polo-like kinase 1 inhibitors and their potential role in...

9
Review Polo-like Kinase 1 Inhibitors and Their Potential Role in Anticancer Therapy, with a Focus on NSCLC Ren e H. Medema 1 , Chia-Chi Lin 2 , and James Chih-Hsin Yang 2,3 Abstract Cytotoxic platinum-doublet chemotherapy that includes antimitotic agents is a current standard of care in advanced non–small cell lung cancer (NSCLC). Microtubule-targeting antimitotics, taxanes, and Vinca alkaloids are effective anticancer therapeutics that affect both dividing and nondividing cells. A new generation of antimitotic agents that target regulatory proteins—mitotic kinases and kinesins—has the potential to overcome the limitations related to the role of tubulin in nondividing cells that are associated with traditional antimitotics. This review concentrates on Polo-like kinase 1, a key regulator of mitosis, outlines a rationale for its development as an anticancer target, and discusses data from preclinical and clinical studies of Plk1 inhibitors with a particular focus on NSCLC. Clin Cancer Res; 17(20); 6459–66. Ó2011 AACR. Current Treatment Options Lung cancer is the second most common malignancy in the United States (219,440 cases) and was the leading cause of cancer-related mortality (159,390 deaths) in 2009 (1). Non–small cell lung cancer (NSCLC) represents 85% of all lung cancers (2); approximately 40% are diagnosed with advanced or metastatic disease (3) with a 5-year survival rate of 14% (4). Chemotherapy with platinum-based doublets (e.g. pac- litaxel/carboplatin, docetaxel/cisplatin, gemcitabine/cis- platin, and pemetrexed/cisplatin) is the standard of care in first-line treatment of advanced NSCLC with median overall survival of approximately 8 months (5) and overall response rate of 19% to 30% (5, 6). Combination of bevacizumab with platinum doublets increases overall response rate and prolongs progression-free survival in selected NSCLC patients (7, 8). Second-line treatments comprise monotherapies, such as docetaxel (9, 10), peme- trexed (11), or erlotinib (12), and the benefits of these agents as maintenance therapy are currently debated (13). Erlotinib is recommended for patients who have failed more than 1 or 2 therapy regimens (12); cytotoxic che- motherapies in such patients are largely ineffective (14). Gefitinib is restricted to second- and third-line patients currently or previously benefiting from gefitinib or to certain conditions, such as the presence of epidermal growth factor receptor (EGFR)-activating mutations. Clin- ical development of erlotinib and gefitinib—tyrosine kinase inhibitors of EGFR—has opened up the possibility of these agents in targeted patients whose tumors harbor activating EGFR gene mutations (15, 16). Significant improvements in response and survival have been reported with erlotinib and gefitinib in such patients (17–21). Novel Approaches Targeting Mitosis The development of novel agents takes advantage of rapidly accumulating knowledge of the pathways altered in cancer cells. Targeting mitosis is a validated approach, and agents that affect the mitotic spindle are well-estab- lished components of many oncotherapeutic regimens, including NSCLC (22). Taxanes (paclitaxel and docetaxel), microtubule-stabilizing agents, Vinca alkaloids (vinorel- bine, vinblastine, etc.), and microtubule-destabilizing agents impair normal mitotic spindle function and block cellular proliferation (23). New approaches to mitosis inhibition target regulatory proteins—mitotic kinases and kinesins—that control the process (Fig. 1) and have the potential to overcome limitations of traditional anti- mitotic agents related to the role of tubulin in normal cells (22). One such target is Polo-like kinase 1 (Plk1), a key regulator in several essential cell-cycle events, including mitotic entry, centrosome maturation, bipolar spindle for- mation, chromosome segregation, anaphase-promoting complex regulation, and cytokinesis (Fig. 2; ref. 24). Plk1 in Cancer Plk1 is the best characterized member of the Plk family (Plk1–5; ref. 25). It is expressed in dividing cells and Authors' Affiliations: 1 Department of Medical Oncology and Cancer Genomics Center, University Medical Center Utrecht, Utrecht, The Nether- lands; 2 Department of Oncology, and 3 Graduate Institute of Oncology and Cancer Research Center, National Taiwan University, Taipei, Taiwan Corresponding Author: James Chih-Hsin Yang, Department of Oncolo- gy, National Taiwan University Hospital, 7 Chung-Shan S. Road, Taipei 100, Taiwan. Phone: 886-2-2312-3456 ext 67511; Fax: 886-2-2371-1174; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-11-0541 Ó2011 American Association for Cancer Research. Clinical Cancer Research www.aacrjournals.org 6459 on November 8, 2018. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Upload: phamtram

Post on 09-Nov-2018

215 views

Category:

Documents


0 download

TRANSCRIPT

Review

Polo-like Kinase 1 Inhibitors and Their Potential Rolein Anticancer Therapy, with a Focus on NSCLC

Ren�e H. Medema1, Chia-Chi Lin2, and James Chih-Hsin Yang2,3

AbstractCytotoxic platinum-doublet chemotherapy that includes antimitotic agents is a current standard of care

in advanced non–small cell lung cancer (NSCLC). Microtubule-targeting antimitotics, taxanes, and Vinca

alkaloids are effective anticancer therapeutics that affect both dividing and nondividing cells. A new

generation of antimitotic agents that target regulatory proteins—mitotic kinases and kinesins—has the

potential to overcome the limitations related to the role of tubulin in nondividing cells that are associated

with traditional antimitotics. This review concentrates on Polo-like kinase 1, a key regulator of mitosis,

outlines a rationale for its development as an anticancer target, and discusses data from preclinical and

clinical studies of Plk1 inhibitors with a particular focus on NSCLC. Clin Cancer Res; 17(20); 6459–66.

�2011 AACR.

Current Treatment Options

Lung cancer is the second most common malignancy inthe United States (219,440 cases) and was the leading causeof cancer-related mortality (159,390 deaths) in 2009 (1).Non–small cell lung cancer (NSCLC) represents 85% of alllung cancers (2); approximately 40% are diagnosed withadvanced or metastatic disease (3) with a 5-year survivalrate of 14% (4).Chemotherapy with platinum-based doublets (e.g. pac-

litaxel/carboplatin, docetaxel/cisplatin, gemcitabine/cis-platin, and pemetrexed/cisplatin) is the standard of carein first-line treatment of advanced NSCLC with medianoverall survival of approximately 8 months (5) and overallresponse rate of 19% to 30% (5, 6). Combination ofbevacizumab with platinum doublets increases overallresponse rate and prolongs progression-free survival inselected NSCLC patients (7, 8). Second-line treatmentscomprise monotherapies, such as docetaxel (9, 10), peme-trexed (11), or erlotinib (12), and the benefits of theseagents as maintenance therapy are currently debated (13).Erlotinib is recommended for patients who have failedmore than 1 or 2 therapy regimens (12); cytotoxic che-motherapies in such patients are largely ineffective (14).Gefitinib is restricted to second- and third-line patients

currently or previously benefiting from gefitinib or tocertain conditions, such as the presence of epidermalgrowth factor receptor (EGFR)-activating mutations. Clin-ical development of erlotinib and gefitinib—tyrosinekinase inhibitors of EGFR—has opened up the possibilityof these agents in targeted patients whose tumors harboractivating EGFR gene mutations (15, 16). Significantimprovements in response and survival have been reportedwith erlotinib and gefitinib in such patients (17–21).

Novel Approaches Targeting Mitosis

The development of novel agents takes advantage ofrapidly accumulating knowledge of the pathways alteredin cancer cells. Targeting mitosis is a validated approach,and agents that affect the mitotic spindle are well-estab-lished components of many oncotherapeutic regimens,including NSCLC (22). Taxanes (paclitaxel and docetaxel),microtubule-stabilizing agents, Vinca alkaloids (vinorel-bine, vinblastine, etc.), and microtubule-destabilizingagents impair normal mitotic spindle function and blockcellular proliferation (23). New approaches to mitosisinhibition target regulatory proteins—mitotic kinasesand kinesins—that control the process (Fig. 1) and havethe potential to overcome limitations of traditional anti-mitotic agents related to the role of tubulin in normal cells(22). One such target is Polo-like kinase 1 (Plk1), a keyregulator in several essential cell-cycle events, includingmitotic entry, centrosome maturation, bipolar spindle for-mation, chromosome segregation, anaphase-promotingcomplex regulation, and cytokinesis (Fig. 2; ref. 24).

Plk1 in Cancer

Plk1 is the best characterized member of the Plk family(Plk1–5; ref. 25). It is expressed in dividing cells and

Authors' Affiliations: 1Department of Medical Oncology and CancerGenomics Center, University Medical Center Utrecht, Utrecht, The Nether-lands; 2Department of Oncology, and 3Graduate Institute of Oncology andCancer Research Center, National Taiwan University, Taipei, Taiwan

Corresponding Author: James Chih-Hsin Yang, Department of Oncolo-gy, National Taiwan University Hospital, 7 Chung-Shan S. Road, Taipei100, Taiwan. Phone: 886-2-2312-3456 ext 67511; Fax: 886-2-2371-1174;E-mail: [email protected]

doi: 10.1158/1078-0432.CCR-11-0541

�2011 American Association for Cancer Research.

ClinicalCancer

Research

www.aacrjournals.org 6459

on November 8, 2018. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

detectable only in proliferating adult tissues (e.g., spleenand testis; ref. 26). High Plk1 levels are a marker ofcellular proliferation (27), observed in NSCLC (28)and other malignancies, including cancers of the breast,colon, endometrium, head and neck, esophagus, ovary,pancreas, prostate, and stomach, as well as glioblastoma,non-Hodgkin lymphoma, leukemia, and melanoma (26,29–31). Plk1 levels in NSCLC tumors correlate inverselywith survival, indicating that Plk1 may have prognostic

value (28); this correlation has also been reported inother tumor types (26, 30, 31). In addition, high tumormitotic rate and risk of metastatic disease have beenassociated with high Plk1 levels (32), implying a rolefor Plk1 in aggressive cancer.

Mechanistic studies using RNA interference (RNAi) haveprovided additional insight into the role of Plk1 in cancercells and the molecular consequences of Plk1 inhibition.Plk1 depletion using siRNA in human cancer cell linesinterrupts spindle assembly and maturation resulting inmitotic (Polo) arrest characterized by scattered chromo-somes on a monopolar spindle and apoptosis (33, 34).This apoptosis is p53 independent; moreover, p53-null or-depleted cells are more sensitive to Plk1 depletion thanwild-type cells (35, 36), suggesting that cancers such asNSCLC with p53 loss of function may be particularlysensitive to Plk1 inhibitors.

Genome-wide RNAi screening has identified Plk1 asan essential survival factor in Ras-driven cells that expe-rience Ras-induced mitotic stress; such cells may beselectively sensitized to Plk-1 inhibition (37). K-ras–activating mutations are common in NSCLC (38) andmay serve as an indicator of better response to Plk1inhibitors. K-ras mutations are found less frequently inthe squamous cell carcinoma histologic subtype (�5%)and more commonly in the adenocarcinoma histologic

Translational Relevance

Targeting the mitotic spindle apparatus leading todisruption of mitosis is a validated approach in cancertreatment. Several novel agents that inhibit proteinregulators of mitosis are undergoing clinical investiga-tion. This review describes inhibitors of a regulator ofmitotic spindle formation, Polo-like kinase 1 (Plk1),introduces the rationale for targeting Plk1 in cancertherapy, and discusses preclinical and early clinical datafrom studies of Plk1 inhibitors, with a particular focuson non–small cell lung cancer (NSCLC). The potentialrole of this novel class of agents in the treatment ofNSCLC and future challenges in their clinical develop-ment are also discussed.

Anaphase Prometaphase

Metaphase

Prophase

Entry intomitosis

Microtubule captureand spindle–pole

separation

Chromosomealignment

Spindle checkpoint

Plk1

Plk1

Plk1CENPE

KSPPlk3

Aurora B

Aurora B

Aurora A

Aurora A

Aurora B

Telophase andcytokinesis

Interphase

Figure 1. Protein effectors of mitosis. KSP, kinesin spindle protein; CENPE, centromeric protein E. Adapted by permission from Macmillan Publishers Ltd.(22). Copyright � 2007.

Medema et al.

Clin Cancer Res; 17(20) October 15, 2011 Clinical Cancer Research6460

on November 8, 2018. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

subtype (�15%–30%; �25% in Western countries, butless common in Asian populations) of NSCLC.Short hairpin RNAs against Plk1 reduced the growth of

NSCLC A549 cells in mouse tumor xenografts and sup-pressed Plk1 tumor expression (39). Systemic treatmentwith siRNA against Plk1 inhibited the growth of A549 cellsin the mouse liver, showing a role for Plk1 in lung cancerliver metastasis (40). These in vivo studies have confirmedthe anticancer effects of Plk1 depletion in vitro and providea rationale for pharmacologic exploration of Plk1 as anovel target.

Preclinical Studies of Plk1 Inhibitors

Plk1 contains a highly conserved N-terminal proteinkinase domain and a C-terminal "Polo box domain"(PBD) that is required for Plk1 to dock to its intracellulartargets (24, 41). These functionally critical sites havebeen explored in the development of drugs targetingPlk1 (42).

Inhibitors targeting the Polo box domain of Plk1Given the large number of substrates and cellular func-

tions of Plk1, attempts have been made to improve theselectivity of Plk1 inhibitory drugs by targeting the PBD ofPlk1. This has the potential benefit of a higher selectivity forPlk1 over the related Plks and may allow for interference insome, but not all, cellular functions of Plk1. Selective small

drug-like inhibitors of the PBD of Plk1 are purpurogallin(43), poloxin, and thymoquinone (44).

BI 2536. The compound is a dihydropteridinonederivative that shows more than 10,000-fold higherinhibitory potency (IC50 ¼ 0.83 nmol/L) toward Plk1than the majority of 63 other protein kinases tested(45). BI 2536 inhibited proliferation of all 32 examinedhuman cancer cell lines (EC50 ¼ 2–25 nmol/L; ref. 45).Comparable inhibition was also observed in nontrans-formed, immortalized cells, indicating that BI 2536inhibition affects all proliferating cells. Cells treatedwith BI 2536 are blocked in G2–M and form mono-polar mitotic spindles, a phenotype similar to thatinduced by siRNA against Plk1, suggesting that Plk1is the main cellular target (45). BI 2536 (40–50 mg/kg)administered i.v. twice a week inhibited the growthof mouse tumor xenografts, including NSCLC (A549and NCI-H460), and induced accumulation of "Polo-arrested" tumor cells followed by apoptosis, showing BI2536–mediated mitotic arrest and tumor cell deathin vivo (45).

Although cellular sensitivity to BI 2536 was indepen-dent of tumor suppressor or oncogene mutations (45), arecent report has shown that tumor cells and xenograftswith K-ras mutations were more sensitive to BI 2536than isogenic wild-type cells (37). These findings sup-port the hypothesis that Ras-mutant cells may be moredependent on Plk1 for mitotic progression (37). Specific

Prophase

Interphase

Centrosomematuration

Spindleassembly

Cdk1activation Mitotic

entryPlk1

Anaphase

Telophase

Cytokinesis

Cytokinesis

Cdk1inactivation

APC/Cregulation

MetaphasePrometaphase

Figure 2. Functions of Plk1 during mitosis. Adapted by permission from Macmillan Publishers Ltd. (24). Copyright � 2004.

Plk1 Inhibitors in Treatment of NSCLC

www.aacrjournals.org Clin Cancer Res; 17(20) October 15, 2011 6461

on November 8, 2018. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

pharmacologic agents targeting Plk1 may selectively killRas-mutant cells by exacerbating their mitotic stress andthus benefit patients who bear tumors with activated Rasoncogenes (37).

Volasertib (BI 6727). This dihydropteridinone deriva-tive targets Plk1 with selectivity and efficiency (IC50 ¼ 0.87nmol/L) similar to that of BI 2536 (46). It is also a potentinhibitor of cellular proliferation (EC50 ¼ 11–37 nmol/L)that induces cellular effects (G2–M cell-cycle block, phos-pho-histone H3–positive staining, and monopolar mitoticspindles; ref. 46). Volasertib showed antitumor activity inmouse xenograft models of human cancers, includingNSCLC (NCI-H460), using i.v. or oral administrationschedules of 70 mg/kg/wk or 10 mg/kg/d (46). Notably,it also inhibited taxane-resistant xenograft tumors. Thehigh volume of distribution of volasertib—indicative ofgood tissue penetration, a long terminal half-life in miceand rats, and sustained exposure in tumor tissue (46)—wasan additional factor in the selection of volasertib forpriority clinical development.

GSK461364. This imidazotriazine, ATP-competitive in-hibitor, exhibits more than 1,000-fold higher potency to-ward Plk1 than the majority of 48 other protein kinasestested (47). Cellular treatment withGSK461364 showed notonly signs typical of Plk1 inhibition (mitotic arrest withmonopolar or apolar spindles) but also micronucleationindicative of mitotic slippage, resulting in tetraploidy (47).The extent of mitotic arrest was concentration dependent,and high concentrations (>300 nmol/L) cause a G2 delay.GSK461364 inhibited the growth of a majority of 74 testedcancer cell lines with 89% of the EC50 values lower than 100nmol/L and induced cytotoxic aswell as cytostatic effects thatwere cell line dependent. Intraperitoneal administration ofGSK461364 at various doses induced partial regressions in 2of 3 mouse tumor xenograft models of NSCLC (47).

HMN-176. This stilbene derivative is an active form ofan oral prodrug, HMN-214. HMN-176 disrupts mitoticspindle assembly, inhibits centrosome-dependent micro-tubule nucleation, and is considered an "anticentrosome"agent (48). It does not directly inhibit the enzymaticactivity of Plk1 but rather affects subcellular distributionof Plk1 (49). HMN-176 showed potent cytotoxicity against22 human tumor cell lines (mean IC50 ¼ 118 nmol/L; ref.50). HMN-214 reduced the growth of primary NSCLCtumors in human tumor cloning assay (51) and severaltumor xenografts in mice, including NSCLC (50).

ON 01910.Na. This benzyl styryl sulfone analogue isan ATP-noncompetitive, multitargeted inhibitor of severaltyrosine kinases and cyclin-dependent kinase 1 (Cdk1;IC50¼ 18–260 nmol/L). It is reported to have a particularlystrong potency (IC50 ¼ 9–10 nmol/L) toward Plk1 (52),although results from another report suggest that Plk1 maynot be the primary target (45). ON 01910.Na inhibitedproliferation of more than 100 cell lines, including thoseresistant to paclitaxel, with EC50 in the range of 50 to 250nmol/L. It also showed efficacy with minimal toxicity as amonotherapy or in combinations with cytotoxic drugs inxenograft models (52).

Preclinical studies with Plk1 inhibitors showed thefeasibility and selectivity of a pharmacologic approachto targeting Plk1 as well as the anticipated mode ofaction of inhibitors and their potent antiproliferativeeffects in a majority of cancer cell lines and tumor xeno-grafts. These results led to initiation of clinical studiesand further development of Plk1 inhibitors as anti-cancer drugs.

Clinical Studies of Plk1 Inhibitors

BI 2536A phase I first-in-humans study of BI 2536 was con-

ducted in 40 patients with refractory or metastatic solidtumors previously treated with a median of 3 chemother-apy regimens (53). Most were patients with colorectalcancer (42.5%); the inclusion of patients with NSCLCwas not reported. BI 2536 was administered i.v. once every3 weeks following a dose-escalation design (25–250 mg).Reversible neutropenia with infection was the dose-limit-ing toxicity (DLT) that defined 200 mg as the maximumtolerated dose (MTD). Patients treated at the MTD (67.5%of all treated patients) developed grade 3/4 adverse events(Common Terminology Criteria for Adverse Events, ver-sion 3.0) including neutropenia (55.6%) and leukopenia(44.4%; ref. 53). Similar results were obtained in anotherphase I study of 70 patients with advanced solid cancerstreated with BI 2536 on days 1 and 8 of a 3-week treatmentcourse with MTD of 100 mg (54). Of patients treated with200 mg or more in the once-per-3-weeks regimen, 23%showed stable disease for more than 3 months (53). Thiseffect was more common with patients experiencing neu-tropenia (53).

Two clinical trials have investigated BI 2536 in NSCLC: aphase I study in combination with the antifolate agentpemetrexed (55) and a phase II study of BI 2536 mono-therapy (56). In the combination trial, 33 patients withrefractory or metastatic NSCLC who relapsed after first-lineplatinum-based therapy were treated with 500 mg/m2

pemetrexed and escalating BI 2536 (100–325 mg) by i.v.on day 1 every 3 weeks. Grade 4 neutropenia and grade 3rash were DLTs that established 300 mg as the MTD.Reversible grade 3/4 neutropenia occurred in 18%. Themost common drug-related adverse events were mild tomoderate nausea (36%), rash (33%), anorexia (27%),stomatitis (24%), and pruritus (24%). Two patients hada partial response. BI 2536 and pemetrexed exposures werenot affected by coadministration (55).

In the monotherapy phase II trial, 95 patients withadvanced or metastatic NSCLC who had progressed after,or failed, first- or second-line therapy received either asingle i.v. dose of 200 mg on day 1 or a single i.v. doseof 50 to 60 mg on days 1, 2, and 3 of each treatment course(56). Neutropenia was the main grade 4 adverse event, andmost of the reported adverse events transiently affected thehematopoietic system. Twelve patients had grade 3/4 com-plications associated with neutropenia, including sepsis orfebrile neutropenia (1 patient each) and fever or infection

Medema et al.

Clin Cancer Res; 17(20) October 15, 2011 Clinical Cancer Research6462

on November 8, 2018. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

associated with low neutrophil counts (10 patients). Fourpatients (4.2%) had an investigator-assessed partial re-sponse (56).

VolasertibA phase I first-in-humans study of volasertib was con-

ducted in 65 patients with advanced solid tumors,including 10 with NSCLC. Volasertib was administeredi.v. once every 3 weeks following a dose-escalation design(12–450 mg; ref. 57). The study reported neutropenia,thrombocytopenia, and febrile neutropenia as DLTs andan MTD of 400 mg. Reversible and noncumulative hema-totoxicity was the major adverse event. Three partialresponses were observed in patients with urothelial andovarian cancer and melanoma, and stable disease wasreported in 48% (57). The study reported favorable phar-macokinetic parameters (linearity in the therapeutic doserange, large volume of drug distribution, and a half-lifearound 110 hours). A phase I trial of volasertib admin-istered on days 1 and 8 every 21 days is currently ongoing(NCT00969553; ref. 58). These results prompted theinitiation of multiple phase II studies, including a ran-domized trial of volasertib as monotherapy or in com-bination with pemetrexed in second-line NSCLC(NCT00824408).The identification of neutropenia as the dominant

adverse event in studies with BI 2536 and volasertibindicates a mechanism-based inhibition of bone marrowcellular proliferation. In addition, the absence of anyreported cumulative or high-grade mucosal side effectssuggests the feasibility of prolonged BI 2536 or volasertibadministration.Although the clinical development of BI 2536 has

been halted, volasertib is currently being investigatedas a single agent in phase II trials in several differentindications (NCT01023958 and NCT01121406) and as acombination with platinum (NCT00969761), peme-trexed (NCT00824408), or other targeted therapies(NCT01022853 and NCT01206816).

GSK461364A phase I first-in-humans study of GSK461364 was

conducted in 27 patients with advanced solid tumors(59). The agent was administered i.v. following 2 sche-dules with different dosing (50–225 mg on days 1, 8, and15 or 25–100 mg on days 1, 2, 8, 9, 15, and 16) on a 28-day cycle. DLTs included grade 4 neutropenia, sepsis, orpulmonary embolism. Common adverse events includedhematologic toxicity, phlebitis, nausea, and fatigue. Sta-ble disease for more than 5 months was observed in 2patients. GSK461364 increased phospho-histone H3levels in circulating tumor cells, indicating antimitoticeffects of the treatment. The recommended phase II dosefor GSK461364 was 225 mg on days 1, 8, and 15 in a 28-day cycle. Because of the high incidence (20%) of venousthromboembolism, GSK461364 should involve coad-ministration of prophylactic anticoagulation for furtherclinical evaluation (59).

HMN-214Two phase I studies of HMN-214 have been reported

in patients with solid tumors (49, 60). DLTs of pro-longed neutropenia, febrile neutropenia, neutropenicsepsis, electrolyte disturbance, neuropathy, and myalgiawere observed at doses of 24 to 48 mg/m2 for 5 conse-cutive days every 4 weeks. MTD was established at therange of 18 to 30 mg/m2, based on previous patienttreatment load (60).

In another study, 33 patients with advanced cancerreceived oral HMN-214 (3.0–9.9 mg/m2/d) in a continu-ous 21-day dosing schedule every 28 days (49). DLTscomprised severe myalgia and/or bone pain syndromeand hyperglycemia. MTD was 8.0 mg/m2/d. The studyreported 24% of patients with stable disease and indicatedthat future development of HMN-214 would focus onpatient populations with high tumor expression of Plk1.

ON 01910.NaA phase I first-in-humans study of ON 01910.Na was

conducted in 20 patients with advanced solid tumors(none with NSCLC; ref. 61). The agent was administeredi.v. at 80 to 4,370mg by accelerated titration design on days1, 4, 8, 11, 15, and 18 in 28-day cycles. Grade 3 abdominalpain was reported as a DLT at an MTD of 3,120 mg. Themost frequently reported adverse events (grade �3) duringthe first treatment cycle comprised skeletal, abdominal,and tumor pain; nausea and vomiting; urge to defecate;flatulence; and fatigue. Mild hematologic toxicity—anemiaand lymphopenia—was reported in approximately 10% ofpatients, and this contrasts with other Plk1-targeting com-pounds. One partial response was observed in a patientwith ovarian cancer (61).

Although early preclinical studies identified Plk1 asa main target of ON 01910.Na (52), its mode of actionremains unknown (45). Current clinical studies ofthis agent concentrate on ovarian cancer and hemato-logic malignancies, with some ongoing phase I studiesinvestigating ON 01910.Na as monotherapy or in com-bination with chemotherapies in patients with solidtumors. Its development in NSCLC in the near futureis unlikely.

Of the Plk inhibitors discussed here, we believe thatvolasertib has several possible advantages. These includeits high volume of distribution, long terminal half-life, andearly signs of antitumor efficacy in single-agent studies (57,58). However, because all of these agents are at an earlydevelopment stage (Table 1), further clinical data arerequired to determine their potential.

The future investigation of Plk inhibitors should focuson combinations with chemotherapies. First, the tumorresponse to Plk inhibitors as a single agent appearslimited. Second, biomarkers predictive of response toPlk inhibitors as a single agent have not been availableso far. Third, the main side effect of Plk inhibitors isreversible, uncomplicated myelosuppression. Therefore,it is possible to combine a Plk inhibitor with otherchemotherapeutic agent(s).

Plk1 Inhibitors in Treatment of NSCLC

www.aacrjournals.org Clin Cancer Res; 17(20) October 15, 2011 6463

on November 8, 2018. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Conclusions

Monotherapy with Plk1 inhibitors, as demonstrated inphase I/II studies, has shown manageable, noncumulative,and predominantly hematologic toxicity that correlateswith the predicted mode of action of agents. Preliminarysigns of antitumor activity have been shown, althoughmuch work needs to be done to optimize treatment regi-mens, identify agents with potentially synergistic activity,and define markers of tumor response and patient char-acteristics that predict therapeutic success. Future clinicalstudies should include the detection of pharmacodynamicbiomarkers, such as phospho-histone H3, to validate Plk1as the tumor target. In the first-in-humans phase I study ofGSK461364 in advanced solid tumors, Plk1 inhibition intumor cells was confirmed on the basis of the analysis ofphospho-histone H3 expression in circulating tumor cells(59). Other promising biomarker assays for Plk1 inhibitionin vivohave been developed, such as an immunohistochem-ical approach that measures serine 46-phosphorylation ofthe translational controlled tumor protein by Plk1 (62)and an ELISA-based assay that measures phosphorylationof the PBD-binding protein 1 (PBIP1, also known as MLF1or CENP-U) by Plk1 (63). Patient stratification based onPlk1 expression should be considered in future trials;NSCLC patients with poor prognosis whose tumors express

high levels of Plk1 may be good candidates for Plk1-target-ing therapy. Preclinical studies have shown that p53 and/orRas-mutant cells may be more sensitive to Plk1 inhibitors.This hypothesis, relevant for several solid tumors (e.g.,ovarian cancer and urothelial carcinoma), but particularlyfor NSCLC, also needs to be clinically validated.

Disclosure of Potential Conflicts of Interest

The authors received no compensation related to the developmentof the manuscript. Chia-Chi Lin and James Chih-Hsin Yang are onthe Advisory Board of Boehringer Ingelheim and did not receive anyhonorarium.

Authors' Contributions

The authors meet criteria for authorship as recommended by the Inter-national Committee of Medical Journal Editors, were fully responsible forall content and editorial decisions, and were involved in all stages ofmanuscript development.

Grant Support

Writing and editorial assistance was provided by Ogilvy Healthworld,which was contracted by Boehringer Ingelheim Pharma GmbH & Co., KGfor these services.

Received February 27, 2011; revised June 14, 2011; accepted July 14,2011; published online October 14, 2011.

References1. Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics,

2009. CA Cancer J Clin 2009;59:225–49.2. Stinchcombe TE, Socinski MA. Current treatments for advanced

stage non-small cell lung cancer. Proc Am Thorac Soc 2009;6:233–41.

3. Ramalingam S, Belani C. Systemic chemotherapy for advanced non-small cell lung cancer: recent advances and future directions. On-cologist 2008;13 Suppl 1:5–13.

4. World Health Organisation. Lung cancer—the big one 2010 [cited 2010Oct 26]. Available from: http://www.who.int/tobacco/research/cancer/en/

Table 1. Plk inhibitors in clinical development

Compound Company Mode of action DLT Efficacy Status References

BI 2536 BoehringerIngelheim

Plk1 inhibitor Neutropenia PR in phase II Phase II(halted)

53–56

Volasertib(BI 6727)

BoehringerIngelheim

Plk1 inhibitor Neutropenia,thrombocytopenia

DurablePR in phase I

Phase I/II 57, 58

GSK461364 GlaxoSmithKline Plk1 inhibitor Neutropenia,thrombocytopenia,pulmonary emboli

ProlongedSD in phase I

Phase I 59

HMN-214 NipponShinyakuCo. Ltd.

Plk1 interference,not direct inhibitor

Muscle/bone pain,hyperglycemia

ProlongedSD in phase I

Phase I 60

ON 01910.Na OnconovaTherapeutics, Inc.

Plk1 inhibitor,Cdk inhibitor,ERK inhibitor,PI3K inhibitor,AKT inhibitor

Abdominal pain ProlongedSD in phase I

Phase I 61

Abbreviations: PR, partial response; SD, stable disease; ERK extracellular signal-regulated kinase; PI3K, phosphoinositide 3-kinase.

Medema et al.

Clin Cancer Res; 17(20) October 15, 2011 Clinical Cancer Research6464

on November 8, 2018. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

5. Schiller JH, Harrington D, Belani CP, Langer C, Sandler A, Krook J,et al. Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N Engl J Med 2002;346:92–8.

6. Ardizzoni A, Boni L, Tiseo M, Fossella FV, Schiller JH, Paesmans M,et al. Cisplatin- versus carboplatin-based chemotherapy in first-linetreatment of advanced non-small-cell lung cancer: an individualpatient data meta-analysis. J Natl Cancer Inst 2007;99:847–57.

7. Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati A, et al.Paclitaxel-carboplatin alone or with bevacizumab for non-small-celllung cancer. N Engl J Med 2006;355:2542–50.

8. Reck M, von Pawel J, Zatloukal P, Ramlau R, Gorbounova V, Hirsh V,et al. Phase III trial of cisplatin plus gemcitabine with either placebo orbevacizumab as first-line therapy for nonsquamous non-small-celllung cancer: AVAil. J Clin Oncol 2009;27:1227–34.

9. Shepherd FA, Dancey J, Ramlau R, Mattson K, Gralla R, O’RourkeM, et al. Prospective randomized trial of docetaxel versus bestsupportive care in patients with non-small-cell lung cancer pre-viously treated with platinum-based chemotherapy. J Clin Oncol2000;18:2095–103.

10. Fossella FV, DeVore R, Kerr RN, Crawford J, Natale RR, Dunphy F,et al. Randomized phase III trial of docetaxel versus vinorelbine orifosfamide in patients with advanced non-small-cell lung cancerpreviously treated with platinum-containing chemotherapy regimens.The TAX 320 Non-Small Cell Lung Cancer Study Group. J Clin Oncol2000;18:2354–62.

11. Hanna N, Shepherd FA, Fossella FV, Pereira JR, De Marinis F, vonPawel J, et al. Randomized phase III trial of pemetrexed versusdocetaxel in patients with non-small-cell lung cancer previouslytreated with chemotherapy. J Clin Oncol 2004;22:1589–97.

12. Shepherd FA, Rodrigues Pereira J, Ciuleanu T, Tan EH, Hirsh V,Thongprasert S, et al. Erlotinib in previously treated non-small-celllung cancer. N Engl J Med 2005;353:123–32.

13. Velez M, Belalcazar A, Domingo G, Blaya M, Raez LE, Santos ES.Accelerated second-line or maintenance chemotherapy versus treat-ment at disease progression in NSCLC. Expert Rev Anticancer Ther2010;10:549–57.

14. Massarelli E, Andre F, Liu DD, Lee JJ, Wolf M, Fandi A, et al. Aretrospective analysis of the outcome of patients who have receivedtwo prior chemotherapy regimens including platinum and docetaxelfor recurrent non-small-cell lung cancer. Lung Cancer 2003;39:55–61.

15. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA,Brannigan BW, et al. Activating mutations in the epidermal growthfactor receptor underlying responsiveness of non-small-cell lungcancer to gefitinib. N Engl J Med 2004;350:2129–39.

16. Cappuzzo F, Hirsch FR, Rossi E, Bartolini S, Ceresoli GL, Bemis L,et al. Epidermal growth factor receptor gene and protein and gefitinibsensitivity in non-small-cell lung cancer. J Natl Cancer Inst2005;97:643–55.

17. Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, et al.Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. NEngl J Med 2009;361:947–57.

18. Rosell R, Moran T, Queralt C, Porta R, Cardenal F, Camps C, et al.Screening for epidermal growth factor receptor mutations in lungcancer. N Engl J Med 2009;361:958–67.

19. Mitsudomi T, Morita S, Yatabe Y, Negoro S, Okamoto I, Tsurutani J,et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growthfactor receptor (WJTOG3405): an open label, randomised phase 3trial. Lancet Oncol 2010;11:121–8.

20. Yang CH, Yu CJ, Shih JY, Chang YC, Hu FC, Tsai MC, et al. SpecificEGFR mutations predict treatment outcome of stage IIIB/IV patientswith chemotherapy-naive non-small-cell lung cancer receiving first-line gefitinib monotherapy. J Clin Oncol 2008;26:2745–53.

21. Sequist LV, Lynch TJ. EGFR tyrosine kinase inhibitors in lung cancer:an evolving story. Annu Rev Med 2008;59:429–42.

22. Jackson JR, Patrick DR, Dar MM, Huang PS. Targeted anti-mitotictherapies: can we improve on tubulin agents? Nat Rev Cancer2007;7:107–17.

23. Jordan MA, Kamath K. How do microtubule-targeted drugs work? Anoverview. Curr Cancer Drug Targets 2007;7:730–42.

24. Barr FA, Sillje HH, Nigg EA. Polo-like kinases and the orchestration ofcell division. Nat Rev Mol Cell Biol 2004;5:429–40.

25. Andrysik Z, Bernstein WZ, Deng L, Myer DL, Li YQ, Tischfield JA,et al. The novel mouse Polo-like kinase 5 responds to DNAdamage and localizes in the nucleolus. Nucleic Acids Res 2010;38:2931–43.

26. Strebhardt K, Ullrich A. Targeting polo-like kinase 1 for cancer ther-apy. Nat Rev Cancer 2006;6:321–30.

27. Yuan J, Horlin A, Hock B, Stutte HJ, R€ubsamen-Waigmann H, Streb-hardt K. Polo-like kinase, a novel marker for cellular proliferation. Am JPathol 1997;150:1165–72.

28. Wolf G, Elez R, Doermer A, Holtrich U, Ackermann H, Stutte HJ, et al.Prognostic significance of polo-like kinase (PLK) expression in non-small cell lung cancer. Oncogene 1997;14:543–9.

29. Renner AG, Dos Santos C, Recher C, Bailly C, Cr�eancier L, KruczynskiA, et al. Polo-like kinase 1 is overexpressed in acute myeloid leukemiaand its inhibition preferentially targets the proliferation of leukemiccells. Blood 2009;114:659–62.

30. Eckerdt F, Yuan J, Strebhardt K. Polo-like kinases and oncogenesis.Oncogene 2005;24:267–76.

31. Takai N, Hamanaka R, Yoshimatsu J, Miyakawa I. Polo-like kinases(Plks) and cancer. Oncogene 2005;24:287–91.

32. Kneisel L, Strebhardt K, Bernd A, Wolter M, Binder A, Kaufmann R.Expression of polo-like kinase (PLK1) in thin melanomas: a novelmarker of metastatic disease. J Cutan Pathol 2002;29:354–8.

33. Liu X, Erikson RL. Polo-like kinase (Plk)1 depletion induces apo-ptosis in cancer cells. Proc Natl Acad Sci U S A 2003;100:5789–94.

34. Spankuch-Schmitt B, Bereiter-Hahn J, Kaufmann M, Strebhardt K.Effect of RNA silencing of polo-like kinase-1 (PLK1) on apoptosis andspindle formation in human cancer cells. J Natl Cancer Inst2002;94:1863–77.

35. Liu X, Lei M, Erikson RL. Normal cells, but not cancer cells, survivesevere Plk1 depletion. Mol Cell Biol 2006;26:2093–108.

36. Guan R, Tapang P, Leverson JD, Albert D, Giranda VL, Luo Y. Smallinterfering RNA-mediated Polo-like kinase 1 depletion preferentiallyreduces the survival of p53-defective, oncogenic transformed cellsand inhibits tumor growth in animals. Cancer Res 2005;65:2698–704.

37. Luo J, Emanuele MJ, Li D, Creighton CJ, Schlabach MR, WestbrookTF, et al. A genome-wide RNAi screen identifies multiple syntheticlethal interactions with the Ras oncogene. Cell 2009;137:835–48.

38. Riely GJ, Marks J, Pao W. KRAS mutations in non-small cell lungcancer. Proc Am Thorac Soc 2009;6:201–5.

39. Spankuch B, Matthess Y, Knecht R, Zimmer B, Kaufmann M, Streb-hardt K. Cancer inhibition in nude mice after systemic application ofU6 promoter-driven short hairpin RNAs against PLK1. J Natl CancerInst 2004;96:862–72.

40. Kawata E, Ashihara E, Kimura S, Takenaka K, Sato K, Tanaka R, et al.Administration of PLK-1 small interfering RNA with atelocollagenprevents the growth of liver metastases of lung cancer. Mol CancerTher 2008;7:2904–12.

41. Elia AE, Cantley LC, Yaffe MB. Proteomic screen finds pSer/pThr-binding domain localizing Plk1 to mitotic substrates. Science2003;299:1228–31.

42. Yap TA, Molife LR, Blagden SP, de Bono J. Targeting cell cyclekinases and kinesins in anticancer drug development. Expert OpinDrug Discov 2007;2:539–60.

43. Watanabe N, Sekine T, Takagi M, Iwasaki J, Imamoto N, Kawasaki H,et al. Deficiency in chromosome congression by the inhibition of PLK1polo box domain-dependent recognition. J Biol Chem 2009;284:2344–53.

44. Reindl W, Yuan J, Kr€amer A, Strebhardt K, Berg T. Inhibition of polo-like kinase 1 by blocking polo-box domain-dependent protein-proteininteractions. Chem Biol 2008;15:459–66.

45. Steegmaier M, Hoffmann M, Baum A, L�en�art P, Petronczki M, Krss�akM, et al. BI 2536, a potent and selective inhibitor of polo-like kinase 1,inhibits tumor growth in vivo. Curr Biol 2007;17:316–22.

46. Rudolph D, Steegmaier M, Hoffmann M, Grauert M, Baum A, Quant J,et al. BI 6727, a Polo-like kinase inhibitor with improved

Plk1 Inhibitors in Treatment of NSCLC

www.aacrjournals.org Clin Cancer Res; 17(20) October 15, 2011 6465

on November 8, 2018. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

pharmacokinetic profile and broad antitumor activity. Clin Cancer Res2009;15:3094–102.

47. Gilmartin AG, BleamMR, Richter MC, Erskine SG, Kruger RG,MaddenL, et al. Distinct concentration-dependent effects of the polo-likekinase 1-specific inhibitor GSK461364A, including differential effecton apoptosis. Cancer Res 2009;69:6969–77.

48. DiMaio MA, Mikhailov A, Rieder CL, Von Hoff DD, Palazzo RE. Thesmall organic compound HMN-176 delays satisfaction of the spindleassembly checkpoint by inhibiting centrosome-dependent microtu-bule nucleation. Mol Cancer Ther 2009;8:592–601.

49. Garland LL, Taylor C, Pilkington DL, Cohen JL, Von Hoff DD. A phase Ipharmacokinetic study of HMN-214, a novel oral stilbene derivativewith polo-like kinase-1-interacting properties, in patients with ad-vanced solid tumors. Clin Cancer Res 2006;12:5182–9.

50. Takagi M, Honmura T, Watanabe S, Yamaguchi R, Nogawa M,Nishimura I, et al. In vivo antitumor activity of a novel sulfonamide,HMN-214, against human tumor xenografts in mice and the spectrumof cytotoxicity of its active metabolite, HMN-176. Invest New Drugs2003;21:387–99.

51. Medina-Gundrum L, Cerna C, Gomez L, Izbicka E. Investigation ofHMN-176 anticancer activity in human tumor specimens in vitro andthe effects of HMN-176 on differential gene expression. Invest NewDrugs 2005;23:3–9.

52. Gumireddy K, Reddy MV, Cosenza SC, Boominathan R, Baker SJ,Papathi N, et al. ON01910, a non-ATP-competitive small moleculeinhibitor of Plk1, is a potent anticancer agent. Cancer Cell 2005;7:275–86.

53. Mross K, Frost A, Steinbild S, Hedbom S, Rentschler J, Kaiser R, et al.Phase I dose escalation and pharmacokinetic study of BI 2536, anovel Polo-like kinase 1 inhibitor, in patients with advanced solidtumors. J Clin Oncol 2008;26:5511–7.

54. Hofheinz R, Al-Batran S, Hochhaus A, J€ager E, Reichardt VL, FritschH, et al. An open-label, phase I study of the polo-like kinase-1 inhibitorBI 2536, in patients with advanced solid tumors. Clin Cancer Res2010;16:4666–74.

55. Ellis PM, Chu QS, Leighl NB, Laurie SA, Trommeshauser D, Hanft G,et al. A phase I dose escalation trial of BI 2536, a novel Plk1 inhibitor,

with standard dose pemetrexed in previously treated advanced ormetastatic non-small cell lung cancer (NSCLC). J Clin Oncol 2008;26Suppl:abstr 8115.

56. Sebastian M, Reck M, Waller CF, Kortsik C, Frickhofen N, Schuler M,et al. The efficacy and safety of BI 2536, a novel Plk-1 inhibitor, inpatients with stage IIIB/IV non-small cell lung cancer who had re-lapsed after, or failed, chemotherapy: results from an open-label,randomized phase II clinical trial. J Thorac Oncol 2010;5:1060–7.

57. Gil T, Sch€offski P, Awada A, Dumez H, Bartholomeus S, Selleslach J,et al. Final analysis of a phase I single dose-escalation study of thenovel polo-like kinase 1 inhibitor BI 6727 in patients with advancedsolid tumors. J Clin Oncol 2010;28 Suppl 15:abstr 3061.

58. Lin C-C, Su W-C, Yen C-J, Cheng A, Lu Y, Hsu C, et al. Phase I doseescalation study of the polo-like kinase 1 inhibitor volasertib [BI 6727]with two different dosing schedules in patients with advanced solidmalignancies. J Clin Oncol 2011;29 Suppl:abstr 3046.

59. Olmos D, Barker D, Sharma R, Brunetto AT, Yap TA, Taegtmeyer AB,et al. Phase I study of GSK461364, a specific and competitive Polo-like kinase 1 (PLK1) inhibitor in patients with advanced solid malig-nancies. Clin Cancer Res 2011;17:3420–30.

60. Patnaik A, Forero L, Goetz A, Tolcher AW, Beeram M, De Bono JS,et al. HMN-214, a novel oral antimicrotubular agent and inhibitor ofpolo-like- and cyclin-dependent kinases: clinical, pharmacokinetic(PK) and pharmacodynamic (PD) relationships observed in a phaseI trial of a daily � 5 schedule every 28 days. J Clin Oncol 2003;22Suppl:abstr 514.

61. Jimeno A, Li J, Messersmith WA, Rudek MA, Maniar M, Hidalgo M,et al. Phase I study of ON 01910.Na, a novel modulator of the Polo-likekinase 1 pathway, in adult patients with solid tumors. J Clin Oncol2008;26:5504–10.

62. Cucchi U, Gianellini LM, De Ponti A, Sola F, Alzani R, Patton V, et al.Phosphorylation of TCTP as a marker for polo-like kinase-1 activity invivo. Anticancer Res 2010;30:4973–85.

63. Park J-E, Lee L, Park J, Knecht R, Strebhardt K, Yuspa SH, et al. Directquantification of polo-like kinase 1 activity in cells and tissues using ahighly sensitive and specific ELISA assay. Proc Natl Acad Sci U S A2009;106:1725–30.

Medema et al.

Clin Cancer Res; 17(20) October 15, 2011 Clinical Cancer Research6466

on November 8, 2018. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

2011;17:6459-6466. Clin Cancer Res   René H. Medema, Chia-Chi Lin and James Chih-Hsin Yang  Therapy, with a Focus on NSCLCPolo-like Kinase 1 Inhibitors and Their Potential Role in Anticancer

  Updated version

  http://clincancerres.aacrjournals.org/content/17/20/6459

Access the most recent version of this article at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/17/20/6459.full#ref-list-1

This article cites 58 articles, 23 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/17/20/6459.full#related-urls

This article has been cited by 3 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected]

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

  Permissions

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

.http://clincancerres.aacrjournals.org/content/17/20/6459To request permission to re-use all or part of this article, use this link

on November 8, 2018. © 2011 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from