109/TCBB.2016.2614301.

30. Hayes DF, Smerage JB (2010) Circulating tumor cells. Prog Mol Biol Transl Sci 95:95–112.

31. Alix-Panabières C, Pantel K (2013) Circulating tumor cells: Liquid biopsy of cancer. ClinChem 59:110–118.

32. Leonard JT, et al. (2016) Targeting BCL-2 and ABL/LYN in Philadelphia chromosome-positive acute lymphoblastic leukemia. Sci Transl Med 8:354ra114.

33. Carter BZ, et al. (2016) Combined targeting of BCL-2 and BCR-ABL tyrosine kinaseeradicates chronic myeloid leukemia stem cells. Sci Transl Med 8:355ra117.

34. Davids MS, Letai A, Brown JR (2013) Overcoming stroma-mediated treatment re-sistance in chronic lymphocytic leukemia through BCL-2 inhibition. Leuk Lymphoma54:1823–1825.

35. Pan R, et al. (2014) Selective BCL-2 inhibition by ABT-199 causes on-target cell death inacute myeloid leukemia. Cancer Discov 4:362–375.

36. Anderson MA, et al. (2016) The BCL2 selective inhibitor venetoclax induces rapidonset apoptosis of CLL cells in patients via a TP53-independent mechanism. Blood127:3215–3224.

37. Roberts AW, et al. (2016) Targeting BCL2 with venetoclax in relapsed chronic lym-phocytic leukemia. N Engl J Med 374:311–322.

38. Tyner JW, et al. (2009) RNAi screen for rapid therapeutic target identification inleukemia patients. Proc Natl Acad Sci USA 106:8695–8700.

39. Pemovska T, et al. (2013) Individualized systems medicine strategy to tailor treat-ments for patients with chemorefractory acute myeloid leukemia. Cancer Discov 3:1416–1429.

40. Tyner JW, et al. (2013) Kinase pathway dependence in primary human leukemiasdetermined by rapid inhibitor screening. Cancer Res 73:285–296.

41. Pabst C, et al. (2014) Identification of small molecules that support human leukemiastem cell activity ex vivo. Nat Methods 11:436–442.

42. Klco JM, et al. (2013) Genomic impact of transient low-dose decitabine treatment onprimary AML cells. Blood 121:1633–1643.

43. Maddocks K, Jones JA (2016) Bruton tyrosine kinase inhibition in chronic lymphocyticleukemia. Semin Oncol 43:251–259.

44. Galletti G, Caligaris-Cappio F, Bertilaccio MT (2016) B cells and macrophages pursue acommon path toward the development and progression of chronic lymphocyticleukemia. Leukemia 30:2293–2301.

45. Galletti G, et al. (2016) Targeting macrophages sensitizes chronic lymphocytic leu-kemia to apoptosis and inhibits disease progression. Cell Reports 14:1748–1760.

46. Polk A, et al. (2016) Colony-stimulating factor-1 receptor is required for nurse-like cellsurvival in chronic lymphocytic leukemia. Clin Cancer Res 22:6118–6128.

47. Irish JM, et al. (2004) Single cell profiling of potentiated phospho-protein networks incancer cells. Cell 118:217–228.

48. Kornblau SM, et al. (2010) Dynamic single-cell network profiles in acute myelogenousleukemia are associated with patient response to standard induction therapy. ClinCancer Res 16:3721–3733.

49. Del Gaizo Moore V, Letai A (2013) BH3 profiling–measuring integrated function ofthe mitochondrial apoptotic pathway to predict cell fate decisions. Cancer Lett 332:202–205.

50. Touzeau C, et al. (2016) BH3 profiling identifies heterogeneous dependency on Bcl-2family members in multiple myeloma and predicts sensitivity to BH3 mimetics.Leukemia 30:761–764.

51. Friedman AA, et al. (June 27, 2017) Feasibility of ultra-high-throughput functionalscreening of melanoma biopsies for discovery of novel cancer drug combinations. ClinCancer Res, 10.1158/1078-0432.CCR-16-3029.

52. Doudna JA, Charpentier E (2014) Genome editing. The new frontier of genome en-gineering with CRISPR-Cas9. Science 346:1258096.

53. Konopleva M, et al. (2016) Efficacy and biological correlates of response in a phase IIstudy of venetoclax monotherapy in patients with acute myelogenous leukemia.Cancer Discov 6:1106–1117.

54. Altshuler B (1981) Modeling of dose-response relationships. Environ Health Perspect42:23–27.

55. Benjamini Y, Hochberg Y (2000) On the adaptive control of the false discovery rate inmultiple testing with independent statistics. J Educ Behav Stat 25:60–83.

10 of 10 | www.pnas.org/cgi/doi/10.1073/pnas.1703094114 Kurtz et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 3

, 202

0