Abstract

Biomarkers in MedicineVol. 2, No. 6 EditorialFree AccessMolecular markers in breast cancer: no longer a ‘free’ run from laboratory to the clinicAndrea NicoliniAndrea NicoliniUniversity of Pisa, Department of Internal Medicine, via Roma 67, Pisa 56126, Italy. Search for more papers by this authorEmail the corresponding author at a.nicolini@int.med.unipi.itPublished Online:12 Dec 2008https://doi.org/10.2217/17520363.2.6.531AboutSectionsPDF/EPUB ToolsAdd to favoritesDownload CitationsTrack CitationsPermissionsReprints ShareShare onFacebookTwitterLinkedInReddit In Western countries, breast cancer remains the most common cancer and the main reason for cancer-related death in women between 35 and 55 years of age [1,2]. Thus, any advance in the field has a great social impact and is rapidly acquired by the scientific community and clinical oncologists.In the last few decades, the identification of new molecular markers for prognostic as well as diagnostic, predictive and therapeutic purposes has been one of the basic research fields most developed to improve the management of breast cancer patients.Conventional clinicopathological factors for the prognosis of breast cancerBefore the 1950s, only few clinicopathological factors, namely tumor size (cm), node status (positive or negative), grade (grade of differentiation) and evidence of metastatic disease, were used to establish the prognosis of breast cancer patients.Around the 1950s, the International Union Against Cancer (UICC) worked on a project to provide a graphical representation of the anatomical extension of any malignant neoplasm. All the aforementioned clinicopathological factors except for grade were necessary to define a pathological (p) tumor (T), nodes (N) and metastatic involvement (M) based on classification known as pTNM that was considered the only classification appropriate for prognostic purposes and to allow treatment after the removal of primary cancer. The first version of pTNM by UICC was published in 1968. In 1987, the TNM classification of the UICC and that of The American Joint Committee on Cancer (AJCC) joined with the fourth version. Thereafter, pTNM classification was continuously updated up until 2002, when the sixth version was published. In the latest versions, grading and receptor status were also recommended to be considered. Therefore, from the mid-1900s onwards, pTNM and the consequential division by stages were the only classifications widely used to define prognosis of breast cancer patients.New emerging molecular markersThe enormous and rapidly increasing progress of techniques in molecular biology and genetics has permitted basic research to elucidate many molecular pathways involved in tumor growth, invasion and metastasization. Epigenetic changes, such as DNA methylation, have also been shown for several genes involved in cell cycle control, apoptosis, angiogenesis and detoxification processes [3–5]. Therefore, a great number of molecular markers related to specific functions and providing insights into cellular mechanisms, including oncogene or oncosuppressor gene deregulation, neoangiogenesis, cell cycle regulation, adhesion, invasion or extracellular matrix modification became available [6].Others are related to the damage provoked by the tumor in different organs or the immune response of the host to molecules overexpressed or overexposed by the tumor [7–9]. Among these molecular markers, estrogen and progesterone receptors have been proven and widely used. Since its introduction into clinical practice, this measurement was considered to provide additional information for prognosis and to predict probable hormone-responsiveness or not of cancer cells.As well as estrogen and progesterone receptors, a long list of putative molecular markers have been proposed. Among prognostic markers, EGF receptor (EGFR), HER2/neu gene (c-erbB2), HER2 protein (p185) (approximately a third of breast cancers do not express estrogen receptor [ER] and are regulated by growth-factor pathways using other receptors), MIB-1 (i.e., antibodies recognizing antigens related to cell cycle proteins) and p53 (p53-suppressive gene encodes for a 53-KDa nuclear phosphoprotein, wild-type p53, involved in controlling cell cycle regulation, cell differentiation and the surveillance of genomic integrity) are those that have been documented the most [10]. In addition, HER/neu gene overexpression has been shown to select hormone-resistant breast cancers and those that are candidates for immunotherapy with anti-HER2 protein monoclonal antibodies (Herceptin®). In addition, other clinicopathological factors such as microvessel count, bone marrow micrometastatic disease, circulating tumor cells and newer approaches such as the number of dividing cells (the mitotic index) and the percentage of cells in the tumor that are making new DNA, (i.e., S-phase fraction) have been investigated and proposed by different researchers for prognostic purposes [10]. In recent years, microarray technology, with its ability to simultaneously examine thousands of genes, permitted visual recognition of tumors that share the so-called ‘genetic signature’ [11]. Using this technology, breast tumors have been grouped into six categories, defined as basal-like, erbB2-positive, normal breast-like and luminal types A, B and C, with luminal types sharing the expression of ER and ER-related genes, and basal-like and erbB2-positive subtypes associated with the shortest survival times [12].Among risk-assessment markers, the BRCA1 and BRCA2 genes have been found to be associated with familial breast cancer [13], while high concentrations of prolactin, insulin, IGF-1 and androgens (i.e., testosterone and adrenal androgens) have often been detected in subjects who eventually developed breast cancer [14]. Other markers that are detected in serum, such as carcinoembryonic (CEA) and mucine-like antigens, are commonly considered tumor burden-related markers [15]. They have largely been investigated for early detection and monitoring of response to treatment by metastatic disease [16,17]. Further markers of response or progression of metastatic disease during treatment can be expected once a better understanding of the relationship between the breast cancer microenvironment and cell-mediated immunity of the host is achieved [18–20].Presently, a large number of molecular markers and many clinicopathological factors can be assessed. Moreover, this long list is inevitably destined to increase. Theoretically, most of them have the potential to refine the prognosis, while others, such as the BRCA1 and BRCA2 genes or tumor burden-related circulating markers, are indicated for risk assessment or postoperative monitoring. Nevertheless, in spite of this large availability, these two related questions have arisen: ▪ Among the available markers, which of them are actually useful?▪ Among the useful markers, which ones have a favorable cost–effectiveness ratio?In fact, the principal aim of any marker is to drive the decision-making process of the clinical oncologist, which has a relevant impact on healthcare expenses.Business, regulation & clinical utilityA long time has passed since estrogen and progesterone receptor determination entered into standard clinical practice. Thereafter, many others have been investigated and proposed. They initially evolved as single molecular elements and progressed to reflect insights into pathophysiology as a panel of genes and their associated mutations, the so-called ‘genetic profiling’ or ‘signature’, up to the recent possibility of assessing the entire transcriptome or proteome to distinguish diseased from normal tissues or disease categories with different risk profiles. In spite of these enormous advances and concomitant financial investments in the field, the only other molecular marker that has thus entered into clinical guidelines is HER2/neu gene expression. In fact, in 2005, the International Expert Consensus on the Primary Therapy of Early Breast Cancer considered this single molecular marker relevant for defining endocrine responsiveness and risk categories [21]. It should be inferred that the remaining molecular markers are either not useful or insufficient documentation is available. BRCA1 and BRCA2 testing is recommended in selected populations of subjects with high incidence of familial breast cancer, while circulating CEA and CA15.3 can be useful for postoperative monitoring in the first years following removal of primary breast cancer in patients with a high risk of relapse. However, in view of increasing demands by clinical oncologists for a more refined prognosis and management of breast cancer patients with respect to the so-called targeted and/or tailored therapies, it must be recognized that there is a significant lack of regulations by independent authorities.In 1996, a Tumor Marker Grading Utility System to standardize tumor-marker information for clinical utility and to establish an investigational agenda for evolution of new tumor markers was proposed [22]. Following this, improved patient outcomes, and more cost-efficient investigation and application of tumor markers were expected. So far, this program has often not been applied, and efforts to standardize the use of tumor markers in clinical studies have only been considered as anecdotal. It is likely that failure to provide definitive validation, qualification and comparison among markers of the same category has been the main obstacle impeding their integration into current clinical practice and patient management. In 2003, a working group developed a check-list and flow diagram to improve the quality and completedness of studies reporting on diagnostic accuracy [23]. The validation of the Standard for Reporting of Diagnostic Accuracy (STARD) initiative is expected to contribute to a constant improvement in the quality of study designs, including those on cancer markers. More recently, the Pharmaceutical Research and Manufacturers of America and the US FDA created a consortium for the development of biomarkers established under the Foundation for NIH in collaboration with the Centers for Medicare and Medicaid Services, academic institutions and representatives from the private sector, including pharmaceutical, biotechnology and diagnostic companies [24]. This consortium is open to public or private institutions, and will manage biomarker programs to ensure scientific integrity, appropriate resources and compliance with relevant statutes. There is the hope that these are the first steps of a necessary and constant collaboration among academia, industry and regulatory agencies. To optimize biomarker application, it is likely that relational databases of many long-term followed-up patients are required to compare patient molecular profiles to clinicopathological characteristics. Such databases, if appropriately constructed, will permit maximized accessibility, applicability and utility. In this context, it is not impossible to imagine a time when clinical oncologists can select ‘the best markers for the individual patient’.Financial & competing interests disclosureThe author has no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.No writing assistance was utilized in the production of this manuscript.Bibliography1 Cancer Incidence in Five Continents (Volume VIII). Parkin DM, Whelan SL, Ferlay J, Teppo L (Eds). IARC Scientific Publication no. 155. Lyon, France (2002).Google Scholar2 Jemal A, Siegel R, Ward E et al.: Cancer statistics 2006. CA Cancer J. Clin.56(2),106–130 (2006).Crossref, Medline, Google Scholar3 Feinberg AP: Cancer epigenetics takes center stage. Proc. Natl Acad. Sci. USA98(2),392–394 (2001).Crossref, Medline, CAS, Google Scholar4 Jones PA, Laird PW: Cancer epigenetics comes of age. Nat. Genet.21(2),163–167 (1999).Crossref, Medline, CAS, Google Scholar5 Esteller M, Sparks A, Toyota M et al.: Analysis of adenomatous polyposis coli promoter hypermethylation in human cancer. Cancer Res.60(16),4366–4371 (2000).Medline, CAS, Google Scholar6 Arciero C, Somiari SB, Shriver CD et al.: Functional relationship and gene ontology classification of breast cancer biomarkers. Int. J. Biol. Markers18(4),241–272 (2003).Crossref, Medline, CAS, Google Scholar7 Robertson JF, Jaeger W, Syzmendera JJ et al.: The objective measurement of remission and progression in metastatic breast cancer by use of serum tumour markers. European Group for Serum Tumour Markers in Breast Cancer. Eur. J. Cancer35(1),47–53 (1999).Crossref, Medline, CAS, Google Scholar8 Cheung KL, Graves CRL, Robertson JF: Autoantibodies as circulating cancer markers. In: Tumor Marker: Physiology, Pathobiology, Technology, and Clinical Applications. Diamanidis EP, Fritsche HA, Lilja DW, Chan DW, Schwartz MK (Eds). AACC Press, Washington DC, USA 123–133 (2002).Google Scholar9 Soussi T: p53 antibodies in the sera of patients with various types of cancer: a review. Cancer Res.60(7),1777–1788 (2000).Medline, CAS, Google Scholar10 Nicolini A, Ferrari P, Cavazzana A, Carpi A, Berti P, Miccoli P: Conventional and new emerging prognostic factors in breast cancer: an update. Biomarkers Med.1(4),525–554 (2007).▪ Report that extensively examines prognostic factors for operable breast cancer basing upon a systematic review of selected good quality reviews.Link, Google Scholar11 van’t Veer LJ, Paik S, Hayes DF: Gene expression profiling of breast cancer: a new tumor marker. J. Clin. Oncol.23(8),1631–1635 (2005).Crossref, Medline, Google Scholar12 Sorlie T, Perou CM, Tibshirani R et al.: Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc. Natl Acad. Sci. USA98(19),10869–10874 (2001).Crossref, Medline, CAS, Google Scholar13 Iau PT, Macmillan RD, Blamey RW: Germ line mutations associated with breast cancer susceptibility. Eur. J. Cancer37(3),300–321 (2001).Crossref, Medline, CAS, Google Scholar14 Seregni E, Coli A, Mazzucca N: Italian Group RIA-IRMA Test, Italian Association of Nuclear Medicine. Circulating tumour markers in breast cancer. Eur. J. Nucl. Med. Mol. Imaging31(Suppl. 1),S15–S22 (2004).Crossref, Medline, Google Scholar15 Gion M, Daidone MG: Circulating biomarkers from tumour bulk to tumour machinery: promises and pitfalls. Eur. J. Cancer40(17),2613–2622 (2004).Crossref, Medline, CAS, Google Scholar16 Bast RC Jr, Ravdin P, Hayes DF et al.: American Society of Clinical Oncology Tumor Markers Expert Panel. 2000 update of recommendations for the use of tumor markers in breast and colorectal cancer: clinical practice guidelines of the American Society of Clinical Oncology. J. Clin. Oncol.19(6),1865–1878 (2001).Crossref, Medline, Google Scholar17 Nicolini A, Carpi A, Tarro G: Biomolecular markers of breast cancer. Front. Biosci.11,1818–1843 (2006).Crossref, Medline, CAS, Google Scholar18 Nicolini A, Carpi A: β-interferon and interleukin-2 prolong more than three times the survival of 26 consecutive endocrine dependent breast cancer patients with distant metastases: an exploratory trial. Biomed. Pharmacother.59(5),253–263 (2005).Crossref, Medline, CAS, Google Scholar19 Nicolini A, Carpi A, Rossi G: Relationship of cellular immunity, cytokines and CRP with clinical course in breast cancer patients with endocrine-dependent distant metastases treated with immunotherapy. Cancer Lett.251(2),330–338 (2007).Crossref, Medline, CAS, Google Scholar20 Nicolini A, Carpi A: Immune manipulation of advanced breast cancer: an interpretative model of the relationship between immune system and tumor cell biology. Medicinal Res. Rev. (2008) (In Press).Google Scholar21 Goldhirsch A, Glick JH, Gelber RD, Coates AS, Thürlimann B, Senn HJ: Panel members. Meeting highlights: international expert consensus on the primary therapy of early breast cancer 2005. Ann. Oncol.16(10),1569–1583 (2005).▪ Report on an international expert consensus that updated guidelines and recommendations for selection of adjuvant systemic treatment in specific breast cancer patients.Crossref, Medline, CAS, Google Scholar22 Hayes DF, Bast RC, Desch CE et al.: Tumor marker utility grading system: a framework to evaluate clinical utility of tumor markers. J. Natl Cancer Inst.88(20),1456–1466 (1996).Crossref, Medline, CAS, Google Scholar23 Bossuyt PM, Reitsma JB, Bruns DE et al.: Standards for reporting of diagnostic accuracy. Towards complete and accurate reporting of studies of diagnostic accuracy: the STARD initiative. Clin. Biochem.36(1),2–7 (2003).Crossref, Medline, Google Scholar24 Wagner JA, Williams SA, Webster CJ: Biomarkers and surrogate end points for fit-for-purpose development and regulatory evaluation of new drugs. Clin. Pharmacol. Ther.81(1),104–107 (2007).▪▪ Report that deals with the need of a consistent framework for the acceptance and qualification of biomarkers and subsequent application in drug development.Crossref, Medline, CAS, Google ScholarFiguresReferencesRelatedDetails Vol. 2, No. 6 Follow us on social media for the latest updates Metrics History Published online 12 December 2008 Published in print December 2008 Information© Future Medicine LtdFinancial & competing interests disclosureThe author has no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.No writing assistance was utilized in the production of this manuscript.PDF download

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call