Breast Cancer Mechansm

download Breast Cancer Mechansm

of 11

Transcript of Breast Cancer Mechansm

  • sbur

    times higher than before 65, and 150-fold higher than before

    has been acknowledged for decades. It is suggested since

    recently, that tumor promotion is not the only mechanism

    the menopause; in pre-menopausal women, overweight is

    The second group of breast cancer predisposing properties

    deficiency in maintenance of genomic integrity has been

    Drug Discovery Today: Disease Mechanisms Vol. 1, No. 2 2004

    d In

    l Rof estrogens action: some of estrogen metabolites were shown

    to cause DNA damage directly (i.e. contribute in the breast

    cancer initiation). Selected causes of estrogen overload (e.g.

    early menarche or late menopause) are at least in part attrib-

    uted to inherited genetic variations. However, most of deter-

    minants of hyperestrogenia are related to the modern,

    Western lifestyle including low parity, delayed age at first

    recognized only recently (Fig. 1). There are two lines of

    supporting arguments: first, all of the identified breast cancer

    susceptibility genes contribute to the sensing or repair of

    DNA damage; second, there is an impressive reproducibility

    of phenotyping studies demonstrating relationships between

    breast cancer risk and constitutional chromosomal instability

    [57].

    Unlike lung or bladder cancers, none of environmental

    carcinogens has been convincingly linked to breast cancer*Corresponding author: (E.N. Imyanitov) [email protected]

    1740-6765/$ 2004 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.ddmec.2004.09.002 www.drugdiscoverytoday.com 23530 years of age. In addition to advanced age, a few dozens of

    other breast cancer predisposing factors have been identified;

    however, all these diverse risks can be assigned to either of

    two major categories: excessive exposure to estrogens and

    deficiency in maintenance of genomic integrity (Fig. 1) [1].

    The proliferative effect of estrogens on breast epithelium

    associated with anovulatory cycles and reduced probability of

    acquiring breast cancer disease. The adverse impact of oral

    contraception and hormone replacement therapy has been

    confirmed in some but not all epidemiological studies. The

    role of exogenous endocrine disruptors in breast cancer inci-

    dence remains to be proven [14].development of dozens of other targeted treatments

    for breast cancer is underway. Unfortunately, some

    intrinsic features of breast cancer biology compromise

    the efficiency of current therapeutic strategies.

    Introduction

    Breast cancer is the most common malignancy among

    females affecting approximately one out of ten women.

    Ageing of population in the industrialized world is the most

    obvious cause of increased breast cancer occurrence; indeed,

    the risk of developing breast cancer after 65 years of age is 5.8Breast cancer is the most common malignancy among females andaffects approximately one in every ten women worldwide. It is the first

    human tumor for which targeted therapies have been developed. Somenotable successful therapies include tamoxifen, aromatase inhibitors

    both estrogen receptor pathway downregulators and Herceptin, aHER2 antagonist. However, despite some spectacular examples of

    prolonged disease remission in selected women, the statistical survivalbenefit in metastatic breast cancer patients is estimated in months and

    not years. In this article, Evgeny Imyanitov and Kaido Hanson describethe progress of targeted treatments for breast cancer that are already

    underway, and discuss the particular features of breast cancer biologythat compromise the efficiency of current therapeutic drugs.

    delivery, short duration of breastfeeding, overeating, limited

    exercise and so on. Interestingly, the obesity correlates with

    hyperestrogenia and excessive breast cancer risk only afterimpressive success stories include estrogen receptorMECHANISMS

    DRUG DISCOVERY

    TODAY

    DISEASE

    Mechanisms of breaEvgeny N. Imyanitov*, Kaido P. HansonGroup of Molecular Diagnostics, N.N. Petrov Institute of Oncology, St. Peters

    Breast cancer is the first human tumor for which

    targeted therapies have been developed. The most

    pathway downregulators (tamoxifen and aromatase

    inhibitors) and HER2 antagonists (Herceptin). The

    Editors-in-Chief

    Toren Finkel National Heart, Lung and Bloo

    Tamas Bartfai Harold L. Dorris Neurologica

    Cancert cancer

    g 197758, Russia

    Section Editor:Silvio Gutkind National Institute of Dental and CraniofacialResearch, National Institutes of Health, Bethesda, MD, USA

    stitute, National Institutes of Health, USA

    esearch Center and The Scripps Research Institute, USA

  • Drug Discovery Today: Disease Mechanisms | Cancer Vol. 1, No. 2 2004Box 1. Challenging the dogmas in breast cancerresearch: recent unexpected findings

    Viral theory revisited?

    There are several independent reports claiming that in some

    groups of patients roughly a third of breast carcinomas contain

    DNA sequences homologous to the well-known Mouse Mammary

    Tumor Virus (MMTV); in intriguing accordance with the viral

    hypothesis, worldwide breast cancer spread appears to parallel the

    geographical distribution of one species of house mouse, Mus

    domesticus. If the contribution of viruses in breast cancer etiology

    will be confirmed, many aspects of breast cancer prevention and

    treatment will need to be re-analyzed. However, there are

    also some negative reports on this subject [8,9].

    Polyclonal origin of breast cancer?

    Monoclonal origin of breast cancer is believed to be a well-

    established fact. However, recent data cast some doubt on this

    confidence: at least in some cases, breast cancer lump appears to

    arise from several independent progenitors, thus supporting the

    cancer field theory [42]. Interestingly, stromal cells surrounding

    breast cancer epithelium also carry somatic mutations, and the

    spectrum of these genetic alterations is different from those

    observed in cancer cells [22].

    Does all tumor mass possess a danger?

    Recent experiments show that only a minor, specific fractionetiology. The results of the studies on dietary factors have

    been inconclusive as well. Contrary to beliefs of many

    patients, psychological stress is not associated with breast

    cancer risk [1,2,4]. There are surprisingly few reports in

    modern scientific literature assessing the relationship

    between breast trauma and subsequent cancer development.

    Intriguingly, some recent findings have resumed the debate

    on the viral etiology of breast cancer [8,9] (Box 1).

    Contribution of inherited features

    In some instances, breast cancer represents a classical heredi-

    tary disease with mendelian mode of transmission. The best-

    known highly penetrant breast cancer genes are BRCA1

    (GenBank accession number NM_007294) and BRCA2 (Gen-

    Bank accession number NM_000059); they account for

    approximately 20% of familial breast cancer clustering

    [10]. Although early studies carried out on breast cancer

    families suggested that BRCAs are associated with nearly fatal

    risk of the disease, recent reports indicate that their pene-

    trance in unselected breast cancer series approaches to 65%

    for BRCA1 and only to 45% for BRCA2 (by age 70 years) [11].

    In a broad sense, ATM (GenBank accession number

    of cells forming breast cancer lump is highly tumorigenic [43].

    Tumor heterogeneity might compromise the search for new

    therapeutic targets, as the current approaches are almost always

    based on the analysis of crude tumor material. The intrinsic resistance

    of particular intratumoral cell subsets to the treatment might

    explain a discordance between high frequency of short-term partial

    tumor responses and low rate of prolonged breast cancer remissions.

    236 www.drugdiscoverytoday.comNM_000051) and p53 (GenBank accession number

    NM_000546) germ-line mutations, predisposing to ataxia-

    telangiectasia and Li-Fraumeni hereditary syndromes, respec-

    tively, can also be considered as breast cancer-associated

    genetic defects, owing to the increased incidence of breast

    neoplasia in their heterozygous carriers [5].

    There is a growing class of breast cancer-associated genetic

    variations, which are situated in between rare catastrophic

    germ-line mutations and frequent normal gene polymorph-

    isms. Some founder mutations can be classified as middle-

    penetrance polymorphisms, owing to their noticeable occur-

    rence in the population (1%) and modest but clinicallymeaningful influence on the breast cancer risk increase. In

    addition to Jewish founder mutations in BRCA genes, which

    seem to be somewhat less penetrant than non-founder

    defects, the examples include CHEK21100delC (GenBank

    accession number NM_007194) and NBS1657del5 (GenBank

    accession number NM_002485) [7]. As already mentioned

    above, all high- and middle-penetrant genes with proven

    breast cancer-associated significance participate in various

    aspects of maintenance of genomic integrity.

    It is frequently stated that the majority of breast cancer

    cases are related to the disadvantageous genetic passport

    (i.e. unfavorable combination of low-penetrant gene poly-

    morphisms). The attempts to link breast cancer risk to poly-

    morphisms in carcinogen metabolizing enzymes have been

    unsuccessful. There is more hope to detect breast cancer gene-

    disease interactions within the class of hormone metaboli-

    zers, based on convincing evidence for heritability of indivi-

    dual features of hormonal portrait coupled with the proven

    role of hyperestrogenia in breast cancer development. How-

    ever, no reproducible associations have been revealed over

    the decade-long research in this field. Following the success

    in identifying breast cancer-associated germ-line mutations

    within genome safeguards in addition to relying on the

    consistency of the results of phenotyping tests (see the text

    above and Fig. 1), many breast cancer researchers have

    recently re-located the efforts to the analysis of DNA repair

    gene polymorphisms; the outcome of these studies remains

    to be seen [7].

    Somatic events in breast cancer pathogenesis

    Interaction between various breast cancer predisposing fac-

    tors leads to accumulation of somatic mutations in breast

    epithelial cells (Fig. 1). Chromosomal instability manifested

    by enormous number of gross chromosomal abnormalities

    appears to be the most characteristic feature of breast cancer

    genome [12]. Wide-spread hypermethylation of regulatory

    regions of genome is another mandatory peculiarity of breast

    tumors [13]. Single nucleotide instability appears to be less

    common in breast cancer than in other major cancer types,

    such as colorectal or lung cancer; however, limitations incurrently available methodologies force to abstain from the

  • Vol. 1, No. 2 2004 Drug Discovery Today: Disease Mechanisms | Cancerdefinitive conclusion [12,14]. Classical type of microsatel-

    lite instability does not occur in breast cancer, although it was

    demonstrated in a small portion of bilateral breast tumors

    [15]. It was shown recently that many breast carcinomas carry

    alterations in mitochondrial DNA [16].

    Figure 1. Breast cancer mechanisms. Two groups of predisposing factors have a

    and deficiency in maintenance of genomic integrity. During the malignant transfo

    genetic events (mainly gross chromosomal alterations and methylation abnorm

    suppressor genes, which eventually results in the manifestation of The hallm

    properties might serve as potential therapeutic targets.Discrete alterations in breast cancer-specific genes can

    serve both as the triggers of somatic genetic instability and

    as consequences of increased mutation rate. Hundreds of

    reports have been devoted to the cataloging of breast can-

    cer-associated genetic abnormalities (e.g. Somatic Mutations

    major role in breast cancer etiology: excessive breast exposure to estrogens

    rmation process, breast epithelial cells accumulate high number of somatic

    alities). These DNA alterations activate oncogenes and inactivate tumor

    arks of cancer [21]. Molecular determinants of the most essential tumor

    www.drugdiscoverytoday.com 237

  • Drug Discovery Today: Disease Mechanisms | Cancer Vol. 1, No. 2 2004

    or

    D1

    CC

    i-6

    H1

    q (8

    3p

    13q

    21.

    6q1

    3, 1

    2q

    CN

    NES

    , R

    3-3

    T1,

    NX

    httTable 1. Genetic alterations in breast carcinomas

    Type of genetic event Chromosomal regions

    Activating events

    Gene amplification followed by over-expression 8q24: MYC; 11q13: CCN

    Gene over-expression BCL2, B94, Cathepsin D,

    GZMH, hTERT, IGF1R, K

    neurosin, PAI1, PAI2, PO

    Gains of genetic material 1q (1q21, 1q32, 1q41), 8

    Events of uncertain significance

    Losses of heterozygosity (allelic imbalances)

    reflecting either allelic deletion or gain

    of the remaining allele

    1p (1p36.3), 2q (2q22.1),

    8p (8p21.3), 9p (9p21.3),

    18p (18p11.32), 18q (18q

    Inactivating events

    Losses of genetic material 1p (1p3135, 1p36), 6q (

    11q2425), 13q (13q121

    17p (17p13.1, 17p13.3), 2

    Intragenic mutations p53

    Promoter methylation followed by the

    loss of gene expression

    APC, BCSG1, BRCA1, C

    HIN1, HOXA5, Maspin,

    RAR-beta, RASSF1A, RFC

    TMS1, TWIST, ZAC, 14-

    Loss of gene expression ATM, BAX, ITGA6, MGS

    SPR1, TGFBR3, TFAP4, T

    a For more information of these genes, see http://www.gene.ucl.ac.uk/nomenclature/, andin Human Cancers Database; http://www.onco-is.com).

    Conditionally, they can be classified into activating and

    inactivating events (Table 1). Noticeable frequency of ampli-

    fications of selected oncogenes [HER2 (also known as ERBB2)

    (GenBank accession number NM_004448), CCND1 (GenBank

    accession number NM_053056), C-MYC (GenBank accession

    number NM_002467)] followed by their overexpression is a

    distinct feature of breast cancer. In addition to identified

    oncogenes, there are several other regions in the genome

    that repeatedly demonstrate extra copies in breast cancer; it

    remains to be established whether they do contain activated

    oncogenes or simply reflect the background noise of somatic

    chromosomal instability [12]. Recent developments in

    expression profiling have significantly enlarged the list of

    genes overexpressed in breast cancer, however systematic

    picture of upregulated transcripts has yet to be drawn [17].

    In 1990s and early 2000s many researchers tried to locate

    breast cancer-specific losses of heterozygosity (LOH). How-

    ever, the attempts to identify new suppressor genes by LOH

    mapping have largely failed: in addition to enormous varia-

    bility of LOH patterns due to admixture of non-specific allelic

    imbalances, it has turned out that conventional PCR allelo-

    typing can not discriminate between inactivating and acti-

    vating mutations (e.g. loss of the allele versus amplification of

    the remaining allele) or between homozygous deletion and

    retention of both gene copies [18,19]. Intragenic mutations

    are relatively uncommon for breast cancer. The p53

    238 www.drugdiscoverytoday.comgenes involveda Refs.

    , EMS1; 17q1221: HER2, TOP2A; 20q13: AIB1 [12]

    NE, CD63, claudin-7, CRABP2, CTSD, GATA3,

    7, lactoferrin, lipocalin 2, MDM2, MUC1, MYBL2,

    , PS2, Rantes, SIX1, SMARCD2, STMY3, VEGF

    [12,17]

    q24), 11q (11q13), 16p (16p11), 17q (17q11.2, 17q24), 20q (20q13) [12]

    (3p14.2), 4q (4q35.1), 6q (6q25.1), 7q (7q31.2),

    (13q14), 16q (16q22.1, 16q24.3), 17p (17p13.3),

    2), 19p (19p13), 21q (21q11.1)

    [18]

    321, 6q2123.3, 6q2527), 8p (8p21, 8p2223), 11q (11q2223,

    3q14.1), 16q (16q2123.3, 16q24.3),

    (22q13)

    [12]

    [20]

    D2, CDH1, CDH13, DAPK, ER, FHIT, GPC3, GSTP1,

    1, NM23-H1, NOEY2, PR, Prostasin, INK4, CIP1,

    IZ1, SOCS1, SRBC, SYK, TGFBR2, THBS, TIMP3,

    sigma

    [13]

    OXTR, plakophilin 1, KIP1, RIG-like7-1, RB1, RBL2, SPARCL1,

    A, 53BP2

    [12,17]

    p://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=OMIM&itool=toolbar.(GenBank accession number NM_000546) gene is mutated

    only in 20% of breast carcinomas; mutations in RAS family

    oncogenes occur in a negligible fraction of breast cancer [20].

    The genome-wide approaches led to the identification of

    dozens breast cancer-associated deletions; there is also a

    growing list of genes whose transcription is downregulated

    in breast cancer [12,17]. Frequently, the decreased expression

    of suppressor genes is associated with the methylation of

    their promoter regions [13]. However, molecular techniques

    enabling global mapping of hypermethylated regions are not

    yet as efficient as expression profiling or array-based com-

    parative genomic hybridization.

    Various combinations of activating and inactivating muta-

    tions and epigenetic events lead to the spectrum of properties

    of tumor cells, which have been defined as The Hallmarks of

    Cancer in already-classical work of Hanahan and Weinberg

    [21], and include self-sufficiency in growth signals, insensi-

    tivity to antigrowth signals, evasion from apoptosis, limitless

    replicative potential, invasion and metastasis, genomic

    instability and sustained angiogenesis. Similarly to the pro-

    gress in understanding of tumor angiogenesis, it is becoming

    apparent that the contribution of surrounding stroma in

    tumor growth is not a merely passive process, but, instead,

    an active and essential component of neoplastic evolution

    [22,23]. Breast carcinomas share all characteristics of malig-

    nancy, with one important reservation: more than 50% of

    breast cancers retain at least partial dependence from estro-

  • Vol. 1, No. 2 2004 Drug Discovery Today: Disease Mechanisms | Cancergenic growth stimulation. This benign feature of some breast

    cancer accounts for remarkable efficiency of the antiestro-

    genic treatment approaches [24].

    Although the diversity of molecular portraits of breast

    cancer is extraordinary, they appear to fit into the limited

    number of distinct disease subsets. Noticeably, many of breast

    cancer signatures identified by array expression profiling tend

    to group around long-known disease markers, such as estro-

    gen receptor (ER) or HER2 oncogene status [25]. Interestingly,

    host factors appear to play an essential role in determining

    the molecular variant of breast cancer pathogenesis [15].

    The web of signaling cascades

    Owing to the rapid accumulation of data in signal transduc-

    tion research, the comprehensive description of breast can-

    cer-associated signaling pathways and their cross-talk has

    become a difficult challenge. However, surprisingly, in the

    light of potential therapeutic relevance, this complexity is

    not as high as it might appear. First, the spectrum of candi-

    date targets is limited by those molecules, which are essential

    for the maintenance of breast cancer cells; the intervention

    with breast cancer initiating cascades might have some

    potential for breast cancer prevention, but not for the cure

    of already existing disease. Second, while the specific target-

    ing of upregulated molecules has become a realistic approach,

    the opposite (i.e. the substitution of downregulated enzymes)

    is not achievable for the time being. Therefore, applied breast

    cancer research is more interested in tumor-activated path-

    ways than in those that are suppressed in breast cancer cells.

    Third, owing to the multifunctionality of many signaling

    proteins and pathways, it is usually difficult to precisely

    assign the molecule of interest to the defined biological

    outcome(s). For example, the members of HER receptor

    family appear to contribute into all cancer hallmarks (Fig.

    1) [26]. COX2 is mentioned most frequently in the context of

    tumor angiogenesis; however, its involvement in autoproli-

    ferative, antiapoptotic and metastatic cascades has also been

    acknowledged [27]. Although the utmost importance of com-

    prehensive functional description of potential breast cancer

    targets is beyond any doubt, the initial inclusion criteria for

    the target usually are its breast cancer specificity and involve-

    ment in the disease maintenance.

    Two molecules appear to be truly specific for breast cancer,

    and both of them are receptors: ER and HER2. ER is a member

    of the family of nuclear receptors, and is involved in tran-

    scriptional regulation of many essential genes. Activation of

    the HER2 pathway in breast cancer is often associated with an

    upregulation of other members of HER receptor family, espe-

    cially HER1, however HER1 overexpression occurs in breast

    cancer significantly less frequently than in several other

    cancer types. Downstream parts of HER-initiated signaling

    cascades include RASRAFMEKMAPK pathway, whichpotentiates cellular proliferation through modification ofcyclin-dependent cascades, and phosphatidylinositol 3-

    kinase (PI3-K) pathway (AKT, mTOR), which has an essential

    role in the regulation of cell survival. These molecules are not

    indeed specific for breast cancer; they do have regulatory

    functions in normal cells as well, therefore, their targeting

    possesses a risk of adverse effects. In addition to ER and HERs,

    there is an increasing attention to the angiogenic pathways

    triggered by vascular endothelial growth factor (VEGF); these

    cascades appear to be upregulated in many cancer types,

    including breast cancer [12,26,28,29].

    Targeted therapies

    The description of breast cancer-specific molecular targets

    and corresponding therapeutic compounds is presented in

    the Table 2.

    The first example of selective breast cancer therapy

    emerged several decades ago owing to an accidental finding.

    Tamoxifen had been initially tested with the intention to

    develop a contraceptive pill. Although it failed to control

    fertility in humans, its inhibitory effect on breast cancer cell

    growth led to the rapid introduction of this drug in the

    treatment of ER-positive breast cancer. In postmenopausal

    women, it might soon be replaced by inhibitors of aromatase,

    a key enzyme of estrogen synthesis. Aromatase inhibitors

    have shown more favorable results than tamoxifen in several

    randomized breast cancer trials [24]. Another success story

    concerns Trastuzumab (Herceptin), which demonstrated evi-

    dent survival benefit for patients suffering from HER2-posi-

    tive breast cancer [29]. Other targeted approaches such as

    HER1 or pan-HER inactivation [26], interference with down-

    stream participants of receptor tyrosine kinase signaling [29

    33], inhibition of molecules involved in tumor metastasis and

    angiogenesis [29,30,34], as well as some additional strategies

    [27,28,3537], are in early clinical trials or pre-clinical studies

    (Table 2).

    All of the approaches mentioned above are based on the

    suppression of molecules essential for breast cancer mainte-

    nance. The alternative strategy suggests the use of breast

    cancer-specific proteins as the anchors for site-specific deliv-

    ery of toxic compounds [3840] (Table 2). This concept

    certainly enlarges the list of potentially relevant targets,

    because it does not require functional evidence but solely

    relies on the proof of breast cancer-specific expression. With

    some reservations, recently introduced Capecitabine

    (Xeloda) could be considered as a relevant example; indeed,

    being a non-toxic precursor of 5-fluorouracil, Xeloda under-

    goes topical conversion to the cytostatic drug in tumors, due

    to increased expression of thymidine phosphorylase in the

    neoplastic tissue [38]. More general approaches are based on

    the use of breast cancer-specific antibodies conjugated with

    various toxins [39].

    The somatic genomic instability might represent not onlythe dangerous property of tumor phenotype, but also an

    www.drugdiscoverytoday.com 239

  • Dru

    gD

    iscovery

    Today:

    Disease

    Mech

    anism

    s|

    Can

    cerV

    ol.

    1,N

    o.2

    2004

    Table 2. Breast cancer targets and related therapies

    Target Therapy against target Stage of developmenta Advantages and/or disadvantages Who is work g

    on the targe

    Website Refs.b

    Properties of cancer cells as a target

    Genetic instability and/or

    high proliferation

    Conventional cytostatic

    drugs (anthracyclines,

    taxanes, alkylating drugs,

    antimetabolites, vinca alkaloids)

    On the market for

    a long time

    Clinical response can be achieved in

    more than 8090% cases. However,

    disadvantages include severe adverse

    effects, limited duration of response,

    overlapping spectrum of drug resistance,

    limited ability to predict response and/or

    non-response to a given cytostatic compound.

    Many

    pharmaceutica

    companies and

    research group

    [38]

    Molecular targets as tumor-specific anchors for delivery of cytostatic substances

    Thymidine phosphorylase Capecitabine (Xeloda) On the market Thymidine phosphorylase converts Xeloda

    to the cytostatic drug, 5-fluorouracil; high

    tumor specificity is attributed to the

    preferential expression of this enzyme

    in cancer tissues.

    Roche http://www.roche.com [38]

    Breast cancer-specific

    antigens

    Immunoconjugates

    (with cytotoxic drugs,

    radioactive isotopes,

    toxins, attractants of

    tumor-killing cells)

    Phase III Presumably large spectrum of candidate

    molecules, since the target does not

    need to be essential for tumor growth.

    Several researc groups [39]

    Sodiumiodide symporter Radioactive iodide Laboratory studies Adverse effects on thyroid gland are probable. [40]

    Genuine targets (i.e. molecules essential for breast cancer development and maintenance)

    Estrogen receptor pathway ER pathway downregulators show high

    efficiency combined with good safety profile.

    However, more than a third of breast cancers

    are resistant to antiestrogen therapy.

    Selective estrogen

    receptor modulators

    (SERMs)

    Tamoxifen On the market for

    a long time

    Many pharmac tical

    companies

    [24]

    Toremifene (Fareston) On the market Almost complete cross-resistance

    with tamoxifen

    Schering-Ploug Orion http://www.schering-

    plough.com

    http://www.orion.fi

    [24]

    Raloxifene (Evista) Chemoprevention

    trials

    Almost complete cross-resistance

    with tamoxifen

    Eli Lilly http://www.lilly.com [24]

    Selective estrogen

    receptor downregulators

    (SERDs)

    Faslodex (Fulvestrant) On the market AstraZeneca http://www.astrazeneca.com [24]

    240

    ww

    w.d

    rugd

    iscoveryto

    day.co

    min

    t

    l

    s

    h

    eu

    h,

  • Vol.

    1,N

    o.2

    2004

    Dru

    gD

    iscovery

    Today:

    Disease

    Mech

    anism

    s|

    Can

    cer

    Aromatase inhibitors Superior when compared with tamoxifen;

    induce tumor response in a subset of

    tamoxifen-resistant tumors. However,

    efficient only in postmenopausal patients.

    Anastrozole (Arimidex) On the market AstraZeneca http://www.astrazeneca.com [24]

    Letrozole (Femara) On the market Novartis http://www.novartis.com [24]

    Exemestane (Aromasin) On the market Pfizer http://www.pfizer.com [24]

    HER2 (ERBB2) HER2 is activated only in one

    out of four breast tumors.

    Antibodies

    Trastuzumab (Herceptin) On the market High-efficiency combined with good

    safety profile. Notice that HER2+

    tumors represent the most aggressive

    subset of breast cancer.

    Genentech, Ro he http://www.gene.com

    http://www.roche.com

    2C4 (Omnitarg, Pertuzumab) Phase III Genentech, Ro he http://www.gene.com

    http://www.roche.com

    [29]

    Small molecule

    inhibitors

    CP-724714 Phase I Pfizer http://www.pfizer.com [29]

    TAK-165 Phase I Takeda http://www.takeda.com [29]

    HER1 (EGFR) HER1 is rarely overexpressed

    in breast cancers.

    ZD1839 (Iressa, Gefitinib) Phase II AstraZeneca http://www.astrazeneca.com [29]

    OSI-774 (Tarceva, Erlotinib) Phase II OSI, Genentec , Roche http://www.osip.com

    http://www.gene.com

    http://www.roche.com

    [29]

    EKB-569 Phase I Wyeth http://www.wyeth.com [29]

    HER2 and HER1 GW572016 (GW2016) Phase II GlaxoSmithKli http://www.gsk.com [26]

    HER kinases CI-1033 (PD183805) Phase II Pfizer http://www.pfizer.com [26]

    VEGF Bevacizumab (Avastin) Phase II Genentech http://www.gene.com [30]

    VEGFR2 ZD6474 Phase II AstraZeneca http://www.astrazeneca.com [29]

    Farnesyl transferase Trials on several farnesyltransferase

    inhibitors have been discontinued due to

    high-toxicity and lack of clinical effect.

    R115777 (Zarnestra, Tipifarnib) Phase II Johnson and Jo nson http://www.jnj.com [31]

    SCH66336 (Sarasar, lonafarnib) Phase I Schering-Ploug http://www.schering-plough.com [31]

    CDK kinases

    Flavopiridol (Alvocidib) Phase III Aventis [32]

    ww

    w.d

    rugd

    iscoveryto

    day.co

    m241c

    c

    h

    ne

    h

    h

  • Dru

    gD

    iscovery

    Today:

    Disease

    Mech

    anism

    s|

    Can

    cerV

    ol.

    1,N

    o.2

    2004

    Table 2 (Continued).

    Target Therapy against target Stage of developmenta Advantages and/or

    disadvantages

    Who is working

    on the target

    Website Refs.b

    UCN-01 Phase I UCN-01 inhibits several other

    kinases, including PKC.

    Kyowa Hakko Kogyo [32]

    CYC202 (Roscovitine) Phase II Cyclacel [32]

    BMS-387032 Phase I Bristol-Myers Squibb [30]

    mTOR kinase

    CCI-779 (Temsirolimus) Phase IIIII Wyeth http://www.wyeth.com [30]

    RAD001

    (Everolimus, Certican)

    Phase I Novartis http://www.novartis.com [33]

    PKC Bryostatin-1 Clinical trials on other

    cancer types

    GPC Biotech [29]

    RAF Bay 43-9006 Clinical trials on other

    cancer types

    Onyx, Bayer [33]

    MEK CI-1040 (PD184352) Phase II Pfizer http://www.pfizer.com [33]

    Akt Perifosine Phase II Aeterna Zentaris http://www.aeterna.com [33]

    Hsp90 17-AAG Phase I Kosan Biosciences http://www.kosan.com [30]

    Matrix

    metalloproteinases

    High toxicity but limited

    therapeutic efficiency in initial

    clinical trials

    BB-2516 (marimastat) Phase II Vernalis http://www.vernalis.com [34]

    BMS-275291 Phase II Bristol-Myers

    Squibb, Celltech

    http://www.bms.com

    http://www.celltechgroup.com

    c-Kit and/or PDGFR STI571

    (Gleevec, Glivec, Imatinib)

    Phase II [28]

    COX2 Celecoxib (Celebrex) Phase IIIII COX2 is selectively expressed in

    tumors, thus COX2 inhibitors have

    good safety profile.

    Pfizer http://www.pfizer.com [27]

    Histone deacetylases SAHA, PXD101,

    LAQ-824, CI-994, MS-275

    Phase III Several research group and

    pharmaceutical compa s

    [32]

    Telomerase GRN163 Laboratory studies Geron http://www.geron.com [35]

    Prolactin receptor Laboratory studies [36]

    Chemokine receptors Laboratory studies Inhibition of chemokine receptors

    might interfere with metastatic process.

    [37]

    a Stage of development applies mainly for breast cancer but not for other cancer types; this is especially true for phase II/III trials and already marketed drugs. Notice that some of e already licensed compounds have been approved for the

    treatment of metastatic breast tumors, but still remain at the stage of clinical trials for the use in adjuvant therapy and/or chemoprevention.b In addition to the literature references indicated in the table, the information was updated using the websites of pharmaceutical companies, Clinical Trials site of the National Cancer Inst ute (http://www.cancer.gov/clinicaltrials), and the Catalog of

    FDA Approved Drug Products (http://www.accessdata.fda.gov/scripts/cder/drugsatfda).

    242

    ww

    w.d

    rugd

    iscoveryto

    day.co

    ms

    nie

    th

    it

  • Achilles heel of neoplasia. It is probable that the mechanism

    of action of conventional non-specific cytostatics utilizes this

    weakness, at least in part. Unfortunately, most of the cur-

    rently available cytotoxic agents have a low therapeutic index

    [38].

    Towards curable breast cancer: how long

    is the road?

    Modern therapeutic combinations can induce tumor

    response in more than 8090% of breast cancer patients.

    However, despite some spectacular examples of prolonged

    disease remission in selected women, the statistical survival

    benefit in metastatic breast cancer patients is estimated in

    months and not years. The situation is slightly better in

    adjuvant breast cancer treatment, where the therapy saves

    the lives in a certain (still frustratingly small) number of

    otherwise relapsed patients [38].

    It is often suggested that the future of breast cancer therapy

    Vol. 1, No. 2 2004 Drug Discovery Today: Disease Mechanisms | Cancerlies in the use of individualized, molecular portrait-based,

    sophisticated combinations of targeted drugs [41]. In this

    approach, the tumor specificity is achieved by fine adjust-

    ment of the multidrug panel to the unique spectrum of

    survival determinants detected in malignant tissue by expres-

    sion profiling (Fig. 2). It is assumed at the same time, that

    normal tissues do not carry exactly the same set of vital

    molecules, therefore, unlike the cancer cells, they can tolerate

    the cocktail of signal transduction modifiers. This model is in

    concordance with the clinical experience; indeed, combined

    therapy of breast cancer is definitively superior when com-

    pared with the monotherapy. However, there are an impress-

    ive number of entirely distinct therapeutic modalities whose

    combinations have already been used in breast cancer treat-

    ment or trials (e.g. cytotoxic agents with various mechanisms

    Figure 2. The promise of drug combinations. The concept of combined

    cancer therapy implies that all survival determinants of the tumor cell

    (depicpted in the center) can be efficiently blocked by the individually

    matched cocktail of signal transduction modifiers. The antitumor spe-

    cificity is warranted if none of normal cells (depicted at the four corners)

    carries the same set of targets.of action, ER pathway downregulators, HER2 antagonists,

    selective and, previously, non-selective COX2 inhibitors

    and so on). Following the logic described above, one would

    expect higher rate of prolonged breast cancer remissions than

    the one currently observed. There are several concerns related

    to the future of breast cancer therapy.

    With the possible exception of ER, HER2 and COX2, most

    of the currently considered targets are not truly cancer-spe-

    cific because they are involved in the functioning of vital

    human tissues. Not surprisingly, administration of signal

    transduction modifiers (Table 2) often results in noticeable

    adverse effects, sometimes comparable with toxicities of con-

    ventional cytostatics. Interestingly, in attempt to prevent

    cancer cell from rescuing from target inhibition, some com-

    pounds with broader substrate specificity have been recently

    tested (e.g. pan-HER inhibitor CI-1033, dual HER1HER2

    inhibitor GW572016 and CDKsPKC inhibitor UCN-01)

    (Table 2). Although the relaxed drug specificity might render

    higher antitumor activity [29], it also increases the risk of

    undesirable consequences. It is hoped that ongoing efforts in

    expression array profiling will help to increase the list of

    genuine breast cancer-specific targets.

    Another group of difficulties is related to the tumor ability

    to evolve over selective pressure of cancer treatment, and

    inevitably develop drug-resistant phenotype. The intrinsic

    genomic instability of cancer cells certainly facilitates this

    process. However, it is not immediately clear whether the

    treatment-related drug resistance is truly attributed to the

    adaptive genetic flexibility of tumor cell clones, or, vise versa,

    simply reflects the selection of pre-existing fatal cell subsets.

    In other words, it is difficult to differentiate between tumor

    evolution and tumor heterogeneity. Although the tumor

    heterogeneity has been acknowledged for a long time, its

    extent could have been underestimated. To the great surprise

    of scientific audience, some of the recent evidence suggests

    that a notable portion of breast cancer might be polyclonal in

    their origin, thus supporting the theory of tumor field [42].

    Furthermore, there are experimental data demonstrating that

    truly dangerous, potentially metastatic cells constitute only a

    negligible fraction of crude breast cancer lump [43]. Tumor

    heterogeneity might compromise current efforts of molecular

    portraying of neoplastic disease, because they are almost

    always based on the analysis of gross primary tumor mass

    and, therefore, might miss the targets in the most destructive

    cancer subclones (Box 1).

    Choice of the targets: cancer-specific or

    breast-specific?

    The initial phase of the search for cancer targets is usually

    based on the comparison of expression profiles in tumor

    versus corresponding normal tissues. Unfortunately, most

    of the detected variations are usually quantitative but notqualitative; furthermore, most of upregulated target mole-

    www.drugdiscoverytoday.com 243

  • Drug Discovery Today: Disease Mechanisms | Cancer Vol. 1, No. 2 2004cules occur only in a subset of carcinomas. Overall, the

    difference between the cancer cell and its normal precursor

    is much less than that between bacteria and mammalian

    organisms, which makes the search for the magic bullet

    against cancer far more complicated than the development of

    antibacterial therapies [41].

    It has been demonstrated that even transformed cells

    stably retain major lineage-specific molecular markers [44].

    In other words, there can be more difference in expression

    profiles between distinct normal tissues than between paired

    tumor-normal cell counterparts. If this is true, targeted che-

    mical organectomy (i.e. selective ablation of particular cell

    lineage) can be more achievable than specific elimination of

    cancer cells [45]. This approach can have a future for the

    treatment of cancers arising from those organs whose loss is

    compatible with survival of the host (breast, prostate, ovary

    and so on). Interestingly, this strategy has some similarities

    with the bone marrow transplantation, where the first step

    includes non-selective elimination of both malignant and

    normal hemopoietic cell clones. The potential feasibility of

    the chemical organectomy is difficult to assess, because the

    comparative cataloging and functional understanding of

    expression profiles of the normal human tissues appears to

    be less developed compared with the body of data accumu-

    lated in the studies of neoplastic disease.

    Summary and conclusions

    Breast cancer shares all of the key properties of malignancy,

    however, it also has some distinct features. At the clinical

    level, breast cancer is relatively easily detectable even in early

    stages; in many instances, it has a reasonably favorable

    prognosis, therefore, breast cancer patients predominate

    among cancer survivors. At the phenotypic level, breast

    cancer is not as malignant as certain other cancers because

    more than 50% breast cancer retain at least partial estrogen

    dependence. At the genetic level, breast carcinomas are char-

    acterized by an increased number of chromosomal abnorm-

    alities, but a limited frequency of small intragenic mutations.

    In contrast to many other epithelial tumors, several genuine

    breast cancer-specific targets have been identified. The intro-

    duction of appropriate targeted therapies, such as tamoxifen

    and aromatase inhibitors against ER pathway, or Herceptin

    against HER2 receptor, resulted in unprecedented clinical

    benefits. However, although currently available approaches

    enable short-term remission in the majority of breast cancer

    patients, metastatic breast cancer remains largely an incur-

    able disease. It is hoped that individualized, expression pro-

    file-based adjustment of drug combinations will improve the

    treatment efficacy. However, there are some intrinsic proper-

    ties of breast cancer biology, which might complicate the

    discovery of smart drugs. In particular, most of the currently

    considered targets show only quantitative but not qualitativeoverexpression in cancer versus normal cells; this might

    244 www.drugdiscoverytoday.comnegatively affect the therapeutic index of newly developed

    compounds. Some alarming data indicate that the most

    dangerous cell subsets might constitute only a minor portion

    of primary breast tumor; if it is so, the molecular profiling of

    gross tumor mass might misguide the search for the cancer

    treatment. Most of current target searches rely on the catalo-

    ging of molecules that are upregulated in cancer cells com-

    pared with their normal precursors. Because the entire loss of

    breast epithelium is compatible with survival, it might turn

    out that the breast-specific molecular targets have advantage

    over the cancer-specific ones. The feasibility of this concept is

    difficult to assess, even in theory, as a result of the deficiency

    in systematic understanding of molecular physiology of nor-

    mal human cells.

    Acknowledgements

    We thank Ekatherina Kuligina for her invaluable help in the

    preparation of figures. We apologize to those authors whose

    articles could not be cited because of space limitations. This

    work was supported by INTAS (grant 03-51-4234) and RFBR

    (grant 02-04-49890).

    References1 Singletary, S.E. (2003) Rating the risk factors for breast cancer. Ann. Surg.

    237, 474482

    2 McPherson, K. et al. (2000) ABC of breast diseases. Breast cancer-

    epidemiology, risk factors, and genetics. Br. Med. J. 321, 624628

    3 Clemons, M. and Goss, P. (2001) Estrogen and the risk of breast cancer. N.

    Engl. J. Med. 344, 276285

    4 Gerber, B. et al. (2003) Nutrition and lifestyle factors on the risk of

    developing breast cancer. Breast Cancer Res. Treat. 79, 265276

    5 Iau, P.T. et al. (2001) Germ line mutations associated with breast cancer

    susceptibility. Eur. J. Cancer 37, 300321

    6 Colleu-Durel, S. et al. (2004) Alkaline single-cell gel electrophoresis

    (comet assay): a simple technique to show genomic instability in sporadic

    breast cancer. Eur. J. Cancer 40, 445451

    7 Imyanitov, E.N. et al. (2004) Searching for cancer-associated gene poly-

    morphisms: promises and obstacles. Cancer Lett. 204, 314

    8 Mant, C. and Cason, J. (2004) A human murine mammary tumour virus-

    like agent is an unconvincing aetiological agent for human breast cancer.

    Rev. Med. Virol. 14, 169177

    9 Mant, C. et al. (2004) A viral aetiology for breast cancer: time to re-

    examine the postulate. Intervirology 47, 213

    10 Nathanson, K.L. et al. (2001) Breast cancer genetics: what we know and

    what we need. Nat. Med. 7, 552556

    11 Antoniou, A. et al. (2003) Average risks of breast and ovarian cancer

    associated with BRCA1 or BRCA2 mutations detected in case Series

    unselected for family history: a combined analysis of 22 studies. Am. J.

    Hum. Genet 72, 11171130

    12 Lerebours, F. and Lidereau, R. (2002) Molecular alterations in sporadic

    breast cancer. Crit. Rev. Oncol. Hematol. 44, 121141

    13 Widschwendter, M. and Jones, P.A. (2002) DNA methylation and breast

    carcinogenesis. Oncogene 21, 54625482

    14 Okochi, E. et al. (2002) Single nucleotide instability: a wide involvement in

    human and rat mammary carcinogenesis? Mutat. Res. 506507, 101111

    15 Imyanitov, E.N. and Hanson, K.P. (2003) Molecular pathogenesis of

    bilateral breast cancer. Cancer Lett. 191, 17

    16 Bianchi, N.O. et al. (2001) Mitochondrial genome instability in human

    cancers. Mutat. Res. 488, 923

    17 Bertucci, F. et al. (2003) Breast cancer revisited using DNA array-basedgene expression profiling. Int. J. Cancer 103, 565571

  • 18 Miller, B.J. et al. (2003) Pooled analysis of loss of heterozygosity in breast

    cancer: a genome scan provides comparative evidence for multiple tumor

    suppressors and identifies novel candidate regions. Am. J. Hum. Genet. 73,

    748767

    19 Tomlinson, I.P. et al. (2002) Loss of heterozygosity analysis: practically

    and conceptually flawed? Genes Chromosomes Cancer 34, 349353

    20 Gasco, M. et al. (2002) The p53 pathway in breast cancer. Breast Cancer

    Res. 4, 7076

    21 Hanahan, D. and Weinberg, R.A. (2000) The hallmarks of cancer. Cell 100,

    5770

    22 Moinfar, F. et al. (2000) Concurrent and independent genetic alterations in

    the stromal and epithelial cells of mammary carcinoma: implications for

    tumorigenesis. Cancer Res. 60, 25622566

    23 Wiseman, B.S. and Werb, Z. (2002) Stromal effects on mammary gland

    development and breast cancer. Science 296, 10461049

    24 ORegan, R.M. and Jordan, V.C. (2002) The evolution of tamoxifen

    therapy in breast cancer: selective oestrogen-receptor modulators and

    downregulators. Lancet Oncol. 3, 207214

    25 Sorlie, T. et al. (2003) Repeated observation of breast tumor subtypes in

    independent gene expression data sets. Proc. Natl. Acad. Sci. USA 100,

    84188423

    26 Mass, R.D. (2004) The HER receptor family: a rich target for therapeutic

    development. Int. J. Radiat. Oncol. Biol. Phys. 58, 932940

    27 Arun, B. and Goss, P. (2004) The role of COX-2 inhibition in breast cancer

    treatment and prevention. Semin. Oncol. 31 (Suppl. 7), 2229

    28 Cristofanilli, M. and Hortobagyi, G.N. (2002) Molecular targets in breast

    cancer: current status and future directions. Endocr. Relat. Cancer 9, 249

    266

    29 Fischer, O.M. et al. (2003) Beyond Herceptin and Gleevec. Curr. Opin.

    Chem. Biol. 7, 490495

    30 Proceedings of the 40th Annual Meeting of American Society of Clinical

    Oncology, 58 June 2004. New Orleans, USA (http://www.aso.org)

    31 de Bono, J.S. et al. (2003) Farnesyltransferase inhibitors and their potential

    in the treatment of breast carcinoma. Semin. Oncol. 30 (Suppl. 16), 7992

    32 McLaughlin, F. et al. (2003) The cell cycle, chromatin and cancer:

    mechanism-based therapeutics come of age. Drug Discov. Today 8,

    793802

    33 de Bono, J.S. et al. (2003) The future of cytotoxic therapy: selective

    cytotoxicity based on biology is the key. Breast Cancer Res. 5, 154159

    34 Lo, S. and Johnston, S.R. (2003) Novel systemic therapies for breast

    cancer. Surg. Oncol. 12, 277287

    35 Mokbel, K. (2000) The role of telomerase in breast cancer. Eur. J. Surg.

    Oncol. 26, 509514

    36 Clevenger, C.V. et al. (2003) The role of prolactin in mammary carcinoma.

    Endocr. Rev. 24, 127

    37 Muller, A. et al. (2001) Involvement of chemokine receptors in breast

    cancer metastasis. Nature 410, 5056

    38 Esteva, F.J. et al. (2001) Chemotherapy of metastatic breast cancer: what to

    expect in 2001 and beyond. Oncologist 6, 133146

    39 Reff, M.E. and Heard, C. (2001) A review of modifications to recombinant

    antibodies: attempt to increase efficacy in oncology applications. Crit. Rev.

    Oncol. Hematol. 40, 2535

    40 Tazebay, U.H. et al. (2000) The mammary gland iodide transporter is

    expressed during lactation and in breast cancer. Nat. Med. 6, 871878

    41 Blagosklonny, M.V. (2003) Matching targets for selective cancer therapy.

    Drug Discov. Today 8, 11041107

    42 Heim, S. et al. (2001) Are some breast carcinomas polyclonal in origin? J.

    Pathol. 194, 395397

    43 Al-Hajj, M. et al. (2003) Prospective identification of tumorigenic breast

    cancer cells. Proc. Natl. Acad. Sci. USA 100, 39833988

    44 Liu, E.T. (2003) Classification of cancers by expression profiling. Curr.

    Opin. Genet. Dev. 13, 97103

    45 Blagosklonny, M.V. (2003) Tissue-selective therapy of cancer. Br. J.

    Cancer 89, 11471151

    Vol. 1, No. 2 2004 Drug Discovery Today: Disease Mechanisms | Cancerwww.drugdiscoverytoday.com 245

    Mechanisms of breast cancerIntroductionContribution of inherited featuresSomatic events in breast cancer pathogenesisThe web of signaling cascadesTargeted therapiesTowards curable breast cancer: how long is the road?Choice of the targets: cancer-specific or breast-specific?Summary and conclusionsAcknowledgementsReferences