+ All Categories
Home > Documents > Targeted therapies in gynecologic cancers and melanoma

Targeted therapies in gynecologic cancers and melanoma

Date post: 20-Nov-2023
Category:
Upload: lleida
View: 0 times
Download: 0 times
Share this document with a friend
12
Targeted therapies in gynecologic cancers and melanoma Eugenia Ortega, Rosa M. Marti, Andree Yeramian, Anabel Sorolla, Xavier Dolcet, David Llobet, Leandro Abal, Maria Santacana, Judit Pallares, Antonio Llombart–Cussac, Xavier Matias–Guiu From the Departments of Medical Oncology, Dermatology, and Pathology, and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida–IRB Lleida, Lleida, Spain. The article reviews the main molecular pathology alterations of endometrial and ovarian carcinomas and melanoma. Several promising drugs targeting the genes most frequently altered in these tumors are under consideration. The most promising signaling pathways to be targeted for therapies in these tumors are the tyrosine kinase receptor (EGFR, HER2, c-KIT), the RAS/B-RAF/MAPK, the PI3K–mTOR, and apoptosis signaling pathways. © 2008 Elsevier Inc. All rights reserved. KEYWORDS Endometrial carcinoma; Ovarian carcinoma; Melanoma; Target therapies Endometrial carcinoma Molecular features of endometrial carcinoma There are two main clinicopathologic variants of endo- metrial carcinoma (EC) (Tables 1 and 2). According to our current knowledge about EC, type I tumors are low-grade and estrogen-related endometrioid carcinomas (EEC) (Fig- ure 1) that usually develop in perimenopausal women and coexist or are preceded by complex and atypical endome- trial hyperplasia. In contrast, type II tumors are non- endometrioid (mainly papillary serous) carcinomas (NEEC) (Figure 2), very aggressive tumors, unrelated to estrogen stimulation, arising occasionally in endometrial polyps or from precancerous lesions developing in atrophic endome- trium, that mainly occur in older women. cDNA analysis clearly shows that EEC and NEEC exhibit different expres- sion profiles. 1 The molecular alterations involved in the development of EEC (type I) carcinomas are different from those of NEEC (type II) carcinomas. 2–4 Whereas EEC shows microsatellite instability (MI) and mutations in the PTEN, PIK3CA, k- RAS, and -catenin (CTNNB1) genes, 5–9 NEEC exhibit alterations of p53, loss of heterozygosity (LOH) on several chromosomes, as well as other molecular alterations (STK15, p16, E-cadherin and C-erb B2). 10,11 Some of the genes that are abnormal in the two forms of EC may be good targets for therapeutic approaches. Targeted therapies in endometrial carcinoma The PI3K/AKT pathway is the most frequent abnormal signaling pathway in EEC, often resulting from mutations in the tumor suppressor gene PTEN, and activating mutations in PIK3CA. 6,7,12–14 The tumor suppressor gene termed PTEN, located on chromosome 10q23.3, is frequently ab- normal in endometrial carcinomas. LOH at chromosome 10q23 occurs in 40% of EC. Somatic PTEN mutations are also common in EC, and they are almost exclusively re- stricted to EEC, occurring in 37% to 66% of them. PTEN antagonizes the PI3K/AKT pathway by dephosphorylating PIP3, leading to a decreased translocation of AKT to cel- lular membranes, and subsequent downregulation of AKT phosphorylation and activation. PTEN mutations have been detected in endometrial hyperplasias with and without Address reprint requests and correspondence: Xavier Matias–Guiu, Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova, Av Alcalde Rovira Roure 80, 25198 Lleida, Spain. E-mail address: [email protected]. 0740-2570/$ -see front matter © 2008 Elsevier Inc. All rights reserved. doi:10.1053/j.semdp.2008.07.008 Seminars in Diagnostic Pathology (2008) 25, 262-273
Transcript

T

EDA

FL

E

M

mcaucte(sftcs

E

DA

0d

Seminars in Diagnostic Pathology (2008) 25, 262-273

argeted therapies in gynecologic cancers and melanoma

ugenia Ortega, Rosa M. Marti, Andree Yeramian, Anabel Sorolla, Xavier Dolcet,avid Llobet, Leandro Abal, Maria Santacana, Judit Pallares,ntonio Llombart–Cussac, Xavier Matias–Guiu

rom the Departments of Medical Oncology, Dermatology, and Pathology, and Molecular Genetics and Research

aboratory, Hospital Universitari Arnau de Vilanova, University of Lleida–IRB Lleida, Lleida, Spain.

The article reviews the main molecular pathology alterations of endometrial and ovarian carcinomasand melanoma. Several promising drugs targeting the genes most frequently altered in these tumors areunder consideration. The most promising signaling pathways to be targeted for therapies in these tumorsare the tyrosine kinase receptor (EGFR, HER2, c-KIT), the RAS/B-RAF/MAPK, the PI3K–mTOR, andapoptosis signaling pathways.© 2008 Elsevier Inc. All rights reserved.

KEYWORDSEndometrialcarcinoma;Ovarian carcinoma;Melanoma;Target therapies

(iRac(gg

T

stiPn1asaPlp

ndometrial carcinoma

olecular features of endometrial carcinoma

There are two main clinicopathologic variants of endo-etrial carcinoma (EC) (Tables 1 and 2). According to our

urrent knowledge about EC, type I tumors are low-gradend estrogen-related endometrioid carcinomas (EEC) (Fig-re 1) that usually develop in perimenopausal women andoexist or are preceded by complex and atypical endome-rial hyperplasia. In contrast, type II tumors are non-ndometrioid (mainly papillary serous) carcinomas (NEEC)Figure 2), very aggressive tumors, unrelated to estrogentimulation, arising occasionally in endometrial polyps orrom precancerous lesions developing in atrophic endome-rium, that mainly occur in older women. cDNA analysislearly shows that EEC and NEEC exhibit different expres-ion profiles.1

The molecular alterations involved in the development ofEC (type I) carcinomas are different from those of NEEC

Address reprint requests and correspondence: Xavier Matias–Guiu,epartment of Pathology and Molecular Genetics, Hospital Universitarirnau de Vilanova, Av Alcalde Rovira Roure 80, 25198 Lleida, Spain.

dE-mail address: [email protected].

740-2570/$ -see front matter © 2008 Elsevier Inc. All rights reserved.oi:10.1053/j.semdp.2008.07.008

type II) carcinomas.2–4 Whereas EEC shows microsatellitenstability (MI) and mutations in the PTEN, PIK3CA, k-AS, and �-catenin (CTNNB1) genes,5–9 NEEC exhibitlterations of p53, loss of heterozygosity (LOH) on severalhromosomes, as well as other molecular alterationsSTK15, p16, E-cadherin and C-erb B2).10,11 Some of theenes that are abnormal in the two forms of EC may beood targets for therapeutic approaches.

argeted therapies in endometrial carcinoma

The PI3K/AKT pathway is the most frequent abnormalignaling pathway in EEC, often resulting from mutations inhe tumor suppressor gene PTEN, and activating mutationsn PIK3CA.6,7,12–14 The tumor suppressor gene termedTEN, located on chromosome 10q23.3, is frequently ab-ormal in endometrial carcinomas. LOH at chromosome0q23 occurs in 40% of EC. Somatic PTEN mutations arelso common in EC, and they are almost exclusively re-tricted to EEC, occurring in 37% to 66% of them. PTENntagonizes the PI3K/AKT pathway by dephosphorylatingIP3, leading to a decreased translocation of AKT to cel-

ular membranes, and subsequent downregulation of AKThosphorylation and activation. PTEN mutations have been

etected in endometrial hyperplasias with and without

arPievmawcbwhepsPPpPbadao

s

Tptltbrtcpta4migRar

Fc

F

263Ortega et al Target Therapies in Gynecologic Cancers and Melanoma

typia (19% and 21%, respectively), both of them currentlyegarded as precursor lesions of EEC.13 Moreover, identicalTEN mutations have been detected in hyperplasias coex-

sting with EEC, which suggests that PTEN mutations arearly events in the development of EEC. There are contro-ersial data regarding the prognostic significance of PTENutations in EC, but there are some results that suggest

ssociation with favorable prognostic factors. In agreementith Knudson’s two-hit proposal, LOH at 10q23 frequently

oexists with somatic PTEN mutations. The coexistence ofoth alterations leads to activation of the PI3K/AKT path-ay, which plays a key role in the regulation of cellularomeostasis. Activated AKT modulates the expression of sev-ral genes involved in suppression of apoptosis and cell cyclerogression. Moreover, mutations in PIK3CA have been de-cribed in EC and may contribute to the alteration of theI3K/AKT signaling pathway in endometrial carcinoma.7,14

TEN has been shown to play several roles in tumor sup-ression and promotion of apoptosis. The importance of theI3K/PTEN/AKT survival pathway in EC raises the possi-ility that PI3K inhibitors, such as wortmannin and deriv-tives, may be used as potential anticancer agents. In fact, aecrease of AKT phosphorylation and increased apoptosisre seen in mutated PTEN human EC cells in the presencef PI3K inhibitor.15

Of particular interest among AKT targets is the down-tream effector mammalian target of rapamycin (mTOR).

Table 1 Clinical and pathological features of types I andII, endometrial carcinoma

Type I Type II

Age Pre- andperimenopausal

Postmenopausal

Unopposed estrogen Present AbsentHyperplasia precursor Present AbsentGrade Low HighMyometrial invasion Minimal DeepSpecific subtypes Endometrioid

carcinomaNon-endometrioidcarcinoma

Behavior Stable Progressive

Table 2 Molecular features of types I and II, endometrialcarcinoma

Type I/EC Type II/NEEC

Microsat instability 20–40% 0–5%PTEN mutation 35–50% 10%K-RAS mutation 15–30% 0–5%PIK3CA mutation 30% 5%CTNNB1 mutation 25–40% 0–5%p53 alteration 10–20% 90%p16 inactivation 10% 40%E-cadherin inactivation 10–20% 80–90%c-erb B2 amplification 2.5% 26%Cyclins D1 and E amplification 2–5% 26–42%

r

he TOR family of proteins has pleiotropic functions andarticipates in the regulation of the initiation of mRNAranscription and protein translation in response to intracel-ular concentrations of amino acids and other essential nu-rients in the organization of the actin cytoskeleton, mem-rane trafficking, protein degradation, PKC signaling, andibosome biogenesis. mTOR regulates essential transduc-ion pathways and is involved in coupling growth stimuli toell cycle progression. AKT activates mTOR via directhosphorylation of TCS2 and by the inhibition of AMPPK,hereby activating Rheb and mTOR–Raptor activity. Onctivation, mTOR–Raptor activates S6K and inhibitsEBP1 to accelerate mRNA translation. However, theTOR pathway can also be activated by other mechanisms,

ncluding activation of tyrosine kinase receptors (epidermalrowth factor receptors EGFR1-4, PDGFR, KIT, IGFR) andas. Moreover, loss of function of p53 may also result inctivation of mTOR. mTOR inhibitors (rapamycin andapamycin derivatives) have been recently developed as

igure 1 Microscopical appearance of an endometrioid adeno-arcinoma of the endometrium.

igure 2 Microscopical appearance of a nonendometrioid (se-

ous) carcinoma of the endometrium.

pithmm(imdamn

cgac(baaodnslTMRatE((etcNsaacaasrmt

miipca

enabopwa(obo

hrmitmhtbatgcitaBnEriosBdsNspoicAaRtz

alia

264 Seminars in Diagnostic Pathology, Vol 25, No 4, November 2008

otential anticancer agents. Tumors associated with PTENnactivation, like EC, are particularly susceptible to theherapeutic effects of mTOR inhibitors. Several mTOR in-ibitors are available for clinical trials: the prototype rapa-ycin and three rapamycin derivatives, CCI-779 (temsiroli-us), RAD001 (everolimus), and AP23573. Both CCI-779

temsirolimus) and RAD001 (everolimus) are currently be-ng tested in two phase II trials, in recurrent EC.16 PTEN�/�

ice is a good model to test sensitivity of EC to anticancerrugs, because they develop complex atypical hyperplasiand endometrial carcinoma. Pharmacological inhibition ofTOR by CCI-779 in PTEN�/� mice has shown reduced

eoplastic proliferation, tumor size, and S6K activity.17

Tyrosine kinase receptors are also good targets for anti-ancer therapies. The epidermal growth factor family and itsrowth factors are known to play critical roles in cell growthnd differentiation. The epidermal growth factor family isomprised of EGFR (ErbB1), HER-2/neu (ErbB2), HER-3ErbB3), and HER-4 (ErbB4). EGFR and HER-2/neu haveeen shown to be highly expressed in normal endometriumnd overexpressed in endometrial cancer, where it has beenssociated with a poor prognosis.18,19 Increased expressionf EGF-related protein and EGFR may contribute to arug-resistant phenotype. EGFR has intrinsic tyrosine ki-ase activity which is activated on ligand binding. Down-tream serine/threonine kinase cascades are then induced,eading to cell proliferation and the inhibition of apoptosis.yrosine kinase inhibitors prevent the phosphorylation ofAP kinase Erk1/2, MAP kinase MEK 1/2, CDK 1, AKT,AF 1, and Rb-1. Inhibition of EGFR with monoclonalntibodies leads to growth arrest, and a similar and poten-ially synergistic effect is anticipated with inhibition ofGFR tyrosine kinase activity. Potential agents are IressaZD1839), Herceptin (trastuzumab), and LapatinibGW572016). The signaling pathways of EGFR, under theffect of EGF and ZD1839, were evaluated in EC cell lines;he results showed that both EEC and NEEC have theapacity to respond to EGFR inhibition, but the response ofEEC may be limited by the constitutive activation of other

ignaling pathways. ZD1839 has been studied as a singlegent in a phase II trial (GOG 229C) of women withdvanced EC. Preliminary data in 29 patients have shown 1omplete response and stable disease in several patientsfter 6 months. GW572016 (Lapatinib) was used as a singlegent in a similar cohort of patients. Moreover, OSI-774howed partial responses in another phase II study in 23ecurrent or metastatic EC patients, with no previous che-otherapy and no more than 1 cycle of prior hormonal

herapy.The EGFR pathway may be induced by steroid hor-

ones, which is the principal growth-promoting mechanismn the endometrium. Estrogen and progesterone are the mostmportant steroid hormones that modulate endometrial cellroliferation and differentiation, respectively, and their re-eptors (ER and PR) are expressed in about 80% of EEC,

lthough the proportion may decrease with a loss of differ- i

ntiation. NEEC are not responsive to hormones and mayot express ER and PR, therefore they present a constitutivectivation of EGFR. Medroxiprogestererone acetate haseen studied in EC patients. Two large Gynecologic Oncol-gy Group (GOG) trials evaluating oral progestins in theseatients showed an overall response rate of 15% to 25%ith median progression-free survival of less than 4 months

nd overall survival of less than 11 months. Fulvestrantfaslodex), a novel antiestrogen classified as an ER antag-nist without known agonist activity in the endometrium, iseing evaluated in a phase II study in patients with recurrentr metastatic EC.

Apoptosis is a key process in the regulation of cellularomeostasis. Deregulation of apoptosis plays an importantole in development and progression of cancer. There areany evidences suggesting that alteration of apoptosis is

mportant in development and progression of EC. Several ofhe molecular abnormalities that have been detected in ECay be associated with apoptosis deregulation. EEC show a

igh frequency of mutations in PTEN, which lead to constitu-ively active Akt, which in turn suppresses apoptosis triggeredy various stimuli. Moreover, the recent evidence that NF-�Bctivation is frequent in endometrial carcinoma20 may explainhe presence of apoptosis resistance by activation of targetenes such as FLIP and Bcl-XL. p53 alterations, which areharacteristic of NEEC, may also occur in EEC, particularlyn those neoplasms showing overlapping features betweenypes I and II tumors; and they may have an impact inpoptosis at several different levels. Also, members of thecl-2 family of genes are abnormal in endometrial carci-oma. For example, BAX is a target gene for mutations inEC with microsatellite instability and may have a role in

esistance to apoptosis in these tumors.21 Proteasome inhib-tors are currently used as chemotherapeutic drugs becausef their ability to trigger cell growth arrest or apoptosis oneveral tumors. In many different types of tumor cells,ortezomib and other proteasome inhibitors cause celleath by blocking NF-�B activity. However, in EC, protea-ome inhibitors induce cell death, but, instead of blockingF-�B, they increase its transcriptional activity. Protea-

ome inhibitors induce phosphorylation of IKK�/�, phos-horylation and degradation of IkB�, and phosphorylationf p65 NF-�B subunit on serine 536. Proteasome inhibitor-nduced cell death was accompanied by activation ofaspases and apoptotic nuclear morphology22,23 (Figure 3).

key molecule involved in resistance to TRAIL-inducedpoptosis is protein kinase CK2, by modulating FLIP.24,25

esults from our group show that pharmacological inhibi-ion of CK2 by DRB, Apigenin, or Emodin causes sensiti-ation of EC cell lines to TRAIL-induced apoptosis.

Sorafenib (BAY 43-9006, Nexavar) is a potent, orallydministered receptor tyrosine kinase inhibitor with antipro-iferative and antiangiogenic activities. Sorafenib was orig-nally described as an inhibitor of B- and c-RAF kinase, butlso has activity against several receptor tyrosine kinases,

ncluding vascular endothelial growth factor receptor 2

((sgcachmhSb

taetcaoeacsemnsiHisc

csitiwdgg

O

M

tSthncs

F

Feo aved c

265Ortega et al Target Therapies in Gynecologic Cancers and Melanoma

VEGFR2), platelet-derived growth factor receptorPDGFR), FLT3, Ret, and c-Kit. The antitumor activity oforafenib may be attributed to inhibition of tumor angio-enesis (VEGFR and PDGFR) and direct effects on tumorell proliferation/survival (Raf kinase signaling-dependentnd signaling-independent mechanisms). A recent multi-enter phase II trial in patients with advanced/recurrent ECas shown that Sorafenib may result in clinical benefit for ainority of EC patients. Preliminary results from our group

ave shown that pharmacological inhibition of B-RAF byorafenib sensitized EC cells to TRAIL-induced apoptosis,y down-regulating FLIP (unpublished results).

Histone acetylation is one of the mechanisms involved inhe epigenetic control of gene expression. Altered histonecetylation in cancer cells may be responsible for abnormalxpression of oncogenes and tumor suppressor genes. His-one deacetylase inhibitors (HDACI) are promising antican-er drugs. In cancer cells, HDACI cause cell-cycle arrest,poptosis, and differentiation. HDACI cause derepressionf genes whose reactivation would promote an antiprolif-rative effect. Examples of genes upregulated by HDACIre p21, TRAIL-R2, p19ARF, Bmf, and Rap1. Paradoxi-ally, HDACI cause downregulation of important genesuch as thymidylate synthetase, Bcr-Abl, and c-Myc. Inter-stingly, microarray studies have shown that HDACI treat-ent leads to changes in RNA levels of a surprisingly small

umber of genes (around 2–5% of the genome). Sometudies have suggested that HDACI may be more effectiven tumors expressing mutant p53. Preclinical studies usingDACI in endometrial carcinoma cell lines have provided

nteresting results.26,27 For example, HDACI have beenhown to induce differentiation of endometrial carcinomaell lines, which resemble normal endometrial epithelial

Table 3 Molecular features of ovarian carcinoma

Low-grade serous carcinoma B-RAF and K-RAS mutationsHigh-grade serous carcinoma P53, LOH 17q, LOH13qEndometroid carcinoma Microsatellite instability,

PTEN, K-RAS, PIK3CA,CTNNB1 mutations

Mucinous carcinoma K-RAS mutation

igure 3 The proteasome inhibitors MG-132 and bortezomibndometrioid carcinoma of the endometrium. Tumor cells were leftr bortezomib for 24 hours. Pictures show Hoechst 33258 and cle

s

ells in the absence of ovarian steroid hormones. Moreover,ome studies have shown that HDACI had important growthnhibitory effect on EC cell lines, by decreasing the propor-ion of cells in S phase and increasing the proportion of cellsn the G0-G1 and or G2-M phases of the cell cycle. Thereas also upregulation of p21, p27, and E-cadherin, andownregulation of Bcl-2 and cyclin-D1 and -D2. Therowth-suppressor effects seem to be irrespective of the p53ene status.

varian carcinoma

olecular features of ovarian carcinoma

There are different types of ovarian carcinoma, each ofhem showing different molecular features (Table 3).28,29

erous tumors account for 30% to 40% of all ovarianumors. A dualistic model of serous ovarian carcinogenesisas recently been proposed. According to this proposal, theatural history and molecular features of low-grade serousarcinoma are different from those of high-grade ovarianerous carcinomas (Figure 4). Low-grade serous carcinomas

igure 4 Microscopical appearance of a high-grade, ovarian

apoptotic cell death in a tumor cell explant obtained from aned (UN) or treated with 0.5 �mol L�1 or 25 nmol L�1 of MG-132aspase-3 staining.

induceuntret

erous carcinoma.

wtcnmaoBam

taamamsPtooc(rpc

mttbtt

cs

T

p

tsaVia

A

atoolctpmfcnsGp3tat(

E

sg(att

266 Seminars in Diagnostic Pathology, Vol 25, No 4, November 2008

ould develop from preexisting serous borderline tumors,hrough oncogenic activation of K-RAS and B-RAF. Inontrast, high-grade serous carcinomas would develop deovo from the epithelial cells of the ovarian surface. Theain molecular feature of the “high-grade” pathway is

lteration of the p53 tumor suppressor gene. However, LOHn 13q and 17q, involving the loci of BRCA-2 andRCA-1, as well as BRCA-1 promoter hypermethylation,re also frequent in sporadic high-grade serous carcino-as.30,31

Endometrioid and clear cell carcinomas account for 10%o 20% and 6% of ovarian carcinomas, respectively. Theyre morphologically similar to their uterine counterparts,nd they frequently arise from ovarian endometriosis. Theolecular features of ovarian endometrioid carcinomas are

lso similar to those of endometrioid endometrial carcino-as. They are also characterized by the presence of micro-

atellite instability and mutations in PTEN, k-RAS,IK3CA, and CTNNB1 gene.32 However, the frequency of

hese alterations is quite different, with increased prevalencef mutations in the CTNNB1 gene. The molecular featuresf ovarian clear cell carcinoma are under evaluation. In-reased expression of hepatocyte nuclear factor 1�HNF1�) and mutations in TGF�RII (exon 7) have beeneported. In this regard, HNF1� has been proposed as aossible molecular target for therapy in ovarian clear cellarcinoma.33

Ovarian mucinous carcinomas are rare, and the vastajority of ovarian carcinomas with mucinous differentia-

ion are, indeed, metastatic tumors from carcinomas fromhe large bowel, appendix, or pancreas. K-RAS appears toe the most typical molecular feature of ovarian mucinousumors (either of the benign, borderline, or malignantype).34

Moreover, the molecular features of transitional cell car-inomas and undifferentiated carcinomas are poorly under-tood.

argeted therapies in ovarian carcinoma

Numerous potential therapeutic targets have been pro-

Table 4 Targeted therapies proposed for ovarian carcinoma

Agent Type Targ

Bevazizumab (Avastin) mAb VEG

Gefitinib (Iressa) TKI EGFErlotinib (Tarceva) TKI EGF

Imatinib (Gleevec) TKI c-ABTrastuzumab (Herceptin) mAb HERPertuzumab (Omnitarg) mAb HERCetuximab (Erbitux) mAb EGFLapatinib (Tykerb) ITK EGF

HER

osed based on prognostic studies, but large randomized r

rials assessing relevant agents are now only in the planningtages (Table 4). Drugs targeting some of the most commonlterations observed in ovarian cancer are those affectingEGF or its receptors (since VEGF production is common

n ovarian cancer and is the basis for development of ascitis)nd EGFR.

ntiangiogenic agents

A solid tumor depends on angiogenesis to grow in sizend to disseminate. VEGF is one of the main factors relatedo vascular proliferation. Overexpression of VEGF has beenbserved in 53% to 97% of ovarian cancers. Althoughverexpression correlates with poor survival in other ma-ignancies, including colorectal, gastric, breast, and prostateancer, its prognostic value in ovarian cancer is still con-roversial. Therefore, antiangiogenic agents represent aromising anticancer strategy. Bevacizumab, a humanizedurine monoclonal antibody against VEGF, blocks all iso-

orms of VEGF. Several phase II trials have investigated thelinical benefit of bevacizumab, either alone or in combi-ation with commonly used chemotherapy regimens in per-istent or recurrent epithelial ovarian cancer patients. TheOG 170D study, using bevacizumab as monotherapy in 62atients with persistent or recurrent ovarian cancer, showedcomplete responses and 8 partial responses with 54.8% of

he patients experiencing stable disease. This drug is actu-lly being tested in phase III first-line trials, either as main-enance therapy or in combination with chemotherapyGOG 218 and ICON 7) in ovarian cancer patients.35

GFR family

The EGFR superfamily is made up of four distinct buttructurally similar tyrosine kinase receptors: epidermalrowth factor receptor (EGFR/HER1/ErbB1), HER2ErbB2), HER3 (ErbB3), and HER4 (ErbB4). Activation ofreceptor occurs by dimerization between two identical or

wo different receptors. This results in activation of theyrosine kinase domain with subsequent activation of inter-

chanism Route Phase

IV IIIII ongoing

sible inhibitor oral IIsible inhibitor oral II

III ongoingIT, PDGFR oral II

IV IIIV IIIV IIoral I

et/me

F

R/reverR/rever

L, c-K-2-2RR-2

elated intracellular signaling pathways, such as the phos-

pMewiEorhposratPias2temEtstrciceaprhcbcsnao

momtbeatchct

cschcmavHmGro2fAaodmzizEbtttpPgmriHmspp

iptoeglidw

tanu

267Ortega et al Target Therapies in Gynecologic Cancers and Melanoma

hatidylinositol 3-kinase (PI3K)–AKT pathway and theAPK pathway. These mechanisms are involved in differ-

nt cellular processes, and their disregulation is associatedith malignant transformation, being responsible for driv-

ng proliferation, invasion, and survival of cancer cells.GFR overexpression has been found in 12% to 82% ofvarian cancers, whereas aberrant HER2 expression waseported in 5% to 66% of ovarian carcinomas. Some studiesave shown that overexpression of EGFR is an independentrognosis factor for poor outcome. Understanding the rolef the EGFR family in cancer has led to the development ofeveral strategies, including development of inhibitors ofeceptor tyrosine kinase domain (TKI), and monoclonalntibodies (mAbs) against the extracellular domain.36 Bothypes of agents result in downregulation of the MAPK, andI3K/AKT pathways. The TKI gefitinib (ZD 1839, Iressa),

n a phase II study combined with paclitaxel and carboplatins second-line therapy in 40 patients platinum-resistant orensitive ovarian cancer, showed overall responses rates of5% and 71% in the resistant and sensitive groups, respec-ively. A number of phase II trails have evaluated thefficacy of gefitinib as a single agent in ovarian cancer, withinimal activity in patients without activating mutations inGFR. Two independent research groups have identified

hat the majority of patients responding to gefitinib in otherolid tumors had specific activating mutations within theyrosine kinase domain, whereas patients who had wild-typeeceptors did not have response. Both of these groups con-luded that specific mutations in the EGFR gene led toncreased growth factor signaling through this pathway andonferred susceptibility to the TKI gefitinib, as well asrlotinib, but do not seem to predict response to EGFRntibodies. Another TKI, erlotinib (OSI-774, Tarceva), in ahase II clinical trial in platinum-resistant ovarian cancer,eported 42% of stable disease in 34 patients, and 2 (5.8%)ad partial responses. A phase III trial of docetaxel andarboplatin with or without erlotinib in ovarian cancer iseing done by Scottish Randomized Trial in Ovarian Can-er. Cetuximab (Erbitux), an anti-EGFR antibody, hashown, in preliminary data from a phase II trial in combi-ation with paclitaxel and carboplatin, that it is well toler-ted in 31 previously untreated patients with advancedvarian cancer.

The HER-2 (c-erbB2) proto-oncogene encodes a trans-embrane receptor, which is overexpressed in several types

f human carcinomas and is an attractive therapeutic target.Abs to HER-2 can inhibit the proliferation of tumor cells

hat overexpress this gene. A humanized monoclonal anti-ody, Herceptin (trastuzumab), has beneficial therapeuticffects in primary breast carcinomas patients with HER-2mplification, particularly when combined with chemo-herapeutic drugs. However, only 25% of primary ovarianarcinomas show HER-2 overexpression by immuno-istochemistry. Moreover, unlike breast cancer, it isontroversial to what extent HER-2 amplification and pro-

ein overexpression correlates with prognosis in ovarian o

ancer. It has, however, been reported that HER-2 expres-ion is more frequent in ovarian carcinomas relapsing afterhemotherapy, suggesting that cells expressing this proteinave a selective growth advantage over HER-2-negativeells. This finding suggests that patients with ovarian canceray benefit from treatment with anti-HER-2 monoclonal

ntibodies plus chemotherapeutic drugs, both at an ad-anced stage (when the majority of tumor cells expressER-2) and at an earlier time, to eliminate the potentiallyore malignant HER-2-positive tumor cells. A phase IIOG trial showed, among 95 patients with persistent or

ecurrent ovarian cancer with HER2 overexpression, anverall response rate of 7.3% with 1 complete response and

partial responses.37 Interestingly, no correlation wasound between HER2 expression with clinical outcome.lso, there was no correlation between gene amplification

nd HER2 overexpression, suggesting that the biologic rolef HER2 in ovarian cancer may differ from that previouslyemonstrated in breast cancer. A different HER-2-targetedonoclonal antibody is pertuzumab. In contrast to trastu-

umab, pertuzumab sterically blocks HER-2 heterodimer-zation with other HER receptors. In other words, pertu-umab blocks ligand-activated signaling from HER-2/GFR and HER-2/HER-3 heterodimers. The pertuzumabinding site of extracellular domain II does not overlap withhe epitope on HER-2, which is the one recognized byrastuzumab. In a phase II trial, 130 patients with previouslyreated ovarian cancer received pertuzumab intravenouslylus gemcitabine or placebo. The adjusted hazard ratio forFS was 0.67 in favor of pertuzumab, with a median pro-ression-free survival (PFS) of 3.0 months versus 2.6onths. More recently, Gordon and coworkers reported the

esults of a phase II trial using pertuzumab as a single agentn 123 patients with ovarian cancer.38 PhosphorylatedER2 expression was studied, suggesting that pertuzumabay be active in ovarian cancer and that HER2 activation

tatus, in the absence of HER 2 overexpression, may beotentially predictive for identifying tumors responsive toertuzumab.

Lapatinib is an orally active small molecule that revers-bly inhibits EGFR and HER2 tyrosine kinases. Varioushase I trials have been conducted in patients with solidumors including ovarian cancer, with preliminary evidencef antitumor activity in heavily pretreated patients. How-ver, no phase II studies have been reported to date inynecological malignancies. The potential advantages ofapatinib over other tyrosine kinase inhibitors are that itnhibits two receptors and thus may inhibit multiple receptorimers and their corresponding downstream signaling path-ays.Imatinib mesylate (STI-571, Gleevec), an adenosine

riphosphate-binding selective inhibitor of BCL/ABL, KIT,nd PDGFR, has been evaluated in some phase II trials. Inormal ovarian surface epithelium, c-ABL was expressedniversally. PDGFR-� was expressed in the majority (93%)

f samples of normal ovarian epithelium, whereas the c-KIT

plcnnoqcimoitaotr

E

lwc

T

oBmspsHilBt

T

fatRMtot4pog

Is

M

M

a1taisnapaen

tCwndciCG1mdf

pR

268 Seminars in Diagnostic Pathology, Vol 25, No 4, November 2008

rotein was undetectable in normal ovarian surface epithe-ium. Overall, c-ABL was expressed in 71% of serousarcinomas [more frequently in the low-grade serous carci-omas (81%) compared with the high-grade serous carci-omas (65%)]. PDGFR-� expression was observed in 81%f serous carcinomas overall and was observed more fre-uently in higher-grade tumors. c-KIT immunohistochemi-al staining was absent in low-grade tumors but was presentn 26% of high-grade serous carcinomas. In conclusion, theajority of ovarian serous carcinomas express one or more

f the kinases targeted by the tyrosine kinase inhibitor,matinib mesylate, suggesting the potential usefulness ofhis drug in the treatment of ovarian carcinoma. However, in

phase II clinical trial, including patients with recurrentvarian cancer whose tumors overexpressed at least one ofhe known molecular targets of imatinib, there were noesponses during a median follow-up of 6.6 months.

strogen receptor signaling pathway

Estrogen growth factors, including EGF, insulin, insulin-ike growth factor-1, and TGF-�, activate estrogen receptor,hich can activate the PI3K/AKT cell survival pathway and

an also activate the EGF family of receptors.

argeting the apoptosis pathway

G3139 (Oblimersen) is an antisense phosphorothioateligodeoxynucleotide that suppresses BCL2 expression.CL2 family proteins play a central role in controlling theitochondria pathway. This family includes proteins that

uppress apoptosis, such as BCL2. Overproduction of BCL2rotein prevents programmed cell death, enhances meta-tatic potential, and promotes resistance to chemotherapy.igh expression of BCL2 proto-oncogene is found in var-

ous solid tumors, including ovarian cancer. Moreover, mo-ecular targeting of BCL2 and Bcl-XL proteins by syntheticCL2 homology 3 domain peptide has shown to enhance

he efficacy of chemotherapy.39

argeting signal transduction

Aberrant or overactive signal transduction has beenound to enhance proliferation, invasiveness, metastasis,nd angiogenesis, and to confer poor response to conven-ional cytotoxic agents. One of these pathways is the RAS/AF/MAPK. Constitutive activation of the RAS/RAF/APK pathway components has been seen in primary

umor cell. In this way, RAS gene mutations have beenbserved in 18% of ovarian cancers, especially in mucinousumors, but also in low-grade serous carcinomas. BAY3-9006 (sorafenib) is currently in evaluation in severalhase II assays for the treatment of recurrent or refractoryvarian cancer alone or with paclitaxel, carboplatin, and

emcitabine. An in vitro and clinical/pharmacological phase

study has recently tested the combination of irinotecan andorafenib.40

elanoma

olecular features of melanoma

Development of malignant melanoma is associated withllelic deletions (LOH) at several chromosomes, includingp, 6q, 9p, 10q, 11q, and 17q. Moreover, there are some locihat correspond to familial susceptibility genes (1p36, 9p21,nd 12q14) which contain tumor suppressor genes that aremportant in melanoma development. There are evidencesuggesting that transition from normal melanocyte to mela-ocytic dysplastic nevus involves the loss of genes at 1p, 9p,nd 10q. Moreover, the progression from melanocytic dys-lastic nevus to the radial growth phase of malignant mel-noma involves further losses on 9p, 10q, and 6q, and thevolution to the vertical growth phase of malignant mela-oma involves 1p, 11q, and 17q.41

Genes controlling cell cycle machinery are very impor-ant in melanoma tumor development and progression. TheDKN2A gene (p16) is located on chromosome 9p21,ithin a region that is frequently deleted in sporadic mela-omas. Moreover, CDKN2A germline mutations have beenetected in many melanoma kindreds. CDKN2A is a pivotalell cycle regulator that binds to CDK4 and CDK6 andnhibits their ability to phosphorylate pRB. Inactivation ofDKN2A increases cell division by altering control of the

1-S checkpoint. CDK4 gene, located on chromosome2q14, shows recurrent focal amplification, and germlineutations in CDK4 have been detected in melanoma kin-

reds. Moreover, CCND1, a binding partner of CDK4, isrequently amplified in melanomas.42,43

Several genes have been shown to be abnormally ex-ressed in melanoma (Table 5); these include B-RAF, N-AS, PTEN, c-KIT, CCND1, and CDKN2A. However, the

Table 5 Molecular features of malignant melanoma

CDKN2A/p16 Deleted in somatic tumors; germlinemutations in familial tumors

CDK4 Amplified in somatic tumors; germlinemutations in familial tumors

CCND1 Amplified in sporadic tumorsB-RAF mutation Melanomas without chronic sun-

induced change (together with PTENloss)

N-RAS mutation Melanomas without chronic sun-induced change

PTEN loss Melanomas without chronic sun-induced change (together withB-RAF mutation)

c-KIT mutation Mutated in acral and mucosalmelanomas

frstt(tAttwuimaa

T

ttsasbmftTiStcNaismp

T

cptasutciwaiaVNmirBis

F

269Ortega et al Target Therapies in Gynecologic Cancers and Melanoma

requency of abnormalities in these genes seems to beelated to the natural history of the tumor. Melanomas onkin without chronic sun-induced damage, which representhe most common type of melanoma, had frequent muta-ions in B-RAF, together with losses of chromosome 10PTEN), or mutations in N-RAS alone. Interestingly, muta-ions in B-RAF and N-RAS are mutually exclusive events.s mentioned below, these alterations lead to activation of

he RAS/B-RAF/MAPK pathway, and could be responsiveo therapeutic interventions targeting such signaling path-ay. On the other side, other types of melanoma do notsually have mutations in B-RAF or N-RAS, but havencreased numbers of copies of CCND1 or CDK4. Further-ore, losses at the CDKN2A locus (p16) are also frequent,

nd they occur more commonly in mucosal and acral mel-noma, which also exhibit mutations in c-Kit.43,44

argeted therapies in melanoma

Malignant melanoma is extremely resistant to chemo-herapy. Empirical use of chemotherapy and/or immuno-herapy in the treatment of patients with melanoma hashown that the majority of melanomas are resistant to mostgents.45 For that reason, it is very important to look for newtrategies (Table 6). There are several molecules that maye good targets for therapy.46 It is now appreciated thatelanomas have defects that include loss of regulatory

unctions or the gain of proliferative or antiapoptotic func-ions that are no longer under normal regulatory control.here are numerous signaling pathways that play a signif-

cant role in melanoma tumor development and progression.ome of them are activated as a result of activating muta-

ions (B-RAF, N-RAS, PTEN), whereas others show in-reased expression of some of the key members (Bcl-2,F-kb, CDK2, cyclin D1). Many agents, such as mAb,

ntisense oligonucleotids, and TKI, are under investigationn melanoma. Unfortunately, most of these are phase I/IItudies, and the real efficacy of agents oriented to theolecular target can only be assessed with well-designed

Table 6 Targeted therapies proposed for malignant melanoma

Drug Function

Sorafenib Kinase inhibitor(BAY 43-9006)

PD0325901AZD0325901Oblimersen Antisense oligonucleotideTemsirolimus mTOR inhibitorCCI-779Everolimus mTOR inhibitorRAD001Velcade Proteasome inhibitorPS-341

hase III trials. B

he RAS/RAF/MAPK pathway

The mitogen-activated protein kinase (MAPK) signalingascade, which normally regulates cell growth, survival, androliferation, appears to be activated in melanoma.47 Two ofhe most important members of the pathway are frequentlybnormal in melanoma; mutations in N-RAS or B-RAF areeen in approximately 90% of cases. RAS is an importantpstream regulator of the MAPK and PI3K pathways and washe first oncogene to be identified in melanoma. In normalells, RAS proteins are activated by cell-surface receptor bind-ng of growth factors. The RAS proteins then activate RAFs,hich phosphorylate and activate MEK, leading to subsequent

ctivation of the extracellular signal-regulated kinase, ERKs,nvolved in cell-cycle progression and proliferation. B-RAFlso contributes to neoangiogenesis by stimulating autocrineEGF secretion. In normal cells, B-RAF is downstream fromRAS, but in melanoma can be constitutively activated by autation in codon V600 (Figure 5). This mutation has been

dentified in 66% of malignant melanomas, and this mutationepresents more than 80% of all B-RAF mutations. Because-RAF mutations can increase chemotherapy resistance, RAF

nhibitors such as sorafenib are being evaluated.48–50 Althoughorafenib has demonstrated limited activity as a single agent,

igure 5 Phenogram showing the typical V600 mutation in

Target Phase

Raf kinases II (� dacarbazine)VEGFR III ongoingPDGFRMEK II ongoingMEK II ongoingBcl-2 IIImTOR IIPTEN/PI3K/AKTmTOR IPTEN/PI3K/AKTNK-�B II

-RAF.

ps(mactiwatit2ci

T

Pnmmlayc7monpdbC

T

dmtatswm

cst1sfn

ttaemdsttioSi

hsbsufSpmeo

mwmrPa

T

Fl

270 Seminars in Diagnostic Pathology, Vol 25, No 4, November 2008

hase II data from patients with advanced melanoma haveuggested that this drug in combination with chemotherapycarboplatin, paclitaxel, temozolomide, and dacarbazine) isuch more active.51 Phase III trials of sorafenib in melanoma

re under development, and additional B-RAF inhibitors areurrently in preclinical development. In vitro data have shownhat melanomas with B-RAF mutations may be selectivelynhibited by two MEK inhibitors, AZD6244 and PD0325901,hich are currently being tested in clinical trials. Interestingly,

nthrax toxin inactivates both MEK1 and MEK2, providinghe basis for study in the treatment of melanoma.52 However,t should be noted that one potential problem of MEK inhibi-ion is that MEK is also activated by tumor progression locus

(TPL2), a protein kinase that regulates innate immunity, sohronic MEK inhibition might have negative effects on innatemmunity.

he PI3K pathway

Another signaling pathway important in melanoma is theI3K pathway. PI3K is mutated in 3% of metastasic mela-omas, and PTEN is inactivated in up to 20% of advancedelanomas. Moreover, AKT is overexpressed in 60% ofelanomas. In melanoma, the accumulation of phosphory-

ated AKT suppresses apoptosis, thereby enhancing survivalnd promoting proliferation of malignant cells.53 Phosphor-lated AKT is associated with tumor progression and de-reased survival and has been observed in 54% of nevi, in1.3% of primary melanomas, and in 71% of metastaticelanomas. In addition, overexpression of AKT has been

bserved in severely dysplastic nevi and metastatic mela-omas, but not in benign nevi, and correlates adversely withatient survival. Agents that target PI3K, AKT, and otherownstream components of this pathway such as mTOR areeing developed.54 Among these, mTOR inhibitors such asCI-779 and RAD001 are the most advanced.

argeting cell surface receptors: c-kit

The proto-oncogene c-KIT is necessary for melanocyteevelopment, differentiation, proliferation, survival, andigration. c-KIT mutations occasionally occur in some

ypes of lentiginous melanoma (mucosal, acral lentiginous,nd lentigo malignant melanoma), but are very infrequent inhe most common types of cutaneous melanoma. Manytudies have linked the loss of normal c-KIT expressionith the development of primary and metastatic melano-as.44

Some preclinical studies have provided evidences that-KIT may be a good target for therapy in melanoma. Atudy done by our group demonstrated a clear antiprolifera-ive effect of imatinib mesylate (STI-571, Gleevec) on 8 of9 (42.1%) human cutaneous melanoma-derived cell lines,upporting a possible use of STI571 as potential treatmentor a subset of patients with disseminated malignant mela-

oma55 (Figure 6). However, the mechanism of the inhibi- f

ory effect of STI571 on melanoma cells was not clear sincehere was not good correlation between response to STI571nd immunohistochemical expression of c-Kit. The clinicalfficacy of STI571 in patients with disseminated cutaneousalignant melanoma is under debate. Results from three

ifferent phase II trials have been reported. They havehown lack of clinical benefit in the vast majority of pa-ients, although a striking objective response has been ob-ained in individual cases. Moreover, the efficacy of STI571n a c-Kit-negative, cutaneous melanoma cell line of murinerigin in a mouse model has also raised the speculation thatTI571 could have some molecular targets, other than c-Kit,

n cutaneous malignant melanoma.56

Promising data about STI571 related to c-Kit expressionave recently been reported in melanoma of uveal origin. Iteems that c-Kit expression in uveal melanoma (analyzedy tissue microarrays) is present in 87% of cases, beingtrong in 69% of them. Moreover, depletion of c-Kit inveal melanoma cell lines expressing c-Kit and its growthactor (SCF) strongly inhibits melanoma cell proliferation.ome clinical effects of STI571 have been reported in twoatients with disseminated ocular melanoma. Since c-KITutations have not been detected in uveal melanomas, the

ffect of STI571 has to be explained by specific inhibitionf tyrosine kinase activity of wild type c-Kit.57

At present, STI-571 and other c-KIT inhibitors (sunitinibalate, SU011248) are currently being evaluated in patientsith metastatic mucosal and acral melanomas. It is worthentioning that these drugs also target other tyrosine kinase

eceptors. For example, imatinib mesylate also inhibitsDGFR and ABL, whereas sunitinib blocks PDGFR, RET,nd VEGFR.

argeting survival signals: NF-�B and BCL-2

Nuclear factor kappa B (NF-��) is a transcription

igure 6 c-Kit expression in 95% of cells of JG melanoma celline which responded to STI571 treatment (flow cytometry).

actor that occupies a pivotal point in survival and growth

pAaepttsrbhotibcmabp1owbtdipwvegpd

mtspalIvd

Oi

A

nbcVftiatwItgaacbsd

kiabScTtaic

I

im

Foc

271Ortega et al Target Therapies in Gynecologic Cancers and Melanoma

athways and can be activated through either the PI3K/kt or the MEK/ERK pathway. NF-�B mediates anti-

poptotic, proliferative, metastatic, and proangiogenicffects primarily through inducing gene expression ofroteins critical to these activities. Constitutive activa-ion of the NF-�� pathway has been identified in manyumor types, including melanoma. Proteasome inhibitors,uch as PS-341 (Bortezomib, Velcade), selectively andeversibly inhibit the 26S proteasome and prevent thereakdown of many regulatory proteins through the in-ibition of the ubiquitination-proteasome process.58 Onef these proteins whose breakdown is impaired is I�B,he protein that inactivates NF-�B. In some tumor types,nhibition of the proteasome leads to NF-�B inactivation,ut in other types of tumor (for example, endometrialarcinoma), proteasome inhibitors activate NF-�B. Inelanoma, Velcade has been shown to inhibit NFKB

ctivity and reduce cell growth in vitro. In a study doney our group,59 the effect of four structurally differentroteasome inhibitors (Bortezomib, PS-341, ALLN, MG-32 and epoximicin) was assessed on 16 human cutane-us melanoma-derived cell lines, which are original andere obtained from patients who were treated by 2 mem-ers of the clinical team. Proteasome inhibitors inhibitedhe in vitro growth of melanoma cells, and this effect wasue either to a reduction in cell proliferation rate or annduction of both caspase-dependent and caspase-inde-endent cell death (Figure 7). Moreover, release of AIFas observed in the absence of detectable caspase acti-ation. Also, caspase 2 processing was observed in Bort-zomib-induced cell death. Interestingly, Velcade syner-izes with temozolamide in human melanoma xenografts,roviding the rationale for using Velcade to overcomerug resistance.58

BCL-2, which suppresses apoptosis, is overexpressed inelanoma, suggesting a direct association with chemoresis-

ance. This idea is supported by studies showing that anti-ense suppression of BCL-2 increases melanoma cell apo-tosis and sensitivity to chemotherapy.60 Oblimersen, anntisense oligonucleotide, suppresses BCL-2 and has shownimited promise in initial melanoma clinical trials. A phaseII study has shown some benefit in progression-free sur-ival and overall response when used in combination withacarbazine, but not in overall survival. At present,

igure 7 The proteasome inhibitors MG-132 and bortezomib indr were treated with 0.5 �mol L�1 or 25 nmol L�1 of MG-132

aspase-3 staining.

blimersen is the only target agent tested in a phase III trialn melanoma.61,62

ngiogenesis

Angiogenesis has proven to be a critical step in mela-oma transformation. A number of angiogenic factors haveeen shown to be released by melanoma cells and/or hostells within the tumor microenvironment. These includeEGF, PDGF, interleukin-8, and basic fibroblastic growth

actor. Several human melanoma xenografts models showhat these factors play a role in tumor progression. Antibod-es against VEGF are effective at blocking tumor growthnd metastasis. Serum levels of VEGF in melanoma pa-ients increase with clinical stage and can be predictive oforse prognosis. However, there are no encouraging phase

I data, and because sorafenib, which is thought to workhrough an antiangiogenic mechanism, provides only a mar-inal response in melanoma, it is possible that targetingngiogenesis alone will be an ineffective treatment for mel-noma. In this way, available results from phase II trialsonfirm it. Carson and coworkers evaluated a regimen ofevacizumab alone or in combination with low-dose IFN-�,howing responses only in patients receiving the tworugs.63

SU011248, AG013736, and ZD6474 are all oral tyrosineinase inhibitors that have already shown promising resultsn other cancers, including clear cell renal carcinoma. Theyre inhibitors for the VEGFR2 receptor signaling but alsolock one or more other receptors. Targets for bothU011248 and AG13736 include VEGFR2, PDGF, and-kit, whereas ZD6474 blocks both VEGFR2 and EGFR1.he ability of these agents to inhibit multiple targets makes

hem of interest to investigate in patients with melanomalone, with chemotherapy, or with other targeted agents. Its anticipated that many such studies will be done in theoming years.46

mmune escape

Negative regulation of antitumor immunity in melanomas a focus of continuing research. Tumors may evade im-unosurveillance (“tumor escape”) through downregulation

optosis in M17 melanoma cell line. M17 cells were left untreatedezomib for 24 hours. Pictures show Hoechst 33258 and cleaved

uce apor bort

oridstiiuttiAtoCtlsa

H

gtwsvu

A

F1RtSti

R

1

1

1

1

1

1

1

1

1

1

2

2

2

2

2

2

2

272 Seminars in Diagnostic Pathology, Vol 25, No 4, November 2008

f various immunoregulatory components via tumor-de-ived soluble factors, alterations in signal transduction, andmmunologic tolerance. In addition, many tumor cells pro-uce antigens that are also expressed on normal cells. Con-equently, these cells are recognized as “self” and are notargeted for immune destruction. Tumors also often secretemmunosuppressing factors as they grow, thereby prevent-ng immune activation. Many of the inhibitory processessed by melanoma cells to evade immune system are po-ential therapeutic targets. The cytotoxic T-lymphocyte an-igen 4 (CTLA4) is a promising novel therapeutic target ands a CD28-family receptor that inhibits T-cell function.ntibodies directed against this receptor block CTLA4,

riggering the immune system by reversing the “invisibility”f several cancer antigens. The ultimate effect of anti-TLA4 therapy is to initiate an immune response against

he invading tumor cells, leading to tumor cytotoxicity viaymphocyte-mediated cell death. Anti-CTLA4 therapy hashown activity and prolonged responses in metastatic mel-noma.64,65

ormones

Melanoma cells express receptors for a number ofrowth factors, including hormones. Our group has recentlyested the effects of somatostatin analogues in correlationith the expression of somatostatin receptors, and the re-

ults show that somatostatin analogues have some impact oniability of melanoma cells, suggesting that they may besed in combination with other anticancer drugs.66

cknowledgments

This work is supported by Grants FISPI060832,ISPI060577, FISPI070304, FISPI070276, SAF2002-0529-E, Marató de TV3 2005-47, 2004XT00090, andD06/0020/1034. XD holds a postdoctoral fellowship from

he Fondo de Investigaciones Sanitarias, Ministerio deanidad y Consumo (CP05/00028). DLL holds a predoc-

oral fellowship from the Fondo de Investigaciones Sanitar-as, Ministerio de Sanidad y Consumo (FI05/00191).

eferences

1. Moreno–Bueno G, Sánchez–Estévez C, Cassia R, et al: Differentialgene expression profile in endometrioid and nonendometrioid endo-metrial carcinoma: STK15 is frequently overexpressed and amplifiedin nonendometrioid carcinomas. Cancer Res 63:5697-5702, 2003

2. Bockman JV: Two pathogenetic types of endometrial carcinoma.Gynecol Oncol 15:10-17, 1983

3. Lax SF, Kurman RJ: A dualistic model for endometrial carcinogenesisbased on immunohistochemical and molecular genetic analyses. VerhDtsch Ges Pathol 81:228-232, 1997

4. Matias–Guiu X, Catasus L, Bussaglia E, et al: Molecular pathology of

endometrial hyperplasia and carcinoma. Hum Pathol 32:569-577, 2001

5. Catasús Ll, Machin P, Matias–Guiu X, et al: Microsatellite instabilityin endometrial carcinomas clinicopathologic correlations in a series of42 cases. Human Pathol 29:1160-1164, 1998

6. Bussaglia E, del Rio E, Matias–Guiu X, et al: PTEN mutations inendometrial carcinomas. A molecular and clinicopathologic analysisof 38 cases. Hum Pathol 31:312-317, 2000

7. Velasco A, Bussaglia E, Pallares J, et al: PIK3CA gene mutations inendometrial carcinoma: correlation with PTEN and K-RAS alterations.Hum Pathol 37:1465-1472, 2006

8. Machin P, Catasus L, Pons C, et al: CTNNB1 mutations and beta-catenin expression in endometrial carcinomas. Hum Pathol 33:206-212, 2002

9. Moreno–Bueno G, Hardisson D, Sánchez C, et al: Abnormalities of theAPC/beta-catenin pathway in endometrial cancer. Oncogene 21:7981-7990, 2002

0. Tashiro H, Isacson C, Levine R, et al: p53 gene mutations are commonin uterine serous carcinoma and occurs early in their pathogenesis.Am J Pathol 150:177-185, 1997

1. Sherman ME, Bur ME, Kurman RJ: p53 in endometrial cancer and itsputative precursors: evidence for diverse pathways of tumorigenesis.Hum Pathol 26:1268-1274, 1995

2. Mutter GL, Lin MC, Fitzgerald JT, et al: Altered PTEN expression asa diagnostic marker for the earliest endometrial precancers. J NatlCancer Inst 92:924-931, 2000

3. Levine RL, Cargile CB, Blazes MS, et al: PTEN mutations andmicrosatellite instability in complex atypical hyperplasia, a precursorlesion to uterine endometrioid carcinoma. Cancer Res 58:3254-3258,1998

4. Oda K, Stokoe D, Taketani Y, et al: High frequency of coexistentmutations of PIK3CA and PTEN genes in endometrial carcinoma.Cancer Res 65:10669-10673, 2005

5. Gagnon V, St-Germain ME, Parent S, et al: Akt activity in endometrialcancer cells: regulation of cell survival through cIAP-1. Int J Oncol23:803-810, 2003

6. Chon HS, Hu W, Kavanagh JJ: Targeted therapies in gynecologiccancers. Curr Cancer Drug Targets 6:333-363, 2006

7. Podsypanina K, Lee RT, Politis C, et al: An inhibitor of mTORreduces neoplasia and normalizes p70/S6 kinase activity in Pten�/�mice. Proc Natl Acad Sci U S A 98:10320-10325, 2001

8. Khalifa MA, Abdoh AA, Mannel RS, et al: Prognostic utility ofepidermal growth factor receptor overexpression in endometrial ade-nocarcinoma. Cancer 73:370-376, 1994

9. Morrison C, Zanagnolo V, Ramirez N, et al: HER-2 is an independentprognostic factor in endometrial cancer: association with outcome in alarge cohort of surgically staged patients. J Clin Oncol 24:2376-2385,2006

0. Pallares J, Martinez–Guitarte JL, Dolcet X, et al: Abnormalities inNF-kB family and related proteins in endometrial carcinoma. A tissuemicroarray study. J Pathol 13:569-577, 2004

1. Catasús L, Matias–Guiu X, Machin P, et al: BAX somatic frameshiftmutations in endometrioid adenocarcinomas of the endometrium: ev-idence for a tumor progression role in endometrial carcinomas withmicrosatellite instability. Lab Invest 78:1439-1444, 1998

2. Dolcet X, Llobet D, Encinas M, et al: Proteasome inhibitors inducedeath but activate NF-kappaB on endometrial carcinoma cell lines andprimary culture explants. J Biol Chem 281:22118-22130, 2006

3. Llobet D, Eritja N, Encinas M, et al: Antioxidants block proteasomeinhibitor function in endometrial carcinoma cells. Anticancer Drugs19:115-124, 2008

4. Dolcet X, Llobet D, Pallares J, et al: FLIP is frequently expressed inendometrial carcinoma and has a role in resistance to TRAIL-inducedapoptosis. Lab Invest 85:885-894, 2005

5. Llobet D, Eritja N, Encinas M, et al: CK2 controls TRAIL and Fassensitivity by regulating FLIP levels in endometrial carcinoma cells.Oncogene 27:2513-2524, 2008

6. Gunin AG, Kapitova IN, Suslonova NV: Effects of histone deacetylaseinhibitors on estradiol-induced proliferation and hyperplasia formation

in the mouse uterus. J Endocrinol 185:539-549, 2005

2

2

2

3

3

3

3

3

3

3

3

3

3

4

4

4

4

4

4

4

4

4

4

5

5

5

5

5

5

5

5

5

5

6

6

6

6

6

6

6

273Ortega et al Target Therapies in Gynecologic Cancers and Melanoma

7. Takai N, Desmond JC, Kumagai T, et al: Histone deacetylase inhibi-tors have a profound antigrowth activity in endometrial cancer cells.Clin Cancer Res 10:1141-1149, 2004

8. Matias–Guiu X, Prat J: Molecular pathology of ovarian carcinomas.Virchows Arch 433:103-111, 1998

9. Kurman RJ, Shih IeM: Pathogenesis of ovarian cancer: lessons frommorphology and molecular biology and their clinical implications. IntJ Gynecol Pathol 27:151-160, 2008

0. Esteller M, Silva JM, Dominguez G, et al: Promoter hypermethylationand BRCA1 inactivation in sporadic breast and ovarian tumors. J NatlCancer Inst 92:564-569, 2000

1. Gras E, Cortes J, Diez O, et al: Loss of heterozygosity on chromosome13q12-q14, BRCA-2 mutations and lack of BRCA-2 promoter hyper-methylation in sporadic epithelial ovarian tumors. Cancer 92:787-795,2001

2. Gras E, Catasus L, Argüelles R, et al: Microsatellite instability,MLH-1 promoter hypermethylation, and frameshift mutations at cod-ing mononucleotide repeat microsatellites in ovarian tumors. Cancer92:2829-2836, 2001

3. Tsuchiya A, Sakamoto M, Yasuda J, et al: Expression profiling inovarian clear cell carcinoma: identification of hepatocyte nuclear fac-tor-1 beta as a molecular marker and a possible molecular target fortherapy of ovarian clear cell carcinoma. Am J Pathol 163:2503-2512,2003

4. Cuatrecasas M, Villanueva A, Matias–Guiu X, et al: K-ras mutationsin mucinous ovarian tumors: a clinicopathologic and molecular studyof 95 cases. Cancer 79:1581-1586, 1997

5. Burger RA, Sill M, Monk BJ, et al: Phase II trial of bevacizumab inpersisntent or recurrent epithelail ovarian cancer or primary peritonealcancer. A Gynecologic Oncology Group study (abstract). J Clin Oncol23:457s, 2005

6. Palayekar MJ, Herzog TJ: The emerging role of epidermal growthfactor receptor inhibitors in ovarian cancer. Int J Gynecol Cancer 2007Dec 5 [Epub ahead of print]

7. Bookman MA, Darcy KM, Clarke–Pearson D, et al: Evaluation ofmonoclonal humanized anti-HER2 antibody, trastuzumab, in patientswith recurrent or refractory ovarian or primary peritoneal carcinomawith overexpression of HER2: a phase II trial of the GynecologicOncology Group. J Clin Oncol 21:283-290, 2003

8. Gordon MS, Matei D, Aghajanian C, et al: Clinical activity of pertu-zumab (rhuMAb 2C4), a HER dimerization inhibitor, in advancedovarian cancer: potential predictive relationship with tumor HER2activation status. J Clin Oncol 24:4324-4332, 2006

9. Dharap SS, Chandna P, Wang Y, et al: Molecular targeting of BCL2and BCLXL proteins by synthetic BCL2 homology 3 domain peptideenhances the efficacy of chemotherapy. J Pharmacol Exp Ther 316:992-998, 2006

0. Mross K, Steinbild S, Baas F, et al: Results from an in vitro and aclinical/pharmacological phase I study with the combination irinotecanand sorafenib. Eur J Cancer 43:55-63, 2007

1. Hussein MR: Genetic pathways to melanoma tumorogenesis. J ClinPathol 57:797-801, 2004

2. Newton Bishop JA, Gruis NA: Genetics: what advice for patients whopresent with a family history of melanoma? Semin Oncol 34:452-459,2007

3. Curtin JA, Fridlyand J, Tageshita T, et al: Distinct sets of geneticalterations in melanoma. N Engl J Med 353:2135-2147, 2005

4. Curtin JA, Busam K, Pinkel D, et al: Somatic activation of KIT indistinct subtypes of melanoma. J Clin Oncol 24:4340-4346, 2006

5. Thompson JF, Scolyer RA, Kefford RF: Cutaneous melanoma. Lancet

365:687-701, 2005

6. Lorigan P, Eisen T, Hauschild A: Systemic therapy for metastaticmalignant melanoma–from deeply disappointing to bright future? ExpDermatol 17:383-394, 2008

7. Fecher LA, Amaravadi RK, Flaherty KT: The MAPK pathway inmelanoma. Curr Opin Oncol 20:183-189, 2008

8. Gray Shopfer VC, da Rocha Dias S, Marais R: The role of B-RAF inmelanoma. Cancer Metastasis Rev 24:165-183, 2005

9. Panka DJ, Wang W, Atkins MB, et al: The Raf inhibitor BAY 43-9006(Sorafenib) induces caspase-independent apoptosis in melanoma cells.Cancer Res 66:1611-1619, 2006

0. Eisen T, Ahmad T, Flahery KT, et al: Sorafenib in advanced mela-noma: a Phase II randomised discontinuation trial analysis. Br JCancer 95:581-586, 2006

1. McDermott DF, Sosman JA, Gonzalez R, et al: Double-blind random-ized phase II study of the combination of sorafenib and dacarbazine inpatients with advanced melanoma: a report from the 11715 StudyGroup. J Clin Oncol 26:2178-2185, 2008

2. Abi–Habib RJ, Urieto JO, Liu S, et al: BRAF status and mitogen-activated protein/extracellular signal-regulated kinase kinase 1/2 ac-tivity indicate sensitivity of melanoma cells to anthrax lethal toxin.Mol Cancer Ther 4:1303-1310, 2005

3. Robertson GP: Functional and therapeutic significance of Akt dereg-ulation in malignant melanoma. Cancer Metastasis Rev 24:273-285,2005

4. Chan S: Targeting the mammalian target of rapamycin (mTOR): a newapproach to treating cancer. Br J Cancer 91:1420-1424, 2004

5. Mayorga ME, Sanchis D, Perez de Santos AM, et al: Antiproliferativeeffect of STI571 on cultured human cutaneous melanoma-derived celllines. Melanoma Res 16:127-135, 2006

6. Grossman D: Imatinib mesylate for melanoma: will a new target berevealed? J Invest Dermatol 123:xi-xiii, 2004

7. Lefevre G, Glotin AL, Calipel A, et al: Roles of stem cell factor/c-kitand effects of Glivec(R)/STI571 in human uveal melanoma cell tu-morigenesis. J Biol Chem 23:31769-31779, 2004

8. Amiri KI, Horton LW, LaFleur BJ, et al: Augmenting chemosensitiv-ity of malignant melanoma tumors via proteasome inhibition: impli-cation for bortezomib (VELCADE. PS-341) as a therapeutic agent formalignant melanoma. Cancer Res 64:4912-4918, 2004

9. Sorolla A, Yeramian A, Dolcet X, et al: Effect of proteasome inhibi-tors on proliferation and apoptosis of human cutaneous melanoma-derived cell lines. Br J Dermatol 158:496-504, 2008

0. Bedikian AY, Millward M, Pehamberger H, et al: Bcl-2 antisense(oblimersen sodium) plus dacarbazine in patients with advanced mel-anoma: the Oblimersen Melanoma Study Group. J Clin Oncol 24:4738-4745, 2006

1. Moreira JN, Santos A, Simões S: Bcl-2-targeted antisense therapy(Oblimersen sodium): towards clinical reality. Rev Recent Clin Trials1:217-235, 2006

2. Tarhini AA, Kirkwood JM: Oblimersen in the treatment of metastaticmelanoma. Future Oncol 3:263-271, 2007

3. Varker KA, Biber JE, Kefauver C, et al: A randomized phase 2 trial ofbevacizumab with or without daily low-dose interferon alfa-2b inmetastatic malignant melanoma. Ann Surg Oncol 14: 2367-2376, 2007

4. Tarhini AA, Kirkwood JM: Tremelimumab, a fully human monoclonalIgG2 antibody against CTLA4 for the potential treatment of cancer.Curr Opin Mol Ther 9:505-514, 2007

5. Lens M, Ferrucci PF, Testori A: Anti-CTLA4 monoclonal antibodyipilimumab in the treatment of metastatic melanoma: recent findings.Recent Patents Anticancer Drug Discov 3:105-113, 2008

6. Marti RM, Mayorga ME, Llecha N, et al: Expression of somatostatinreceptors (SSTR) in melanoma cell lines. Effect of somatostatin ana-

logues on their proliferation. Arch Dermatol Res 299:290, 2007

Recommended