+ All Categories
Home > Documents > Palbociclib (PD 0332991): targeting the cell cycle machinery in breast cancer

Palbociclib (PD 0332991): targeting the cell cycle machinery in breast cancer

Date post: 23-Dec-2016
Category:
Upload: dino
View: 214 times
Download: 0 times
Share this document with a friend
14
1. Introduction 2. Palbociclib preclinical studies 3. Palbociclib clinical development 4. Conclusions 5. Expert opinion Drug Evaluation Palbociclib (PD 0332991): targeting the cell cycle machinery in breast cancer Andrea Rocca , Alberto Farolfi, Sara Bravaccini, Alessio Schirone & Dino Amadori Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Department of Medical Oncology, Meldola, Italy Introduction: The cyclin D-cyclin-dependent kinases 4 and 6 (CDK4/6)-retino- blastoma (Rb) pathway, governing the cell cycle restriction point, is frequently altered in breast cancer and is a potentially relevant target for anticancer therapy. Palbociclib (PD 0332991), a potent and selective inhibitor of CDK4 and CDK6, inhibits proliferation of several Rb-positive cancer cell lines and xenograft models. Areas covered: The basic features and abnormalities of the cell cycle in breast cancer are described, along with their involvement in estrogen signaling and endocrine resistance. The pharmacological features of palboci- clib, its activity in preclinical models of breast cancer and the potential determinants of response are then illustrated, and its clinical development in breast cancer described. A literature search on the topic was conducted through PubMed and the proceedings of the main cancer congresses of recent years. Expert opinion: The combination of palbociclib with endocrine agents is a very promising treatment and Phase III clinical trials are ongoing to confirm its efficacy. Further, potentially useful combinations are those with drugs targeting mitogenic signaling pathways, such as HER2- and PI3K-inhibitors. Combination with chemotherapy seems more problematic, as antagonism has been reported in preclinical models. The identification of predictive factors, already explored in preclinical studies, must be further refined and validated in clinical trials. Keywords: breast cancer, cell cycle, cyclin-dependent kinase inhibitors, endocrine resistance, palbociclib Expert Opin. Pharmacother. (2014) 15(3):407-420 1. Introduction Breast cancer is the most frequent cancer among women, both in developed and developing regions, with an estimated 1.38 million new cancer cases diagnosed in 2008 (23% of all cancers) [1]. About 200 women per 100,000 are diagnosed with breast cancer every year in the USA, two-thirds of whom have estrogen receptor (ER)-positive tumors. ER-positive cancers are expected to increase in the near term (2009 through 2016), whereas ER-negative tumors show a more encouraging trend, with a steady decrease of nearly 2% per year [2]. Treatment of metastatic breast cancer is palliative in nature, and advances in our knowledge of the biology of this disease are fundamental to the identification of new targets and the development of new active treatments. Sustained cell proliferation is a hallmark of cancer [3], and cell cycle control is altered in virtually all cancer cells, often as a result of abnormal oncogene products or deletion or inactivation of tumor suppressor genes. Many anticancer agents aim 10.1517/14656566.2014.870555 © 2014 Informa UK, Ltd. ISSN 1465-6566, e-ISSN 1744-7666 407 All rights reserved: reproduction in whole or in part not permitted Expert Opin. Pharmacother. Downloaded from informahealthcare.com by University of Sydney on 01/27/14 For personal use only.
Transcript

1. Introduction

2. Palbociclib preclinical studies

3. Palbociclib clinical

development

4. Conclusions

5. Expert opinion

Drug Evaluation

Palbociclib (PD 0332991): targetingthe cell cycle machinery in breastcancerAndrea Rocca†, Alberto Farolfi, Sara Bravaccini, Alessio Schirone &Dino Amadori†Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS,

Department of Medical Oncology, Meldola, Italy

Introduction: The cyclin D-cyclin-dependent kinases 4 and 6 (CDK4/6)-retino-

blastoma (Rb) pathway, governing the cell cycle restriction point, is frequently

altered in breast cancer and is a potentially relevant target for anticancer

therapy. Palbociclib (PD 0332991), a potent and selective inhibitor of

CDK4 and CDK6, inhibits proliferation of several Rb-positive cancer cell lines

and xenograft models.

Areas covered: The basic features and abnormalities of the cell cycle in

breast cancer are described, along with their involvement in estrogen

signaling and endocrine resistance. The pharmacological features of palboci-

clib, its activity in preclinical models of breast cancer and the potential

determinants of response are then illustrated, and its clinical development

in breast cancer described. A literature search on the topic was conducted

through PubMed and the proceedings of the main cancer congresses of recent

years.

Expert opinion: The combination of palbociclib with endocrine agents is a

very promising treatment and Phase III clinical trials are ongoing to confirm

its efficacy. Further, potentially useful combinations are those with drugs

targeting mitogenic signaling pathways, such as HER2- and PI3K-inhibitors.

Combination with chemotherapy seems more problematic, as antagonism

has been reported in preclinical models. The identification of predictive

factors, already explored in preclinical studies, must be further refined and

validated in clinical trials.

Keywords: breast cancer, cell cycle, cyclin-dependent kinase inhibitors, endocrine resistance,

palbociclib

Expert Opin. Pharmacother. (2014) 15(3):407-420

1. Introduction

Breast cancer is the most frequent cancer among women, both in developed anddeveloping regions, with an estimated 1.38 million new cancer cases diagnosed in2008 (23% of all cancers) [1]. About 200 women per 100,000 are diagnosed withbreast cancer every year in the USA, two-thirds of whom have estrogen receptor(ER)-positive tumors. ER-positive cancers are expected to increase in the nearterm (2009 through 2016), whereas ER-negative tumors show a more encouragingtrend, with a steady decrease of nearly 2% per year [2].

Treatment of metastatic breast cancer is palliative in nature, and advances in ourknowledge of the biology of this disease are fundamental to the identification of newtargets and the development of new active treatments.

Sustained cell proliferation is a hallmark of cancer [3], and cell cycle control isaltered in virtually all cancer cells, often as a result of abnormal oncogene productsor deletion or inactivation of tumor suppressor genes. Many anticancer agents aim

10.1517/14656566.2014.870555 © 2014 Informa UK, Ltd. ISSN 1465-6566, e-ISSN 1744-7666 407All rights reserved: reproduction in whole or in part not permitted

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 01

/27/

14Fo

r pe

rson

al u

se o

nly.

at disrupting the cell proliferation process, often ultimatelytriggering apoptosis. Improved knowledge of the cell cyclemachinery has led to identifying different families of kinaseswith specific important roles in the cell cycle [4,5], which arepotential targets for cancer therapy [6-8]. Among these, thecyclin-dependent kinases (CDKs) have a prominent role.This review summarizes the basic features of the cell cycle

and its abnormalities in breast cancer, describes their involve-ment in estrogen signaling and endocrine resistance and illus-trates the preclinical and clinical activity of palbociclib (PD0332991) (Box 1), a potent and selective inhibitor of CDKs4 and 6.

1.1 Cell cycle machinery and controlA broad molecular apparatus executes the cell cycle, and itsregulation is likewise complex [4,5,9,10]. The mammalian cellcycle is composed of four phases: a first preparatory G1 (gap1) phase, the S phase during which DNA synthesis takesplace, a second gap G2 phase and the mitotic M phase, fol-lowed by cytokinesis and the formation of two daughter cells.Although newborn cells re-enter the cell cycle, at some pointduring late G1 phase (called restriction or R point) [11], thecell cycle regulatory machinery must make a decision on theultimate fate of the cell: continue cycling, or exit active prolif-eration and enter a quiescent (G0) state [12]. The progressionof the cell cycle during G1 phase is the result of a balancebetween growth factors and growth inhibitors present in theextracellular environment, and this balance will ultimatelyaffect the decision of the cell cycle regulatory machinery, aprevalence of growth factors portending active cycling and aprevalence of growth inhibitors inducing entry into G0. TheG1 phase is therefore the most critical site of cell cycle control:if the cell passes the restriction point, then the rest of the cyclewill proceed automatically until cell division, independently

of growth- and inhibitory-factors, unless major damage occursto the dividing cell [12].

Major players in cell cycle regulation are a family of serine/threonine kinases, called cyclin-dependent kinases or CDKs,which act in conjunction with their regulatory subunits calledcyclins. In human cells, at least 13 CDKs have been described(only some of which are involved in the cell cycle) that inter-act with at least 25 cyclins, each CDK binding only one or afew types of cyclins [9,10]. As their name indicates, most cyclinsare synthesized in a cyclic way, only at specific times duringcell cycle, while CDK levels are more stable. As the kinaseactivity of CDKs is present only when they bind their cyclinpartner, the levels of the different types of cyclins affect theCDK-cyclin complexes that are active during specific phasesof the cell cycle. During G1 phase, mitogenic growth factors,acting through different signaling cascades (such as the Ras/Raf/MAPK pathway, NF-kB pathway, Jak/STAT pathway,hedgehog pathway, Wnt/b-catenin pathway, steroid hor-mones), induce the expression of D-type cyclins (D1,D2 and D3) that preferentially bind and activate two closelyrelated CDKs: CDK4 and CDK6, often referred to collec-tively as CDK4/6 due to their similar function [13].CDK4/6-cyclin D complexes sustain progression of cell cyclethrough the G1 phase and, in late G1, induce cyclin E synthe-sis and the formation of CDK2-cyclin E complexes, whichcontribute to progression through the R point and entryinto S phase (Figure 1) [14]. CDK4/6-cyclin D complexes aretherefore important mediators of cell cycle regulatory deci-sions, which make them potentially strategic targets for anti-cancer treatment. After crossing the R point, sequentialactivation of the other cyclins governs the cell throughoutthe entire cell cycle: E-type cyclins, binding CDK2, controlentry into S phase; A-type cyclins bind CDK2 for S-phaseprogression and subsequently CDK1 during G2; finallyB-type cyclins, binding CDK1, govern entry into mitosis.

The CDK4/6-cyclin D complexes carry out their functionon cell cycle by phosphorylating and thus inactivating theso-called pocket proteins: Rb (the product of the Rb --retinoblastoma -- tumor suppressor gene) and its relatedproteins p107 (RBL1) and p130 (RBL2). Rb and its cousinproteins, when present in an unphosphorylated state at thebeginning of G1, bind to transcription factors, primarily ofthe E2F family, inhibiting their transcriptional activity. E2Ftranscription factors are bound to the promoters of their targetgenes whose products mediate S-phase entry and mitosis [15,16];these include factors involved in subsequent cell cycle progres-sion, such as cyclin E, cyclin A and the CDK activator mem-bers of the Cdc25 phosphatase family [17]; factors involved inDNA replication, such as DNA polymerase a, proliferatingcell nuclear antigen (PCNA) and minichromosome mainte-nance 7 (MCM7); factors involved in DNA damage repair,such as Rad51; and factors involved in mitosis, such as cyclinB1 and CDK1. The CDK4/6-cyclin D complexes produceprogressive phosphorylation of the pocket proteins at serineand threonine residues during G1, leading to their gradual

Box 1. Drug summary.

Drug name Palbociclib (PD 0332991)Phase IIIIndication First-line advanced breast cancerPharmacologydescription

Pyridopyrimidine-derived potentand highly selective inhibitor ofcyclin-dependent kinases 4 and6 (CDK4/6)

Route ofadministration

Oral

Chemical formula 6-Acetyl-8-cyclopentyl-5-methyl-2-((5-(piperazin-1-yl)pyridin-2-yl)amino)pyrido[2,3-d]pyrimidin-7(8H)-one

Pivotal trial(s) [80-86]

Pharmaprojects -- copyright to Citeline Drug Intelligence (an Informa

business). Readers are referred to Pipeline (http://informa-pipeline.

citeline.com) and Citeline (http://informa.citeline.com).

A. Rocca et al.

408 Expert Opin. Pharmacother. (2014) 15(3)

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 01

/27/

14Fo

r pe

rson

al u

se o

nly.

inactivation, which in turn, restores E2F transcriptional activ-ity. This induces expression of E-type cyclins and Cdc25 phos-phatase, leading to the formation of CDK2-cyclin E complexesduring late G1. These complexes further phosphorylate thepocket proteins, leading to their complete inactivation andpromoting progression through the R point [14,18]. Rb is con-sidered the ‘guardian’ of the R-point gate because of its centralrole in G1-S transition.

Although CDK2 and CDK4/6 are deeply involved in thecell cycle process, knockout studies in mice have shown thatthey are not essential for the cell cycle of most cell types,and CDK1 is sufficient to drive cell division in most cellularlineages [9,10]. While knockdown of CDK1 causes cell cyclearrest, preventing early embryonic development, ablation ofCDK4/6 impairs proliferation of hematopoietic precursors,leading to late embryonic death. On the other hand, thereis also evidence that some tumor cells may have specificrequirements for individual CDKs [10].

Further regulators of the cell cycle machinery are the CDKinhibitors (CdkIs). The INK4 (inhibitors of CDK4) proteins,including p16INK4A, p15INK4B, p18INK4C and p19INK4D,block the formation of cyclin D-CDK4/6 complexes [19].The Cip/Kip (kinase inhibitor protein) family of CdkIs,including p21Cip1, p27Kip1 and p57Kip2, inhibit all the othercyclin-CDK complexes. On the contrary, p21Cip1 andp27Kip1 stimulate the assembly, activation and nuclear locali-zation of cyclin D-CDK4/6 complexes [20]. During the

G1 phase, p27Kip1 and p21Cip1 form ternary complexes withthe growing number of cyclin D-CDK4/6 complexes;p27Kip1 and p21Cip1 are thus sequestered by CDK4/6 andsubtracted from cyclin E-CDK2 complexes, leading to theiractivation. With a positive feedback, cyclin E-CDK2 com-plexes phosphorylate p27Kip1, leading to its degradation [21].The increase in cyclin E-CDK2 complexes favors progressionof the cell cycle beyond the restriction point.

CDKs are also involved in DNA-damage checkpoints.These regulatory mechanisms induce late G1 arrest or lateG2 arrest in response to DNA damage to enable its repairbefore starting DNA synthesis or mitosis. They act by induc-ing the expression of CDK inhibitors such as p21 [22] or byinhibiting CDK activators such as the Cdc25 phospha-tases [23]. Deregulated CDK activation in cancer cells favorsgenomic and chromosomal instability [10].

1.2 Alterations of key cell cycle molecules in breast

cancerAltered expression and/or dysfunction of several moleculesinvolved in the cell cycle process have been described in breastcancer. Amplification of the cyclin D1 gene (CCND1) hasbeen found in ~ 15 -- 20% of human mammary carcino-mas [24,25] and cyclin D1 overexpression in up to 50%, inboth early and advanced stages of the disease [26,27]. Despiteits central role in breast cancer pathogenesis, the prognostic

M

G2

S

G1

Cip/Kip family

Rb

p21Cip1

p16INK4

p27Kip1

INK4 family

Ras/Raf/MAPKNF-κBJak/STATWnt/β-cateninEstrogen receptor

E2F

CDK4/6 CDK4/6Cyclin D Cyclin D

p p

pRb

STOP

CDK2 Cyclin E

E2F

Figure 1. Cyclin D-cyclin dependent kinase 4/6-Rb protein: a key pathway in cell cycle progression.CDK: Cyclin dependent kinase; Cip/Kip: Kinase inhibitor protein family; E2F: Elongation factor 2; INK4: Inhibitors of CDK4; P: Phosphate; Rb: Retinoblastoma

protein; STAT: Signal transducer and activator of transcription.

Palbociclib (PD 0332991)

Expert Opin. Pharmacother. (2014) 15(3) 409

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 01

/27/

14Fo

r pe

rson

al u

se o

nly.

significance of cyclin D1 overexpression is less clear [28].Cyclin D1-null mice have been shown to be resistant to breastcancers induced by the neu and ras oncogenes, while remain-ing sensitive to other oncogenic pathways, such as thosedriven by c-myc or Wnt-1 [29]. This has been attributed tothe fact that the neu-ras pathway is closely connected to thecell-cycle machinery by cyclin D1, and suggests potentialactivity of anti-cyclin D1-CDK4/6 agents in these tumors [29].The critical role of cyclin D1 in breast cancer has been

shown to be related to its ability to activate CDK4, and thecontinued presence of CDK4-associated kinase activity seemsnecessary to maintain breast tumorigenesis [30]. Amplificationof CDK4 gene is reported in about 15% of breast cancers,resulting in CDK4 protein overexpression, and is associatedwith high Ki-67 labeling index [31].Several breast cancer cell lines show homozygous deletion

of p16INK4A or p15INK4B, or low expression of these CDKinhibitors, while others have high expression levels associatedwith deletion or inactivation of Rb [32]. P21Cip1 expressionis frequently reduced as a consequence of TP53 mutation [32]

or MYC overexpression [33], and p27Kip1 expression is reducedas a result of HER2 amplification [34].Rb gene deletion or mutation is found in 20 -- 30% of breast

cancers, but Rb inactivation due to cyclin D1 overexpressionor p16INK4A inactivation is present in the majority of breastcancers [35]. While immunohistochemical studies of Rb expres-sion have yielded contrasting results, an Rb-loss gene signaturecan identify tumors with dysregulated Rb [35]. Maximal dereg-ulation of Rb target gene expression is observed in ER-negativebreast cancer, where it is devoid of relevant prognostic effectbut is associated with a better response to chemotherapy.Immunohistochemically detected Rb loss confirms this associ-ation in triple-negative breast cancer [36]. In ER-positive breastcancer Rb deregulation is rarer but confers poor prognosis [35].

1.3 Cell cycle, estrogen activity and endocrine

resistanceEstrogens stimulate cellular proliferation in the female repro-ductive tract and mammary gland, and play an important rolein breast cancer carcinogenesis [37] and breast cancer progres-sion. In normal breast epithelium [38], as well as inMCF-7 breast cancer cell lines [39], 17-b-estradiol binds tonuclear estrogen receptor alpha (ER-a), a ligand-dependenttranscription factor, inducing the expression of several genesincluding CCND1, coding for cyclin D1, and therefore, acti-vating CDK4/6 with subsequent Rb inactivation. Estradiol-bound ER-a can also bind other transcription factors, suchas members of the activation protein 1 (AP1), specificityprotein 1 (SP1) and NF-kB, to induce transcription of differ-ent sets of genes [40]. In addition, estradiol inhibitsthe expression of the p21Cip1 and p27Kip1 CDK inhibitorsand induces the expression of the CDK-activating phospha-tase Cdc25A [41], independently of D cyclin-CDK4function, both mechanisms contributing to sustain cell

proliferation. Furthermore, cyclin D1 has been shown tointeract directly with ER-a, activating its transcriptional func-tion independently of CDKs and estrogens [42].

Inhibiting ER-a inMCF-7 breast cancer cells using selectiveER modulators (SERMs) such as tamoxifen leads to cell cyclearrest in G1 and apoptosis as a result of reduced expressionof cyclin D and B, increased expression of p53 and p21Cip1

and loss of the prosurvival protein Bcl-2 [43]. The selectiveER downregulator (SERD) fulvestrant shows similar activitybut also induces accumulation of p130-E2F4 complexescharacteristic of quiescence and G0 arrest [44].

Aberrant expression of several molecules involved in cellcycle regulation and in estrogen and tamoxifen action mayinduce endocrine resistance. Overexpression of MYC sup-presses p21Cip1expression, favoring the formation of cyclinE1-CDK2 complexes and tamoxifen resistance [33]. CyclinD1 overexpression favors formation of cyclin D1-CDK4/6complexes, which sequester p21Cip1 and p27Kip1, allowingactivation of cyclin E1-CDK2 complexes and inducing tamox-ifen resistance, as shown in breast cancer cell lines [45] and inpatients with breast cancer [46-48]. Cyclin E1 overexpressionhas also been implicated in endocrine resistance [49]. Rb inacti-vation has resulted in tamoxifen and fulvestrant resistance inxenograft breast cancer models [50], and gene expression profil-ing studies on human breast cancer specimens have shown anassociation between Rb-dysfunction signatures and luminalB breast cancer subtype [51] or increased recurrence risk follow-ing tamoxifen therapy [50]. Reduced expression of p27 [34]

and cytoplasmic localization of p21Cip1 [52] have also beenassociated with endocrine resistance.

Other mechanisms of endocrine resistance, not primarilyinvolving alterations of molecules of the cell cycle machinery,may still implicate cyclin D-CDK4/6 activity as a mediatorof cellular proliferation [40,53] and therefore as a potential ther-apeutic target. Ligand-independent activation of ER-a mayoccur as a result of activation of receptor tyrosine kinases thatinduce phosphorylation of ER-a or its coregulators. Bidirec-tional cross-talk between ER-a and members of the epidermalgrowth factor receptor family, such as EGFR and HER2 [54,55],or the insulin-like growth factor receptor (IGFR) [56], havebeen extensively associated with the development of endocrineresistance. The same occurs with alterations of components oftheir downstream signaling pathways, such as MAPK/ERKand PI3K/Akt [57]. Estrogens also bind to G-protein coupledreceptor GPR30, located on the cell membrane, which medi-ates nongenomic effects, assembling with c-Src and otherproteins and activating Akt or ERK and their downstreamcascades (Figure 2) [58].

Several other mechanisms of endocrine resistance have beendescribed, including increased activity of transcription factorssuch as AP1 and NF-kB, alterations in ER-a coregulators,increased expression of survival molecules such as Bcl2, anddecreased expression of death factors such as BAK, BIKor caspases, aberrant miRNA expression and epigeneticalterations [40,53].

A. Rocca et al.

410 Expert Opin. Pharmacother. (2014) 15(3)

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 01

/27/

14Fo

r pe

rson

al u

se o

nly.

Resistance to aromatase inhibitors is not as well understoodat the molecular level, may differ, in part, from resistance toSERMs or SERDs and seems to involve the activation of cel-lular stress response and apoptosis [59]. In long-term estrogen-deprived breast cancer cell lines exhibiting hyperactivation ofthe PI3K pathway, the unbound ER has been shown to playa role in hormone-independent growth of these cells by acti-vating E2F transcriptional activity [60], a process mediatedby CDK4.

Given the plethora of mechanisms underlying endocrineresistance, the position of CDK4/6 downstream of multiplegrowth factors pathways and the preserved high CDK4/6activity and CDK4/6 tumor addiction in many cases [61], tar-geting CDK4/6 appears a promising strategy to overcomeendocrine resistance [62]. It has been shown that, in differentMCF7-derived models of spontaneous and acquired endo-crine-resistance, treatment with fulvestrant, although effec-tively downregulating ER-a, does not reduce the expressionof cyclin D and as a result does not lead to Rb dephosphory-lation and activation and to cell cycle arrest [51]. Treatment ofthese cell lines with the CDK4/6 inhibitor palbociclib hasresulted in effective Rb dephosphorylation and profoundinhibition of cell cycle progression, across all of the models

employed. In contrast to the quiescent state induced by fulves-trant in sensitive cells, palbociclib has been shown to inducecellular senescence, an irreversible cell-cycle arrest, indicatedby large, flat cellular morphology and b-galactosidaseexpression [51].

2. Palbociclib preclinical studies

As first-generation pan-CDK inhibitors showed modest clini-cal activity as single agents and considerable toxicity, subse-quent efforts have led to the development of more potentand selective small molecule CDK inhibitors [63,64].

2.1 Preclinical pharmacologyPalbociclib is an orally active, potent and highly selectiveinhibitor of CDK4 and CDK6, with IC50 values for CDK4/cyclinD1, CDK4/cyclinD3 and CDK6/cyclinD2 of 11, 9and 15 nmol/l, respectively, and low or absent activity againsta panel of 36 additional protein kinases, including CDK2,CDK1 and several tyrosine kinases and serine-threonine kin-ases. It is a pyridopyrimidine derivative, inhibiting CDK4/6by binding to the ATP site [65]. As Rb phosphorylation byCDK4 and CDK6 occurs specifically on serine residues

ER

ER Cyclin DCyclin DCyclin ECDC25

MAPK

E EE

GPR30

Cell membraneSrc

PI3K

Receptor tyrosinekinases

AKT

ER

Sp1 Ap1Nucleus

PP

PP

ERK

E

EE

E

ERE

Figure 2. Pathways of estrogen signaling.Akt: Ap1, activation protein 1; CDC25: Cell division cycle 25 protein; E: Estrogen; ER: Estrogen receptor; ERK: Extracellular signal-regulated kinase;

GPR30: G-protein coupled receptor 30; P: Phosphate; Sp1: Specificity protein 1.

Palbociclib (PD 0332991)

Expert Opin. Pharmacother. (2014) 15(3) 411

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 01

/27/

14Fo

r pe

rson

al u

se o

nly.

Ser780 and Ser795, the phosphorylation status of these sitesserves as a specific biomarker of CDK4/6 inhibition by palbo-ciclib. In cell cultures, reduction of Rb phosphorylationbegins 4 h after exposure, reaches a maximum at 16 h and iscompletely reversible after removal of the drug. Palbociclibis a potent inhibitor of cell proliferation, preventing progres-sion of the cell cycle from G1 into the S phase in Rb-positivecells of different tumor types, but showing no activity againstRb-negative tumor cells in vitro or in vivo [65]. This fact,combined with the exclusive G1 arrest even at very high con-centrations, is consistent with selective CDK4/6 inhibition asits only mechanism of action [65].In mouse xenograft models, palbociclib showed significant

antitumor activity in several tumor types, including breastcancer xenografts. In mice bearing MDA-MB-435 breast car-cinoma, antitumor activity was present only at doses yieldingcomplete suppression of Rb Ser780 phosphorylation through-out the entire treatment period, whereas resumption ofSer780 phosphorylation during the intervals between doseswas associated with treatment failure, indicating that totaltarget inhibition must be maintained between doses toachieve tumor regression. Substantial differences in dose(up to eightfold) were necessary to produce comparable effi-cacy in xenograft models with different sensitivity to thedrug [65]. Although palbociclib has a cytostatic effect ontumor cell cultures in vitro and does not induce apopto-sis [65,66], it has led to tumor regression in vivo, includingsome durable complete remissions. This could be attributedto the presence of a fraction of cells spontaneously dyingwithin tumors, or to the need for CDK4/6 as a survivalfactor by some tumors [65].

2.2 Sensitivity and resistance to palbociclibRb-negative breast cancer cell lines are resistant to palbocicliband are characterized, apart from loss of Rb, by an upregula-tion of p16INK4A and no appreciable cyclin D1 protein expres-sion. It appears therefore that they do not respond topalbociclib simply because they lack the palbociclib target,that is, active cyclin D-CDK4/6, because CDK4/6 is alreadyinhibited by the overexpression of p16INK4A

[67].Sensitivity to palbociclib was assessed in a panel of 44

human breast cancer cell lines, representative of the differentbreast cancer subtypes, to identify predictors of response [66].ER-positive cell lines with luminal features were the mostsensitive, and three-quarters of the genes highly expressedin sensitive lines were luminal markers, whereas none werebasal markers. Of 16 HER2-amplified cell lines, 10 were sen-sitive, and these generally had luminal (ER-positive) features.Cell lines with basal features were the most resistant. Highlevels of cyclin D1 and Rb, and low levels of p16 were pre-dictors of sensitivity to palbociclib. Although the drug hadno effect on total Rb levels, Rb phosphorylation decreasedafter drug exposure in sensitive cells. Resistant cells usuallyhad low baseline levels of Rb, and in the few resistant lineswith detectable levels of Rb, its phosphorylation did not

decrease after drug exposure. Therefore, the presence of Rbappears to predict response to palbociclib in luminal, ER-positive breast cancer cell lines but not in basal cell lines. Ithas been speculated that failure of palbociclib to dephos-phorylate Rb could be due to CDK4/6 mutations, to agreater dependence on CDK1/2-cyclin E interactions or toa loss of negative regulators of CDKs, but these hypothesesremain to be confirmed in breast cancer [66]. However, resis-tance to palbociclib due to p27 downregulation and CDK2reactivation has been demonstrated in models of acute mye-logenous leukemia [68]. Resistance to palbociclib in breastcancer cell lines with basal features appears frequently relatedto a lack of Rb, and loss of Rb has been described in basal-likebreast cancer [69] and can result in epithelial-mesenchymaltransition [70].

Further investigation of the mechanisms of response andresistance to palbociclib was conducted on a panel of breastcancer cell lines using Rb knockdown experiments [67]. Rb sta-tus appeared to play a prominent role in acute response to pal-bociclib, but compensatory mechanisms controlling E2Factivity would also seem to influence response, especiallylong-term. Although Rb knockdown resulted in a modestincrease in baseline proliferation, palbociclib still exerted a par-tial cytostatic effect (albeit much lower than in Rb-proficientcells), suggesting that Rb protein is not essential for responseto palbociclib. On the other hand, E2F overexpression bytransduction yielded complete resistance to palbociclib, inde-pendently of Rb status. This suggests that E2F transcriptionalcontrol may be partly independent of Rb protein and poten-tially mediated by p107 protein, and that palbociclib mightcause partial repression of E2F-target genes by activatingp107. Nonetheless, while Rb-proficient cells may become tem-porarily resistant to palbociclib after prolonged exposure, theyusually remain sensitive to deferred second rounds of treat-ment. Loss of Rb function normally marks the evolution totrue, long-term resistance. Cell populations retrieved afterlong-term exposure to palbociclib often show elevated p107protein expression as well as increased CDK2 protein and/orloss of p21Cip1 and p27Kip1. In this context, loss of Rb functionleads to increased transcription of the E2F-target genes cyclinA and E, which activate CDK2 and drive cell cycle indepen-dently of CDK4/6 in some tumor cells [67]. On the contrary,Rb function is necessary for the induction of senescence bypalbociclib, wherein tumor cells permanently exit the cellcycle [67].

CDK4 inhibition has also been reported to induce apoptosisin colon cancer cell models by causing degradation of theNF-kB suppressor protein IkB, with subsequent translocationof RelA (principal component of NF-kB) from the cytoplasmto the nucleoplasm and the nucleolus, and repression of NF-kB-driven transcription of anti-apoptotic genes [71]. It has alsobeen shown that inhibition of CDK4 sensitizes pancreatic can-cer cells to TRAIL-induced apoptosis via downregulation ofsurvivin [72]. The role of these processes in breast cancer remainsto be assessed.

A. Rocca et al.

412 Expert Opin. Pharmacother. (2014) 15(3)

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 01

/27/

14Fo

r pe

rson

al u

se o

nly.

2.3 Combination of palbociclib with other targeted

agentsCombinations of palbociclib with trastuzumab or withtamoxifen were tested in HER2-amplified cell lines and inER-positive cell lines, respectively [66]. Both combinationswere synergistic, with a mean combination index < 1 acrossclinically relevant concentrations of the drugs. Furthermore,tamoxifen-resistant MCF7 cell lines were sensitive to palboci-clib monotherapy, and palbociclib partially restored sensitiv-ity to tamoxifen in resistant lines [66]. This is consistent withother observations of the potentially enhanced endocrinesensitivity of CDK inhibition [73].

A sequential combination of palbociclib with a PI3K inhib-itor (GS-1101, inhibiting PI3K-d, whose expression isrestricted to cells of the hematopoietic lineage) yielded arobust apoptotic response in lymphoma cell lines [74]; studiesare also warranted in combination with other PI3K inhibitorsin breast cancer.

2.4 Combination of palbociclib with

chemotherapeutic agentsAlthough the synergism between palbociclib and endocrineagents such as tamoxifen or anti-HER2 drugs such as trastu-zumab is well documented in preclinical models, the associa-tion of palbociclib with chemotherapeutic agents is moreproblematic. Indeed, most chemotherapeutic agents act spe-cifically on proliferating cells, and their combination with acytostatic agent may be ineffective. In genetically engineeredmouse models of a HER2-positive, Rb-competent breast can-cer (MMTV-c-neu), palbociclib showed antitumor activity asa single agent, but the combination of palbociclib with carbo-platin or with doxorubicin proved less active than single-agentcarboplatin or doxorubicin [75]. This antagonism was not seenin engineered models of a basal-like, Rb-incompetent breastcancer, where palbociclib showed no antitumor effect as a sin-gle agent and did not reduce the activity of carboplatin whengiven in combination. The same authors demonstrated theability of palbociclib to protect an immortalized human fibro-blast cell line from the toxicity of a variety of DNA-damagingagents, such as carboplatin, doxorubicin, etoposide and camp-tothecin, or from the antimitotic agent paclitaxel, and toprotect the bone marrow from carboplatin-induced myelo-suppression in mice, highlighting the potential use of palboci-clib as a chemoprotectant of normal tissues to overcome thedose-limiting toxicity of many chemotherapeutic agents.

Further experiments in Rb-proficient, triple-negative breastcancer cell lines and xenograft mice models showed that,although the combination of palbociclib with doxorubicinyielded an additive cytostatic effect, palbociclib antagonizeddoxorubicin-mediated cytotoxicity, greatly reducing theinduction of apoptosis [76]. As a result, palbociclib maintainedviability of Rb-proficient cells treated with doxorubicin,resulting in a recurrent population of cells after doxorubicinexposure. Again, these effects were not seen in Rb-deficient,

triple-negative breast cancer models. Anthracyclines induceDNA damage, and palbociclib can shift DNA repair fromhomologous recombination to non-homologous end joining,a more error-prone mechanism that could contribute totumor progression [77]. Pretreatment or co-treatment oftriple-negative breast cancer cell lines with palbociclib alsoshowed antagonism to paclitaxel, a microtubule-stabilizingagent that acts by promoting mitotic catastrophe [77]. In con-trast, a short exposure to palbociclib to synchronize cells priorto paclitaxel treatment resulted in increased cytotoxicity.

2.5 Further potentially useful or deleterious effectsPalbociclib has been shown to reverse epithelial dysplasia asso-ciated with abnormal activation of the cyclin D-CDK4/6-Rbpathway, highlighting a potential role of this molecule as achemopreventive agent [78].

Other studies raise some potential concerns about CDK4/6inhibition in specific experimental models. Palbociclib hasbeen shown to induce epithelial-mesenchymal transition andenhance invasiveness in pancreatic cancer cell lines by activatingSmad-dependent TGF-b signaling. Senescent fibroblast overex-pressing CDK inhibitors also promote tumor growth via theparacrine production of high-energy mitochondrial fuels, suchas L-lactate [79].

3. Palbociclib clinical development

A Phase I dose escalation study of palbociclib was conductedwith two different schedules: daily treatment for 2 weeksfollowed by 1 week off treatment (schedule 2/1) [80] and dailytreatment for 3 weeks followed by 1 week off (schedule3/1) [81]. Eligible patients had Rb-positive (assessed by immu-nohistochemistry) solid tumors or non-Hodgkin’s lymphomarefractory to standard therapy or for which no standardtherapy was available.

For the first schedule, a total of 33 patients were enrolled.The dose escalation sequence progressed from 100 to150 mg/day, and then to 225 mg/day, the maximum admin-istered dose at which two dose-limiting toxicities (DLTs)occurred: one case of grade 4 neutropenia and thrombocyto-penia and another of grade 3 neutropenia, resulting in a delayin the initiation of cycle 2. The dose was then reduced to200 mg/day, and this dose level was expanded to a total of20 patients and selected as the MTD. Four DLTs occurredat this dose level, all consisting in grade 3 neutropenia, withor without grade 3 thrombocytopenia, delaying the initiationof the subsequent cycle. Non-hematological toxicity wasgenerally mild, with only five grade 3 adverse events (AEs)reported overall and no treatment-related grade 4 toxicity.The most common non-hematological AEs included fatigue,nausea, diarrhea and constipation, with some patients alsoexperiencing vomiting. Of 31 patients assessable for response,1 patient with testicular cancer had a partial response and anadditional 9 (29%) with different types of tumors had stable

Palbociclib (PD 0332991)

Expert Opin. Pharmacother. (2014) 15(3) 413

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 01

/27/

14Fo

r pe

rson

al u

se o

nly.

disease, which in 3 cases lasted beyond 10 cycles oftreatment [80].Forty-one patients were enrolled for the second schedule,

with a sequential dose escalation ranging from 25 to150 mg/day [81]. Again, the major AEs were related to myelo-suppression. Overall, 5 patients experienced DLTs, allconsisting of neutropenia, grade 4 in 2 cases and grade 3 in3 cases, the latter resulting in a delay in the subsequent cycle.Non-hematological toxicity was generally mild and, like theschedule 2/1, included, fatigue, nausea, vomiting andconstipation. No clinically significant effects on cardiac repo-larization were reported (with either schedule). The recom-mended Phase II dose was 125 mg/day. Among 37 patientsevaluable for response, disease stabilization for at least twocycles was achieved in 13 cases (35%) and maintained for atleast 10 cycles in 6 patients, including one with breast cancerand high levels of Rb-positive cells. There were no partialresponses according to RECIST criteria.Pharmacokinetic parameters for palbociclib with the two

schedules are reported in Table 1 [80,81]. The main parametersshowed low-to-moderate inter-patient variability, with a gen-erally dose-dependent increase in exposure (assessment basedon maximum observed plasma concentration [Cmax] and thearea under the plasma concentration-time curve from 0 to10 h [AUC0-10]) over the dose range studied. At steady state,palbociclib was absorbed with a median Tmax (time to firstoccurrence of Cmax) of ~ 4 -- 5.5 h. The mean drug apparentvolume of distribution (Vz/F) was significantly greater thantotal body water, suggesting an extensive penetration intoperipheral tissues and substantial tissue binding. Palbociclibwas eliminated slowly, with a mean terminal half-life (t1/2)of ~ 26 h and a mean apparent clearance of 80 -- 90 l/h. Renalexcretion was a minor route of elimination with a mean of1.8% of unchanged palbociclib found in urine.A pharmacodynamic model correlating the AUC with

nadir values of absolute neutrophil count (ANC) and plateletcount showed a non-linear relationship, with increasing drugexposure resulting in a saturable decrease from baseline forboth ANC and platelets [80,81].Based on the results from the Phase I study, the schedule

3/1 at a dose of 125 mg/day was selected for further clinicaldevelopment. Preliminary results of a Phase II, single-armstudy of palbociclib (schedule 3/1) in patients with advancedbreast cancer expressing the Rb protein have only beenreported in abstract form and were updated at the2013 ASCO Annual Meeting [82]. Of the 37 patients enrolled,30 had hormone receptor-positive tumors (HER2-positive intwo cases) and showed 2 (7%) partial responses and 17 (57%)disease stabilizations lasting longer than 6 months in 3 cases(10%); 11 patients (36%) had disease progression. Triple-negative tumors were seen in 6 patients, and 5 of themshowed disease progression at first assessment, whereas only1 had disease stabilization, lasting longer than 6 months.Median progression-free survival (PFS) was 3.8 months inhormone receptor-positive patients (95% CI 2.3 -- 7.7) and

1.9 months in triple-negative patients (95% CI 0.9 -- ¥). Tox-icities were consistent with previous studies, with grade3/4 AEs limited to neutropenia and thrombocytopenia. Thistrial confirms results from preclinical studies, showing betteractivity of palbociclib in hormone-receptor-positive andHER2-positive tumors compared with triple-negative ones.

Given the important role of both estrogens and CDKs inthe genesis and progression of breast cancer, the preclinicalresults showing preferential activity of palbociclib in luminaltumors, and the synergism between palbociclib and tamoxifendemonstrated in vitro, clinical trials were planned to test thecombination of palbociclib with letrozole. In a Phase IBstudy [83], 12 post-menopausal patients with ER-positive,HER2-negative advanced breast cancer pretreated with che-motherapy (67%) or endocrine therapy (50%) received palbo-ciclib 125 mg/day (schedule 3/1) and letrozole 2.5 mg/daycontinuously. The median duration of treatment was6 months (range 2 -- 13). Treatment was well tolerated, withthree DLTs (grade 4 neutropenia in two cases and dose inter-ruption in one) and leuko-neutropenia and fatigue as themost common side-effects. The pharmacokinetic evaluationsuggested that there was no interaction between palbocicliband letrozole. Out of 9 patients with measurable disease,3 experienced a partial response and 9 showed stable disease.

A randomized Phase II study was then conducted in post-menopausal patients with ER-positive, HER2-negativeadvanced breast cancer who were randomized to receive letro-zole 2.5 mg/day alone or in combination with palbociclib125 mg/day (schedule 3/1) as first-line therapy for advanceddisease (TRIO-18 trial). The trial consisted of two parts: inthe first part, patient selection was based only on ER andHER2 status, whereas in the second part, CCND1 amplifica-tion and/or p16 loss, assessed by fluorescence in situ hybrid-ization, was required for eligibility. Patients were stratifiedby disease site and disease-free interval. Part 1, enrolling66 patients, showed a significant improvement in PFS withthe combination of palbociclib plus letrozole compared withletrozole alone (HR = 0.35; 95% CI, 0.17 -- 0.72;p = 0.006) [84,85]. Objective response rates (27 vs 23%) andclinical benefit (partial responses plus stable disease‡ 24 weeks) rates (59 vs 44%) were also improved in patientstreated with the combination [84]. The most commontreatment-related adverse events in the combination armwere neutropenia, leukopenia and fatigue. In an exploratoryanalysis, CCND1 and p16 status did not add a further predic-tive value over ER expression alone. Thus, after a further99 patients had been randomized in part 2, results from thesecond interim analysis were presented at the 2012 San Anto-nio Breast Cancer Symposium, combining parts 1 and 2 ofthe study, for a total of 165 patients, 84 randomized in thecombination arm and 81 in the letrozole-alone arm. Baselinecharacteristics were well balanced between the two arms.A PFS of 26.1 months was observed for patients receivingpalbociclib plus letrozole, compared with 7.5 months forthose treated with letrozole alone (HR = 0.37; 95% CI,

A. Rocca et al.

414 Expert Opin. Pharmacother. (2014) 15(3)

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 01

/27/

14Fo

r pe

rson

al u

se o

nly.

0.21 -- 0.63; p < 0.001). The objective response rate was 34%in the combination arm and 26% in the letrozole-alone arm,while clinical benefit rates were 70 and 44%, respectively. Thetoxicity profile for the combination was favorable, the mostcommon AEs being uncomplicated neutropenia, leukopenia,anemia and fatigue [85].

On the basis of these extremely promising results, enroll-ment is ongoing in a randomized, multicenter, double-blind,first-line Phase III study of palbociclib plus letrozole com-pared with letrozole/placebo in postmenopausal women withER-positive, HER2-negative advanced breast cancer whohave not received any prior systemic anticancer treatmentfor advanced disease [86].

Other Phase II or III studies are planned or ongoing withpalbociclib in combination with endocrine agents in patientswith hormone receptor (HR)-positive, HER2-negative disease:in combination with fulvestrant after endocrine failure in met-astatic breast cancer (NCT01942135); in combination withstandard endocrine treatment in patients with residual diseaseafter neoadjuvant chemotherapy and surgery for primarybreast cancer (PENELOPE-B, NCT01864746); and in com-bination with anastrozole as neoadjuvant therapy in patientswith stage II or III ER-positive, HER2-negative breast cancer(without PI3K hotspot mutations) (NCT01723774).A Phase I study is ongoing to establish the MTD and the rec-ommended Phase II dose of a combination of palbocicliband paclitaxel (NCT01320592).

4. Conclusions

Deregulation of cell cycle control is a prominent feature of can-cer, and the cyclin D-CDK4/6-Rb pathway, governing the cellcycle restriction point, is often altered in breast cancer, contrib-uting to tumor progression and to the development of endo-crine resistance. Palbociclib, a small molecule and highlyselective, reversible inhibitor of CDK4 and CDK6, inhibitsprogression of the cell cycle from G1 into the S phase in Rb-proficient cells, exerting cytostatic activity on different tumortypes in vitro and in vivo. It is not active on Rb-negative,p16INK4A overexpressing tumor cells.

Palbociclib has shown considerable activity in luminal, ER-positive breast cancer cell lines and xenograft models, includ-ing some luminal HER2-positive lines, and little or no activ-ity in breast cancer models with basal features. Combinationsof palbociclib with trastuzumab or tamoxifen have provensynergistic in HER2-amplified and in ER-positive cell lines,respectively.

Based on these preclinical findings, palbociclib was testedin a randomized Phase II study in combination with letrozole,versus letrozole alone, as first-line therapy in patients withadvanced ER-positive, HER2-negative breast cancer, leadingto a substantial improvement in PFS. Palbociclib alone hasalso shown some activity in patients with ER-positive breastcancer expressing Rb protein.

Although palbociclib appears to be a very promising agentin breast cancer, preliminary results must now be confirmedin Phase III trials, which are ongoing. Further clinical studiesare currently exploring its activity or efficacy in other diseasesettings and in combination with other agents. Translationaland basic research is focusing mainly on the detection of pre-dictive biomarkers that could identify patients who are likelyto benefit from this agent.

5. Expert opinion

The cyclin D-CDK4/6-Rb pathway represents a master regula-tor of cell cycle and is downstream of multiple mitogenic cas-cades, representing a relevant target for anticancer therapy.Preclinical evaluation shows that palbociclib, a reversibleinhibitor of CDK4 and CDK6, is active in breast cancer mod-els, especially those with luminal features, including someHER2-positive tumors. Synergistic results have also beenreported for palbociclib when used in combination withtamoxifen and trastuzumab, while a potential antagonism hasbeen noted with several chemotherapeutic agents.

Preliminary results from clinical trials confirm the preferen-tial activity of the drug in ER-positive tumors [82] and itspotential synergism with letrozole [85].

An important characteristic that should be taken intoaccount for the clinical development of palbociclib is itspure cytostatic action exerted in most tumor models, that is,

Table 1. Pharmacokinetic parameters for palbociclib by schedule after multiple doses.

Parameter 200 mg/day (schedule 2/1) 125 mg/day (schedule 3/1)

Cmax (ng/ml), mean (%CV) 174 (17) 86 (34)Tmax (h), median (range) 4 (2 -- 7) 4 (1 -- 10)AUC(0 -- 10) (ng·h/ml), mean (%CV) 1395 (23) 724 (38)Vz/F (l) 3241 2793t1/2 (h), mean 26.7 26CL/F (l/h), mean 88.5 80.6Rac, median 2.4 2.2

Reported values for Cmax, Tmax and AUC were measured at day 8 of the first cycle; Vz/F, t1/2, CL/F and Rac were estimated following repeated daily dosing to

steady-state for both schedules.

CL/F: Apparent clearance; CV: Coefficient of variation; Rac: Drug accumulation ratio; Vz/F: Apparent volume of distribution during the terminal phase.

Palbociclib (PD 0332991)

Expert Opin. Pharmacother. (2014) 15(3) 415

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 01

/27/

14Fo

r pe

rson

al u

se o

nly.

no induction of apoptosis, but a capacity to induce cellularsenescence. This may explain the low objective response ratesand higher rates of disease stabilization obtained when palbo-ciclib has been used as a single agent in clinical trials [82,87].Although tumor remissions have been observed in preclinicalstudies, the use of palbociclib alone may be more appropriateas ‘maintenance’ therapy, whereas treatment in combinationwith other agents may be needed to obtain maximal, rapidtumor shrinkage.Preclinical studies clearly point at Rb-proficiency, cyclin

D1 overexpression or gene amplification and loss or reducedexpression of p16 as predictors of response. Nonetheless, anexploratory analysis from the TRIO-18 trial showed that nei-ther CCND1 amplification nor p16 loss added any furtherpredictive value over ER expression alone [85]. There couldbe several reasons for this discrepancy. CCND1 amplificationis only responsible for a minority of cases of cyclin D1 over-expression. In some tumor models, the activation of cyclinE-CDK2 complexes may compensate for the lack or inactiva-tion of cyclin D-CDK4/6 complexes, abolishing the predic-tive value of cyclin D. Palbociclib has also been reported toexert some activity in experimental models after Rb knock-down, possibly due to compensation of Rb protein functionby p107 protein, which may be activated by palbociclib [66].Thus, prediction of response to palbociclib needs to be furtherinvestigated as a constellation of molecules is involved. An Rbgene expression signature could perhaps be more informative,albeit also more complicated, than the assessment of singlemolecules.The combination of palbociclib with endocrine agents

appears to be a highly promising area of application. Theimpressive results from the TRIO-18 trial are awaiting confir-mation in the ongoing Phase III trial [86]. However, the trialdoes not include an arm of letrozole followed by palbociclibor an arm in which palbociclib is added to letrozole only atthe moment of progression. The need for combined treatmentmay differ in tumors with intrinsic endocrine resistance com-pared to those that develop acquired resistance. A randomizedtrial testing the addition of lapatinib to letrozole showed, inthe subgroup of HER2-negative tumors, a trend towards pro-longed PFS only in patients who relapsed < 6 months after

tamoxifen discontinuation, but no effect in those who pro-gressed after ‡ 6 months or who had not received prior adju-vant tamoxifen [88]. Other drugs have also produced a benefitwhen combined with endocrine agents with the intent ofovercoming or preventing endocrine resistance: trastuzumabadded to anastrozole in patients with HER2/hormonereceptor-copositive tumors [89]; everolimus combined withexemestane in patients with HER2-negative breast cancerwho had disease recurrence or progression while receiving anon-steroidal aromatase inhibitor [90]; the histone deacetylaseinhibitor entinostat added to exemestane in the same diseasesetting [91]. The usefulness of the sequential administrationof these combinations and the best sequence remain to beascertained. However, confirmation of an important advan-tage in the metastatic setting could lead to their developmentin the adjuvant setting.

Combinations of palbociclib and other targeted therapiessuch as anti-HER2 agents, PI3K inhibitors or mTOR inhibi-tors could represent potentially important areas of research.The association of palbociclib with chemotherapy, however,appears more problematic because of the potential antago-nism. An ongoing Phase I trial of palbociclib in associationwith paclitaxel could provide an important insight into thepotential of such associations. Experiments carried out ontumor cell synchronization also indicate the possible useful-ness of cyclic palbociclib in combination with metronomicchemotherapies [78].

Although palbociclib has been shown to offer protectionfrom chemotherapy-induced myelosuppression, it would benecessary to accurately identify CDK4/6-independent tumorsin which palbociclib would not antagonize the activity of che-motherapy before implementing this strategy in clinical prac-tice. The ability of palbociclib to reverse epithelial dysplasiaalso highlights a possible chemopreventive role in selectedwomen at high risk for breast cancer.

Declaration of interest

The authors state no conflict of interest and have received nopayment in preparation of this manuscript.

A. Rocca et al.

416 Expert Opin. Pharmacother. (2014) 15(3)

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 01

/27/

14Fo

r pe

rson

al u

se o

nly.

BibliographyPapers of special note have been highlighted as

either of interest (�) or of considerable interest(��) to readers.

1. Cancer Incidence and Mortality

Worldwide: IARC CancerBase No. 10

[Internet]. GLOBOCAN 2008 v2.0.

Lyon, France: International Agency for

Research on Cancer; 2010;

Available from: http://globocan.iarc.fr

[Last accessed on 14 October 2013]

2. Anderson WF, Katki HA, Rosenberg PS.

Incidence of breast cancer in the United

States: current and future trends. J Natl

Cancer Inst 2011;18:1397-402

3. Hanahan D, Weinberg RA. Hallmarks of

cancer: the next generation. Cell

2011;144:646-74

4. Malumbres M. Physiological relevance of

cell cycle kinases. Physiol Rev

2011;91:973-1007

5. Bayliss R, Fry A, Haq T, Yeoh S. On

the molecular mechanisms of mitotic

kinase activation. Open Biol

2012;2:120136

6. Shapiro GI. Cyclin-dependent kinase

pathways as targets for cancer treatment.

J Clin Oncol 2006;24:1770-83

7. Collins I, Garrett MD. Targeting the cell

division cycle in cancer: CDK and cell

cycle checkpoint kinase inhibitors.

Curr Opin Pharmacol 2005;5:366-73

8. Manchado E, Guillamot M,

Malumbres M. Killing cells by targeting

mitosis. Cell Death Differ

2012;19:369-77

9. Satyanarayana A, Kaldis P. Mammalian

cell-cycle regulation: several Cdks,

numerous cyclins and diverse

compensatory mechanisms. Oncogene

2009;28:2925-39

10. Malumbres M, Barbacid M. Cell cycle,

CDKs and cancer: a changing paradigm.

Nat Rev Cancer 2009;9:153-66

11. Pardee AB. A restriction point for

control of normal animal cell

proliferation. Proc Natl Acad Sci USA

1974;71:1286-90

12. Weinberg RA. The retinoblastoma

protein and cell cycle control. Cell

1995;81:323-30

13. Malumbres M, Barbacid M. To cycle or

not to cycle: a critical decision in cancer.

Nat Rev Cancer 2001;1:222-31

14. Harbour JW, Luo RX, Dei Santi A,

et al. Cdk phosphorylation triggers

sequential intramolecular interactions

that progressively block Rb functions as

cells move through G1. Cell

1999;98:859-69

15. Knudsen ES, Knudsen KE. Tailoring to

RB: tumour suppressor status and

therapeutic response. Nat Rev Cancer

2008;8:714-24

16. Henley SA, Dick FA. The retinoblastoma

family of proteins and their regulatory

functions in the mammalian cell division

cycle. Cell Div 2012;7:10

17. Chen X, Prywes R. Serum-induced

expression of the cdc25A gene by relief

of E2F-mediated repression.

Mol Cell Biol 1999;19:4695-702

18. Lundberg AS, Weinberg RA. Functional

inactivation of the retinoblastoma protein

requires sequential modification by at

least two distinct cyclin-cdk complexes.

Mol Cell Biol 1998;18:753-61

19. Sherr CJ, Roberts JM. CDK inhibitors:

positive and negative regulators of G1-

phase progression. Genes Dev

1999;13:1501-12

20. LaBaer J, Garrett MD, Stevenson LF,

et al. New functional activities for the

p21 family of CDK inhibitors.

Genes Dev 1997;11:847-62

21. Slingerland J, Pagano M. Regulation of

the cdk inhibitor p27 and its

deregulation in cancer. J Cell Physiol

2000;183:10-17

22. Li R, Waga S, Hannon GJ, et al.

Differential effects by the p21 CDK

inhibitor on PCNA-dependent

DNA replication and repair. Nature

1994;371:534-7

23. Mailand N, Falck J, Lukas C, et al.

Rapid destruction of human Cdc25A in

response to DNA damage. Science

2000;288:1425-9

24. Buckley MF, Sweeney KJ, Hamilton JA,

et al. Expression and amplification of

cyclin genes in human breast cancer.

Oncogene 1993;8:2127-33

25. Dickson C, Fantl V, Gillett C, et al.

Amplification of chromosome band

11q13 and a role for cyclin D1 in

human breast cancer. Cancer Lett

1995;90:43-50

26. Bartkova J, Lukas J, Muller H, et al.

Cyclin D1 protein expression and

function in human breast cancer.

Int J Cancer 1994;57:353-61

27. Gillett C, Fantl V, Smith R, et al.

Amplification and overexpression of

cyclin D1 in breast cancer detected by

immunohistochemical staining.

Cancer Res 1994;54:1812-17

28. Arnold A, Papanikolaou A. Cyclin D1 in

breast cancer pathogenesis. J Clin Oncol

2005;23:4215-24

29. Yu Q, Geng Y, Sicinski P. Specific

protection against breast cancers by

cyclin D1 ablation. Nature

2001;411:1017-21

30. Yu Q, Sicinska E, Geng Y, et al.

Requirement for CDK4 kinase function

in breast cancer. Cancer Cell

2006;9:23-32

31. An HX, Beckmann MW,

Reifenberger G, et al. Gene amplification

and overexpression of CDK4 in sporadic

breast carcinomas is associated with high

tumor cell proliferation. Am J Pathol

1999;154:113-18

32. Musgrove EA, Lilischkis R, Cornish AL,

et al. Expression of the cyclin-dependent

kinase inhibitors p16INK4, p15INK4B

and p21WAF1/CIP1 in human breast

cancer. Int J Cancer 1995;63:584-91

33. Mukherjee S, Conrad SE. c-Myc

suppresses p21WAF1/CIP1 expression

during estrogen signaling and

antiestrogen resistance in human breast

cancer cells. J Biol Chem

2005;280:17617-25

34. Chu IM, Hengst L, Slingerland JM. The

Cdk inhibitor p27 in human cancer:

prognostic potential and relevance to

anticancer therapy. Nat Rev Cancer

2008;8:253-67

35. Ertel A, Dean JL, Rui H, et al.

RB-pathway disruption in breast cancer:

differential association with disease

subtypes, disease-specific prognosis and

therapeutic response. Cell Cycle

2010;9:4153-63

36. Trere D, Brighenti E, Donati G, et al.

High prevalence of retinoblastoma

protein loss in triple-negative breast

cancers and its association with a good

prognosis in patients treated with

adjuvant chemotherapy. Ann Oncol

2009;20:1818-23

37. Yue W, Yager JD, Wang JP, et al.

Estrogen receptor-dependent and

independent mechanisms of breast cancer

carcinogenesis. Steroids 2013;78:161-70

Palbociclib (PD 0332991)

Expert Opin. Pharmacother. (2014) 15(3) 417

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 01

/27/

14Fo

r pe

rson

al u

se o

nly.

38. Said TK, Conneely OM, Medina D,

et al. Progesterone, in addition to

estrogen, induces cyclin D1 expression in

the murine mammary epithelial cell, in

vivo. Endocrinology 1997;138:3933-9

39. Altucci L, Addeo R, Cicatiello L, et al.

17beta-Estradiol induces cyclin D1 gene

transcription, p36D1-p34cdk4 complex

activation and p105Rb phosphorylation

during mitogenic stimulation of

G(1)-arrested human breast cancer cells.

Oncogene 1996;12:2315-24

40. Musgrove EA, Sutherland RL. Biological

determinants of endocrine resistance in

breast cancer. Nat Rev Cancer

2009;9:631-43

41. Foster JS, Henley DC, Bukovsky A,

et al. Multifaceted regulation of cell cycle

progression by estrogen: regulation of

Cdk inhibitors and Cdc25A independent

of cyclin D1-Cdk4 function.

Mol Cell Biol 2001;21:794-810

42. Zwijsen RM, Wientjens E,

Klompmaker R, et al. CDK-independent

activation of estrogen receptor by cyclin

D1. Cell 1997;88:405-15

43. Ichikawa A, Ando J, Suda K. G1 arrest

and expression of cyclin-dependent

kinase inhibitors in tamoxifen-treated

MCF-7 human breast cancer cells.

Hum Cell 2008;21:28-37

44. Carroll JS, Prall OW, Musgrove EA,

Sutherland RL. A pure estrogen

antagonist inhibits cyclin E-Cdk2 activity

in MCF-7 breast cancer cells and induces

accumulation of p130-E2F4 complexes

characteristic of quiescence. J Biol Chem

2000;275:38221-9

45. Hui R, Finney GL, Carroll JS, et al.

Constitutive overexpression of cyclin

D1 but not cyclin E confers acute

resistance to antiestrogens in T-47D

breast cancer cells. Cancer Res

2002;62:6916-23

46. Stendahl M, Kronblad A, Ryden L, et al.

Cyclin D1 overexpression is a negative

predictive factor for tamoxifen response

in postmenopausal breast cancer patients.

Br J Cancer 2004;90:1942-8

47. Jirstrom K, Stendahl M, Ryden L, et al.

Adverse effect of adjuvant tamoxifen in

premenopausal breast cancer with cyclin

D1 gene amplification. Cancer Res

2005;65:8009-16

48. Rudas M, Lehnert M, Huynh A, et al.

Austrian Breast and Colorectal Cancer

Study Group. Cyclin D1 expression in

breast cancer patients receiving adjuvant

tamoxifen-based therapy.

Clin Cancer Res 2008;14:1767-74

49. Butt AJ, McNeil CM, Musgrove EA,

Sutherland RL. Downstream targets of

growth factor and oestrogen signalling

and endocrine resistance: the potential

roles of c-Myc, cyclin D1 and cyclin E.

Endocr Relat Cancer

2005;12(Suppl 1):S47-59

50. Bosco EE, Wang Y, Xu H, et al. The

retinoblastoma tumor suppressor

modifies the therapeutic response of

breast cancer. J Clin Invest

2007;117:218-28

51. Thangavel C, Dean JL, Ertel A, et al.

Therapeutically activating RB:

reestablishing cell cycle control in

endocrine therapy-resistant breast cancer.

Endocr Relat Cancer 2011;18:333-45

52. Perez-Tenorio G, Berglund F,

Esguerra Merca A, et al. Cytoplasmic

p21WAF1/CIP1 correlates with Akt

activation and poor response to

tamoxifen in breast cancer. Int J Oncol

2006;28:1031-42

53. Bianco S, Gevry N. Endocrine resistance

in breast cancer: from cellular signaling

pathways to epigenetic mechanisms.

Transcription 2012;3:165-70

54. McClelland RA, Barrow D, Madden TA,

et al. Enhanced epidermal growth factor

receptor signaling in MCF7 breast cancer

cells after long-term culture in the

presence of the pure antiestrogen ICI

182,780 (Faslodex). Endocrinology

2001;142:2776-88

55. Knowlden JM, Hutcheson IR, Jones HE,

et al. Elevated levels of epidermal growth

factor receptor/c-erbB2 heterodimers

mediate an autocrine growth regulatory

pathway in tamoxifen-resistant

MCF-7 cells. Endocrinology

2003;144:1032-44

56. Miller TW, Perez-Torres M,

Narasanna A, et al. Loss of Phosphatase

and Tensin homologue deleted on

chromosome 10 engages ErbB3 and

insulin-like growth factor-I receptor

signaling to promote antiestrogen

resistance in breast cancer. Cancer Res

2009;69:4192-201

57. Miller TW, Balko JM, Arteaga CL.

Phosphatidylinositol 3-kinase and

antiestrogen resistance in breast cancer.

J Clin Oncol 2011;29:4452-61

58. Ignatov A, Ignatov T, Weissenborn C,

et al. G-protein-coupled estrogen

receptor GPR30 and tamoxifen resistance

in breast cancer. Breast Cancer Res Treat

2011;128:457-66

59. Riggins RB, Bouton AH, Liu MC,

Clarke R. Antiestrogens, aromatase

inhibitors, and apoptosis in breast cancer.

Vitam Horm 2005;71:201-37

60. Miller TW, Balko JM, Fox EM, et al.

ERa-dependent E2F transcription can

mediate resistance to estrogen deprivation

in human breast cancer. Cancer Discov

2011;1:338-51

61. Paternot S, Bockstaele L, Bisteau X, et al.

Rb inactivation in cell cycle and cancer:

the puzzle of highly regulated activating

phosphorylation of CDK4 versus

constitutively active CDK-activating

kinase. Cell Cycle 2010;9:689-99

62. Lange CA, Yee D. Killing the second

messenger: targeting loss of cell cycle

control in endocrine-resistant breast

cancer. Endocr Relat Cancer

2011;18:C19-24

63. Collins I, Garrett MD. Targeting the cell

division cycle in cancer: CDK and cell

cycle checkpoint kinase inhibitors.

Curr Opin Pharmacol 2005;5:366-73

64. Canavese M, Santo L, Raje N. Cyclin

dependent kinases in cancer: potential for

therapeutic intervention.

Cancer Biol Ther 2012;13:451-7

65. Fry DW, Harvey PJ, Keller PR, et al.

Specific inhibition of cyclin-dependent

kinase 4/6 by PD 0332991 and

associated antitumor activity in human

tumor xenografts. Mol Cancer Ther

2004;3:1427-38

. The first preclinical study showing

in vitro and in vivo antitumor activity

in different neoplasms.

66. Finn RS, Dering J, Conklin D, et al. PD

0332991, a selective cyclin D kinase

4/6 inhibitor, preferentially inhibits

proliferation of luminal estrogen

receptor-positive human breast cancer cell

lines in vitro. Breast Cancer Res

2009;11:R77

. First report of differential activity of

palbociclib in different breast cancer

cell lines, representing the spectrum of

breast cancer subtypes.

67. Dean JL, Thangavel C, McClendon AK,

et al. Therapeutic CDK4/6 inhibition in

breast cancer: key mechanisms of

A. Rocca et al.

418 Expert Opin. Pharmacother. (2014) 15(3)

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 01

/27/

14Fo

r pe

rson

al u

se o

nly.

response and failure. Oncogene

2010;29:4018-32

.. The paper highlights potential

predictors of response and potential

mechanisms of resistance

to palbociclib.

68. Wang L, Wang J, Blaser BW, et al.

Pharmacologic inhibition of CDK4/6:

mechanistic evidence for selective activity

or acquired resistance in acute myeloid

leukemia. Blood 2007;110:2075-83

69. Gauthier ML, Berman HK, Miller C,

et al. Abrogated response to cellular stress

identifies DCIS associated with

subsequent tumor events and defines

basal-like breast tumors. Cancer Cell

2007;12:479-91

70. Arima Y, Inoue Y, Shibata T, et al. Rb

depletion results in deregulation of

E-cadherin and induction of cellular

phenotypic changes that are characteristic

of the epithelial-to-mesenchymal

transition. Cancer Res 2008;68:5104-12

71. Thoms HC, Dunlop MG, Stark LA.

CDK4 inhibitors and apoptosis: a novel

mechanism requiring nucleolar targeting

of RelA. Cell Cycle 2007;6:1293-7

72. Retzer-Lidl M, Schmid RM,

Schneider G. Inhibition of

CDK4 impairs proliferation of pancreatic

cancer cells and sensitizes towards

TRAIL-induced apoptosis via

downregulation of survivin. Int J Cancer

2007;121:66-75

73. Wesierska-Gadek J, Gritsch D,

Zulehner N, et al. Roscovitine, a selective

CDK inhibitor, reduces the basal and

estrogen-induced phosphorylation of ER-

a in human ER-positive breast cancer

cells. J Cell Biochem 2011;112:761-72

74. Chiron D, Martin P, Di Liberto M,

et al. Induction of prolonged early

G1 arrest by CDK4/CDK6 inhibition

reprograms lymphoma cells for durable

PI3Kd inhibition through PIK3IP1.

Cell Cycle 2013;12:1892-900

75. Roberts PJ, Bisi JE, Strum JC, et al.

Multiple roles of cyclin-dependent kinase

4/6 inhibitors in cancer therapy. J Natl

Cancer Inst 2012;104:476-87

. First report of potential antagonism

with chemotherapeutic agents.

76. McClendon AK, Dean JL,

Rivadeneira DB, et al. CDK4/

6 inhibition antagonizes the cytotoxic

response to anthracycline therapy.

Cell Cycle 2012;11:2747-55

. The paper shows further evidence of

antagonism between palbociclib and

doxorubicin in triple negative breast

cancer cell lines and xenografts.

77. Dean JL, McClendon AK, Knudsen ES.

Modification of the DNA damage

response by therapeutic CDK4/6

inhibition. J Biol Chem

2012;287:29075-87

. The paper demonstrates the

antagonism between CDK4/6

inhibition and cytotoxics acting with

different mechanisms, highlighting the

role of DNA-repair pathways.

78. Cabrera MC, Dıaz-Cruz ES,

Kallakury BV, et al. The CDK4/

6 inhibitor PD0332991 reverses

epithelial dysplasia associated with

abnormal activation of the

cyclin-CDK-Rb pathway. Cancer Prev

Res (Phila) 2012;5:810-21

79. Capparelli C, Chiavarina B,

Whitaker-Menezes D, et al. CDK

inhibitors (p16/p19/p21) induce

senescence and autophagy in cancer-

associated fibroblasts, "fueling" tumor

growth via paracrine interactions, without

an increase in neo-angiogenesis.

Cell Cycle 2012;11:3599-610

80. Schwartz GK, LoRusso PM,

Dickson MA, et al. Phase I study of PD

0332991, a cyclin-dependent kinase

inhibitor, administered in 3-week cycles

(Schedule 2/1). Br J Cancer

2011;104:1862-8

. First in human Phase I dose escalation

trial with two weeks on -- one week

off schedule.

81. Flaherty KT, Lorusso PM, Demichele A,

et al. Phase I, dose-escalation trial of the

oral cyclin-dependent kinase

4/6 inhibitor PD 0332991, administered

using a 21-day schedule in patients with

advanced cancer. Clin Cancer Res

2012;18:568-76

. First in human Phase I dose escalation

trial with three weeks on -- one week

off schedule.

82. DeMichele A, Clark AS, Heitjan D,

et al. A phase II trial of an oral CDK

4/6 inhibitor, PD0332991, in advanced

breast cancer. J Clin Oncol

2013;31s:abstract 519

83. Slamon DJ, Hurvitz SA, Applebaum S,

et al. Phase I study of PD 0332991,

cyclin-D kinase (CDK) 4/6 inhibitor in

combination with letrozole for first-line

treatment of patients with ER-positive,

HER2-negative breast cancer [abstract

3060]. J Clin Oncol 2010;28:15s

84. Finn RS, Crown JP, Boer K, et al.

Results of a randomized phase 2 study of

PD 0332991, a cyclin-dependent kinase

(CDK) 4/6 inhibitor, in combination

with letrozole vs letrozole alone for first-

line treatment of ER+/HER2- advanced

breast cancer (BC) [abstract 100O].

Ann Oncol 2012;23:ii43-5

85. Finn RS, Crown JP, Lang I, et al.

Results of a randomized phase 2 study of

PD 0332991, a cyclin-dependent kinase

(CDK) 4/6 inhibitor, in combination

with letrozole vs letrozole alone for first-

line treatment of ER+/HER2- advanced

breast cancer (BC). Cancer Res

2012;72:abstract nr S1-6

86. Finn RS, Dieras V, Gelmon KA, et al.

A randomized, multicenter, double-blind

phase III study of palbociclib (PD-

0332991), an oral CDK 4/6 inhibitor,

plus letrozole versus placebo plus

letrozole for the treatment of

postmenopausal women with ER(+),

HER2(--) breast cancer who have not

received any prior systemic anticancer

treatment for advanced disease.

J Clin Oncol 2013;31s:abstract TPS652

87. Dickson MA, Tap WD, Keohan ML,

et al. Phase II trial of the

CDK4 inhibitor PD0332991 in patients

with advanced CDK4-amplified well-

differentiated or dedifferentiated

liposarcoma. J Clin Oncol

2013;31:2024-8

88. Johnston S, Pippen J Jr, Pivot X, et al.

Lapatinib combined with letrozole versus

letrozole and placebo as first-line therapy

for postmenopausal hormone receptor-

positive metastatic breast cancer.

J Clin Oncol 2009;27:5538-46

89. Kaufman B, Mackey JR, Clemens MR,

et al. Trastuzumab plus anastrozole

versus anastrozole alone for the treatment

of postmenopausal women with human

epidermal growth factor receptor

2-positive, hormone receptor-positive

metastatic breast cancer: results from the

randomized phase III TAnDEM study.

J Clin Oncol 2009;27:5529-37

90. Baselga J, Campone M, Piccart M, et al.

Everolimus in postmenopausal

hormone-receptor-positive advanced

breast cancer. N Engl J Med

2012;366:520-9

Palbociclib (PD 0332991)

Expert Opin. Pharmacother. (2014) 15(3) 419

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 01

/27/

14Fo

r pe

rson

al u

se o

nly.

91. Yardley DA, Ismail-Khan RR,

Melichar B, et al. Randomized phase II,

double-blind, placebo-controlled study of

exemestane with or without entinostat in

postmenopausal women with locally

recurrent or metastatic estrogen

receptor-positive breast cancer

progressing on treatment with a

nonsteroidal aromatase inhibitor.

J Clin Oncol 2013;31:2128-35

AffiliationAndrea Rocca†1, Alberto Farolfi1,

Sara Bravaccini2, Alessio Schirone1 &

Dino Amadori1

†Author for correspondence1Istituto Scientifico Romagnolo per lo Studio e la

Cura dei Tumori (IRST) IRCCS,

Department of Medical Oncology, Meldola, Italy

Tel: +39 0543 739100;

Fax: +39 0543 739151;

E-mail: [email protected] Scientifico Romagnolo per lo Studio e la

Cura dei Tumori (IRST) IRCCS,

Biosciences Laboratory, Meldola, Italy

A. Rocca et al.

420 Expert Opin. Pharmacother. (2014) 15(3)

Exp

ert O

pin.

Pha

rmac

othe

r. D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

Sydn

ey o

n 01

/27/

14Fo

r pe

rson

al u

se o

nly.


Recommended