Clinical proteomics in breast cancer ISBN/EAN: 978-90-393-4994-6 © 2009 Marie-Christine Gast, Den Haag Cover design: Printed by: Initium- grafisch en interactief, Utrecht, The Netherlands Gildeprint Drukkerijen BV, Enschede, The Netherlands Clinical proteomics in breast cancer “Clinical proteomics” in borstkanker (met een samenvatting in het Nederlands) PROEFSCHRIFT ter verkrijging van de graad van doctor aan de Universiteit Utrecht op gezag van de rector magnificus, prof. dr J.C. Stoof, ingevolge het besluit van het college voor promoties in het openbaar te verdedigen op donderdag 12 februari 2009 des middags te 12.45 uur door Maria Christine Willemine Gast geboren op 23 februari 1978 te Woerden Promotoren: Promotoren: Prof. dr J.H. Beijnen Prof. dr J.H.M. Schellens The laboratory research described in this thesis was performed at the Department of Pharmacy & Pharmacology, Slotervaart Hospital / The Netherlands Cancer Institute, Amsterdam, The Netherlands The research in this thesis was financially supported by the Dutch Cancer Society (project NKI 2005-3421). Publication of this thesis was financially supported by: Dutch Cancer Society, Amsterdam, The Netherlands The Netherlands Laboratory for Anticancer Drug Formulation, Amsterdam, The Netherlands Genzyme Nederland, Naarden, The Netherlands Contents Preface 9 Chapter 1 Introduction 1.1 Clinical proteomics in breast cancer: a review Chapter 2 Technical aspects 2.1 Comparing the old and new generation SELDI-TOF MS: Consequence for serum protein profiling 39 2.2 Serum protein profiling using SELDI-TOF MS: Influence of sample storage duration 59 Chapter 3 Protein profiling of serum 3.1 Serum protein profiling for diagnosis of breast cancer using SELDI-TOF MS 83 3.2 SELDI-TOF MS serum protein profiles in breast cancer: Assessment of robustness and validity 101 3.3 Haptoglobin phenotype is not a predictor of recurrence free survival in high-risk primary breast cancer patients 121 3.4 Post-operative proteomic profiles may predict recurrence free survival in high-risk primary breast cancer 143 Chapter 4 Protein profiling of tissue 4.1 Detection of breast cancer by SELDI-TOF MS tissue and serum protein profiling 13 163 perspectivess Conclusions and perspective 187 Summary 197 Samenvatting 201 Dankwoord Dankw oord 209 Curriculum Vitae 213 List of publications 215 Preface Preface Breast cancer imposes a significant healthcare burden on women worldwide. For example, in the USA, breast cancer currently is estimated to be the most commonly diagnosed neoplasm in women, accounting for more than a quarter of all new female cancer cases (1). In addition, preceded only by lung cancer, breast cancer is at present the second leading cause of cancer deaths (1). Despite the substantial progress made in cancer therapy, the five-year survival rate of breast cancer still is inversely proportional to its stage at the time of diagnosis (2). Hence, short of prevention, detection of breast cancer at an early, still curable, stage would offer the best route to decrease its mortality rates. However, since many patients present with advanced disease, the currently applied diagnostic screening tools (e.g., mammography) obviously do not suffice for adequate breast cancer diagnosis. In addition, despite the survival benefit achieved by locoregional treatment and adjuvant systemic therapy, many breast cancer patients will eventually develop metastatic relapse and die (3), while a small percentage of patients would have survived without these treatment modalities. Evidently, the currently applied prognostic and predictive markers (e.g., age, hormone receptor status) lack adequate performance as well. Hence, better markers for early diagnosis, accurate prognosis and treatment prediction, applied either individually or in conjunction with existing modalities, are warranted to improve breast cancer care. Although (breast) cancer is, for a large part, a genetic disease, it is currently understood that gene analysis by itself does not provide a complete picture of the actual state of an individual. Instead, the functional “end-units” of the genome, the proteome, will offer a more dynamic and accurate reflection of a biological status. The clinical relevance of these proteins as cancer biomarkers is augmented by their ease of access in blood, being a readily accessible biological matrix that allows for repeated collection. In fact, several blood proteins are already in use a breast cancer markers (e.g., Cancer Antigen 15.3 and 27.29) (4). Their lack of adequate performance, however, precludes their use as singular breast cancer markers. Conversely, a panel of protein markers is expected to better reflect breast cancer complexity, yielding improved sensitivities and specificities. The search for this biomarker panel has been boosted by recent developments in mass spectrometry, resulting in a.o. the surface-enhanced laser desorption/ionisation time-offlight mass spectrometry (SELDI-TOF MS) technology (5). Enabling the simultaneous detection of a large part of the (blood) proteome in a high-throughput fashion, this technology holds promise as a screening tool for discovery of cancer biomarkers. A landmark paper in this respect was written by Petricoin et al. (6), providing the first report on SELDI-TOF MS serum protein profiling for identification of (ovarian) cancer patients. The objectives of this thesis were the evaluation and application of SELDI-TOF MS protein profiling for detection of serum and tissue protein profiles that could yield new biomarkers for diagnosis and prognosis of breast cancer. First, we provide an overview 9 Preface of protein profiling studies performed in breast cancer, and evaluate the potential of proteins identified by this research for clinical use as breast cancer biomarkers (Chapter 1). Subsequently, both technical (Chapter 2.1) and pre-analytical (Chapter 2.2) aspects related to protein profiling research were investigated. The SELDI-TOF MS technology was then used for protein profiling of serum, searching for novel markers that can be applied in diagnosis (Chapter 3.1) or prognosis (Chapter 3.3 and 3.4) of breast cancer, and determining the reproducibility of diagnostic serum protein profiles (Chapter 3.2). Finally, to augment insight into the pathophysiological mechanisms associated with, or underlying, breast cancer, the SELDI-TOF MS technology was applied in protein profiling of breast tissue (Chapter 4). References (1) Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T et al. Cancer statistics, 2008. CA Cancer J Clin 2008; 58(2):71-96. (2) Ries L, Melbert D, Krapcho M, Stinchcomb D, Howlader N, Horner M et al. SEER Cancer Statistics Review, 1975-2005, National Cancer Institute. Bethesda, MD, http://seer.cancer.gov/csr/1975_2005/ , based on November 2007 SEER data submission, posted on the SEER website. 2008. (3) Polychemotherapy for early breast cancer: an overview of the randomised trials. Early Breast Cancer Trialists' Collaborative Group. Lancet 1998; 352(9132):930-942. (4) Harris L, Fritsche H, Mennel R, Norton L, Ravdin P, Taube S et al. American Society of Clinical Oncology 2007 update of recommendations for the use of tumor markers in breast cancer. J Clin Oncol 2007; 25(33):5287-5312. (5) Hutchens TW, Yip TT. New desorption strategies for the mass spectrometric analysis of macromolecules. Rapid Commun Mass Spectrom 1993; 7:576-580. (6) Petricoin EF, Ardekani AM, Hitt BA, Levine PJ, Fusaro VA, Steinberg SM et al. Use of proteomic patterns in serum to identify ovarian cancer. Lancet 2002; 359(9306):572-577. 10 Chapter Introduction 1 Chapter Clinical proteomics in breast cancer: a review Marie-Christine W. Gast Jan H.M. Schellens Jos H. Beijnen Breast Cancer Res Treatm 2008; in press 1.1 Chapter 1.1 Abstract Breast cancer imposes a significant healthcare burden on women worldwide. Early detection is of paramount importance in reducing mortality, yet the diagnosis of breast cancer is hampered by the lack of an adequate detection method. In addition, better breast cancer prognostication may improve selection of patients eligible for adjuvant therapy. Hence, new markers for early diagnosis, accurate prognosis and prediction of response to treatment are warranted to improve breast cancer care. Since proteomics can bridge the gap between the genetic alterations underlying cancer and cellular physiology, much is expected from proteome analyses for the detection of better protein biomarkers. Recent technical advances in mass spectrometry, such as matrix-assisted laser desorption/ionisation time-of-flight mass spectrometry (MALDI-TOF MS) and its variant surface-enhanced laser desorption/ionisation (SELDI-) TOF MS, have enabled high-throughput proteome analysis. In the current review, we give a comprehensive overview of the results of expression proteomics (i.e., protein profiling) research performed in breast cancer using these two platforms. Many protein peaks have been reported to bear significant diagnostic, prognostic or predictive value, however, only few candidate markers have been structurally identified yet. In addition, although of pivotal importance in preventing overfitting of data and systematic bias by preanalytical parameters, validation of biomarker candidates by other, quantitative, methods and/or in new populations is very limited. Moreover, none of the identified candidate biomarkers has been investigated for their utility as breast cancer markers in large, prospective, clinical settings. As such, the candidate biomarkers discussed in this overview have not been validated sufficiently to be used for clinical patient care. Nonetheless, regarding the promising results up to now, MALDI- and SELDI-TOF MS protein profiling studies could eventually fulfil the great promise that protein biomarkers have for improving cancer patient outcome, provided that these studies are performed with adequate statistical power and analytical rigour. 14 Clinical proteomics in breast cancer Introduction Breast cancer imposes a significant healthcare burden to women worldwide. For example, in the USA, breast cancer is estimated to be the most commonly diagnosed neoplasm in women in 2008, as it will account for 26% of all new female cancer cases (1). In addition, preceded only by lung cancer, breast cancer is expected to be the second leading cause of USA cancer deaths in 2008 (1). The five-year survival rates of breast cancer decrease from 98% for localised disease to 26% for late stage disease (2). Hence, short of prevention, detection of breast cancer at an early, still curable stage would offer the best route to decrease its mortality rates. However, since only 63% of breast cancers are still confined to the breast at the time of diagnosis (1), the currently applied diagnostic screening tools (e.g., mammography) obviously do not suffice for adequate breast cancer diagnosis. In addition, despite the survival benefit achieved by locoregional treatment and adjuvant systemic therapy, 30-50% of breast cancer patients will eventually develop metastatic relapse and die (3), while a small percentage of patients would have survived without these treatment modalities. Evidently, the currently applied prognostic and predictive markers (e.g., age, hormone receptor status) lack adequate performance as well. Hence, better markers for early diagnosis, accurate prognosis and prediction of response to treatment are warranted to improve breast cancer care. We now comprehend that cancer arises from successive genetic changes, by which a number of cellular processes, including growth control, senescence, apoptosis, angiogenesis, and metastasis, are altered (4;5). Consequently, researchers initially searched for markers by employing genomic and transcriptomic approaches, providing new biomarkers (e.g., (6-9)) and expanding our insight into the genetic basis of cancer. It is, however, currently understood that gene analysis by itself provides an incomplete picture. Due to alternative splicing of both mRNA and proteins, combined with more than 100 unique post-translational modifications, one gene can give rise to multiple protein species (10). Hence, compared to the genome, the proteome can provide a more dynamic and accurate reflection of both the intrinsic genetic programme of the cell and the impact of its immediate environment (11). Since proteome analysis can provide the link between gene sequence and cellular physiology (12), proteomics is expected to complement gene analyses for evaluating disease development, prognosis, and response to treatment (13). Until recently, the search for novel protein biomarkers has been dominated by twodimensional gel electrophoresis (14), a major disadvantage of which is its lack of real high-throughput capability. However, recent advances in analytical technologies, such as protein microarrays and mass spectrometry (MS), have enabled large-scale proteomic analyses (15). Due to their relative simplicity of sample preparation, high analytical sensitivity and speed of data acquisition, two MS-based technologies in particular, i.e., matrix-assisted laser desorption/ionisation time-of-flight (MALDI-TOF) MS (16) and its 15 Chapter 1.1 variant surface-enhanced laser desorption/ionisation (SELDI-) TOF MS (17;18) have been widely deployed for cancer biomarker discovery (19). In both laser desorption/ionisation (LDI) platforms, biological samples (e.g., serum, tissue lysate) are co-crystallised with an energy absorbing matrix on a sample probe surface. Subsequent irradiation with brief laser pulses sublimates and ionises the proteins out of their crystalline matrix, after which an electric field migrates the charged proteins to the time-of-flight mass analyser. Herein, proteins are separated based on their mass, as the time to detector impact (TOF) is proportional to protein mass per charge (m/z). The two LDI platforms differ in their sample probe surfaces. In MALDI, the probe surface merely presents the sample to the mass spectrometer, warranting off-line sample fractionation and clean-up to produce usable MS signals. In contrast, the probe surfaces utilised by SELDI are comprised of various chromatographic surfaces, enabling their active role in sample fractionation (Figure 1). In the current overview, we focus on the expression proteomics (i.e., protein profiling) studies performed using the two LDI platforms in the search for novel breast cancer biomarkers. We will discuss the studies performed thus far for discovery of diagnostic, prognostic and predictive biomarkers, and evaluate the potential of the discriminating proteins identified in this research for clinical use as breast cancer biomarkers. Diagnostic protein profiling studies Short of prevention, detection at an early stage remains the best route to decrease breast cancer related mortality. Hence, the majority of MALDI/SELDI protein profiling studies performed in breast cancer has searched for novel diagnostic markers (Table 1). All diagnostic protein profiling studies were performed in vivo, investigating various biological matrices, including serum, plasma and tissue, but also nipple aspirate fluid, ductal lavage fluid and saliva. Protein profiling of tissue As tissue proteins will reflect the earliest changes caused by the successive genetic mutations that lead to breast cancer, it has been hypothesised that the concentration of potential biomarkers is highest in the tumour and its immediate microenvironment (19). Although tissue provides an invaluable sample source, tissue sampling through biopsies is highly invasive, thereby limiting the number of diagnostic tissue protein profiling studies performed thus far. Analysis of tumour tissue lysates by SELDI-TOF MS revealed several peaks that were significantly associated with lymph node status (20) or cancer subtype (i.e., lobular and ductal carcinoma) (21). However, the search for tumour originating proteins can be complicated by the high cellular heterogeneity of whole tumour tissue specimens. This can be reduced by laser capture microdissection (LCM), enabling selective capture of a 16 Clinical proteomics in breast cancer specific subset of cells (22). Following microdissection, captured cells can be mounted directly on a MALDI target, thereby preserving their spatial conformation for imaging MS (23;24). Using LCM, Umar et al. (24) detected 9 differentially expressed tryptic peptides (not structurally identified) following analysis of stromal and tumour cells collected from five tissue specimens. In addition, Sanders et al. (23) identified ubiquitin and S100-A8 to be decreased in tumour (n = 122) compared to normal tissue (n = 167), whereas S100-A6 was found increased. Their split-sample approach allowed a successful within-study validation of the three potential markers (23). As both ubiquitin and S100A6 were also found to decrease in lysates of human breast cancer cell lines following chemotherapy induced apoptosis (25), these proteins may provide insight into the pathogenesis of breast cancer upon further investigation. Despite the clear potential of (tumour) tissue to yield cancer-specific diagnostic biomarkers, their routine clinical application is seriously hampered by the intricacies associated with tissue sampling. Although this can be avoided by assessment of tumourderived markers in easier accessible biological matrices such as serum, this type of validation has not been performed in breast cancer yet. Protein profiling of serum and plasma Since whole blood is considered to provide a dynamic reflection of physiological and pathological status, human plasma and serum represent the most extensively studied biological matrices in the quest for (breast) cancer biomarkers (26). Constantly perfusing and percolating the human body, the blood compartment endows a proteinrich information archive (27). Besides the expected circulatory proteins, this archive also contains specific tumour-secreted proteins, normal tissue- and plasma-proteins digested by tumour-secreted proteases, and proteins produced by local and distant responses to the tumour (11;28;29). Moreover, whole blood is an easy to sample, readily accessible matrix that allows repeated collection, thereby augmenting the clinical relevance of candidate blood-borne biomarkers (28;30). Several MALDI-TOF MS and SELDI-TOF MS peaks (not structurally identified) have been reported to differentiate between serum or plasma of breast cancer patients, patients with benign breast disease and/or healthy controls (31-36). Since a small percentage (7 to 10%) of breast cancers is attributable to hereditary syndromes (e.g., BRCA-1, -2 mutations), Becker et al. (37) investigated whether the BRCA-1 mutation was reflected by the serum proteome. Multiple SELDI-TOF MS peaks were significantly different in expression between breast cancer patients with and without the BRCA-1 mutation (37). However, as none of these peaks were structurally identified, their association to the BRCA-1 gene remains unclear. Moreover, none of the peaks reported by these studies have been validated by analysis of an independent sample set. Yet validation is of utmost importance to ascertain reproducibility and prevent systematic bias and overfitting of data. This is highlighted by a study of our group (38), in which the potential markers for breast cancer and lymph node status, reported by Vlahou et al. 17 Chapter 1.1 (39) and Laronga et al. (34), respectively, could not be confirmed following analysis of an independent sample set. In contrast, Belluco et al. (40) report excellent performance of their seven-peak classifier (not structurally identified) following validation by an independent sample set analysed 14 months after their initial discovery study. Figure 1 Schematic representation of the MALDI- and SELDI-TOF MS principle (adapted from (15)). A) Protein profiling by MALDI-TOF MS: 1. samples (μl volume) are fractionated off-line using for instance magnetic beads coated with a chromatographic surface (e.g., hydrophilic, hydrophobic, cationic, anionic, or immobilised metal affinity capture moiety), 2. addition of energy absorbing matrix (e.g., α-cyano-4-hydroxycinnamic acid) to (fractionated) samples, 3. application of mixed specimen to inert target plate for laser irradiation in C. B) Protein profiling by SELDI-TOF MS: 1. application of sample (μl volume) from, for example, cancer and control patients to an 8-spot array with a chromatographic surface (e.g., hydrophilic, hydrophobic, cationic, anionic, or immobilised metal affinity capture moiety) in appropriate binding buffer, 2. on-chip sample cleanup using various wash-buffers, 3. application of energy absorbing matrix (e.g., sinapinic acid) for desorption / ionisation of proteins by laser irradiation in a laser desorption/ionisation time-of-flight analyser (C). C) Schematic representation of laser desorption/ionisation (LDI) time-of-flight (TOF) analyser: the MALDI target plate or SELDI array is inserted in the MALDI or SELDI instrument. Subsequent laser irradiation desorbs and ionises bound proteins, after which an electric field migrates the charged proteins to the TOF analyser. Herein, proteins are separated based on their mass, as the time to detector impact (TOF) is proportional to the protein mass per charge (m/z = constant * t2). Thus, small proteins (c) fly faster than large ones (a), and multiple charged ones (b) faster than single-charged ones (a). D) Representative example of SELDI-TOF mass spectra of sera from female healthy controls (HC) and breast cancer patients (BC). On the x-axis the protein m/z is displayed, and the y-axis depicts its abundance. Expression differences are visible between breast cancer and control sera at m/z 3980 and m/z 4292 (first arrow, ITIH4 fragments), and m/z 8939 (second arrow, C3adesArg). B A 1 sample fractionation 1 D 2 2 3 3 60 40 20 0 60 40 20 0 60 40 20 0 60 40 20 0 + 18 Mirror Laser + a b c Mass analyser (TOF MS) Detector Target C 4000 6000 8000 HC HC BC BC 4000 6000 8000 Training Platform LCM, IMS LCM, IMS LCM lysate Lysis C8 fractionation C18 fractionation WCX fractionation IMAC fractionation WCX fractionation Albumin depletion SAX fractionation - Pretreatment IMAC Cu IMAC, WCX, SAX H4 IMAC Cu, SAX IMAC Cu, SAX IMAC Cu, SAX IMAC Cu IMAC Ni IMAC Ni IMAC Ni IMAC Ni IMAC Ni Immunoassay Immunoassay IMAC Cu IMAC Cu H4 NP20, H4, SAX NP20, H4, SAX IMAC Cu, WCX IMAC Cu, WCX IMAC Cu NP20 Condition 5 62 65 20 78 21 46 48 76 49 45 16 15 155 103 20 19 61 29 12 20 25 23 23 5 38 46 42 25 83 - Samples (n) BC BD 3a 84 29 33 28 77 33 47 15 15 155 41 41 40 61 15 15 13 23b 23b,5 5 63 HC Protein profiling studies performed in breast cancer by MALDI- and SELDI-TOF MS. Diagnostic studies Tissue MALDI MALDI SELDI SELDI Serum MALDI MALDI MALDI MALDI MALDI SELDI SELDI SELDI SELDI SELDI SELDI SELDI SELDI SELDI SELDI SELDI SELDI Plasma SELDI SELDI NAF SELDI SELDI SELDI SELDI SELDI SELDI SELDI Matrix Table 1 n.p. MALDI n.p. n.p. n.p. n.p. n.p. n.p. n.p. n.p. n.p. n.p. SELDI n.p. SELDI n.p. SELDI SELDI SELDI n.p. n.p. SELDI n.p. n.p. n.p. n.p. n.p. n.p. SELDI n.p. Validation Platform 0 0 37 13 0 0 0 60 47 93 49 48 28 9 Samples (n) BC BD 7b 0 46 27 48 48 83 HC No Yes No No No Yes No No No No No No No No No No Yes No No Yes Yes Yes No No Yes No No No Yes No ID (24) (23) (20) (21) (32) (41) (36) (31) (42) (33) (39) (34) (38) (37) (40) (43) (44) (45) (46) (47) (48) (49) (35) (50) (51) (52) (53) (54) (55) (56) Ref. Clinical proteomics in breast cancer 19 20 SELDI SELDI SELDI Lysis (Medium) Lysis H4 IMAC Cu WCX WCX IMAC Cu IMAC Cu, WCX IMAC Cu IMAC Cu IMAC Cu, SAX SAX - IMAC Cu, WCX SAX WCX 3 2 3 6 24 27 105 60 81 63 87 21 16 3 - - - - n.p. n.p. n.p. n.p. n.p. IHC on TMA n.p. n.p. n.p. 1D GE n.p. n.p. n.p. n.p. 21b,44 16b 3 - Platform Validation HC 0 0 371 BD 547a BC Samples (n) 0 0 HC No Yes Yes Yes No Yes Yes Yes Yes Yes Yes No No No ID (67) (68) (25) (69) (35) (60) (61) (62) (63) (64) (65;66) (57) (58) (59) Ref. Abbreviations: 1D GE: 1 dimensional gel electrophoresis, BC: breast cancer, BD: benign breast disease patient, CSF: cerebrospinal fluid, DLF: ductal lavage fluid, H4: reversed phase array, HC: healthy control, ID: structural identification of candidate biomarkers, IHC: immunohistochemistry, IMAC: immobilised metal affinity capture (fractionation or array), IMS: imaging mass spectrometry, LCM: laser capture microdissection, NAF: nipple aspirate fluid, n.p.: not performed, NP20: normal phase array, SAX: strong anion exchange (fractionation or array), TMA: tissue microarray, WCX: weak cation exchange (fractionation or array). a: tissue sample obtained from tissue adjacent to tumourous tissue, b: NAF / DLF sample obtained from non-cancerous contralateral breast. Predictive studies Cell line SELDI SELDI SELDI Serum SELDI Plasma SELDI Tryptic digestion Lysis Lysis Lysis SAX fractionation - BC BD Samples Samples (n) Condition Platform Pretreatment Training Protein profiling studies performed in breast cancer by MALDI- and SELDI-TOF MS (continued) Prognostic studies Cell line SELDI Tissue SELDI SELDI Serum SELDI SELDI CSF MALDI NAF DLF Saliva Matrix Table 1 Chapter 1.1 Clinical proteomics in breast cancer Li et al. (43) observed three serum peaks to distinguish patients from controls: one (4.3 kDa) decreased and two (8.1 kDa and 8.9 kDa) increased in patients. These peaks were structurally identified as a fragment of inter-alpha-trypsin inhibitor heavy chain H4 (ITIH4, 4.3 kDa), C3a des-arginine (C3adesArg, 8.9 kDa) and a C-terminal truncated form thereof (C3adesArg∆8, 8.1 kDa) (44). Subsequent analysis of an independent sample set could only confirm the increased 8.1 kDa and 8.9 kDa C3a fragments (44). However, the 8.1 kDa C3adesArg∆8 was found to lack significance in a second (45) and third validation study (46). The latter study also reported a decreased 8.9 kDa C3adesArg expression in breast cancer (46), whereas in all previous studies, this fragment was found increased (43-45). Beyond these four studies, C3adesArg has been found associated with survival, as its expression decreased in metastatic relapse (63). In addition, the 4.3 kDa ITIH4 fragment was one of the several ITIH4 fragments found increased in breast cancer by Song et al. (48). Similar ITIH4 fragments, observed by Villanueva et al. (41) and Fung et al. (47), were found either increased in cancer (41), or devoid of discriminative power (47). Regarding the inconsistent regulation observed across multiple studies, the definitive value of the different ITIH4 fragments, C3adesArg, and C3adesArg∆8 in the diagnosis of breast cancer cannot be determined yet. In addition to the various ITIH4 fragments, several fragments of fibrinopeptide A, fibrinogen alpha, C3f, C4a, apolipoprotein A-IV, bradykinin, factor XIII, and transthyretin were found to provide accurate class discrimination (41). Generated by exoprotease activities superimposed on the ex vivo coagulation and complementdegradation pathways, these fragments are proposed to bear cancer-type specificity. It has, however, been argued that this ´peptidome signature´ merely reflects the hypercoaguable state of the blood of cancer patients (70) and not necessarily a cancerspecific signature (19). Although the peptidome signature has not been validated yet, two fragments thereof (i.e., the ITIH4 fragment discussed above, and a fibrinogen fragment) have been encountered in other studies as well (43;47;48). The fibrinogen fragment, though increased in the breast cancer serum peptidome, was found decreased in breast cancer plasma and reverted to normal values after surgical extirpation of the tumour (49). The difference between study results most likely originates from the biological matrix investigated, as plasma differs from serum by inhibition of the coagulation cascade, by which fibrinogen is generated. Also of interest are the results of the ‘Classification competition on clinical mass spectrometry proteomic diagnosis data’ (42). For this competition, sera of breast cancer patients (n = 76) and healthy controls (n = 77) were analysed by MALDI-TOF MS. Data were subsequently analysed by ten competition participants for construction of diagnostic classifiers (71-80). Surprisingly, though the various bioinformatic methods applied resulted in highly divergent classification models, reported performances (ranging from 83% to 89%) were very similar. However, as these results are based on a single dataset, validation by analysis of an independent study population most likely 21 Chapter 1.1 will reveal differences between the various bioinformatic methods and their resulting classification models. Serum and plasma protein profiling studies by MALDI-TOF or SELDI-TOF MS have yielded numerous protein peaks with a significantly different expression between breast cancer and healthy control. However, although elucidation of protein identities is essential for insights into the molecular mechanisms involved in breast cancer, thus far, only a small percentage of reported peaks has been structurally identified. Moreover, since most studies did not investigate other cancer types or patients with benign breast disease, the specificity of reported markers for breast cancer still has to be addressed. Furthermore, although of pivotal importance, only few potential markers have been validated by analysis of independent sample sets. As these studies generally yielded contradictory results, further research is needed to determine the potential of identified markers in breast cancer diagnosis. Protein profiling of nipple aspirate fluid and ductal lavage fluid Most breast cancers (70-80%) are thought to arise from the epithelial cells lining the mammary ducts (13). The breast epithelium exfoliates cells as a renewal of tissue and secretes fluid into the ductal-lobular system of the breast. While this fluid exits each breast through six to nine orifices at the nipple, it can be collected by either of two noninvasive methods; aspiration or ductal lavage. Both nipple aspirate fluid (NAF) and ductal lavage fluid (DLF) are traditionally used for cytological assessment (56;58), but their vicinity to the breast epithelium renders them valuable matrices for diagnostic protein profiling studies as well. Although several discriminating protein peaks were detected when comparing equal volumes of NAF or DLF from breast cancer patients and healthy controls by SELDITOF MS (50;55;56;58), large variations in the spectra between different samples within one diagnostic group were observed (50;55;58). Likely originating from the wide protein content range of NAF (1-90 mg/ml (56)), further studies have normalised the protein content prior to analysis. Nonetheless, despite normalisation, Sauter et al. (51;52) could not confirm the initially observed diagnostic potential of three SELDITOF MS peaks (identified as haemoglobin beta chain isoforms) in a second, larger, study population. In contrast, despite the very limited sample size, the differential expression of human neutrophil peptides 1-3 observed in NAF (n = 10) was confirmed by analyses of pooled DLF samples from cancerous (n = 9) and unrelated healthy (n = 7) breasts (55). As the breasts are a paired organ system, NAF samples from both the cancerous and non-cancerous breast of patients with unilateral breast cancer have been compared as well. Surprisingly, although different between patients, protein expression patterns were highly similar in both breasts of each patient (53;54;57). Comparison of either the cancerous or the contralateral breast to unrelated healthy controls, however, yielded several significantly different peaks (54;57). 22 Clinical proteomics in breast cancer Despite limited sample sizes and lack of validation studies, NAF protein profiling did distinguish between women with and without breast cancer. However, as identification of the cancer-bearing breast was not possible, protein profiling of NAF may have more value in breast cancer risk assessment and disease monitoring than as a diagnostic tool (57). Evidently, further research is needed to assess the value of the intraductal approach in breast cancer diagnosis. Protein profiling of saliva The use of saliva in diagnosis of systemic diseases such as breast cancer has been demonstrated by the detection of increased levels of solubilised c-erbB-2 and CA15.3 in breast cancer patients compared to healthy controls (33). Investigating saliva for diagnostic purposes has several key advantages, including its noninvasive collection, the possibility of repeated sampling, and the ease of sample handling and processing. Nonetheless, thus far, only one feasibility study has been performed in saliva. Using SELDI-TOF MS, five high molecular weight peaks were found to be overexpressed in breast cancer (n = 3) compared to control (n = 3) (59). Although these peaks were neither structurally identified nor validated in larger sample sets, this study does show the potential of using saliva for diagnostic purposes. Prognostic protein profiling studies Compared to diagnostic studies, protein profiling studies aimed at discovering novel markers to improve breast cancer prognostication are rather limited (Table 1). Investigating post-operative sera of 83 high-risk breast cancer patients by SELDI-TOF MS, Goncalves et al. (63) constructed a 40-protein signature that correctly predicted outcome in 83% of patients. Major components of this signature included haptoglobin alpha-1, complement component C3a, transferrin, and apolipoprotein A-I and C-I (Table 2). These results should be interpreted cautiously, as the number of proteins used for classification is rather high in comparison with the limited study population, indicating probable over-fitting of the data. Moreover, results have not been validated in independent sample sets. The importance of validation is emphasised by a study performed by our group (64). Using SELDI-TOF MS, we discovered a strong association between haptoglobin phenotype and recurrence free survival in sera of 63 high-risk primary breast cancer patients. However, as results were not confirmed following validation by haptoglobin phenotyping of a six-fold larger sample set (n = 371), this observation most likely resulted from a type I error (i.e., false positive) (64). In a third SELDI-TOF MS study, performed in breast cancer tissue (n = 60), high levels of ubiquitin and/or low levels of ferritin light chain were found associated with a good prognosis (62). Although results have not been confirmed by analysis of independent 23 Chapter 1.1 sample sets, ubiquitin has also been found differently expressed in breast cancer by three other studies investigating tissue specimens (23) and cell lines (25;60). Lastly, cerebrospinal fluid (CSF) has also been explored for prognostic markers (65;66). CSF is specific for the central nervous system (81), contains less total protein than serum and provides a low fluid-volume-to-organ ratio, thereby augmenting biomarker discovery (30). As collection of CSF by invasive lumbar puncture is not applicable to healthy controls, this matrix has thus far only been investigated for prognostic purposes. In search for markers indicative of leptomeningeal metastases (LM), whole CSF samples of 106 breast cancer patients were digested with trypsin (65). Following MALDI-TOF MS analysis of the resulting peptides, a classifier with 77% accuracy in determining LM status was constructed (65). The discriminative tryptic peptides were derived of several proteins (66). Three of these proteins (i.e., apolipoprotein A-I, haptoglobin and transferrin) have also been found associated to clinical outcome in serum (63). Currently, breast cancer prognosis is assessed by a.o. TNM classification, assigning breast tumours to different stages based on depth of tumour invasion and presence of metastases. However, considering the heterogeneity in outcome of patients diagnosed with equivalent TNM stage, this classification system is suboptimal in tumour characterisation. Instead, tumour staging on the molecular level could be more accurate. Indeed, microarray-based gene expression profiling studies have identified five major molecular breast cancer subtypes (i.e., luminal A and B, ERBB2-overexpressing, basallike, and normal-like), showing distinct clinical courses and responses to therapeutic agents (82;83). Hence, in search for prognostic markers, two studies have investigated the correlation between SELDI-TOF MS protein profiles of tumour tissue lysates (n = 105) (61) and breast cancer cell lines (n = 27) (60) with the previously reported breast cancer subtypes. Although discrepancies between cells grown in vivo and in vitro exist due to adaptation to cell culture conditions, breast cancer cell lines have been shown to accurately reflect the genomic, transcriptional, and biological heterogeneity found in primary tumours (84). As such, they appear to be a good surrogate matrix for tumour tissues, enabling proteome comparisons without introducing interfering factors. Indeed, in both studies, patient subgroups identified by hierarchical clustering of SELDI-TOF MS protein profiles were analogous to the molecular breast cancer subtypes (60;61). Of the several differentially expressed protein peaks detected, heat shock protein (HSP) 27 and annexin V were identified as over-expressed in the luminal A type tumour tissue lysates (61), while S100-A9 and a C-terminal truncated form of ubiquitin were found differentially expressed between the luminal-like and basal-like cell lines (60). Of note, subsequent immunohistochemical analysis of S100-A9 in tumour specimens of 547 early breast cancer patients confirmed its association with basal subtypes, as well as its value as an indicator of poor prognosis (60). The in vivo prognostic potential of HSP 27 and annexin V should be assessed by validation in clinical samples. Similar to the diagnostic studies, the prognostic studies published thus far generally investigated only a limited number of samples. Combined with the large number of 24 Clinical proteomics in breast cancer features generated by the resulting protein profiles, datasets are frequently subjected to multiple testing. Hence, candidate biomarkers are prone to be false positive, rendering validation of pivotal importance to assess their true clinical performance. Nonetheless, thus far, only two validation studies have been performed. All studies have, however, structurally identified (part of) the candidate prognostic markers. The markers identified across serum and CSF (e.g., apolipoprotein A-I, haptoglobin and transferrin) were highly abundant, non-specific, host-response generated proteins. In addition, many of the proteins identified in tissue and cell lines (e.g., annexin V, S100-A9) are in fact normal cellular proteins. However, as their precise role in breast cancer remain to be elucidated, further research is needed to determine their value for breast cancer prognostication. Predictive protein profiling studies Although accurate prediction of chemosensitivity in cancer therapy would enable individualised therapy, thus avoiding toxic side effects and eliminating the use of ineffective agents, protein profiling studies searching for markers for response prediction and treatment monitoring of breast cancer are scarce. Several SELDI-TOF MS peaks (not structurally identified) were found indicative of treatment regimen for chemosensitive and -resistant breast cancer cell lines following exposure to doxorubicin or paclitaxel (67). In addition, Dowling et al. (68) found an increase of a 7.6 kDa bovine transferrin fragment in serum-free conditioned medium of paclitaxel resistant human breast cancer cell lines, corresponding to the increased expression of the transferrin receptor they observed in whole cell lysates. Although these results were not translated to a human in vivo setting, other studies have indeed reported an association between increased serum and CSF transferrin levels and poor clinical outcome (63;65;66). Similarly, while ubiquitin and S100-A6 were found to decrease in lysates of human breast cancer cell lines following chemotherapy induced apoptosis (25), an aberrant expression of both proteins has also been reported in breast cancer tissue (23;62). Nonetheless, regarding the very limited number of samples investigated in the various studies, screening of larger cohorts and validation of the preclinical data in clinical samples is warranted before these potential markers can be used to improve therapeutic accuracy in clinical practice. In vivo studies have been performed as well (35;69). In serum, both high molecular weight kininogen and apolipoprotein A-II were found significantly decreased in expression following docetaxel-induced shock (69). Likewise, in plasma, a SELDI-TOF MS peak at m/z 2790 (not structurally identified) was found to significantly increase following (neo)adjuvant paclitaxel infusion (35). As it remains to be elucidated whether identified proteins are treatment-responsive, originate from micrometastatic carcinoma, or merely result from a general host-response to cytotoxic therapy, the definitive value of identified proteins as predictive markers can not be established yet. 25 Chapter 1.1 Discussion and conclusion Thus far, the majority of LDI protein profiling studies performed in breast cancer has searched for novel diagnostic markers, while the search for new prognostic and predictive biomarkers is limited to only few studies. The studies discussed in the current overview together have reported hundreds of mass-to-charge values, intensities of which were found to contain significant diagnostic, prognostic or predictive value. However, although indispensable for providing insight into the pathophysiological mechanisms associated with, or underlying, breast cancer, and development of absolute quantitative assays, only very few of these mass-to-charge ratios have been structurally identified yet. Moreover, the candidate markers that have been identified constitute of normal cellular proteins and high abundant blood proteins involved in coagulation and the acute phase response. Since their biology cannot be linked directly to tumour biochemistry, one of the ultimate aims of (LDI) protein profiling studies, i.e., increasing knowledge of the molecular mechanisms involved in cancer by identification of discriminative (full-length) proteins generated exclusively by cancer cells, has not been fulfilled yet. Moreover, many of the identified candidate breast cancer markers have been found to bear diagnostic potential for other cancer types as well (e.g., C3adesArg in colorectal cancer (85), apolipoprotein A-I in ovarian cancer (86)), indicating a general lack of tumour-specificity. However, as cancer cells are deranged host cells, and most cancers of epithelial origin share similar molecular features (81), it may prove difficult to find a true cancer-specific protein that is expressed exclusively by one type of malignant cells. On the other hand, as such proteins are expected to be among the least abundant proteins, they could well be below the detection limit of the current (LDI) methods. Hence, these specific tumour-secreted proteins might actually exist, but could simply have eluded detection thus far. Nonetheless, identification of specific tumour-secreted proteins is no prerequisite for improving breast cancer care, as better breast cancer diagnosis, prognosis, and prediction can also be accomplished by surrogate biomarkers of disease. A class of proteins currently recognised for their surrogate biomarker potential is the (proteolytic fragments of) high-abundant circulatory proteins. These fragments are hypothesised to be generated by cancer type-specific exoprotease activity, superimposed on the ex vivo coagulation and complement degradation proteolytic pathways. In addition, these fragments can also result from the proteases specifically expressed by malignant cells within the tumour microenvironment for tumour invasion and metastasis (87;88), as they proteolytically process the acute phase proteins that are generated by the host response to the tumour. Since these modified host response proteins generally are present at substantially higher circulatory concentrations than the enzymes that process them upon their exposure to the tumour microenvironment, they can be detected in blood by current (LDI) methods for diagnostic purposes (47). Although in breast cancer, this concept has been investigated for a.o. serum ITIH4, the various studies have 26 Clinical proteomics in breast cancer reported contradictory results, a finding not entirely unforeseen regarding the biological matrix commonly investigated (i.e., serum). Since serum is generated by coagulation, its proteome is prone to the proteases involved in this cascade, as well as to those involved in the complement cascade, activated upon clotting. Various preanalytical parameters, such as sampling device, clotting temperature, and storage time, can thus all exert a distinct influence on the serum proteome. Hence, the concept of cancer type-specific (host response) protein fragments generated by tumour-secreted proteases still awaits confirmation by validation studies that adhere to rigorous sample handling protocols. The need for such validation studies is, however, not limited to the reported host response protein fragments. Regardless of their identity, the majority of markers has been reported by single breast cancer studies, in which only limited numbers of samples were investigated, thereby compromising the generalisibility of results. Moreover, as the number of generated features (i.e., protein MS peaks) usually by far exceeds the number of samples investigated, proteomic (LDI) datasets are frequently subjected to multiple testing. As such, many candidate biomarkers are prone to be false positive. Hence, to prevent overfitting of data, as well as systematic bias by above-mentioned pre-analytical parameters, validation of biomarker candidates by other, quantitative, methods and/or in new study populations is of pivotal importance. Yet, thus far, such validation studies have been performed for only few of the candidate biomarkers detected in LDI studies (i.e., serum C3adesArg, C3adesArg∆8, ITIH4 fragments, haptoglobin alpha-1, plasma m/z 2660 fibrinogen, and tissue S100-A9). As these studies generally yielded contradictory results (except for the m/z 2660 fibrinogen fragment and S100A9), further research is needed to determine the true value of these markers in breast cancer management. The few validation studies performed thus far are all of retrospective nature. In fact, none of the identified candidate markers has been investigated for their utility as breast cancer biomarkers in a larger, prospective, clinical setting. As such, none of the candidate biomarkers discussed in this overview has been validated sufficiently to be used for clinical patient care. Yet, the move from the discovery phase to the pre-clinical and subsequent clinical validation phase is mandatory, as the sole purpose of a biomarker lies in its application. Nonetheless, overseeing the results of MALDI- and SELDI-TOF MS protein profiling studies up to now, the two platforms hold promise as high-throughput screening tools for discovery of novel breast cancer markers. Provided that these studies are performed with adequate statistical power and analytical rigour, they could eventually fulfil the great promise that protein biomarkers have for improving cancer patient outcome. 27 28 6 1627 1704 2602 Factor XIII Ferritin light chain 942 - 1865 C4a (fragments) 19809 8900 8926 8919 8941 8936 C3a des-R anaphylatoxin (C3adesArg) C3f (fragments) 8100 8116 8129 8129 C3a C-terminal fragment (C3adesArg∆8) 6647 904, 1061 t.p. 2508 9285 Bradykinin (fragments) Apolipoprotein E Apolipoprotein C-I Apolipoprotein A-IV Apolipoprotein A-II 28284 Apolipoprotein A-I t.p. 33327 t.p. α-1-antichymotrypsin Annexin V Platform (m/z) MALDI SELDI Tissue Serum Serum Serum Serum Serum Serum Serum Serum Serum Serum Serum Serum Serum CSF Serum Serum Serum Serum CSF Tissue CSF Matrix + + + - + + + - + + n.s. n.s. + - - + - + + + Relapse Cancer Cancer Cancer Cancer Cancer Cancer Cancer Relapse Cancer Cancer Cancer Cancer Cancer Metastasis Relapse Cancer Shock Relapse Metastasis Luminal subtype Metastasis Expression +/in +/- Acute phase protein, iron homeostasis Blood coagulation Complement activation Complement activation Complement activation Complement activation Inflammation mediator Lipid metabolism Acute phase protein, lipid metabolism Lipid metabolism Lipid metabolism Lipid metabolism Tumour proliferation / metastasis?, anticoagulant protein Acute phase protein, serine protease inhibitor Function Candidate biomarkers in breast cancer identified by MALDI- and SELDI-TOF MS protein profiling studies. Biomarker identity Table 2 (62) (41) (41) (41) (43) (44) (45) (46) (63) (43) (44) (45) (46) (41) (65;66) (63) (41) (69) (63) (65;66) (61) (65;66) Ref. Chapter 1.1 905 - 1537 t.p. Fibrinopeptide A (fragments) Haptoglobin (alpha 1) S100-A6 (isoforms) Prostaglandin D2 synthase Kininogen HMW 10094 t.p. 10900 7790 4300 4300 4286 4276 2271 3272 2271 4293 ITIH4 ITIH4 3375 3490 Human Neutrophil Peptide 1-3 998 -2358 15940 Haemoglobin beta chain (isoforms) Haemopexin 27152 9192 9192 Heat shock protein 27 t.p. 2379, 2659 Fibrinogen alpha (fragments) 2661 Platform (m/z) MALDI SELDI Cell line Tissue CSF Serum - - - + + Serum Serum + n.s. + + + + + + - +/-* + - Apoptosis Cancer Metastasis Shock Cancer Cancer Cancer Cancer Cancer Cancer Cancer Cancer Metastasis Cancer Luminal subtype Metastasis Relapse Relapse Cancer Cancer Cancer Expression +/in +/- Serum Serum Serum Serum Serum NAF CSF NAF Tissue CSF Serum Serum Serum Serum Plasma Matrix (65;66) (25) (23) Ca2+-binding protein, growth factor (69) (41) (48) (43) (44) (45) (46) (47) (55) (65;66) (51) (61) (65;66) (63) (64) (41) (41) (49) Ref. Catalyses prostaglandin conversion Blood coagulation, bradykinin release Acute phase reactant? Antibiotic, fungicide and antiviral Haeme binding and transport, acute phase protein Oxygen transport Stress resistance, actin organisation Acute phase protein, haemoglobin binding Blood coagulation Blood coagulation Function Candidate biomarkers in breast cancer identified by MALDI- and SELDI-TOF MS protein profiling studies (continued). Biomarker identity Table 2 Clinical proteomics in breast cancer 297 30 Ubiquitin Transthyretin (fragment) Transferrin (bovine) 8568 2451 t.p. Cell line Tissue Cell line Tissue 8507 8560 Serum CSF Medium Serum CSF Cell line Tissue Matrix 8445 7600 81763 Transferrin (human) t.p. 13300 10842 S100-A8 S100-A9 Platform (m/z) MALDI SELDI + + + - + + + - + Basal-like subtype Metastasis Apoptosis Cancer Cancer Metastasis Resistance Relapse Metastasis Basal-like subtype Cancer Expression +/in +/- (60) Ca2+-binding protein, inflammation (dimer with S100-A8) Protein modifier Thyroid hormone-binding protein, acute phase reactant Iron binding & transport, cell proliferation (62) (25) (23) (60) (41) (65;66) (68) (63) (65;66) (23) Ca2+-binding protein, inflammation (dimer with S100-A9) Acute phase reactant, iron binding & transport, cell proliferation Ref. Function Candidate biomarkers in breast cancer identified by MALDI- and SELDI-TOF MS protein profiling studies (continued). Biomarker identity Table 2 Chapter 1.1 Clinical proteomics in breast cancer References (1) Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T et al. Cancer statistics, 2008. CA Cancer J Clin 2008; 58(2):71-96. (2) Ries LA, Melbert D, Krapcho M, Mariotto A, Miller BA, Feuer EJ et al. SEER Cancer Statistics Review, 1975-2004. http://seer.cancer.gov/csr/1975_2004/ . 2008. (3) Polychemotherapy for early breast cancer: an overview of the randomised trials. Early Breast Cancer Trialists' Collaborative Group. Lancet 1998; 352(9132):930-942. (4) Mommers EC, van Diest PJ, Leonhart AM, Meijer CJ, Baak JP. Balance of cell proliferation and apoptosis in breast carcinogenesis. Breast Cancer Res Treat 1999; 58(2):163-169. (5) Reis-Filho JS, Lakhani SR. The diagnosis and management of pre-invasive breast disease: genetic alterations in pre-invasive lesions. Breast Cancer Res 2003; 5(6):313-319. (6) 't Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature 2002; 415(6871):530-536. (7) Wang Y, Klijn JG, Zhang Y, Sieuwerts AM, Look MP, Yang F et al. Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet 2005; 365(9460):671-679. (8) Miki Y, Swensen J, Shattuck-Eidens D, Futreal PA, Harshman K, Tavtigian S et al. A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1. Science 1994; 266(5182):66-71. (9) Vogelstein B, Kinzler KW. Has the breast cancer gene been found? Cell 1994; 79(1):1-3. (10) Banks RE, Dunn MJ, Hochstrasser DF, Sanchez JC, Blackstock W, Pappin DJ et al. Proteomics: new perspectives, new biomedical opportunities. Lancet 2000; 356(9243):1749-1756. (11) Aebersold R, Anderson L, Caprioli R, Druker B, Hartwell L, Smith R. Perspective: a program to improve protein biomarker discovery for cancer. J Proteome Res 2005; 4(4):1104-1109. (12) Dove A. Proteomics: translating genomics into products? Nat Biotechnol 1999; 17(3):233-236. (13) Clarke W, Zhang Z, Chan DW. The application of clinical proteomics to cancer and other diseases. Clin Chem Lab Med 2003; 41(12):1562-1570. (14) Hondermarck H, Vercoutter-Edouart AS, Revillion F, Lemoine J, Yazidi-Belkoura I, Nurcombe V et al. Proteomics of breast cancer for marker discovery and signal pathway profiling. Proteomics 2001; 1(10):1216-1232. (15) Engwegen JY, Gast MC, Schellens JH, Beijnen JH. Clinical proteomics: searching for better tumour markers with SELDI-TOF mass spectrometry. Trends Pharmacol Sci 2006; 27(5):251-259. (16) Hortin GL. The MALDI-TOF mass spectrometric view of the plasma proteome and peptidome. Clin Chem 2006; 52(7):1223-1237. (17) Hutchens TW, Yip TT. New desorption strategies for the mass spectrometric analysis of macromolecules. Rapid Commun Mass Spectrom 1993; 7:576-580. (18) Merchant M, Weinberger SR. Recent advancements in surface-enhanced laser desorption/ionizationtime of flight-mass spectrometry. Electrophoresis 2000; 21(6):1164-1177. (19) Simpson RJ, Bernhard OK, Greening DW, Moritz RL. Proteomics-driven cancer biomarker discovery: looking to the future. Curr Opin Chem Biol 2008; 12(1):72-77. (20) Nakagawa T, Huang SK, Martinez SR, Tran AN, Elashoff D, Ye X et al. Proteomic profiling of primary breast cancer predicts axillary lymph node metastasis. Cancer Res 2006; 66(24):11825-11830. (21) Traub F, Feist H, Kreipe HH, Pich A. SELDI-MS-based expression profiling of ductal invasive and lobular invasive human breast carcinomas. Pathol Res Pract 2005; 201(12):763-770. 31 Chapter 1.1 (22) Cowherd SM, Espina VA, Petricoin EF, III, Liotta LA. Proteomic analysis of human breast cancer tissue with laser-capture microdissection and reverse-phase protein microarrays. Clin Breast Cancer 2004; 5(5):385-392. (23) Sanders ME, Dias EC, Xu BJ, Mobley JA, Billheimer D, Roder H et al. Differentiating proteomic biomarkers in breast cancer by laser capture microdissection and MALDI MS. J Proteome Res 2008; 7(4):1500-1507. (24) Umar A, Dalebout JC, Timmermans AM, Foekens JA, Luider TM. Method optimisation for peptide profiling of microdissected breast carcinoma tissue by matrix-assisted laser desorption/ionisation-time of flight and matrix-assisted laser desorption/ionisation-time of flight/time of flight-mass spectrometry. Proteomics 2005; 5(10):2680-2688. (25) Leong S, Christopherson RI, Baxter RC. Profiling of apoptotic changes in human breast cancer cells using SELDI-TOF mass spectrometry. Cell Physiol Biochem 2007; 20(5):579-590. (26) Hanash SM, Pitteri SJ, Faca VM. Mining the plasma proteome for cancer biomarkers. Nature 2008; 452(7187):571-579. (27) Petricoin EF, Zoon KC, Kohn EC, Barrett JC, Liotta LA. Clinical proteomics: translating benchside promise into bedside reality. Nat Rev Drug Discov 2002; 1(9):683-695. (28) Conrads TP, Zhou M, Petricoin EF, III, Liotta L, Veenstra TD. Cancer diagnosis using proteomic patterns. Expert Rev Mol Diagn 2003; 3(4):411-420. (29) Grizzle WE, Semmes OJ, Bigbee WL, Zhu L, Malik G, Oelschlager DK et al. The need from review and understanding of SELDI/MALDI mass spectroscopy data prior to analysis. Cancer Informatics 2005; 1:8697. (30) Good DM, Thongboonkerd V, Novak J, Bascands JL, Schanstra JP, Coon JJ et al. Body fluid proteomics for biomarker discovery: lessons from the past hold the key to success in the future. J Proteome Res 2007; 6(12):4549-4555. (31) Callesen AK, Vach W, Jorgensen PE, Cold S, Tan Q, Depont CR et al. Combined experimental and statistical strategy for mass spectrometry based serum protein profiling for diagnosis of breast cancer: a case-control study. J Proteome Res 2008; 7(4):1419-1426. (32) de Noo ME, Deelder A, van der WM, Ozalp A, Mertens B, Tollenaar R. MALDI-TOF serum protein profiling for the detection of breast cancer. Onkologie 2006; 29(11):501-506. (33) Hu Y, Zhang S, Yu J, Liu J, Zheng S. SELDI-TOF-MS: the proteomics and bioinformatics approaches in the diagnosis of breast cancer. Breast 2005; 14(4):250-255. (34) Laronga C, Becker S, Watson P, Gregory B, Cazares L, Lynch H et al. SELDI-TOF serum profiling for prognostic and diagnostic classification of breast cancers. Dis Markers 2003; 19(4-5):229-238. (35) Pusztai L, Gregory BW, Baggerly KA, Peng B, Koomen J, Kuerer HM et al. Pharmacoproteomic analysis of prechemotherapy and postchemotherapy plasma samples from patients receiving neoadjuvant or adjuvant chemotherapy for breast carcinoma. Cancer 2004; 100(9):1814-1822. (36) Shin S, Cazares L, Schneider H, Mitchell S, Laronga C, Semmes OJ et al. Serum biomarkers to differentiate benign and malignant mammographic lesions. J Am Coll Surg 2007; 204(5):1065-1071. (37) Becker S, Cazares LH, Watson P, Lynch H, Semmes OJ, Drake RR et al. Surfaced-enhanced laser desorption/ionization time-of-flight (SELDI-TOF) differentiation of serum protein profiles of BRCA-1 and sporadic breast cancer. Ann Surg Oncol 2004; 11(10):907-914. (38) Gast MC, Bonfrer JM, van Dulken EJ, de Kock L, Rutgers EJ, Schellens JH et al. SELDI-TOF MS serum protein profiles in breast cancer: assessment of robustness and validity. Cancer Biomark 2006; 2(6):235248. (39) Vlahou A, Laronga C, Wilson L, Gregory B, Fournier K, McGaughey D et al. A novel approach toward development of a rapid blood test for breast cancer. Clin Breast Cancer 2003; 4(3):203-209. 32 Clinical proteomics in breast cancer (40) Belluco C, Petricoin EF, Mammano E, Facchiano F, Ross-Rucker S, Nitti D et al. Serum Proteomic Analysis Identifies a Highly Sensitive and Specific Discriminatory Pattern in Stage 1 Breast Cancer. Ann Surg Oncol 2007; 14(9):2470-2476. (41) Villanueva J, Shaffer DR, Philip J, Chaparro CA, Erdjument-Bromage H, Olshen AB et al. Differential exoprotease activities confer tumor-specific serum peptidome patterns. J Clin Invest 2006; 116(1):271284. (42) van der Werff MP, Mertens B, de Noo ME, Bladergroen MR, Dalebout HC, Tollenaar RA et al. Casecontrol breast cancer study of MALDI-TOF proteomic mass spectrometry data on serum samples. Stat Appl Genet Mol Biol 2008; 7:Article 2. (43) Li J, Zhang Z, Rosenzweig J, Wang YY, Chan DW. Proteomics and bioinformatics approaches for identification of serum biomarkers to detect breast cancer. Clin Chem 2002; 48(8):1296-1304. (44) Li J, Orlandi R, White CN, Rosenzweig J, Zhao J, Seregni E et al. Independent Validation of Candidate Breast Cancer Serum Biomarkers Identified by Mass Spectrometry. Clin Chem 2005; 51(12):2229-2235. (45) Mathelin C, Cromer A, Wendling C, Tomasetto C, Rio MC. Serum biomarkers for detection of breast cancers: a prospective study. Breast Cancer Res Treat 2005;1-8. (46) van Winden AWJ, Gast MCW, Beijnen JH, Rutgers EJ, Grobbee DE, Peeters PHM et al. Validation of previously identified serum biomarkers for breast cancer with SELDI-TOF MS: a case control study. BMC Medical Genomics 2008; Accepted for publication. (47) Fung ET, Yip TT, Lomas L, Wang Z, Yip C, Meng XY et al. Classification of cancer types by measuring variants of host response proteins using SELDI serum assays. Int J Cancer 2005; 115(5):783-789. (48) Song J, Patel M, Rosenzweig CN, Chan-Li Y, Sokoll LJ, Fung ET et al. Quantification of fragments of human serum inter-alpha-trypsin inhibitor heavy chain 4 by a surface-enhanced laser desorption/ionization-based immunoassay. Clin Chem 2006; 52(6):1045-1053. (49) Shi Q, Harris LN, Lu X, Li X, Hwang J, Gentleman R et al. Declining plasma fibrinogen alpha fragment identifies HER2-positive breast cancer patients and reverts to normal levels after surgery. J Proteome Res 2006; 5(11):2947-2955. (50) Paweletz CP, Trock B, Pennanen M, Tsangaris T, Magnant C, Liotta LA et al. Proteomic patterns of nipple aspirate fluids obtained by SELDI-TOF: potential for new biomarkers to aid in the diagnosis of breast cancer. Dis Markers 2001; 17(4):301-307. (51) Sauter ER, Zhu W, Fan XJ, Wassell RP, Chervoneva I, Du Bois GC. Proteomic analysis of nipple aspirate fluid to detect biologic markers of breast cancer. Br J Cancer 2002; 86(9):1440-1443. (52) Sauter ER, Shan S, Hewett JE, Speckman P, Du Bois GC. Proteomic analysis of nipple aspirate fluid using SELDI-TOF-MS. Int J Cancer 2005; 114(5):791-796. (53) Kuerer HM, Coombes KR, Chen JN, Xiao L, Clarke C, Fritsche H et al. Association between ductal fluid proteomic expression profiles and the presence of lymph node metastases in women with breast cancer. Surgery 2004; 136(5):1061-1069. (54) Pawlik TM, Fritsche H, Coombes KR, Xiao L, Krishnamurthy S, Hunt KK et al. Significant differences in nipple aspirate fluid protein expression between healthy women and those with breast cancer demonstrated by time-of-flight mass spectrometry. Breast Cancer Res Treat 2005; 89(2):149-157. (55) Li J, Zhao J, Yu X, Lange J, Kuerer H, Krishnamurthy S et al. Identification of biomarkers for breast cancer in nipple aspiration and ductal lavage fluid. Clin Cancer Res 2005; 11(23):8312-8320. (56) He J, Gornbein J, Shen D, Lu M, Rovai LE, Shau H et al. Detection of breast cancer biomarkers in nipple aspirate fluid by SELDI-TOF and their identification by combined liquid chromatography-tandem mass spectrometry. Int J Oncol 2007; 30(1):145-154. (57) Noble JL, Dua RS, Coulton GR, Isacke CM, Gui GP. A comparative proteinomic analysis of nipple aspiration fluid from healthy women and women with breast cancer. Eur J Cancer 2007; 43(16):23152320. 33 Chapter 1.1 (58) Mendrinos S, Nolen JD, Styblo T, Carlson G, Pohl J, Lewis M et al. Cytologic findings and protein expression profiles associated with ductal carcinoma of the breast in ductal lavage specimens using surface-enhanced laser desorption and ionization-time of flight mass spectrometry. Cancer 2005; 105(3):178-183. (59) Streckfus CF, Bigler LR, Zwick M. The use of surface-enhanced laser desorption/ionization time-offlight mass spectrometry to detect putative breast cancer markers in saliva: a feasibility study. J Oral Pathol Med 2006; 35(5):292-300. (60) Goncalves A, Charafe-Jauffret E, Bertucci F, Audebert S, Toiron Y, Esterni B et al. Protein profiling of human breast tumor cells identifies novel biomarkers associated with molecular subtypes. Mol Cell Proteomics 2008; 7(8):1420-1433. (61) Brozkova K, Budinska E, Bouchal P, Hernychova L, Knoflickova D, Valik D et al. Surface-enhanced laser desorption/ionization time-of-flight proteomic profiling of breast carcinomas identifies clinicopathologically relevant groups of patients similar to previously defined clusters from cDNA expression. Breast Cancer Res 2008; 10(3):R48. (62) Ricolleau G, Charbonnel C, Lode L, Loussouarn D, Joalland MP, Bogumil R et al. Surface-enhanced laser desorption/ionization time of flight mass spectrometry protein profiling identifies ubiquitin and ferritin light chain as prognostic biomarkers in node-negative breast cancer tumors. Proteomics 2006; 6(6):19631975. (63) Goncalves A, Esterni B, Bertucci F, Sauvan R, Chabannon C, Cubizolles M et al. Postoperative serum proteomic profiles may predict metastatic relapse in high-risk primary breast cancer patients receiving adjuvant chemotherapy. Oncogene 2006; 25(7):981-989. (64) Gast MC, van Tinteren H, Bontenbal M, van Hoesel QC, Nooij MA, Rodenhuis S et al. Haptoglobin phenotype is not a predictor of recurrence free survival in high-risk primary breast cancer patients. BMC Cancer 2009; 8:389. (65) Dekker LJ, Boogerd W, Stockhammer G, Dalebout JC, Siccama I, Zheng P et al. MALDI-TOF mass spectrometry analysis of cerebrospinal fluid tryptic peptide profiles to diagnose leptomeningeal metastases in patients with breast cancer. Mol Cell Proteomics 2005; 4(9):1341-1349. (66) Rompp A, Dekker L, Taban I, Jenster G, Boogerd W, Bonfrer H et al. Identification of leptomeningeal metastasis-related proteins in cerebrospinal fluid of patients with breast cancer by a combination of MALDI-TOF, MALDI-FTICR and nanoLC-FTICR MS. Proteomics 2007; 7(3):474-481. (67) Mian S, Ball G, Hornbuckle J, Holding F, Carmichael J, Ellis I et al. A prototype methodology combining surface-enhanced laser desorption/ionization protein chip technology and artificial neural network algorithms to predict the chemoresponsiveness of breast cancer cell lines exposed to Paclitaxel and Doxorubicin under in vitro conditions. Proteomics 2003; 3(9):1725-1737. (68) Dowling P, Maurya P, Meleady P, Glynn SA, Dowd AJ, Henry M et al. Purification and identification of a 7.6-kDa protein in media conditioned by superinvasive cancer cells. Anticancer Res 2007; 27(3A):1309-1317. (69) Heike Y, Hosokawa M, Osumi S, Fujii D, Aogi K, Takigawa N et al. Identification of serum proteins related to adverse effects induced by docetaxel infusion from protein expression profiles of serum using SELDI ProteinChip system. Anticancer Res 2005; 25(2B):1197-1203. (70) Goldsmith GH. Hemostatic changes in patients with malignancy. Int J Hematol 2001; 73(2):151-156. (71) Barrett JH, Cairns DA. Application of the random forest classification method to peaks detected from mass spectrometric proteomic profiles of cancer patients and controls. Stat Appl Genet Mol Biol 2008; 7:Article 4. (72) Heidema AG, Nagelkerke N. Developing a discrimination rule between breast cancer patients and controls using proteomics mass spectrometric data: a three-step approach. Stat Appl Genet Mol Biol 2008; 7:Article 5. (73) Fearn T. Principal component discriminant analysis. Stat Appl Genet Mol Biol 2008; 7:Article 6. 34 Clinical proteomics in breast cancer (74) Datta S. Classification of breast cancer versus normal samples from mass spectrometry profiles using linear discriminant analysis of important features selected by random forest. Stat Appl Genet Mol Biol 2008; 7:Article 7. (75) Hoefsloot HC, Smit S, Smilde AK. A classification model for the Leiden proteomics competition. Stat Appl Genet Mol Biol 2008; 7:Article 8. (76) Strimenopoulou F, Brown PJ. Empirical Bayes logistic regression. Stat Appl Genet Mol Biol 2008; 7:Article 9. (77) Goeman JJ. Autocorrelated logistic ridge regression for prediction based on proteomics spectra. Stat Appl Genet Mol Biol 2008; 7:Article 10. (78) Pham TV, van de Wiel MA, Jimenez CR. Support vector machine approach to separate control and breast cancer serum samples. Stat Appl Genet Mol Biol 2008; 7:Article 11. (79) Valkenborg D, Van Sanden S, Lin D, Kasim A, Zhu Q, Haldermans P et al. A cross-validation study to select a classification procedure for clinical diagnosis based on proteomic mass spectrometry. Stat Appl Genet Mol Biol 2008; 7:Article 12. (80) Gammerman A, Nouretdinov I, Burford B, Chervonenkis A, Vovk V, Luo Z. Clinical mass spectrometry proteomic diagnosis by conformal predictors. Stat Appl Genet Mol Biol 2008; 7(2):Article 13. (81) Hu S, Loo JA, Wong DT. Human body fluid proteome analysis. Proteomics 2006; 6(23):6326-6353. (82) Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A 2001; 98(19):10869-10874. (83) Perou CM, Sorlie T, Eisen MB, van de RM, Jeffrey SS, Rees CA et al. Molecular portraits of human breast tumours. Nature 2000; 406(6797):747-752. (84) Neve RM, Chin K, Fridlyand J, Yeh J, Baehner FL, Fevr T et al. A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell 2006; 10(6):515-527. (85) Habermann JK, Roblick UJ, Luke BT, Prieto DA, Finlay WJ, Podust VN et al. Increased serum levels of complement C3a anaphylatoxin indicate the presence of colorectal tumors. Gastroenterology 2006; 131(4):1020-1029. (86) Zhang Z, Bast RC, Jr., Yu Y, Li J, Sokoll LJ, Rai AJ et al. Three biomarkers identified from serum proteomic analysis for the detection of early stage ovarian cancer. Cancer Res 2004; 64(16):5882-5890. (87) Bank U, Kruger S, Langner J, Roessner A. Review: peptidases and peptidase inhibitors in the pathogenesis of diseases. Disturbances in the ubiquitin-mediated proteolytic system. Proteaseantiprotease imbalance in inflammatory reactions. Role of cathepsins in tumour progression. Adv Exp Med Biol 2000; 477:349-378. (88) Blasi F. Proteolysis, cell adhesion, chemotaxis, and invasiveness are regulated by the u-PA-u-PAR-PAI-1 system. Thromb Haemost 1999; 82(2):298-304. 35 Chapter Technical aspects 2 Chapter Comparing the old and new generation SELDI-TOF MS: consequence for serum protein profiling Marie-Christine W. Gast Judith Y.M.N. Engwegen Jan H.M. Schellens Jos H. Beijnen BMC Med Genomics 2008;1:4 2.1 Chapter 2.1 Abstract Although the PBS-IIc SELDI-TOF MS apparatus has been extensively used in the search for better biomarkers, issues have been raised concerning the semi-quantitative nature of the technique and its reproducibility. To overcome these limitations, a new SELDITOF MS instrument has been introduced: the PCS 4000 series. Changes in this apparatus compared to the older one are a.o. an increased dynamic range of the detector, an adjusted configuration of the detector sensitivity, a raster scan that ensures more complete desorption coverage and an improved detector attenuation mechanism. In the current study, we evaluated the performance of the old PBS-IIc and new PCS 4000 series generation SELDI-TOF MS apparatus. To this end, two different sample sets were profiled after which the same ProteinChip arrays were analysed successively by both instruments. Generated spectra were analysed by the associated software packages. The performance of both instruments was evaluated by assessment of the number of peaks detected in the two sample sets, the biomarker potential and reproducibility of generated peak clusters, and the number of peaks detected following serum fractionation. We could not confirm the claimed improved performance of the new PCS 4000 instrument, as assessed by the number of peaks detected, the biomarker potential and the reproducibility. However, the PCS 4000 instrument did prove to be of superior performance in peak detection following profiling of serum fractions. As serum fractionation facilitates detection of low abundant proteins through reduction of the dynamic range of serum proteins, it is now increasingly applied in the search for new potential biomarkers. Hence, although the new PCS 4000 instrument did not differ from the old PBS-IIc apparatus in the analysis of crude serum, its superior performance after serum fractionation does hold promise for improved biomarker detection and identification. 40 Comparison of SELDI-TOF MS apparatus Introduction The development of mass spectrometry (MS) for the analysis of complex protein mixtures has greatly enhanced the possibility of large-scale protein profiling studies. Protein profiling studies are generally performed using a top-down approach starting with a mixture of intact proteins and peptides. After sample pre-fractionation, e.g., by two-dimensional polyacrylamide gel electrophoresis (2D-PAGE), proteins are identified either by peptide mass fingerprinting using tryptic digestion and/or tandem MS. Mass spectrometry for protein profiling is particularly important for the low-molecularweight fraction of the proteome, since the use of immunological assays is limited due to a lack of antibodies for these peptides. Up until recently, real high-throughput technologies for mass spectrometric protein profiling have been lacking. Two recent applications of matrix-assisted laser desorption/ionisation time-of-flight mass spectrometry (MALDI-TOF MS) combine sample pre-fractionation with MS, facilitating the analysis of many samples at the time. A magnetic beads-based assay using beads with different chromatographic affinities is available from Bruker Daltonics (1). Alternatively, surface-enhanced laser desorption/ionisation time-of-flight mass spectrometry (SELDI-TOF MS; Bio-Rad Laboratories, Hercules, CA) can be used to profile biological matrices on arrays with different surface chemistries. A ProteinChip Interface is available for hybrid quadrupole - time of flight mass spectrometers (PCIQqTOF; e.g., QSTAR, Applied Biosystems / MDS SCIEX, Foster City, CA, USA), permitting QqTOF analysis of ProteinChip arrays. Although QqTOF platforms have both tandem MS capability and superior mass accuracy, it suffers from decreased sensitivity and a limited data acquisition range (up to 4 kDa), compared to the SELDITOF MS platform (up to 200 kDa). The SELDI-TOF MS technology has been extensively used for the assessment of tissue, serum and plasma to find diagnostic, prognostic or therapy-predictive biomarkers for diseases, especially cancer (2-8). However, issues have been raised concerning the semiquantitative nature of the technique and its reproducibility (9-11). The first generation SELDI-TOF MS instruments (PBS-II and PBS-IIc) generate spectra with a fixed maximum signal, which is set to 100. Protein abundances exceeding this maximum saturate the detector and are cut off to 100, neglecting the excess abundance and leading to underestimated peak intensities from both the saturated peak and its following peak, as the detector remains saturated for some time (12). Furthermore, settings for laser intensity and detector sensitivity are not easily optimised to generate unsaturated spectra for all the samples to be measured. To overcome these limitations a new SELDITOF MS instrument has been introduced: the PCS 4000 series. Changes in this apparatus compared to the older ones are: 1) the increased dynamic range of the detector, so that saturation is less likely to occur, 2) the special configuration for sensitivity in the high mass range for better detection of proteins > 100 kDa, 3) a socalled Synchronised Optical Laser Extraction, which scans each spot in a raster to 41 Chapter 2.1 ensure complete desorption coverage, 4) a detector attenuation mechanism, enabling signal reduction up to a specified mass and preventing saturation by matrix molecules. Furthermore, instead of using arbitrary units, peak intensities are scaled in µA, corresponding to the real electric current generated by the impact of ions onto the detector. Laser intensity settings are in nJ (13). These improvements should lead to better reproducibility of peak intensities and detection of more peaks. Yet, the ultimate gain would be that this leads to more and better biomarker candidates. We chose to assess these claims by serum protein profiling of two different cohorts of cancer patients and matched controls on both the PBS-IIc and the PCS 4000 SELDI-TOF MS. The data generated on each platform were analysed by the associated software packages. Furthermore, the PBS-IIc generated data were analysed by the software package associated with the PCS 4000 apparatus, to assess the influence of the different software packages. The numbers of detected and significantly different peaks on both instruments were compared, as was the potential of each data set to yield a reliable classification of patients and controls. Furthermore, the reproducibilities of the instruments were compared. Lastly, we also profiled serum fractions and assessed the difference in number of peaks detected between the PBS-IIc and PCS 4000 instruments. Materials and Methods Chemicals All used chemicals were obtained from Sigma, St. Louis, MO, USA, unless stated otherwise. Patient samples The performances of both apparatus were assessed with two distinct sample sets. A first set of 45 sera from colorectal cancer (CRC) patients and 43 matched controls (CON) was prospectively collected between July 2003 and October 2005 (referred to as the CRC set). The second set consisted of 45 sera from breast cancer (BC) patients and 46 matched normal women (CON), collected between January 2003 and July 2005 (referred to as the BC set). Both sets were obtained at the Netherlands Cancer Institute, in Amsterdam, The Netherlands. Sample collection was performed with individuals' informed consent after approval by the institutional review boards. Serum fractionation Serum samples from three normal women were fractionated in duplicate on QhyperD beads with a strong anion exchange moiety (Bio-Rad Labs), according to manufacturer’s protocol. Sample fractionation was performed with a Biomek 3000 Laboratory Automation Workstation (Beckman Coulter Inc.). First, sera were denatured with 9 M 42 Comparison of SELDI-TOF MS apparatus urea / 2% 3[(3-cholamidepropyl)-dimethylammonio]-propane sulfonate (CHAPS). After binding of denatured serum to the beads, the flow-through was collected and bound proteins were subsequently eluted with buffers with pH from 9 to 3. Remaining proteins were finally eluted with an organic buffer. Protein profiling For profiling of whole serum each sample was analysed according to previously developed protocols (4). CRC samples and their matched controls were first denatured with 9 M urea / 2% CHAPS / 1% dithiotreitol. Then, each sample was applied in triplicate on CM10 arrays (weak cation exchange chromatography) with 20 mM sodium phosphate pH 5 / 0.1% TritonX-100 as a binding buffer and 20 mM sodium phosphate pH 5 as a wash buffer. BC samples and their matched controls were denatured in 9 M urea / 2% CHAPS, after which each sample was applied in duplicate on IMAC30 arrays (immobilised metal affinity capture chromatography). Prior to sample application, IMAC30 arrays were charged twice with 50 µL 100 mM nickel sulphate (Braun, Emmenbrücke, Germany), followed by three rinses with deionised water. Phosphate Buffered Saline (PBS; 0.01 M) pH 7.4 / 0.5 M sodium chloride / 0.1% TritonX-100 was applied as a binding buffer and PBS pH 7.4 / 0.5 M sodium chloride as a wash buffer. For both sample sets, a 50% sinapinic acid (SPA; Bio-Rad Labs) solution in 50% acetonitrile (ACN) / 0.5% trifluoroacetic acid (TFA) was used as energy absorbing matrix. Profiling of fractionated serum was performed on both CM10 chips and IMAC30 arrays. Binding and wash buffers were 100 mM sodium acetate pH 4 and 50 mM HEPES buffer for CM10. For IMAC30, 100 mM copper sulphate was used as charging solution, 100 mM sodium acetate as neutralizing buffer and 100 mM sodium phosphate pH 7 / 0.5 M sodium chloride as binding and wash buffer (all: Bio-Rad Labs). A solution of 50% SPA in 50% ACN / 0.5% TFA was used as matrix. During all profiling experiments arrays were assembled in 96-well format bioprocessors (Bio-Rad Labs), which were placed on a platform shaker at 350 rpm. Arrays were equilibrated twice with 200 µL of binding buffer, incubated with denatured sample or QHyperD serum fraction for 30 min and, after binding, washed twice with binding buffer, followed by two washes with wash buffer. Lastly, arrays were rinsed with deionised water. After air-drying, two times 1 µL of matrix was applied to the array spots. SELDI-TOF MS analysis of all data sets was performed with both the PBS-IIc and the PCS 4000 ProteinChip Reader (Bio-Rad Labs). Data acquisition and processing were optimised for each sample set separately. Each spot was read twice, once with the PCS 4000 and once with the PBS-IIc instrument. Measurement settings for each apparatus and sample set are summarised in Table 1. M/z values were calibrated externally with All-in-One peptide standard (Bio-Rad Labs). 43 Chapter 2.1 bioinformatics Statistics and bio informatics To account for possible differences in data processing by the different software packages, data from the PBS-IIc were analysed with the ProteinChip Software, version 3.1 (Bio-Rad Labs) as well as with Ciphergen Express™ version 3.0.6. (Bio-Rad Labs). PCS 4000 data were only processed with the latter package. The PBS-IIc-generated spectra analysed by the ProteinChip Software and Ciphergen Express™ respectively will further be referred to as “data set 1a” and “data set 1b”. The PCS 4000-generated spectra, analysed by Ciphergen Express™ will be referred to as “data set 2”. Table 1 Settings for protein profiling and data processing. CRC sample set PBS-IIc PCS 4000 BC sample set PBS-IIc PCS 4000 CRC: n = 45 CON: n = 43 CM10 20 mM NaAc pH 5 3 BC: n = 45 CON: n = 46 IMAC30 Ni PBS pH 7.4 + 0.5 M NaCl 2 SELDI analysis parameters Samples Array type Binding conditions Replicates SELDI acquisition parameters m/z range Laser intensity Detector sensitivity Deflector/detector attenuation Laser shots kept 0-200 kDa 155 6 2000 Da 65 Not-assessable spectra Cluster settings: First pass Second pass Cluster mass window Present in 0-200 kDa 3500 nJ n.a. 2000 Da 530 0-200 kDa 155 5 1000 Da 105 CRC: 2 / 135 CON: 4 / 129 S/N 5 S/N 2 0.3% 45% Valley depth 5 Valley depth 2 0.3% 45% 0-200 kDa 3500 nJ n.a 1000 Da 530 BC: 4 / 90 CON: 0 / 92 S/N 5 S/N 2 0.3% 30% Valley depth 5 Valley depth 2 0.3% 30% Abbreviations: BC: breast cancer, CM10: weak cation exchange ProteinChip array, CON: control, CRC: colorectal cancer, IMAC30: Immobilised metal affinity capture ProteinChip array, n.a.: not applicable, PBS: phospate buffered saline, S/N: Signal to noise ratio. Spectra from the CRC and BC sets were analysed separately. Acquired spectra from each set were compiled and analysed as a whole. Both the ProteinChip and Ciphergen Express™ software spectra were baseline subtracted with the following settings: smooth before fitting baseline: 25 points, fitting width: 10 times expected peak width. Filtering was “on” using an average width of 0.2 times the expected peak width. The noise was calculated from 2000 or 1000 to 200,000 Da for the CRC and BC set respectively. Spectra were normalised to the total ion current in the same m/z range. For peak clustering with the ProteinChip Software, the Biomarker Wizard (BMW; Bio-Rad Labs) 44 Comparison of SELDI-TOF MS apparatus application was used. For clustering with the Ciphergen Express™ software (Bio-Rad Labs), identical clustering conditions were defined (see Table 1). In each set, peaks were auto-detected starting from 2000 Da. For the CRC and BC set, peak intensities from the triplicate (CRC) and duplicate (BC) analyses were averaged and mean peak intensities between groups compared by the non-parametric Mann-Whitney U (MWU) test (p < 0.01 considered statistically significant). For the CRC set, the median CV in each sample set was calculated from the CV’s of the triplicate analyses for all clustered peaks in each data set as well as for all common peaks present in each of the three data sets. For the BC set, Spearman’s rank correlation coefficient was calculated on peak intensities in each duplicate analysis for all three data sets. The majority of peaks (> 50%) detected spectrum wide in the three data sets are of relatively low average intensity (< 5), increasing the chance of finding potential biomarkers in the low intensity range. Hence, the reproducibility of peaks in the low intensity range is of special interest. However, correlation analyses are influenced by outliers (i.e., the few high intensity peaks detected), even when using non-parametric statistics. We therefore chose to assess the reproducibility in subsets of peaks, starting with inclusion of the 10%, 20%, 30%, etc. of peaks with lowest intensity, and ending with inclusion of all peaks detected. Spearman’s rank correlation coefficient and corresponding p-values were subsequently plotted per subset of peaks. Classification performance of the data sets obtained with both apparatus was assessed by building classification trees with the Biomarker Patterns Software (BPS; Bio-Rad Labs). Trees were generated with the gini method and the minimal cost tree was chosen in both the CRC and BC sample set. A ten-fold cross validation was used to estimate the sensitivity and specificity for each tree. For the CM10 and IMAC30 serum fractionation sets, baseline correction and noise calculation was performed as described for the CRC and BC set. For each duplicate fraction, peaks were auto-detected by the ProteinChip software or Ciphergen Express™ by the settings described in Table 1. The number of peaks in each fraction was assessed, as well as the number of unique peaks across all fractions. Results Protein profiling CRC set Six spectra did not contain a protein profile and were thus not assessable (Table 1). For data set 1a normalisation factors as estimated by the apparatus-associated software of the assessable spectra were 0.67 to 3.39 (log -0.18 to 0.53) and for data set 1b 0.62 to 2.4 (log -0.21 to 0.38). For data set 2 values ranged from 0.33 to 4.8 (log -0.49 to 0.68). Since the spectra with aberrant normalisation factors (>2 SD from mean of log normalisation factor) were mostly not from the same samples for the two apparatus, none were 45 Chapter 2.1 excluded, to ensure an equal comparison of both machines. This concerned 13 and 11 spectra from data set 1a and 1b, and 14 from data set 2. Comparing CRC vs. CON, 32 clusters were generated for data set 1a (Table 2). In contrast, despite similar settings for processing and the same spectra, only 27 clusters were generated for data set 1b. With the PCS 4000 (data set 2) 48 clusters could be detected. Although the number of detected peaks was highest for data set 2, data set 1a yielded a similar number of significantly different peaks. Detailed peak cluster information can be found in Table 3. Overall, the significantly different peaks in all data sets were largely similar. Table 2 Peak clustering results for the CRC and BC sample set. Number of peaks peaks detected in: Data set 1a * Also detected in data set 1b Also detected in data set 2 Data set 1b † Also detected in data set 2 Data set 2 ‡ CRC set all 32 31 31 27 26 48 p < 0.01 19 13 15 14 11 20 BC set all 81 30 43 31 29 59 p < 0.01 47 22 28 22 21 45 Abbreviations: BC: breast cancer, CRC: colorectal cancer. * Data set 1a: PBS-IIc generated data, analysed by the ProteinChip software, † data set 1b: PBS-IIc generated data, analysed by Ciphergen Express, ‡ data set 2: PCS 4000 generated data, analysed by Ciphergen Express. Classification trees were built with all clustered peaks in each data set and with the subset of the 25 clusters that were detected in all three data sets (Table 4). The best tree was generated with data set 2, with m/z 4446 as single classifier and sensitivity and specificity ≥ 80%. Since this peak was not detected in data set 1a and 1b, other peaks were used as classifiers in these sets, respectively m/z 15930 and 32308. The classification trees constructed on the subset of 25 common clusters in data sets 1a and 1b applied the same cluster (m/z 32308). The best classifier of data set 2 made use of apparently the same cluster (m/z 32394), and had a better performance as single classifier in this set than in set 1a, but a similar performance as in set 1b. Table 3 Data set 1a 2746* 3979* 4160 4179* 4290* 4481 4605 46 Peak cluster information for CRC and BC sample set. CRC sample set Data set 1b 2745* 4162 4181* 4474 Data set 2 2746* 3163 3406* 3978* 4159 4182* 4287* 4303 4446* 4480 4607 Data set 1a 2027 2146* 2154* 2235* 2277* 2647 2675* 2731* 2747 2760 2775 BC sample set Data set 1b Data set 2 2028 2146* 2760* Comparison of SELDI-TOF MS apparatus Table 3 Data set 1a 5723* 5913* 6443 6459* 6641 6655* Peak cluster information for CRC and BC sample set (continued). CRC sample set Data set 1b 5724* 6443 6460* 6640 6654* Data set 2 4961 5719* 5915* 6442 6458* 6643 6659* 6687 6846 7778 7982* 8079* 6860* 7779 7982* 8968* 9186 8962* 9187 9307* 9307* 13779* 14077* 15121 15930* 16105* 13778* 14077* 15121 15919* 16105* 23426* 28098* 32308* 23426 28093 32308* 51034 56408 67003 79100 51006 56337 67003 79040 6865* 7778 7990* 8074* 8159 8889 8962* 9210 9315 9360 9409 9593 10072* 12889 13796 14053 15168 15982* 16139* 16334* 18617* 23486 28216 32394* 39727 51116 56685 67239 80075 Data set 1a 2794 2888* 2960* 2968* 3091 3107 3151* 3168* 3282* 3296* 3431 3451 3689* 3781* 3824 3891* 3898 3916* 3965* 3980* 3995* 4078 4137 4155 4204* 4218* 4292* 4308* 4334* 4449* 4464* 4484* 4497* 4513* 4653 4669 4691 4798 5076 5090 5274* 5348* 5363* 5554* 5815 5916* 5932* 6100* 6122* 6142* 6667 6848 6965* 6990* 7482 7778 BC sample set Data set 1b Data set 2 2961* 3165* 3281* 3164* 3281* 3683* 3891* 3963* 3980* 3997* 3962* 3979* 3994* 4138 4218* 4292* 4308* 4218* 4289* 4304* 4447* 4463* 4484* 4444* 4458* 4482* 4652 4650 5078 5348* 5364* 5917* 5932* 6097* 6122* 5348* 5360* 5549* 5810 5915* 5929* 6676 6972* 6966* 7778 7775 47 Chapter 2.1 Table 3 Data set 1a Peak cluster information for CRC and BC sample set (continued). CRC CRC sample set Data set 1b Data set 2 Data set 1a 7939* 7985 8155 8948* 9161* 9302 BC sample set Data set 1b 8155 8955* 9303 13925* 13925* 33475* 33583* 43108 43015 60776 66702* 79724 60804 66711* 79393 109494 133447 133435 149723 Data set 2 7982 8150 8946* 9151* 9299 9526 11096* 11747* 13919* 14124* 22284* 28221 30502* 33490* 40059* 43098* 50704* 60889* 67142* 80323* 89750* 91037 93689* 104178* 110344* 136611* 149579* 177011 Abbreviations: BC: breast cancer, CRC: colorectal cancer. *MWU test; p < 0.01. Protein profiling BC set Following array reading with both the PCS 4000 and PBS-IIc apparatus, two spectra did not contain a protein profile. Along with their duplicate reading, these spectra were excluded from further analyses (Table 1). The normalisation factors for data set 1a were 0.52 to 2.15 (log -0.29 to 0.33). Using Ciphergen Express™ software, normalisation factors of all spectra ranged from 0.51 to 2.25 (log -0.29 to 0.35) for the PBS-IIc generated spectra and from 0.44 to 2.69 (log -0.36 to 0.43) for the PCS 4000 generated spectra. In total, 9 and 8 spectra from data set 1a and 1b respectively and 10 spectra from data set 2 had an aberrant normalisation factor (>2 SD from mean of log normalisation factor). As the majority of these spectra were from different samples for the 3 data sets, none were excluded, to ensure equal comparison of both apparatus. In data set 1a and 1b respectively, a total of 81 and 31 clusters were detected. The ProteinChip software detected 51 clusters that were not detected by Ciphergen Express™ in the same data set. Except for one cluster (> 100 kDa), these unique clusters were all < 10 kDa in mass and < 4 in intensity. In the data set 2, a total of 59 peak clusters was detected. Fifteen of these clusters (all > 9 kDa) were not detected in either 48 Comparison of SELDI-TOF MS apparatus data set 1a or 1b. Tables 2 and 3, respectively, provide an overview of peak clustering results and detailed peak cluster information. Table 4 Characteristics of the classification trees constructed on the CRC sample set. Tree characteristics Data set Clusters 1a All Common 1b All Common 2 All Common (#) (32) (25) (27) (25) (48) (25) Node 1 m/z 15930 m/z 15930 m/z 32308 m/z 32308 m/z 4446 m/z 32394 ≤ 35.576 ≤ 35.576 ≤ 0.676 ≤ 0.676 ≤ 1.136 ≤ 0.149 Node 2 m/z 51034 m/z 51034 - ≤ 1.372 ≤ 1.372 - Tree performance* Sens (%) Spec (%) 68.8 62.8 68.8 62.8 75.6 73.8 75.6 73.8 82.2 90.5 73.3 81.0 Abbreviations: sens: sensitivity, spec: specificity. *Tree performance as determined by 10-fold cross validation. Classification trees were generated on all peaks detected in data set 1a, 1b or 2, and on the subset of peaks detected across all three data sets (Table 5). All optimum decision trees constructed on data set 1a and 1b, using either all peaks detected or only the common peaks, applied m/z 3964 as single classifier, with data set 1b yielding the best performance of ~ 80%. The trees constructed on data set 2 made use of different clusters, either considering all peaks detected (m/z 9151 and m/z 5360) or the common peaks detected (m/z 3979 and m/z 4218). However, the tree constructed on data set 1b generally had the best performance. Table 5 Characteristics of the classification trees constructed on the BC sample set. Tree characteristics Data set Clusters 1a All Common 1b All Common 2 All Common (#) (81) (28) (31) (28) (59) (28) Node 1 m/z 3964 m/z 3964 m/z 3964 m/z 3964 m/z 9151 m/z 3979 ≤ 4.010 ≤ 4.010 ≤ 3.855 ≤ 3.855 ≤ 1.614 ≤ 32.163 Node 2 m/z 5360 m/z 4218 ≤ 17.86 ≤ 4.648 Tree performance* Sens (%) Spec (%) 74.4 73.9 74.4 73.9 83.7 78.3 83.7 78.3 72.1 78.3 62.8 71.7 Abbreviations: sens: sensitivity, spec: specificity. *Tree performance as determined by 10-fold cross validation. Reproducibility CRC set For each data set on each apparatus the inter-chip reproducibility was assessed by calculating the median CV across all samples from replicate peak intensities of all clustered peaks and of the subset of 25 common peaks in the three data sets. The median CV of all peaks and all common peaks was lowest for data set 2 and highest for data set 1a (Table 6). Considering all peaks, the CV was significantly different for the data sets (Kruskall Wallis test; p = 0.012), but not when considering only the common peaks to each data set (Kruskall Wallis test; p = 0.3). 49 Chapter 2.1 Table 6 Reproducibility of the CRC data sets. Peak clusters All peaks Common peaks All peaks (CON) All peaks (CRC) Data set 1a 28.30% 25.48% 28.52% 27.68% Median CV of Data set 1b 22.61% 23.06% 21.76% 23.38% Data set 2 20.62% 21.71% 18.50% 23.33% Reproducibility BC set For the BC sample set, Spearman’s rank correlation coefficient was calculated on peak intensities in each duplicate analysis for successive subsets of peaks including the 10%, 20%, 30% to 100% of peaks with lowest intensity, for all three data sets. As depicted in Figure 1, a correlation coefficient > 0.8 was only reached after inclusion of 80% of lowest peaks in data set 1a, while in the other two data sets, this coefficient was already reached at inclusion of < 20% of lowest peaks. Similar results were obtained when considering the significance of correlation (Figure 1). However, when considering only the common peaks detected across all three data sets, results obtained were highly similar for the three data sets (Figure 2). Serum fractionation The numbers of clusters detected on CM10 and IMAC arrays for each sample in each acquired fraction are summarised in Table 7. Some of the clusters are occurring in several fractions. Ignoring these overlapping clusters, on average twice as many peaks were detected in the PCS 4000 generated spectra compared to the PBS-IIc generated spectra (analysed either by the ProteinChip software or Ciphergen Express™). This is also illustrated in Figure 3. Discussion Although the PBS-IIc SELDI-TOF MS apparatus has been extensively used in the search for better biomarkers, issues have been raised concerning the semi-quantitative nature of the technique and its reproducibility. To overcome these limitations, a new SELDITOF MS instrument has been introduced: the PCS 4000 series. In the current study, we compared the performances of the old PBS-IIc and new PCS 4000 series generation SELDI-TOF MS apparatus, by analysis of two sample sets. Peak detection For the CRC sample set, most peaks were detected with the new PCS 4000 series using the Ciphergen Express™ software, indicating a better sensitivity and less detector saturation of this apparatus. The latter allows for the application of increased laser intensities, after which proteins will desorb more comprehensively, resulting in 50 Comparison of SELDI-TOF MS apparatus detection of more peaks. However, for the BC sample set, most peaks were detected with the PBS-IIc instrument using the ProteinChip software, indicating the opposite. Interestingly, in both sample sets, fewer peaks were detected by Ciphergen Express™ than by the ProteinChip software in the spectra generated with the PBS-IIc, despite the fact that both software packages use the same algorithm with similar settings to generate peak clusters. Apparently, the spectrum processing algorithms underlying the visible settings are different for both software packages. Figure 1 Plots of Spearman’s rank correlation coefficient and p-values for all peaks detected in the BC data sets. Depicted are the mean (red) and median (black) values of all peaks detected in the three data sets of the BC sample set. PBS: data set 1a (PBS-IIc generated data, analysed by ProteinChip software), PCS: data set 2 (PCS 4000 generated data, analysed by Ciphergen Express™), PBS/PCS: data set 1b (PBS-IIc generated data, analysed by Ciphergen Express™). In the BC set, all peaks detected in the PBS-IIc generated spectra by the ProteinChip software, but missed by Ciphergen Express™ were < 4 in intensity. As peaks are detected by means of their signal-to-noise ratio, detection of these low intensity peaks becomes critical when either the noise increases or the signal decreases due to over- 51 Chapter 2.1 estimation of the baseline. Conceivably, the algorithm for noise and/or baseline estimation between both software packages has been changed. Due to the detector attenuation of the PCS 4000 instrument, matrix blanking has improved compared to the PBS-IIc. Hence, less chemical noise is expected when measuring with the PCS 4000 instrument, to which the algorithm applied in noise calculation might have been adapted. As such, for spectra generated with the PBS-IIc (in which relatively more chemical noise is present), the Ciphergen Express™ software will estimate the noise too high or the signal too low, the latter being the consequence of the baseline being estimated too high. Either way results in fewer detected peaks. Table 7 Data set* et* Peak clustering results for the serum fractions profiled on CM10 and IMAC30 arrays. Serum sample 1 CM10 1a 1b 2 FT+pH9 43 24 60 pH7 16 6 45 pH5 42 19 51 pH4 22 22 46 pH3 20 20 46 Organic 22 17 58 Total 165 108 306 Unique 103 78 167 IMAC30 1a 1b 23 22 9 9 15 15 24 22 16 14 31 30 118 112 85 82 Serum sample 2 CM10 2 1a 1b 2 52 59 23 57 30 28 9 46 32 37 16 49 53 19 17 50 42 10 8 48 40 17 12 61 249 170 85 311 158 106 61 162 IMAC30 1a 1b 28 22 9 7 24 16 34 24 16 13 22 15 133 97 82 72 Serum sample 3 CM10 2 1a 1b 2 43 29 19 47 24 9 5 51 29 21 15 56 54 23 19 54 36 19 16 58 36 24 20 53 222 125 94 319 135 82 71 163 IMAC30 1a 1b 24 15 7 7 17 11 23 21 18 13 19 20 108 87 67 53 2 47 26 28 46 38 42 227 128 * Data set 1a: PBS-IIc generated data, analysed by the ProteinChip software, data set 1b: PBS-IIc generated data, analysed by Ciphergen Express, data set 2: PCS 4000 generated data, analysed by Ciphergen Express. The difference between peaks detected by either software package in the PBS-IIc generated spectra was more pronounced in the BC set than in the CRC set. These two data sets differed in their deflector / detector attenuation settings (CRC: 2000 Da, BC: 1000 Da), but in both sets, the noise was calculated between 2 and 200 kDa. However, as matrix peaks are generally observed up to 2000 Da, their contribution to the noise will most likely increase with decreasing deflector settings. Hence, the difference in deflector settings could have caused higher noise estimation in the BC set compared to the CRC set. Combined with the probable noise overestimation by Ciphergen Express™ in PBS-IIc generated spectra, and the fact that relative to the CRC data sets, the BC data sets contained more low intensity peaks (30 and 70%, respectively), which were mainly present in the <10 kDa range, this might explain the more pronounced difference in number of peaks detected in the PBS-IIc generated BC data set by both software packages. The difference in deflector / detector attenuation settings might also explain why, contrary to the CRC set, in the BC set more peaks were detected by the ProteinChip software in the PBS-IIc spectra than by Ciphergen Express™ in the PCS 4000 spectra. Compared to the ProteinChip software, the noise calculation algorithm in Ciphergen Express™ apparently is more sensitive to the noise in the low molecular weight range. Due to the difference in detector attenuation settings, this low molecular weight range 52 Comparison of SELDI-TOF MS apparatus will contain a higher signal in the BC spectra than in the CRC spectra. Consequently, the noise is estimated higher and less peaks are detected. This hypothesis is supported by the observation that all peaks detected in the PBS-IIc spectra, but not in the PCS 4000 spectra were < 3 in intensity. One of the alleged improvements of the PCS 4000 compared to its PBS-IIc predecessor is its special configuration for sensitivity in the high mass range that allows detection of proteins above 100 kDa. Indeed, in the BC set, four > 100 kDa peaks were detected exclusively in the PCS 4000 generated spectra, compared to two peaks in the PBS-IIc generated spectra. Moreover, all peaks that were detected exclusively in the PCS 4000 spectra by Ciphergen Express™ were above 10 kDa. However, in none of the CRC data sets any proteins > 100 kDa were detected, indicating no better sensitivity for proteins in the higher mass range for the PCS 4000 series. Most peaks detected only in data set 2 were in the 2-10 kDa range. The differences in detection of high molecular weight peaks could, however, be caused by the different array types used for the analyses of both sample sets. Classification As the ultimate gain of the improved performance of the PCS 4000 instrument would be detection of more and better biomarker candidates, we also assessed the classification potential of the data sets generated by both machines. For the CRC set, the improved performance of the new instrument was indeed reflected in the classifiers constructed, as the best classification was obtained with the data set generated by the PCS 4000 instrument, using the total number of peaks detected. When using the subset of peaks detected in all three data sets, the performance of the classifier build on data set 1b and 2 was similar. For the BC data set, results were less unambiguous. While for data set 1a and 1b only one classifier was applied in the different optimum decision trees constructed, best performance was achieved in data set 1b. Apparently, the different spectrum processing algorithms underlying both software packages also contribute to the alleged improved performance of the PCS 4000 instrument. However, application of both the PCS 4000 and Ciphergen Express™ yielded no better classifiers. Hence, for the BC set, the superior performance of the PCS 4000 instrument in providing better biomarker candidates could not be confirmed. It can, however, not be precluded that our data sets do not contain any real biomarkers. Reproducibility For the CRC set, the reproducibility of peak intensities was largely similar across data sets, although a non-significant trend could be seen to a lower CV for data set 2 compared to 1a and 1b. Thus, the spot scanning in a raster and the less detector saturation with the PCS 4000 series does not seem to result in a significant better reproducibility. The fact that significant differences in CV were seen when all peaks were considered indicates that the surplus of peaks detected in data set 2 consists of 53 Chapter 2.1 more robust peaks than the ones also detected in the other data sets, causing the median CV to drop. Reproducibility of the PCS 4000 instrument as measured by the CV has been stated to be < 20% using an external standard (13). It is not known to us in which m/z range this reproducibility was obtained and whether this was with manual or robotic sample handling. However, our observed median CV is well in concordance with this value, especially taking the manual sample handling into account. Figure 2 Plots of Spearman’s rank correlation coefficient and p-values for common peaks detected in the BC data sets. Depicted are the mean (red) and median (black) values of common peaks detected across all three data sets of the BC sample set. PBS: data set 1a (PBS-IIc generated data, analysed by ProteinChip software), PCS: data set 2 (PCS 4000 generated data, analysed by Ciphergen Express™), PBS/PCS: data set 1b (PBS-IIc generated data, analysed by Ciphergen Express™). Reproducibility in the BC data sets was assessed by calculation of Spearman’s rank correlation coefficient on duplicate intensities of the 10 to 100% peaks with lowest intensity. When all peaks detected were included in this calculation, usage of the PCS 4000 and Ciphergen Express™ software package led to a better performance, as statistically significantly (p < 0.05) good correlations (R > 0.8) were already achieved 54 Comparison of SELDI-TOF MS apparatus upon inclusion of only 20% of lowest peaks, compared to the 80% of lowest peaks necessary to achieve comparable results in the PBS-IIc generated data set. However, when correcting for the excess of low intensity peaks detected in data set 1a relative to data set 2 by considering only the peaks detected across all three data sets, results obtained were highly similar for the three data sets. Thus, the improved features of the PCS 4000 instrument relative to the PBS-IIc apparatus do not lead to an improved reproducibility, as already observed in the CRC data sets. Serum fractionation Analysis of the PBS-IIc generated spectra by Ciphergen Express™ generally yielded the lowest number of peaks detected. Hence, the performance of the PCS 4000 in serum fractionation is indeed superior compared to the PBS-IIc instrument, reflecting the improved spot coverage and increased detector sensitivity. These observations are highly similar to the results obtained following peak detection in the three CRC data sets. Figure 3 Spectra of serum fractions analysed on CM10 arrays and measured on the PBS-IIc and PCS 4000 instrument. A: flow through/pH 9 fractions, B: pH 7 fractions. Although deflector / detector attenuation settings were different for the fractionation spectra on IMAC and CM10 chips, peak clustering results were highly similar for the two array types used, contrary to the results obtained in the CRC and BC sample sets. This could be due to the fact that these spectra have a higher noise level than spectra from crude serum (data not shown), limiting the influence of the different noise estimation between both software packages. Moreover, the number of peaks < 10 kDa is similar in the fractionation spectra from the IMAC and CM10 chips, contrary to the spectra from the CRC and BC set, which could also cause less influence of the noise estimation on peak detection. 55 Chapter 2.1 Conclusion In conclusion, regarding the number of peaks detected, the biomarker potential and the reproducibility of the two sample sets investigated by both the old (PBS-IIc) and new (PCS 4000) generation SELDI-TOF MS apparatus, we could not confirm the alleged improved performance of the PCS 4000 instrument over the PBS-IIc apparatus. However, the PCS 4000 instrument did prove to be of superior performance in peak detection following profiling of serum fractions. Until now, the majority of studies in which SELDI-TOF MS was applied in crude serum protein profiling for biomarker discovery generally reported high abundant, non-disease-specific proteins as potential biomarkers. However, the large dynamic range of crude serum hampers detection of the allegedly high-informative low abundant serum proteins. As serum fractionation facilitates detection of low abundant proteins through reduction of this dynamic range, it is increasingly applied in the search for new potential biomarkers. Hence, although the new PCS 4000 instrument did not differ from the old PBS-IIc apparatus in the analysis of crude serum, its superior performance of fractionated serum samples does hold promise for improved biomarker detection and identification. Acknowledgement The authors gratefully acknowledge Ciphergen Biosystems for use of the PCS 4000 SELDI-TOF MS, and the Department of Clinical Chemistry, University Hospital Maastricht for use of the Ciphergen Express™ software package. Wouter Meuleman is greatly acknowledged for help with data analysis. References (1) (2) (3) (4) (5) (6) 56 http://www.bruker.nl/daltonics/home_daltonics.html. 2007. Adam BL, Qu Y, Davis JW, Ward MD, Clements MA, Cazares LH et al. Serum protein fingerprinting coupled with a pattern-matching algorithm distinguishes prostate cancer from benign prostate hyperplasia and healthy men. Cancer Res 2002; 62(13):3609-3614. Caspersen MB, Sorensen NM, Schrohl AS, Iversen P, Nielsen HJ, Brunner N. Investigation of tissue inhibitor of metalloproteinases 1 in plasma from colorectal cancer patients and blood donors by surfaceenhanced laser desorption/ionization time-of-flight mass spectrometry. Int J Biol Markers 2007; 22(2):89-94. Engwegen JY, Helgason HH, Cats A, Harris N, Bonfrer JM, Schellens JH et al. Identification of serum proteins discriminating colorectal cancer patients and healthy controls using surface-enhanced laser desorption ionisation-time of flight mass spectrometry. World J Gastroenterol 2006; 12(10):1536-1544. Li J, Zhao J, Yu X, Lange J, Kuerer H, Krishnamurthy S et al. Identification of biomarkers for breast cancer in nipple aspiration and ductal lavage fluid. Clin Cancer Res 2005; 11(23):8312-8320. Schultz IJ, De Kok JB, Witjes JA, Babjuk M, Willems JL, Wester K et al. Simultaneous proteomic and genomic analysis of primary Ta urothelial cell carcinomas for the prediction of tumor recurrence. Anticancer Res 2007; 27(2):1051-1058. Comparison of SELDI-TOF MS apparatus (7) (8) (9) (10) (11) (12) (13) Zhang Z, Bast RC, Jr., Yu Y, Li J, Sokoll LJ, Rai AJ et al. Three biomarkers identified from serum proteomic analysis for the detection of early stage ovarian cancer. Cancer Res 2004; 64(16):5882-5890. Freed GL, Cazares LH, Fichandler CE, Fuller TW, Sawyer CA, Stack BC, Jr. et al. Differential Capture of Serum Proteins for Expression Profiling and Biomarker Discovery in Pre- and Posttreatment Head and Neck Cancer Samples. Laryngoscope 2008; 118(1):61-68. Diamandis EP. Point: Proteomic patterns in biological fluids: do they represent the future of cancer diagnostics? Clin Chem 2003; 49(8):1272-1275. Baggerly KA, Morris JS, Coombes KR. Reproducibility of SELDI-TOF protein patterns in serum: comparing datasets from different experiments. Bioinformatics 2004; 20(5):777-785. Ransohoff DF. Lessons from controversy: ovarian cancer screening and serum proteomics. J Natl Cancer Inst 2005; 97(4):315-319. Malyarenko DI, Cooke WE, Adam BL, Malik G, Chen H, Tracy ER et al. Enhancement of sensitivity and resolution of surface-enhanced laser desorption/ionization time-of-flight mass spectrometric records for serum peptides using time-series analysis techniques. Clin Chem 2005; 51(1):65-74. ProteinChip System, Series 4000, Product Note (www.bio-rad.com). 2005. 57 Chapter Serum protein profiling using SELDI-TOF MS: influence of sample storage duration Marie-Christine W. Gast Carla H. van Gils Lodewijk F.A. Wessels Nathan Harris Johannes M.G. Bonfrer Emiel J. Th. Rutgers Jan H.M. Schellens Jos H. Beijnen Submitted for publication 2.2 Chapter 2.2 Abstract In the last two decades, great efforts have been made in the search for better serum protein biomarkers that can be applied in screening, diagnosis and prognosis of cancer. One of the technologies used extensively for protein profiling is surface-enhanced laser desorption/ionisation time-of-flight mass spectrometry (SELDI-TOF MS). However, issues have been raised concerning the robustness and validity of alleged serum markers discovered by SELDI-TOF MS. Pre-analytical variables, such as sample collection, processing and storage temperature, have been shown to exert a profound effect on protein profiles, irrespective of true biological variation. However, little is known about the possible effects of the pre-analytical variable ‘sample storage duration’ on the serum proteome. We therefore aimed to investigate the effects of extended storage duration on the serum protein profile. To this end, archival sera of 140 breast cancer patients, stored at -30°C for 1 to 11 years, were profiled by SELDI-TOF MS using Immobilized Metal Affinity Capture arrays, a condition applied in the majority of biomarker discovery studies performed thus far in breast cancer. Spectrum-wide, 76 peak clusters were detected, 14 of which were found significantly associated to sample storage duration, following five distinct patterns. These clusters were structurally identified as C3a des-arginine anaphylatoxin and multiple fragments of albumin and fibrinogen. These proteins have, however, also been described previously as potential cancer markers, rendering them specific to both disease and sample handling issues. Hence, to prevent experimental variation to be interpreted erroneously as disease associated variation, assessment of potential confounding by pre-analytical parameters (such as storage time) is a prerequisite in biomarker discovery and validation studies. Moreover, regarding the different (nonlinear) patterns by which peak intensities were found associated to storage duration, merely linear corrections for sample storage duration will not necessarily suffice. 60 Influence of sample storage duration on serum protein profiles Introduction In the last two decades, much effort has been devoted to the search for improved markers that can be applied in screening, diagnosis and prognosis of cancer. With cancer being a genetic disease, genetic markers were initially pursued by the investigation of the cancer genome. It is, however, currently understood that gene analysis by itself provides an incomplete picture. Due to alternative splicing of both mRNA and proteins, combined with more than 100 unique post-translational modifications, one gene can give rise to multiple protein species (1). Hence, compared to the genome, the proteome can provide a more dynamic and accurate reflection of both the intrinsic genetic programme of the cell and the impact of its immediate environment (2). Since proteome analysis can provide the link between gene sequence and cellular physiology (3), proteomics is expected to complement gene analyses for the detection of novel cancer markers (4). In search of those markers, several different methods based on mass spectrometry (MS) have been applied to interrogate the proteome. One of the technologies used extensively for protein profiling is surface-enhanced laser desorption/ionisation timeof-flight mass spectrometry (SELDI-TOF MS) (5). This technology combines retentive chromatography with laser desorption/ionisation MS instrumentation, enabling highthroughput mass profiling of highly complex biological samples such as serum. During subsequent analyses, spectral patterns are compared across sample groups to find discerning masses or differential changes in peak intensities. The majority of SELDI studies reported thus far have investigated serum, though the technology is equally effective in analysing tissue lysates, for instance (6). Serum, however, is an easy to sample, readily accessible protein-rich body fluid, perfusing all tissues of the body and thus, theoretically, providing a good reflection of the human proteome (7). In addition, existing serum banks could readily provide serum from a large number of patients, enabling studies to be carried out in a timely fashion, as sample collection otherwise would have taken years (8). Indeed, many reports have described the successful application of SELDI-TOF MS in the discovery of potential serum markers for different types of cancer, such as ovarian (9), colorectal (10), and thyroid carcinoma (11). However, issues have been raised concerning the robustness and validity of alleged serum markers discovered by SELDI-TOF MS. A potential drawback of analysing highdimensional proteomic (SELDI-TOF MS) data for disease associated biomarkers is the propensity to discover patterns resulting from pre-analytical artefacts in a given sample set, rather than from the pathology of interest (12). Indeed, several lines of evidence indicate that pre-analytical variables, such as sample collection, processing and storage temperature, can exert a profound effect on protein profiles, regardless of true biological variation (13-15). 61 Chapter 2.2 However, yet little is known about the possible effects of the pre-analytical variable ‘sample storage duration’ on the serum proteome. Although this parameter has been investigated in two studies, only very few sera (n ≤ 12), stored for relatively short periods of time (1-3 months) were profiled (16;17). Clinical studies generally exceed these storage durations, since study sera either originate from sample banks (18-20), or are collected prospectively over a period of years (21). Therefore, we previously set out to study the effects of longer storage duration periods (0 to 16 months), with a larger sample size (n = 150) (22). Nonetheless, even this extended storage interval is generally surpassed by clinical proteomics studies, as, for instance, McLerran et al. (23) have reported a collection interval of more than 20 years. Their prostate cancer study provides a clear example of the potentially detrimental effects of this long-term storage duration on clinical proteomics studies. Following analysis of prospectively collected sera, their initial results (obtained in archival sera) could not be confirmed. It was not until they subjected their initial study to extensive post-study data analysis, that they discovered their study to be biased by, amongst other, sample storage duration, as the cases had a much longer sample storage duration compared to the controls (23). Although the study of McLerran is not unique in analysing sera that originate from a serum bank, the influence of storage duration on reported serum protein profiles is seldomly investigated. In the current study, we therefore aimed to investigate the effects of extended storage duration (1 to 11 years) on the serum protein profile. To this end, archival sera of 140 breast cancer patients were profiled by SELDI-TOF MS with Immobilized Metal Affinity Capture (IMAC30) arrays, as these settings are employed in the majority of serum biomarker discovery studies performed in breast cancer (18;19;21;24-28). Peak clusters found significantly associated with sample storage duration were structurally identified. Materials and methods Study population Archival sera of 140 breast cancer patients, collected between January 1993 and December 2002, were analysed in our laboratory using standardised analytical procedures. All sera were collected prior to any therapy, with individuals’ informed consent, after approval by the Institutional review boards. All sera originated from the Netherlands Cancer Institute serum bank, where they had been collected and stored for 9 to 128 months at -30°C according to standard procedures. Chemicals All used chemicals were obtained from Sigma, St. Louis, MO, USA, unless stated otherwise. 62 Influence of sample storage duration on serum protein profiles Serum protein profiling Serum protein profiling was performed using the ProteinChip SELDI Reader (Bio-Rad Labs, Hercules, CA, USA) with IMAC30 arrays. Sera were analysed in three batches (n = 39, 47, and 54 in Batch 1, 2, and 3, respectively), on three consecutive days. Throughout the assay, arrays were assembled in a 96-well bioprocessor, which was shaken on a platform shaker at 350 rpm. Arrays were charged twice with 50 μl 100 mM nickel sulphate (Merck, Darmstadt, Germany) for 15 min, followed by three rinses with deionised water (Braun, Emmenbrücke, Germany) and two equilibrations with 200 µl Phosphate Buffered Saline (PBS; 0.01 M) pH 7.4 / 0.5 M sodium chloride / 0.1% TritonX-100 (binding buffer; sodium chloride from Merck) for 5 min. Sera thawed on ice and denatured by 1:10 dilution in 9 M urea / 2% 3-[(3-cholamidopropyl)dimethylammonio-]-1-propanesulfonic acid (CHAPS). Pretreated samples were diluted 1:10 in binding buffer and randomly applied in singular to the arrays. After a 30 min incubation, the arrays were washed twice with binding buffer and twice with PBS pH 7.4 / 0.5 M sodium chloride for 5 min. Following a quick rinse with deionised water, arrays were air-dried. A saturated solution of sinapinic acid (Bio-Rad Labs) in 50% acetonitrile (ACN; Biosolve, Valkenswaard, The Netherlands) / 0.5% trifluoroacetic acid (TFA; Merck) was applied twice (0.6 μl) to the arrays as the matrix. Following airdrying, the arrays were analysed using the ProteinChip SELDI (PBSIIc) Reader. Data were collected between 0 and 100 kDa, averaging 65 laser shots with intensity 200, detector sensitivity 9, and a focus lag time of 636 ns (m/z 7000). For mass accuracy, the instrument was calibrated on the day of measurements with All-in-One peptide standard (Bio-Rad Labs). Statistics and bioinformatics Spectra of the three batches were processed separately by the ProteinChip Software v3.1 (Bio-Rad Labs). Spectra were baseline subtracted, after which they were normalised to the total ion current. Spectra with normalisation factors > 2 or < 0.5 were excluded from further analysis. Following spectrum pre-processing, the Biomarker Wizard (BMW) software package was applied for peak detection. Peaks were auto-detected when occurring in at least 30% of spectra and when having a signal-to-noise ratio (S/N) of at least 7. Peak clusters were completed with peaks with a S/N of at least 5 in a cluster mass window of 0.4%, and peak information was subsequently exported as spreadsheet files. The batches were analysed on three consecutive days, a parameter known to influence spectral data (29;30). As such, merging peak intensity data of the three sets would lead to spurious results. To this end, the intensities of peaks occurring across all three sample sets were log transformed to obtain normal distributions. Next, the log transformed peak intensities were converted to standard Z-values per sample set, by subtracting the mean and dividing by the standard deviation. The log-Z transformed data of the three sets were subsequently merged in one file. 63 Chapter 2.2 To investigate (non-)linear effects of sample storage time on peak expression, samples were split according to four categories of sample storage duration (≤ 32 months, n = 16; 32-64 months, n = 59; 64-96 months, n = 48; > 96 months, n = 17). Mean peak intensity differences between the four categories were subsequently investigated by means of ANOVA analyses. Resulting p-values were corrected for multiple testing using the Bonferroni correction, by multiplying p-values with the number of peak clusters detected and tested (n = 76). Next, we investigated whether the relationship between peak intensity and sample storage time was influenced by patients’ age and / or stage of disease. To this end, samples were split according to tertiles of patients’ age (≤ 49.4, 49.5 -61.8, and > 61.8 years), or according to stage of disease (Stage 2A, and 2B). Mean peak intensity differences between the categories were subsequently investigated by ANOVA, and T-test statistics, respectively. For storage time-associated clusters found significantly related to patients’ age and / or stage of disease, the relationship between peak intensity and storage duration was investigated in subgroups of age (i.e., < and > median age) and stage (i.e., Stage 2A, and 2B). Statistical analyses were performed by using SPSS statistical software, version 13.0 (SPSS Inc., Chicago, IL, USA). Protein purification and identification A serum sample (500 μl) containing the proteins we found significantly associated with sample storage duration was denatured in 9 M urea / 2% CHAPS / 50 mM Tris-HCl pH 9. The sample was subsequently fractionated on Q Ceramic HyperD beads with a strong anion exchange moiety (Biosepra Inc., Marlborough, MA, USA). After binding of denatured serum to the beads, the flow through was collected and bound proteins were subsequently eluted with buffers with pH from 9 to 3. The fractions containing the proteins of interest were further purified by size fractionation, using Microcon 50 kDa MW spin concentrators (YM50, Millipore, Billerica, MA, USA) with increasing concentrations of ACN / TFA 0.1%. The filtrates containing the proteins of interest were subsequently de-salted by application on reversed phase RP18 beads (Varian Inc., Palo Alto, CA, USA), followed by elution with increasing concentrations of ACN / TFA 0.1%. The purification process was monitored by profiling each fraction on IMAC30 Ni arrays and NP20 arrays (a non-selective, silica chromatographic surface). Eluates containing the proteins of interest were dried and redissolved in loading buffer for SDSPAGE. Gel electrophoresis was performed on Novex NuPage gels (18% Tris-Glycine gel; Invitrogen, San Diego, CA, USA). Following staining with colloidal Coomassie staining (Simply Blue; Invitrogen), protein bands of interest were excised and collected. The proteins within the excised bands were eluted by washing twice with 30% ACN / 100 mM ammonium bicarbonate, followed by dehydration in 100% ACN. Next, gel bands were heated at 50°C for 5 min and eluted with 45% formic acid / 30% ACN / 10% isopropanol under sonification for 30 min. After leaving the eluates overnight at room temperature, they were profiled on NP20 arrays. Eluates were subsequently dried, resuspended in 20 ng/µl trypsin (Promega, Madison, WI, USA) in 10% ACN / 25 mM 64 Influence of sample storage duration on serum protein profiles ammonium bicarbonate, followed by incubation at room temperature for 4 h. The tryptic digests were profiled on NP20 chips, using 1 μl 20% alpha-cyano-4-hydroxy cinnaminic acid (Bio-Rad Labs) in 50% ACN / 0.5% TFA as matrix. Peptides in the digests were investigated with the NCBI database using the ProFound search engine at http://prowl.rockefeller.edu/prowl-cgi/profound.exe with the following search parameters: standard cleavage rules for trypsin, 1 missed cleavage allowed. Confirmation of protein identity was provided by sequencing tryptic digest peptides by quadrupole-TOF (Q-TOF) MS (Applied Biosystems / MSD Sciex, Foster City, CA, USA) fitted with a ProteinChip Interface (PCI-1000). Fragment ion spectra resulting from QTOF analyses were taken to search the SwissProt 44.2 database (Homo Sapiens: 11072 sequences) using the MASCOT search engine at www.matrixscience.com (Matrix Science Ltd., London, UK), with the following search parameters: monoisotopic precursor mass tolerance: 40 ppm, fragment mass tolerance: 0.2 Da, variable modifications: methionine oxidation, and trypsin cleavage site. Protein identity was furthermore confirmed by immunoassay on ProteinA beads, using the appropriate antibodies. Beads were loaded with antibody in PBS, washed twice with PBS, incubated for 30 min with whole serum, and washed 5 times with PBS and once with deionised water. Bound proteins were subsequently eluted using 0.1 M acetic acid, and eluates were profiled on NP20 arrays. The extent of non-specific binding was tested using a murine IgG antibody (Bio-Rad Labs). For all identification experiments, a serum sample lacking the protein of interest was run concurrently as a negative control. Table 1 Patient and sample characteristics of the study population. Parameter Patients N 140 Age (years), median [IQR] 52.6 [45.7-66.0] Stage* 2A 2B 3A Unknown 48 78 10 4 Diagnosis* IDC ILC IDC & ILC Other 104 23 4 9 Sample storage time (months), median [IQR] 61.3 [49.1-76.4] Sample collection interval Jan-’93 - Dec-‘02 Abbreviations: IQR: interquartile range. * Pathologically determined stage and diagnosis; IDC: invasive ductal carcinoma; ILC: invasive lobular carcinoma; other: mucinous, tubular, mixed, unknown. 65 Chapter 2.2 Results Study population Patient and sample characteristics are summarized in Table 1. The majority of breast cancer patients (> 80%) was diagnosed with Stage 2A and Stage 2B invasive ductal carcinoma. Median patients’ age was 52.6 years. Serum protein profiling Representative SELDI-TOF MS spectra are presented in Figure 1. Following serum protein profiling and spectrum pre-processing by the ProteinChip Software v3.1, spectra of two breast cancer patients (Batch 3) were discarded due to aberrant normalisation factors. Spectrum-wide, the Biomarker Wizard detected a total of 76 peak clusters across the spectra of all three batches. Figure 1 Representative example of protein profiles obtained from two breast cancer patient sera, stored for 10 (BC 1) and 120 (BC 2) months. Relative intensity → 5000 7000 8000 5911.0+H 40 20 6000 9000 8939.1+H 5342.4+H BC 1 0 40 BC 2 20 0 5000 6000 7000 8000 9000 Mass-to-charge ratio → Influence of sample storage duration on the serum protein profile Mean log-Z peak intensity differences between the four discrete sample storage time categories were investigated by ANOVA analyses. In total, 14 of the 76 peak clusters detected spectrum-wide were found to differ significantly in mean log-Z intensity between the four categories of sample storage duration (Table 2). None of the 14 peak clusters were found related to patients’ age (ANOVA; p > 0.05) or stage of disease (Ttest; p > 0.05). 66 Influence of sample storage duration on serum protein profiles Overall, five different patterns by which peak intensities were associated with sample storage duration were observed. In Figure 2, these patterns (A-E) are depicted for five representative peak clusters. For the peak clusters m/z 2773, 2789, 3089, and 3104, a positive association with storage time was observed up to a storage duration of approximately 5 years, after which peak intensities gradually decreased with sample storage time (Pattern A). Peak intensities of m/z 4215, 5908, 5929, 6114, and 11091 were observed to continuously decrease over sample storage time (Pattern B), while the intensity of m/z 4441 was found to increase over time (Pattern C). The fourth pattern (Pattern D), observed in peak clusters m/z 4471, and 8939, consists of an initial increase in peak intensity, after which the intensities remain stable over a prolonged period of sample storage. Finally, for both clusters m/z 5341 and 5557, peak intensities were stable up to approximately 8 years of storage, after which peak intensities decreased rapidly (Pattern E). Table 2 Protein Peak cluster information of the 14 peaks found significantly associated with sample storage duration. Peak (m/z) 2773 2789 3089 3104 ANOVA p-value* 0.033 0.001 0.016 0.005 Fibrinogen 4215 5341 5357 5908 5929 6114 11091 < 0.001 0.003 0.046 < 0.001 < 0.001 0.013 0.003 B E E B B B B Unknown fibrinogen fragment m/z 5341 fibrinogen‡ m/z 5341 fibrinogen‡, Ox m/z 5908 fibrinogen‡ m/z 5908 fibrinogen‡, Ox m/z 5908 fibrinogen‡ SPA adduct Unknown fibrinogen fragment C3adesArg 4471 8937 0.018 0.002 D D m/z 8939 C3adesArg‡ double charge m/z 8939 C3adesArg‡ Unknown 4441 0.028 C Unknown Albumin Regulation† Regulation† (pattern) A A A A (Alleged) identity‡ identity‡ m/z 2756 albumin‡, Ox m/z 2756 albumin‡, (Ox)2 m/z 3089 albumin‡ m/z 3089 albumin‡, Ox * Bonferroni corrected p-values from ANOVA test of mean intensity differences between the discrete time intervals, † 5 patterns by which peak intensity was associated to sample storage duration: A: initial increase, followed by a gradual decrease, B: continuous decrease or C: continuous increase, D: initial increase, after which intensities remain stable, and E: stable up to ~8 years of storage time, followed by a decrease. Peptides marked with ‡ were structurally identified. Peptide purification and (tentative) identification The m/z 2756 and 3089 peptides were present in the pH 4 eluate after QhyperD fractionation of whole serum. Following concentration of this fraction on YM50 spin concentrators the two peptides were detected in the 50% ACN eluate. Since the peptides were lost during subsequent purification processes, their amino acid (aa) sequence was determined by direct tandem MS on a PCI-interfaced Q-TOF. The two 67 Chapter 2.2 peptides were identified as N-terminal fragments of albumin (Figure 3). The theoretical mass of the m/z 2756 peptide (24 aa) and the m/z 3089 peptide (27 aa) is 2754.10 Da and 3085.51 Da, respectively, and the pI of both fragments is 6.04. Figure 2 Representative examples of the five Patterns (A-E) by which peak intensities were found associated to sample storage duration (y-axis: Z-log transformed peak intensity, x-axis: sample storage duration in months). A: m/z 3089 C: m/z 4441 B: m/z 5908 2 2 2 1 1 0 0 0 -1 -1 -2 -2 -2 0 - 32 0 - 32 64 - 96 32 - 64 64 - 96 32 - 64 96 - 128 0 - 32 96 - 128 64 - 96 32 - 64 96 - 128 E: m/z 5341 D: m/z 8939 2 3 1 2 0 1 -1 0 -2 -3 -1 -4 0 - 32 64 - 96 32 - 64 96 - 128 0 - 32 64 - 96 32 - 64 96 - 128 Figure 4 depicts the correlation matrix presenting the (absolute) Pearson’s correlation coefficients calculated between the peak intensities of the 14 peaks found significantly associated to storage time. The three peak clusters m/z 2756, 2773, and 2789 were found highly correlated (Figure 4). As the mass deviation between m/z 2773, 2789 and 2756 is approximately 16 and 32 Da, these peptides most likely represent oxidised forms of the m/z 2756 albumin fragment. Similarly, m/z 3104 was found highly correlated to m/z 3089 (Figure 4). Regarding the mass difference of approximately 16 Da, m/z 3104 is likely to represent the oxidised form of the m/z 3089 albumin fragment. These hypothesised identities are endorsed by the observation that all five peak clusters show a similar correlation with sample storage time (i.e., initial increase, followed by a gradual decrease in peak intensity: Pattern A). 68 Influence of sample storage duration on serum protein profiles Following QhyperD fractionation, the m/z 5908 peptide was detected in the flow through. After concentration of this fraction on YM50 spin concentrators, the peptide was found in the flow through. De-salting by use of RP18 beads resulted in elution of the peptide in the 50% ACN / 0.1% TFA eluate. This fraction was again concentrated on YM3 spin concentrators. Profiling of the retentate revealed only peptides < 5908 Da. In prior identification attempts, the m/z 5908 peptide was shown to degrade with increasing manipulation. The detected peptides are therefore likely to originate from breakdown of the m/z 5908 peptide. Direct sequencing of three peptides by tandem MS on a Q-TOF confirmed the peptides to originate from a fibrinogen alpha-E fragment (FGA576-630), 54 aa in length, with theoretical mass 5904.22 Da and pI 8.07 (Figure 5). Figure 3 Structural identification of the m/z 2756 and m/z 3089 peak clusters. Direct sequencing of the m/z 2756 and m/z 3089 peak clusters. MS spectrum of the YM50 50% ACN eluate. All peptides were sequenced with tandem MS using Q-TOF. Results from the MASCOT search for protein identification include start and end positions of the peptide sequence starting from the amino acid terminal of the whole protein, the observed m/z (Mr (obs)), transformed to its experimental mass (Mr(expt)), the calculated mass (Mr(calc)) from the matched peptide sequence, as well as their mass difference (Delta), and the peptide sequence (in grey: the amino acid sequence determined by Q-TOF MS). 2754.5771 100 3969.6741 3970.6267 3818.5620 3968.6443 2753.6125 2755.6064 3085.8679 3819.5469 3084.8691 3816.5674 3086.9119 2744.2961 % 2736.9192 2756.5286 3820.5825 3083.8936 3087.8425 3800.4783 2728.5276 2727.3542 2677.0747 2669.2397 2543.2390 2655.1824 0 2500 2600 2757.6008 3371.8872 3100.8860 2770.1846 3166.9778 2939.7974 3284.0730 3372.8596 3783.3423 2800 2900 3000 3100 3689.3057 3448.2319 3044.8630 2700 3799.5215 3200 3300 3400 3500 3600 3700 3822.5530 3875.6406 3800 3900 m/z, amu MASCOT search results: m/z 2756 and m/z 3089 N-terminal truncated albumin fragments StartMr Mr Mr Delta Sequence StartEnd (obs) (expt) (calc) 25-48 2753.61 2752.60 2752.43 0.17 R.DAHKSEVAHRFKDLGEENFKALVL.I 25-51 3083.83 3082.82 3083.62 -0.80 R.DAHKSEVAHRFKDLGEENFKALVLIAF.A Amino acid sequence of albumin fragments (start: 25 - end: 57, 82% sequence coverage): DAHKSE VAHRFKDLGE ENFKALVLIA FAQYLQQ 69 Chapter 2.2 The m/z 4215, m/z 5341, m/z 5357, m/z 5908, m/z 5929, m/z 6114 and m/z 11091 peak clusters were found highly correlated to each other (Figure 4). Moreover, as the mass of the m/z 5341 corresponds to the theoretical mass of the 49 aa fibrinogen alpha-E fragment FGA576-625, the correlated peptides most likely represent (oxidised) fibrinogen fragments. The m/z 6114 cluster represents the SPA adduct of m/z 5908 FGA576-630. Indeed, except for m/z 5341 and m/z 5357, the (hypothesized) fibrinogen fragments all show a similar correlation with sample storage time (i.e., gradual decrease in peak intensity over sample storage time: Pattern B). Figure 4 Peak intensity correlation matrix for the 14 peaks found associated with sample storage duration (for clarity, Pearsons´ correlation coefficients were converted into absolute values). 1 m/z 2773 0.9 m/z 2789 m/z 3089 0.8 m/z 3104 0.7 m/z 4215 m/z 5341 0.6 m/z 5357 0.5 m/z 5908 0.4 m/z 5929 m/z 6114 0.3 m/z 11091 0.2 m/z 4471 m/z 8937 0.1 m/z 4441 m/z 4441 m/z 8937 m/z 4471 m/z 11091 m/z 6114 m/z 5929 m/z 5908 m/z 5357 m/z 5341 m/z 4215 m/z 3104 m/z 3089 m/z 2789 m/z 2773 Following QhyperD fractionation, the m/z 8939 peptide was eluted in the flow through. This fraction was concentrated on YM50 spin concentrators, and the peptide was found in the 30% ACN eluate. De-salting of the eluate on RP18 beads resulted in elution of the peptide in the 50% ACN / 0.1% TFA eluate, which was subsequently subjected to SDS-PAGE analysis. After staining, a clear band in the 8.9 kDa region was visible, which was excised, Elution of the proteins within the excised bands was followed by 70 Influence of sample storage duration on serum protein profiles tryptic digestion of the eluate. Profiling of the gel-eluate confirmed the presence of the peptide, and peptide mapping of the tryptic digest identified it as complement component 3 precursor (estimated Z-score 1.57, 4% sequence coverage). Amino acid sequencing of 6 peptides in the tryptic digest by tandem MS on a Q-TOF identified the marker as C3a des-arginine anaphylatoxin (C3adesArg, 61% sequence coverage), a 76 amino acid protein with theoretical mass 8939.46 Da and pI 9.54 (Figure 6). This identity was confirmed by an immunoassay, for which a C3a polyclonal antibody (Abcam Ltd, Cambridge, UK) was used. Profiling of the eluates revealed the presence of a peak at m/z 8940. Non-specific binding as determined by binding to murine IgG antibody was very low. The peak intensities of m/z 4471 and m/z 8939 were found highly correlated (Figure 4). Regarding its mass, the m/z 4471 peak most likely represents the doubly charged form of the m/z 8939 peak. The two peak clusters show similar correlations to sample storage duration (i.e., initial increase, after which peak intensities remain stable over time: Pattern D), endorsing the hypothesised identity of m/z 4471. Discussion In the current study, archival sera of 140 breast cancer patients, stored at -30°C for 1 to 11 years, were analysed by SELDI-TOF MS. Of the 76 peak clusters detected spectrumwide, peak intensities of 14 peak clusters were found to be significantly associated with sample storage duration by five different patterns (A - E). These peak clusters were structurally identified as C3adesArg and multiple fragments of albumin and fibrinogen. Their susceptibility to sample handling issues has been discussed in previous studies (8;22;31). A number of these proteins have, however, also been reported as potential cancer markers (10;11;21). Although they are not tumour-derived, it is currently hypothesized that these (cancer specific) serum proteins are generated from a pool of high-abundant founder proteins by tumour specific protease activities (11;32;33). Hence, these cleavage products were found to be specific for both disease and preanalytical sample handling parameters. Evidently, assessment of potential confounding by pre-analytical parameters (such as storage time) is of vital importance, to prevent experimental variation to be interpreted erroneously as disease associated variation. Moreover, regarding the different (non-linear) patterns by which peak intensities were found associated to storage duration, merely linear corrections for sample storage duration will not necessarily suffice. The albumin clusters Four of the 14 significant peak clusters were observed to initially increase in peak intensity up to approximately five years of storage time, after which peak intensities decreased (Pattern A). Two clusters (m/z 3089 and m/z 3105) were structurally 71 Chapter 2.2 identified as albumin fragments, while the other two clusters (m/z 2773 and m/z 2789) most likely correspond to oxidised forms of the structurally identified m/z 2756 albumin fragment. We hypothesise that the observed pattern is the result of continuous in vitro proteolytic degradation of albumin and its fragments. Figure 5 Structural identification of the m/z 5341 and m/z 5908 peak clusters. Direct sequencing of the m/z 5341 and m/z 5908 peak clusters. MS spectrum of the YM3 retentate. Insert: SELDI-TOF MS spectrum of the 50% ACN / 0.1% TFA RP18 eluate (upper spectrum) and YM3 retentate (lower spectrum). All peptides were sequenced with tandem MS using Q-TOF for confirmation. Results from the MASCOT search for protein identification include start and end positions of the peptide sequence starting from the amino acid terminal of the whole protein, the observed m/z (Mr (obs)), transformed to its experimental mass (Mr(expt)), the calculated mass (Mr(calc)) from the matched peptide sequence, as well as their mass difference (Delta), and the peptide sequence (in grey: the amino acid sequence determined by QTOF MS). Relative intensity → 5914.5+H 5347.8+H 8597.0+H 6120.3+H 2768.0+H 4000 6000 8000 2557.8+H 2937.0+H 2773.5+H 3284.5+H 3212.6+H 2341.2+H 1816.1+H 1899.8 2000 3000 4000 5368.9+H 5000 6000 Mass to charge ratio → MASCOT search results: m/z 5908 C-terminal fibrinogen fragment StartStart-End Mr Mr Mr Delta Sequence (obs) (expt) (calc) 576-598 2553.00 2551.99 2552.09 -0.10 K.SSSYSKQFTSSTSYNRGDSTFES.K 576-600 2768.20 2767.19 2767.22 -0.03 K.SSSYSKQFTSSTSYNRGDSTFESKS.Y 576-601 2931.20 2930.19 2930.28 -0.09 K.SSSYSKQFTSSTSYNRGDSTFESKSY.K Amino acid sequence of m/z 5341 fibrinogen fragment (start: 576 - end: 625, 53% sequence coverage): SSSYSKQFTS STSYNRGDST FESKSYKMAD EAGSEADHEG THSTKRGHA Amino acid sequence of m/z 5908 fibrinogen fragment (start: 576 - end: 630, 48% sequence coverage): SSSYSKQFTS STSYNRGDST FESKSYKMAD EAGSEADHEG THSTKRGHAK SRPV 72 Influence of sample storage duration on serum protein profiles Albumin is the most abundant serum protein (30-50 mg/ml), comprising about one-half of the blood serum proteins (34). As such, detection of its proteolytic fragments as markers for sample storage duration is not unexpected. The susceptibility of albumin for proteolytic degradation during prolonged storage at -30°C has been described previously by our group (22). We observed the N-terminal albumin25-57 fragment to be positively correlated to storage duration (1.4 years) at -30°C. Similarly, peaks corresponding to our m/z 2773 and m/z 3104 peak clusters were also found to increase in peak intensity with increasing storage time. The fibrinogen clusters Seven of the 14 peak clusters found significantly associated with sample storage duration were identified as (probable) fibrinogen fragments. The m/z 5908 FGA576-630, its oxidised form at m/z 5929, its SPA adduct at m/z 6114, and the two alleged fibrinogen fragments at m/z 4215 and m/z 11091 all continuously decrease in peak intensity with increasing sample storage time (Pattern B). The m/z 5341 FGA576-625 and its oxidised form at m/z 5357, however, decrease in peak intensity only after approximately six years of storage (Pattern E). These time-dependent changes in peak intensities represent the characteristics of a sequential reaction, in which the fibrinogen fragments are proteolytically degraded into subsequent smaller fragments. Fibrinogen acts as the main factor in the formation of a blood clot by polymerisation to a fibrin network and by enabling platelets to aggregate. (35) Similar to albumin, it is one of the most abundant blood proteins (2-4 mg/ml), and as such, detection of proteolytic fibrinogen fragments by SELDI-TOF MS as indicators for sample storage duration is not surprising. In addition, various fibrinogen fragments have been found correlated to coagulation time. While the m/z 5908 FGA576-630 continuously decreased in peak intensity with coagulation time, the intensities of all other peaks initially increased with coagulation time, after which they either remained stable (m/z 4215, m/z 11091), or decreased (m/z 5341, m/z 6114). (8;31;36) The different fibrinogen fragments have been described in relation to various types of cancer. Villanueva et al. (11) reported a decreased serum m/z 5902 FGA576-630 peak intensity in thyroid cancer compared to normal. In contrast, a 5.9 kDa peak (not structurally identified) has been found increased in cancer vs. control in colorectal (37;38), pancreatic (39), gastric (40), and lung cancer (41;42), and in hypopharyngeal squamous cell carcinoma (HSCC) (43). The two latter studies also reported increased serum m/z 5339, m/z 5927 and m/z 6114 peak intensities in cases compared to controls (42;43). Although the sera of the groups compared in above-mentioned studies allegedly were collected in the same time interval, precise information on storage duration generally is not provided. Proteolytic degradation is, however, known to decelerate with decreasing temperature. Since all sera investigated in these studies were stored at 80°C, influence of storage duration on peak expression may be limited compared to our (-30°C) study. 73 Chapter 2.2 Figure 6 Structural identification of the m/z 8939 peak cluster. Peptide mapping of the m/z 8939 peak cluster. MS spectrum of the m/z 8939 tryptic digest in the gel eluate. All peptides were sequenced with tandem MS using Q-TOF for confirmation. Results from the MASCOT search for protein identification include start and end positions of the peptide sequence starting from the amino acid terminal of the whole protein, the observed m/z (Mr (obs)), transformed to its experimental mass (Mr(expt)), the calculated mass (Mr(calc)) from the matched peptide sequence, as well as their mass difference (Delta), the number of missed cleavage sites for trypsin (Miss), and the peptide sequence (in grey: the amino acid sequence determined by Q-TOF MS). 1588.7266 100 1589.7362 1590.7296 % 1591.7397 1339.5571 1568.6200 1716.8252 1745.8353 960.5211 1095.5529 1037.4445 0 900 1000 1100 1321.5455 1200 1300 1800.8739 1949.8494 1433.6863 1400 1500 1600 1700 2164.0596 1966.8585 1592.7504 1341.5610 1800 1900 2274.1758 1984.8695 2000 2100 2166.0745 2200 2457.1707 2553.3289 2276.1829 2300 2400 2500 2600 2700 2800 2900 m/z, amu MASCOT peptide mapping results: StartStart- End m/z 8939 C3adesArg anaphylatoxin Mr Mr Mr Delta Miss Sequence (obs) (expt) (calc) 672-679 960.52 959.51 959.54 -0.03 1 R.SVQLTEKR.M 713-722 1095.55 1094.54 1094.58 -0.04 1 R.FISLGEACKK.V 699-709 1339.59 1338.58 1338.58 -0.00 2 K.ELRKCCEDGMR.E 692-704 1568.64 1567.63 1567.64 -0.00 2 R.KCCEDGMRENPMR.F 723-735 1588.74 1587.73 1587.74 -0.01 0 K.VFLDCCNYITELR.R 722-735 1716.84 1715.83 1715.84 -0.00 1 K.KVFLDCCNYITELR.R Amino acid sequence of m/z 8939 C3adesArg (start: 672 - end: 747, 61% sequence coverage): SVQLTEKRMDKVGKYPKELRKCCEDGMRENPMRFSCQRRTRFISLGEACKKVFLDCCNYITELRRQHARA SHLGLA 74 Influence of sample storage duration on serum protein profiles The C3adesArg clusters We observed two peak clusters at m/z 8939 and m/z 4471, identified as C3adesArg and its doubly charged form, to initially increase in peak intensity, after which intensities remained constant during the residual time interval studied (Pattern D). The acute phase reactant C3 is the most abundant (1.2 mg/ml) complement protein in serum (44). This protein supports the activation of all three pathways of complement activation (the classic, alternative, and lectin pathway) (45;46). Produced mainly in the liver and adipocytes, it is formed by cleavage of C3 (185 kDa) by C3-convertases into C3b (176 kDa) and C3a (9 kDa) (47). The anaphylatoxin C3a is only short lived in serum, as carboxypeptidases cleave the C-terminal arginine residue, creating the more stable but biologically inactive C3adesArg (8.9 kDa) (46-48). Presumably, the conversion of C3a to C3adesArg becomes complete during the first months of storage, explaining the observed increase in m/z 8939 peak intensity during the first months of storage. As m/z 8939 C3adesArg peak intensities remain stable following the initial increase, the protein appears not susceptible to proteolytic degradation, a finding that is corroborated by the reported stability of C3a(desArg) to extremes of heat and pH (49). Complement can also be activated in vitro, as activation of the coagulation system is followed by activation of platelets, eliciting complement activation (50). Although not structurally identified, Banks et al. (8) indeed reported the intensity of an IMAC-Cu m/z 8939 and m/z 4477 peak to significantly increase with prolonged coagulation times. The 8.9 kDa C3adesArg peak has been described as an alleged biomarker in a number of studies investigating different cancer types using serum SELDI-TOF MS analysis (21;5154). This protein peak has been found increased in different cancer types, such as breast (18;21;24;55), colorectal (51;54), hepatocellular cancer (52;56;57), and chronic lymphoid malignancies (53). In contrast, however, the studies of Hu et al. (58) and Han et al. (42) have reported an 8.9 kDa peak (not structurally identified) to be decreased in breast and lung cancer sera. As information regarding sample collection intervals is rarely provided by reported studies, the extent to which sample storage duration might have biased reported results can not be assessed. Of particular interest though are two studies published by Li et al. (18;21). Their first study reports an 8.9 kDa peak that was increased in breast cancer compared to healthy controls. This peak had a very high diagnostic performance (18). The cancer sera were, however, collected during a (non-specified) longer time interval than the control sera, as mentioned in their validation study, but the association between sample storage duration and peak expression was not investigated. The increase of the 8.9 kDa peak (identified as C3adesArg) in breast cancer was confirmed by analysis of a second, independent sample set, all sera of which were collected within the same 2-year window. Compared to their discovery study, however, the diagnostic performance of the 8.9 kDa peak was much lower, indicating probable bias by storage duration in their discovery study (21). Nonetheless, results of their initial study were indeed reproducible, as proven by the validation study. Their first study sample set was stored 75 Chapter 2.2 at -80°C, a temperature at which the formation of C3adesArg might be limited compared to -30°C. Their validation sample set was, however, stored at -30°C, but as all these sera were procured in the same time interval, the samples of this set are unlikely to be biased by storage duration. Although evidently, results can be reproducible when sample groups differ in storage time, investigators are not absolved from assessment of potential bias by storage parameters. We have not yet structurally identified the last peak cluster at m/z 4441. This peak was found positively associated to sample storage duration. Most likely, this m/z 4441 cluster represents a high-abundant serum protein fragment, formed by continuous non-specific proteolytic activity during storage at -30°C. Conclusion In conclusion, we have identified SELDI-TOF MS peak intensities of C3adesArg and various albumin and fibrinogen fragments to be significantly associated to storage duration in sera of 140 breast cancer patients. Reported proteins, however, have also been described as potential cancer markers in previous reports, rendering them specific to both disease and sample handling issues. Hence, assessment of potential confounding pre-analytical parameters (such as storage time) should be an integral component of biomarker discovery and validation studies, to prevent experimental variation to be interpreted erroneously as disease associated variation. Moreover, regarding the different (non-linear) patterns by which peak intensities were found associated to storage duration, merely linear corrections for sample storage duration will not necessarily suffice. Acknowledgement This study was supported by a grant of the Dutch Cancer Society (project NKI 20053421). References (1) Banks RE, Dunn MJ, Hochstrasser DF, Sanchez JC, Blackstock W, Pappin DJ et al. Proteomics: new perspectives, new biomedical opportunities. Lancet 2000; 356(9243):1749-1756. (2) Aebersold R, Anderson L, Caprioli R, Druker B, Hartwell L, Smith R. Perspective: a program to improve protein biomarker discovery for cancer. J Proteome Res 2005; 4(4):1104-1109. (3) Dove A. Proteomics: translating genomics into products? Nat Biotechnol 1999; 17(3):233-236. (4) Clarke W, Zhang Z, Chan DW. The application of clinical proteomics to cancer and other diseases. Clin Chem Lab Med 2003; 41(12):1562-1570. 76 Influence of sample storage duration on serum protein profiles (5) Hutchens TW, Yip TT. New desorption strategies for the mass spectrometric analysis of macromolecules. Rapid Commun Mass Spectrom 1993; 7:576-580. (6) Issaq HJ, Veenstra TD, Conrads TP, Felschow D. The SELDI-TOF MS approach to proteomics: protein profiling and biomarker identification. Biochem Biophys Res Commun 2002; 292(3):587-592. (7) Anderson NL, Anderson NG. The human plasma proteome: history, character, and diagnostic prospects. Mol Cell Proteomics 2002; 1(11):845-867. (8) Banks RE, Stanley AJ, Cairns DA, Barrett JH, Clarke P, Thompson D et al. Influences of blood sample processing on low-molecular-weight proteome identified by surface-enhanced laser desorption/ionization mass spectrometry. Clin Chem 2005; 51(9):1637-1649. (9) Zhang Z, Bast RC, Jr., Yu Y, Li J, Sokoll LJ, Rai AJ et al. Three biomarkers identified from serum proteomic analysis for the detection of early stage ovarian cancer. Cancer Res 2004; 64(16):5882-5890. (10) Engwegen JY, Helgason HH, Cats A, Harris N, Bonfrer JM, Schellens JH et al. Identification of serum proteins discriminating colorectal cancer patients and healthy controls using surface-enhanced laser desorption ionisation-time of flight mass spectrometry. World J Gastroenterol 2006; 12(10):1536-1544. (11) Villanueva J, Martorella AJ, Lawlor K, Philip J, Fleisher M, Robbins RJ et al. Serum peptidome patterns that distinguish metastatic thyroid carcinoma from cancer-free controls are unbiased by gender and age. Mol Cell Proteomics 2006; 5(10):1840-1852. (12) Ransohoff DF. Lessons from controversy: ovarian cancer screening and serum proteomics. J Natl Cancer Inst 2005; 97(4):315-319. (13) Villanueva J, Philip J, Chaparro CA, Li Y, Toledo-Crow R, DeNoyer L et al. Correcting common errors in identifying cancer-specific serum peptide signatures. J Proteome Res 2005; 4(4):1060-1072. (14) Findeisen P, Sismanidis D, Riedl M, Costina V, Neumaier M. Preanalytical impact of sample handling on proteome profiling experiments with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Clin Chem 2005; 51(12):2409-2411. (15) Albrethsen J, Bogebo R, Olsen J, Raskov H, Gammeltoft S. Preanalytical and analytical variation of surface-enhanced laser desorption-ionization time-of-flight mass spectrometry of human serum. Clin Chem Lab Med 2006; 44(10):1243-1252. (16) Hsieh SY, Chen RK, Pan YH, Lee HL. Systematical evaluation of the effects of sample collection procedures on low-molecular-weight serum/plasma proteome profiling. Proteomics 2006; 6(10):31893198. (17) Rai AJ, Gelfand CA, Haywood BC, Warunek DJ, Yi J, Schuchard MD et al. HUPO Plasma Proteome Project specimen collection and handling: towards the standardization of parameters for plasma proteome samples. Proteomics 2005; 5(13):3262-3277. (18) Li J, Zhang Z, Rosenzweig J, Wang YY, Chan DW. Proteomics and bioinformatics approaches for identification of serum biomarkers to detect breast cancer. Clin Chem 2002; 48(8):1296-1304. (19) Belluco C, Petricoin EF, Mammano E, Facchiano F, Ross-Rucker S, Nitti D et al. Serum Proteomic Analysis Identifies a Highly Sensitive and Specific Discriminatory Pattern in Stage 1 Breast Cancer. Ann Surg Oncol 2007; 14(9):2470-2476. (20) Adam BL, Qu Y, Davis JW, Ward MD, Clements MA, Cazares LH et al. Serum protein fingerprinting coupled with a pattern-matching algorithm distinguishes prostate cancer from benign prostate hyperplasia and healthy men. Cancer Res 2002; 62(13):3609-3614. (21) Li J, Orlandi R, White CN, Rosenzweig J, Zhao J, Seregni E et al. Independent Validation of Candidate Breast Cancer Serum Biomarkers Identified by Mass Spectrometry. Clin Chem 2005; 51(12):2229-2235. (22) Engwegen JY, alberts M, knol JC, Jimenez C.R., Depla ACTM, Tuynman H et al. Influence of variations in sample handling on SELDI-TOF MS serum protein profiles for colorectal cancer. Proteomics Clin Appl 2008; 2(6):936-945. 77 Chapter 2.2 (23) McLerran D, Grizzle WE, Feng Z, Bigbee WL, Banez LL, Cazares LH et al. Analytical Validation of Serum Proteomic Profiling for Diagnosis of Prostate Cancer; Sources of Sample Bias. Clin Chem 2007; 54(1):44-52. (24) Mathelin C, Cromer A, Wendling C, Tomasetto C, Rio MC. Serum biomarkers for detection of breast cancers: a prospective study. Breast Cancer Res Treat 2005;1-8. (25) Becker S, Cazares LH, Watson P, Lynch H, Semmes OJ, Drake RR et al. Surfaced-enhanced laser desorption/ionization time-of-flight (SELDI-TOF) differentiation of serum protein profiles of BRCA-1 and sporadic breast cancer. Ann Surg Oncol 2004; 11(10):907-914. (26) Laronga C, Becker S, Watson P, Gregory B, Cazares L, Lynch H et al. SELDI-TOF serum profiling for prognostic and diagnostic classification of breast cancers. Dis Markers 2003; 19(4-5):229-238. (27) Vlahou A, Laronga C, Wilson L, Gregory B, Fournier K, McGaughey D et al. A novel approach toward development of a rapid blood test for breast cancer. Clin Breast Cancer 2003; 4(3):203-209. (28) Goncalves A, Esterni B, Bertucci F, Sauvan R, Chabannon C, Cubizolles M et al. Postoperative serum proteomic profiles may predict metastatic relapse in high-risk primary breast cancer patients receiving adjuvant chemotherapy. Oncogene 2006; 25(7):981-989. (29) Karsan A, Eigl BJ, Flibotte S, Gelmon K, Switzer P, Hassell P et al. Analytical and preanalytical biases in serum proteomic pattern analysis for breast cancer diagnosis. Clin Chem 2005; 51(8):1525-1528. (30) Hu J, Coombes KR, Morris JS, Baggerly KA. The importance of experimental design in proteomic mass spectrometry experiments: some cautionary tales. Brief Funct Genomic Proteomic 2005; 3(4):322-331. (31) Baumann S, Ceglarek U, Fiedler GM, Lembcke J, Leichtle A, Thiery J. Standardized approach to proteome profiling of human serum based on magnetic bead separation and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Clin Chem 2005; 51(6):973-980. (32) Fung ET, Yip TT, Lomas L, Wang Z, Yip C, Meng XY et al. Classification of cancer types by measuring variants of host response proteins using SELDI serum assays. Int J Cancer 2005; 115(5):783-789. (33) Villanueva J, Shaffer DR, Philip J, Chaparro CA, Erdjument-Bromage H, Olshen AB et al. Differential exoprotease activities confer tumor-specific serum peptidome patterns. J Clin Invest 2006; 116(1):271284. (34) Quinlan GJ, Martin GS, Evans TW. Albumin: biochemical properties and therapeutic potential. Hepatology 2005; 41(6):1211-1219. (35) Mosesson MW. Fibrinogen and fibrin structure and functions. J Thromb Haemost 2005; 3(8):1894-1904. (36) Timms JF, Arslan-Low E, Gentry-Maharaj A, Luo Z, T'Jampens D, Podust VN et al. Preanalytic influence of sample handling on SELDI-TOF serum protein profiles. Clin Chem 2007; 53(4):645-656. (37) Chen YD, Zheng S, Yu JK, Hu X. Artificial neural networks analysis of surface-enhanced laser desorption/ionization mass spectra of serum protein pattern distinguishes colorectal cancer from healthy population. Clin Cancer Res 2004; 10(24):8380-8385. (38) Yu JK, Chen YD, Zheng S. An integrated approach to the detection of colorectal cancer utilizing proteomics and bioinformatics. World J Gastroenterol 2004; 10(21):3127-3131. (39) Koopmann J, Zhang Z, White N, Rosenzweig J, Fedarko N, Jagannath S et al. Serum diagnosis of pancreatic adenocarcinoma using surface-enhanced laser desorption and ionization mass spectrometry. Clin Cancer Res 2004; 10(3):860-868. (40) Liang Y, Fang M, Li J, Liu CB, Rudd JA, Kung HF et al. Serum proteomic patterns for gastric lesions as revealed by SELDI mass spectrometry. Exp Mol Pathol 2006; 81(2):176-180. (41) Xiao X, Liu D, Tang Y, Guo F, Xia L, Liu J et al. Development of proteomic patterns for detecting lung cancer. Dis Markers 2003; 19(1):33-39. (42) Han KQ, Huang G, Gao CF, Wang XL, Ma B, Sun LQ et al. Identification of lung cancer patients by serum protein profiling using surface-enhanced laser desorption/ionization time-of-flight mass spectrometry. Am J Clin Oncol 2008; 31(2):133-139. 78 Influence of sample storage duration on serum protein profiles (43) Zhou L, Cheng L, Tao L, Jia X, Lu Y, Liao P. Detection of hypopharyngeal squamous cell carcinoma using serum proteomics. Acta Otolaryngol 2006; 126(8):853-860. (44) Hugli TE. Human anaphylatoxin (C3a) from the third component of complement. Primary structure. J Biol Chem 1975; 250(21):8293-8301. (45) Bohana-Kashtan O, Ziporen L, Donin N, Kraus S, Fishelson Z. Cell signals transduced by complement. Mol Immunol 2004; 41(6-7):583-597. (46) Sahu A, Sunyer JO, Moore WT, Sarrias MR, Soulika AM, Lambris JD. Structure, functions, and evolution of the third complement component and viral molecular mimicry. Immunol Res 1998; 17(1-2):109-121. (47) de Bruijn MH, Fey GH. Human complement component C3: cDNA coding sequence and derived primary structure. Proc Natl Acad Sci U S A 1985; 82(3):708-712. (48) Nettesheim DG, Edalji RP, Mollison KW, Greer J, Zuiderweg ER. Secondary structure of complement component C3a anaphylatoxin in solution as determined by NMR spectroscopy: differences between crystal and solution conformations. Proc Natl Acad Sci U S A 1988; 85(14):5036-5040. (49) Hugli TE, Morgan WT, Muller-Eberhard HJ. Circular dichroism of C3a anaphylatoxin. Effects of pH, heat, guanidinium chloride, and mercaptoethanol on conformation and function. J Biol Chem 1975; 250(4):1479-1483. (50) Hamad OA, Ekdahl K, Lambris JD, Nilsson B. Complement activation triggered by thrombin receptoractivated platelets. Mol.Immunol. 44, 180. 2007. (51) Habermann JK, Roblick UJ, Luke BT, Prieto DA, Finlay WJ, Podust VN et al. Increased serum levels of complement C3a anaphylatoxin indicate the presence of colorectal tumors. Gastroenterology 2006; 131(4):1020-1029. (52) Lee IN, Chen CH, Sheu JC, Lee HS, Huang GT, Chen DS et al. Identification of complement C3a as a candidate biomarker in human chronic hepatitis C and HCV-related hepatocellular carcinoma using a proteomics approach. Proteomics 2006; 6(9):2865-2873. (53) Miguet L, Bogumil R, Decloquement P, Herbrecht R, Potier N, Mauvieux L et al. Discovery and identification of potential biomarkers in a prospective study of chronic lymphoid malignancies using SELDI-TOF-MS. J Proteome Res 2006; 5(9):2258-2269. (54) Ward DG, Suggett N, Cheng Y, Wei W, Johnson H, Billingham LJ et al. Identification of serum biomarkers for colon cancer by proteomic analysis. Br J Cancer 2006; 94(12):1898-1905. (55) Pusztai L, Gregory BW, Baggerly KA, Peng B, Koomen J, Kuerer HM et al. Pharmacoproteomic analysis of prechemotherapy and postchemotherapy plasma samples from patients receiving neoadjuvant or adjuvant chemotherapy for breast carcinoma. Cancer 2004; 100(9):1814-1822. (56) Poon TC, Yip TT, Chan AT, Yip C, Yip V, Mok TS et al. Comprehensive proteomic profiling identifies serum proteomic signatures for detection of hepatocellular carcinoma and its subtypes. Clin Chem 2003; 49(5):752-760. (57) Schwegler EE, Cazares L, Steel LF, Adam BL, Johnson DA, Semmes OJ et al. SELDI-TOF MS profiling of serum for detection of the progression of chronic hepatitis C to hepatocellular carcinoma. Hepatology 2005; 41(3):634-642. (58) Hu Y, Zhang S, Yu J, Liu J, Zheng S. SELDI-TOF-MS: the proteomics and bioinformatics approaches in the diagnosis of breast cancer. Breast 2005; 14(4):250-255. 79 Chapter Protein profiling of serum 3 Chapter Serum protein profiling for diagnosis of breast cancer using SELDI-TOF MS Marie-Christine W. Gast Carla H. van Gils Lodewijk F.A. Wessels Nathan Harris Johannes M.G. Bonfrer Emiel J. Th. Rutgers Jan H.M. Schellens Jos H. Beijnen Submitted for publication 3.1 Chapter 3.1 Abstract Early detection is of paramount importance in reducing breast cancer related mortality, yet the diagnosis of breast cancer is hampered by a lack of adequate detection methods. In search for novel markers for breast cancer, the surface-enhanced laser desorption/ionisation time-of-flight mass spectrometry (SELDI-TOF MS) technology has been applied with varying success in the investigation of the serum proteome. Both robustness and validity of alleged markers can, however, be jeopardised by demographic and pre-analytical variables, which are known to exert profound effects on protein profiles obtained by SELDI-TOF MS. Although validation as well as structural identification of putative markers can aid in determining their true performance, thus far, this has been performed in only a limited number of studies. We therefore aimed to identify and validate novel serum protein profiles specific for breast cancer and assess the influence of clinical (i.e., subjects’ age) and pre-analytical (i.e., sample storage duration) variables on the constructed classifiers. To this end, sera of breast cancer patients (n = 152) and healthy controls (n = 129) were analysed using the SELDI-TOF MS technology. Cases and controls were randomly and evenly divided into a training and test set. In the training set, 14 peak clusters were found to differ significantly in peak expression between cases and controls. The intensities of none of these peak clusters were influenced by subjects’ age and sample storage duration. Ten peak clusters were also found significantly discriminative in the test set. One peak cluster was structurally identified as C3a des-arginine anaphylatoxin, while 12 other peak clusters were tentatively identified as inter-alpha-trypsin inhibitor heavy chain 4 fragments and a fibrinogen fragment, respectively. Subsequent logistic regression analyses on the training set yielded a classification model with a moderate performance on the test set, corresponding to those reported in previously performed validation studies. As this moderate performance most likely originates from the highly heterogeneous nature of breast cancer, selection of breast cancer subgroups for comparison with healthy controls is expected to improve results of future diagnostic SELDI-TOF MS studies. 84 Diagnostic serum protein profiles for breast cancer Introduction The American Cancer Society has estimated that breast cancer will be the most commonly diagnosed cancer among women in the USA in 2008, as it is expected to account for 26% of all new cancer cases among women (1). Following lung cancer, breast cancer currently is the second leading cause of cancer deaths in women (1). As the 5-year survival rate decreases from 98% for localised disease to 26% for distant stage disease (2), early detection is of paramount importance in reducing breast cancer related mortality. The diagnosis of breast cancer is, however, hampered by a lack of adequate detection methods, resulting in detection of only 63% of breast cancers at an early stage (1). Although mammography currently is the most widely applied imaging test today, its predictive value is lower in women with dense breast tissue and smaller lesions. Moreover, no molecular markers are recommended for the (early) detection of breast cancer hitherto. Currently used serum tumour markers in breast cancer, e.g., Cancer Antigen 15.3, lack adequate sensitivity and specificity to be applicable in early detection, and are therefore approved by the FDA only for monitoring therapy of advanced breast cancer or recurrence (3). The application of a single biomarker in the detection of breast cancer may, however, not be feasible, as a single marker is unlikely to cover the high heterogeneity of breast cancer. Instead, a panel of markers is expected to better reflect breast cancer complexity, yielding an improved sensitivity and specificity. With cancer being, for a large part, a genetic disease, researchers initially searched for biomarkers by employing genomic and transcriptomic approaches. Although this has greatly expanded our insight into the genetic basis of cancer, it is currently understood that the functional “endunits” of the genome, the proteins, cannot be predicted by genetic and transcriptomic data alone. Due to amongst other post-transcriptional mRNA modifications (e.g., alternative splicing) and post-translational protein modifications, one gene can encode multiple proteins, reflecting both the intrinsic genetic programme of the cell and the impact of its immediate environment (4). As such, the proteome provides a more realistic and detailed view of the biological status, offering a richer source of potential biomarkers. One of the techniques currently applied in proteomics research of breast cancer is surface-enhanced laser desorption/ionisation time-of-flight mass spectrometry (SELDITOF MS). Until now, eleven studies have been published in which the SELDI-TOF MS platform was applied with varying success in the identification and validation of serum markers for diagnosis (5-12), prognosis (13), or monitoring of therapy-efficacy (14) or toxicity (15) in breast cancer. However, issues have been raised concerning the robustness and validity of alleged markers discovered by SELDI-TOF MS. A potential drawback of analysing high-dimensional proteomic (SELDI-TOF MS) data for disease associated biomarkers is the propensity to discover patterns among variables that are the result of pre-analytical artefacts in a given sample set, rather than of the pathology of 85 Chapter 3.1 interest (16). Indeed, several lines of evidence indicate that pre-analytical variables, e.g., sample collection, processing and storage, can exert profound effects on protein profiles, regardless of true biological variation. In addition, clinical characteristics, such as patients’ age, could also introduce bias (17). Despite these concerns, only few studies investigating the serum proteome for discovery of breast cancer specific biomarkers investigate the possible influence of pre-analytical and patient-related variables on the expression of potential biomarkers. The raised issues on the validity and robustness of alleged biomarkers can, however, also be addressed by validation and structural identification (16). Nonetheless, thus far, in breast cancer, only two panels of biomarkers discovered by SELDI-TOF MS have been validated by analysis of independent sample sets, resulting in partial (10;11) or no validation (18). Moreover, only few biomarkers discovered by SELDI-TOF MS breast cancer research have been structurally identified. In the current study, we aimed to discover and validate novel serum protein profiles specific for breast cancer. To this end, archival sera of breast cancer patients and healthy controls were analysed using SELDI-TOF MS. Spectral data were merged in one file, after which they were randomly and evenly split into a training and test set. In the training set, we detected 14 discriminating peak clusters, one cluster of which was structurally identified. Furthermore, the relationship between the intensity of the classifier peak clusters and breast cancer status was adjusted for demographic and preanalytical variables (i.e., subjects’ age and sample storage duration). Finally, the samples in the test set were applied for validation purposes. Materials and methods Study population Archival sera of 152 breast cancer patients (BC) and 129 female healthy controls (HC), collected between January 2003 and July 2005, were analysed on different occasions in our laboratory using standardised analytical procedures. All sera were collected prior to any therapy, with individuals’ informed consent after approval by the institutional review boards. All sera originate from the Netherlands Cancer Institute serum bank, where they had been collected and stored for 3 to 50 months at -30°C according to standard procedures. Chemicals All used chemicals were obtained from Sigma, St. Louis, MO, USA, unless stated otherwise. 86 Diagnostic serum protein profiles for breast cancer SELDI--TOF MS protein profiling SELDI Serum protein profiling was performed using the ProteinChip SELDI (PBSIIc) Reader (Bio-Rad Labs, Hercules, CA, USA). Various chip chemistries, binding- and washingprocedures and sample pretreatments were initially evaluated to determine which affinity chemistry and sample pretreatment procedure provided the best serum profiles in terms of number and resolution of proteins. Immobilized Metal Affinity Capture (IMAC30) arrays were selected for further analysis. Samples were analysed in three batches (Batch 1: BC: n = 40, HC: n = 40; Batch 2: BC: n = 43, HC: n = 46; Batch 3: BC: n = 69, HC: n = 43). The samples in Batch 1 were analysed in singular, while the samples in Batch 2 and 3 were analysed in duplicate. Throughout the assay, arrays were assembled in a 96-well bioprocessor, which was shaken on a platform shaker at 300 rpm. Arrays were charged twice with 50 μl 100 mM nickel sulphate (Merck, Darmstadt, Germany) for 15 min, followed by three rinses with deionised water (Braun, Emmenbrücke, Germany) and two equilibrations with 200 μl Phosphate Buffered Saline (PBS; 0.01 M) pH 7.4 / 0.5 M sodium chloride / 0.1% TritonX-100 (binding buffer; sodium chloride from Merck) for 5 min. Unfractionated serum samples were thawed on ice and denatured by 1:10 dilution in 9 M urea / 2% 3-[(3-cholamidopropyl)dimethylammonio-]-1-propanesulfonic acid (CHAPS). Pretreated samples were diluted 1:10 in binding buffer and randomly applied to the arrays. After a 30 min incubation, the arrays were washed twice with binding buffer and twice with PBS pH 7.4 / 0.5 M sodium chloride for 5 min. Following a quick rinse with deionised water, arrays were air-dried. A 50% sinapinic acid (SPA; Bio-Rad Labs) solution in 50% acetonitrile (ACN; Biosolve, Valkenswaard, The Netherlands) / 0.5% trifluoroacetic acid (TFA; Merck) was applied twice (1.0 μl) to the arrays as the matrix. Following air-drying, the arrays were analysed using the ProteinChip SELDI (PBS IIc) Reader. As the three batches were analysed on different occasions (with PBS IIc Reader maintenance in between), data acquisition was optimised for each sample set separately (data not shown), to obtain similar spectra. For mass accuracy, the instrument was calibrated on each day of measurements with All-in-One peptide standard (Bio-Rad Labs). Statistics and bioinformatics Spectra were processed per batch by the ProteinChip Software v3.1 (Bio-Rad Labs). Spectra were baseline subtracted, followed by normalisation to the total ion current. Spectra with normalisation factors > 2 or < 0.5 were excluded from further analysis. The Biomarker Wizard (BMW) software package was applied for peak detection. BMW settings were optimised for each batch separately (data not shown), to ascertain correct detection of real peaks (instead of peaks that merely represent noise). Peak information was subsequently exported as spreadsheet files, and peak intensities from the duplicate analyses in Batch 2 and 3 were averaged. The three batches were analysed on three 87 Chapter 3.1 separate occasions, a parameter known to influence spectral data (19;20). As such, merging peak intensity data of the three batches could lead to spurious results. To this end, first, the intensities of peaks occurring across all three batches were log transformed to obtain normal distributions. Next, the log transformed peak intensities were converted to standard Z-values per batch, by subtracting the mean and dividing by the standard deviation. The log-Z transformed data of the three batches were merged in one file. After this, cases and controls were randomly divided over a training (BC: n = 76, HC: n = 65) and test (BC: n = 76, HC: n = 64) set. In the training set, the parametric T-test was applied for the comparison of the mean log-Z transformed peak intensities between cases and controls. Resulting p-values were corrected for multiple testing by the Bonferroni method, by multiplying p-values with the number of peak clusters detected and tested. To estimate the influence of subjects’ age and storage duration on the relationship between the 14 discriminating peak clusters and breast cancer status, logistic regression analyses were performed on the training set. First, we calculated a crude odds ratio per peak cluster, using a univariate model (i.e., by inclusion of only one peak cluster as continuous variable). Next, multivariate odds ratios adjusted for subjects’ age (categorized according to tertiles: ≤ 51.3 years, 51.3-61.4 years, > 61.4 years), and storage duration (categorized according to tertiles: ≤ 14.5 months, 14.5-31.7 months, > 31.7 months) were calculated. Both parameters were considered confounders if the adjusted odds ratio was 10% different from the crude odds ratio. To investigate the relationship between a combination of the log-Z transformed peak intensities and the presence of breast cancer, crude odds ratios for each of the peak intensities (as continuous variables) were estimated in a logistic regression model with the inclusion of all peak clusters detected based on forward entry (p < 0.05). Again, to investigate whether the relationship between peak intensities and the presence of breast cancer could be explained by subjects’ age and / or sample storage duration, the odds ratios were adjusted for these parameters. The classification performance of the logistic regression model was evaluated by estimation of the area under the Receiver Operating Characteristic (ROC) curve (AUC) and accompanying 95% confidence interval. The model was subsequently applied to the test set for validation purposes. All statistical analyses were performed using SPSS statistical software, version 13.0 (SPSS Inc., Chicago, IL, USA). purification Peptide purifica tion and identification Structural identification of potential biomarkers was performed previously. Briefly, potential markers were purified from serum using anion-exchange chromatographic, size exclusion, and gel-electrophoresis techniques, following by trypsin digestion. The peptide map of the digest, acquired on the ProteinChip SELDI (PBS IIc) Reader, was investigated with the NCBI database using the ProFound search engine at http://prowl.rockefeller.edu/prowl-cgi/profound.exe. Confirmation of protein identity 88 Diagnostic serum protein profiles for breast cancer was provided by sequencing tryptic digest peptides by quadrupole-TOF (Q-TOF) MS (Applied Biosystems / MSD Sciex, Foster City, CA, USA) fitted with a ProteinChip Interface (PCI-1000). Fragment ion spectra were taken to search the SwissProt 44.2 database (Homo Sapiens: 11072 sequences) using the MASCOT search engine at www.matrixscience.com (Matrix Science Ltd., London, UK). Protein identity was further confirmed by immunoassay on ProteinA beads. Results Study population Patient and sample characteristics are summarized in Table 1. The healthy controls were significantly younger than the breast cancer patients at time of sample procurement (Mann-Whitney U test (MWU); p < 0.001). The majority of breast cancer patients had invasive ductal carcinoma (76%) and was diagnosed with Stage 2 (63%) disease. The median sample storage duration was slightly longer for breast cancer sera (median: 26.0 months) than for the healthy control sera (median: 20.1 months) (MWU; p = 0.018). Table 1 Patient and sample characteristics of the study population. Parameter N Breast cancer 152 Healthy control 129 Age (years), median [IQR] 61.1 [50.3-67.0] 52.0 [42.0-57.7] Stage* n.a. 0 1 2A / 2B 3A / 3C 7 30 68 / 28 13 / 6 DCIS IDC ILC IDC & ILC Other 6 116 16 5 9 Diagnosis* n.a. Sample storage time (months), median [IQR] Sample collection interval 26.0 [14.1-36.7] Apr-’03 - Jul-’05 20.1 [12.6-31.9] Jan-’03 - Jul-‘05 Abbreviations: DCIS: ductal carcinoma in situ, IDC: invasive ductal carcinoma, ILC: invasive lobular carcinoma, IQR: interquartile range, n.a.: not applicable. * Pathologically determined stage and diagnosis; other: mucinous, tubular, mixed, unknown. 89 Chapter 3.1 SELDI--TOF MS protein profiling SELDI Representative SELDI-TOF MS spectra are presented in Figure 1. Following spectrum pre-processing and normalisation, 73 (BC: n = 36; HC: n = 37), 89 (BC: n = 43; HC: n = 46), and 111 samples (BC: n = 68; HC: n = 43) were left for analysis in Batch 1, 2, and 3, respectively. The Biomarker Wizard detected 57 peak clusters across all three batches. In the training set, 14 peak clusters were found significantly different in expression between breast cancer and control (T-test; Bonferroni corrected p < 0.05, Table 2). Except for the m/z 4219 and m/z 11745 peak clusters, intensities were found decreased in breast cancer compared to control (Table 2: logistic regression, odds ratio < 1). Following correction for subjects’ age and sample storage duration, the adjusted odds ratios of three peak clusters (m/z 2733, 3965, and 4219) differed by more than 10% from the crude odds ratios. All three peaks remain, however, significantly related to breast cancer status. Ten of these 14 peak clusters were found significantly different in peak expression between breast cancer and control in the test set as well. Figure 1 Representative example of protein profiles obtained from a healthy control (HC) and a breast cancer patient (BC). 4000 Relative intensity → 40 20 6000 8000 8939.6+H 4289.7+H 3979.8+H HC 3281.3+H 0 40 BC 20 0 4000 6000 8000 Mass-to-charge ratio → Next, multivariate logistic regression analyses were performed on the training set. Following forward entry inclusion of all peak clusters detected spectrum-wide, four peak clusters (m/z 4219, 4309, 5350, and 29183) were incorporated in the model, resulting in a ROC AUC of 0.813 (85% CI: 0.742-0.884) (Table 3). Two peak clusters (m/z 4219 and 4309) were already found significantly different in peak expression between breast cancer and healthy control. Of the four peak clusters included in this model, only m/z 4219 had an adjusted odds ratio that differed more than 10% from the crude odds ratio. Similar to the univariate analyses, however, after adjustment this peak 90 Diagnostic serum protein profiles for breast cancer cluster was even more strongly related to breast cancer status. The multivariate model classified the samples in the training set with a sensitivity and specificity of 74.3% and 71.9%, respectively. Model performance was lower following validation on the test set (ROC AUC: 0.713 (95% CI: 0.626-0.800); sensitivity: 72.6%, specificity: 61.3%). Table 2 Characteristics of the 14 clusters that differ significantly in expression between breast cancer and healthy control in the training set. Cluster T-test Training set p-value* 0.011 < 0.001 < 0.001 < 0.001 < 0.001 0.004 0.005 0.007 0.003 0.046 0.002 < 0.001 < 0.001 0.003 (m/z) 2733 3166 3282 3299 3691 3782 3965 3980 3997 4219 4292 4309 8940 11745 Test set p-value* 0.047 0.013 0.005 < 0.001 n.s. 0.004 0.004 n.s. n.s. n.s. 0.028 0.007 0.004 0.008 Logistic regression analyses Training set † Training set ‡ (adjusted) OR (95% CI) OR (95% CI) 0.45 (0.28-0.70) 0.50 (0.31-0.81) 0.40 (0.26-0.62) 0.41 (0.25-0.67) 0.41 (0.26-0.64) 0.44 (0.28-0.70) 0.42 (0.27-0.64) 0.41 (0.26-0.66) 0.37 (0.23-0.59) 0.37 (0.22-0.63) 0.44 (0.28-0.68) 0.48 (0.30-0.75) 0.45 (0.29-0.69) 0.52 (0.33-0.81) 0.47 (0.31-0.71) 0.49 (0.31-0.77) 0.44 (0.29-0.67) 0.44 (0.27-0.70) 1.86 (1.27-2.72) 2.40 (1.51-3.80) 0.39 (0.24-0.65) 0.42 (0.25-0.71) 0.32 (0.20-0.54) 0.32 (0.18-0.56) 0.37 (0.24-0.57) 0.35 (0.22-0.58) 2.21 (1.46-3.33) 2.22 (1.39-3.56) Test set † OR 0.52 0.50 0.49 0.40 0.58 0.49 0.49 0.65 0.62 1.80 0.50 0.48 0.48 2.18 (95% CI) (0.34-0.79) (0.34-0.74) (0.34-0.72) (0.26-0.61) (0.40-0.85) (0.33-0.71) (0.33-0.71) (0.45-0.94) (0.44-0.87) (1.23-2.64) (0.33-0.76) (0.32-0.72) (0.33-0.71) (1.43-3.34) Abbreviations: 95% CI: 95% Confidence Interval, n.s.: not significant, OR: odds ratio. † Crude logistic regression analyses, by inclusion of one peak cluster (continuous), ‡ adjusted logistic regression analyses (training set only), by inclusion of one peak cluster (continuous), subjects’ age (categorical), and sample storage duration (categorical), * Bonferroni corrected p-values. Peptide purification and identification One of the 14 peak clusters found significantly different between breast cancer and control was m/z 8940, which we previously identified as complement component 3 precursor by peptide mapping (ProFound; estimated Z-score 1.57, 4% sequence coverage). Amino acid sequencing of 6 peptides in the tryptic digest by tandem MS on a Q-TOF identified the marker as C3a des-arginine anaphylatoxin (C3adesArg, 61% sequence coverage), a 76 amino acid protein with theoretical mass 8939.46 Da and pI 9.54. This identity was confirmed by an immunoassay, for which ProteinA beads were loaded with a C3a polyclonal antibody (Abcam Ltd, Cambridge, UK). Figure 2 depicts the correlation matrix presenting the (absolute) Pearson’s correlation coefficients calculated between the peak intensities of the 14 peaks found significantly different in expression between breast cancer and healthy control. To preclude bias by group, all Pearsons’ correlation analyses were performed in the healthy controls of the total study population. As 11 peak clusters were found highly correlated to each other (Pearson’s R > 0.63, Figure 2), we hypothesize these clusters to represent multiple fragments of one founder protein. Using data from previous publications, we suggest 91 Chapter 3.1 this founder protein to be inter-alpha-trypsin inhibitor heavy chain 4 (ITIH4). Eight of the alleged ITIH4 peak clusters had an observed mass corresponding to the theoretical mass of the different ITIH4 fragments described in the literature (Table 4). The peak clusters at m/z 4219 and 11745 were not correlated to any of the significantly different peak clusters. The m/z 4219 and m/z 5350 peak clusters, selected in the multivariable logistic regression analysis, were previously identified as (putative) fibrinogen fragments by our group. Table 3 Multivariate logistic regression analyses in the training set, by forward entry inclusion of all peak clusters detected, before and after adjustment for subjects’ age and sample storage duration. Multivariate model OR (95% CI) 1.94 (1.24-3.04) 0.26 (0.14-0.48) 0.62 (0.39-0.97) 0.53 (0.33-0.83) Variable m/z 4219 m/z 4309 m/z 5350 m/z 28183 p-value 0.004 < 0.001 0.035 0.006 Multivariate model, adjusted OR (95% CI) p-value 2.78 (1.59-4.86) < 0.001 0.26 (0.13-0.52) < 0.001 0.60 (0.36-1.01) 0.054 0.49 (0.29-0.85) 0.011 Performance ROC AUC Training set Test set 0.813 0.713 Sensitivity Specificity 74.3% 71.9% Sensitivity Specificity 72.6% 61.3% (0.742-0.884) (0.626-0.800) Training set Test set Abbreviations: AUC: area under the Receiver Operating Characteristic (ROC) curve, 95% CI: 95% Confidence Interval, OR: odds ratio, ROC: Receiver Operating Characteristics curve. Discussion In the current study, sera of breast cancer patients (n = 152) and healthy controls (n = 129) were analysed using the SELDI-TOF MS technology. Spectra were divided into a training and test set, and 14 peak clusters were found to differ significantly in peak expression between breast cancer and healthy control in the training set. Ten of these 14 peak clusters could also be validated in the test set. We previously identified one peak cluster as C3adesArg, while 12 other peak clusters were tentatively identified as ITIH4 fragments and a fibrinogen fragment, respectively. A classification model was subsequently generated by multivariate logistic regression analysis on the training set. Its performance on the test set was similar to those reported by previously performed independent validation studies (10;11;18;21). Hence, our split-sample approach yielded reliable estimates of performance. Nonetheless, the diagnostic performances reported thus far are only moderate. The identification of a general diagnostic biomarker is, however, seriously challenged by the molecular characteristics of breast cancer, which 92 Diagnostic serum protein profiles for breast cancer are highly heterogeneous (22-24). As such, selection of breast cancer subgroups for comparison with healthy controls is expected to improve results of future diagnostic SELDI-TOF MS studies. Table 4 Structural identities of eight alleged ITIH4 peak clusters that significantly differ in expression between breast cancer and healthy control in the training set. Mr (obs) (m/z) Mr (calc) (Da) Structural identity of putative putative ITIH4 fragment Ref. 2725.06 StartAmino acid sequence End 662-688 R.PGVLSSRQLGLPGPPDVPDHAAYHPF.R 2733 3166 (25-27) 3157.58 617-644 R.NVHSGSTFFKYYLQGAKIPKPEASFSPR.R (25;26;28) 3282 3273.72 658-688 R.MNFRPGVLSSRQLGLPGPPDVPDHAAYHPF.R (25-27) 3299 3289.72 658-688 R.MNFRPGVLSSRQLGLPGPPDVPDHAAYHPF.R * 3965 3957.46 654-690 A.AGSRMNFRPGVLSSRQLGLPGPPDVPDHAAYHPFRR.L (27;28) 3980 3973.46 654-690 A.AGSRMNFRPGVLSSRQLGLPGPPDVPDHAAYHPFRR.L * (27;28) 4292 4284.83 650-690 R.QAGAAGSRMNFRPGVLSSRQLGLPGPPDVPDHAAYHPFRR.L (28) 4309 4300.83 650-690 R.QAGAAGSRMNFRPGVLSSRQLGLPGPPDVPDHAAYHPFRR.L * (28) Abbreviations: ITIH4: inter-alpha-trypsin inhibitor heavy chain 4, Mr (obs): observed mass-to-charge ratio, Mr (calc): calculated mass from the matched peptide sequence. * Met-Ox fragment Complement C3adesArg We discovered the expression of the serum m/z 8940 C3adesArg peak to be significantly decreased in breast cancer compared to controls in both the training and test set. Complement C3 is the most abundant (1.2 mg/ml) complement protein in serum (29), supporting the activation of all three pathways of complement activation (the classic, alternative, and lectin pathway) (30;31). Produced mainly in the liver and adipocytes, C3a is formed by cleavage of C3 (185 kDa) by C3-convertases into C3b (176 kDa) and C3a (8.9 kDa) (32). The anaphylatoxin C3a is only short lived in serum as carboxypeptidases cleave the C-terminal arginine residue, creating the more stable, but biologically inactive C3adesArg (8.9 kDa) (31-33). As C3 is a positive acute phase reactant (34), elevated serum levels of C3 (and hence, C3a(desArg)) in cancer compared to control are anticipated. Indeed, elevated serum C3 levels have been described in various cancer types, including neuroblastoma (35), lung cancer (36), and cancer of the digestive tract (37). Likewise, increased serum C3adesArg levels, determined by SELDI-TOF MS, have been reported in breast- (9;10), hepatocellular- (38), and colorectal cancer (39;40), and chronic lymphoid malignancies (41). We, on the other hand, observed decreased C3adesArg levels in breast cancer in the current study population, as well as in a subset hereof, which we analysed for validation of the 8.9 kDa marker reported by Li et al. (21). Other studies have described decreased 8.9 kDa peak intensities in breast- (7;42), and lung cancer (43). Moreover, Li et al. observed decreased SELDI-TOF MS C3adesArg peak intensities in sera of metastatic breast 93 Chapter 3.1 cancer patients (10). Their finding is corroborated by the decreased serum C3 levels reported in patients with metastatic breast-, gastric-, and colorectal cancer (37) and brain tumours (35). Hence, complement activation seems an early event during tumourigenesis. This, however, can not explain the results of the current study, as we included only sera of patients with locally invasive breast cancer. An other possible explanation for the observed inconsistencies in 8.9 kDa C3adesArg regulation can be the in vitro complement activation, caused by coagulation induced platelet activation (44). Banks et al. (45) indeed reported the intensity of an IMAC3 m/z 8939 peak (not structurally identified though) to significantly increase with prolonged coagulation times. Coagulation time is, however, an unlikely confounder, as studies generally apply standardised collection protocols for both cancer and control samples. C3adesArg levels can also be affected by sample storage time. In a previous study, we found the m/z 8939 C3adesArg peak intensity positively correlated to sample storage time during the first three years of storage, after which intensities remained stable. Although in the current study, the breast cancer sera were stored for a slightly longer period than the control sera, storage time of both sample groups was less than three years. Moreover, as the m/z 8939 peak performance was not influenced by adjustment for sample storage duration, this parameter is unlikely to have confounded results of the current study. ITIH4 fragments Of the 14 peaks we found significantly different in expression between breast cancer and healthy controls, 11 were identified as putative ITIH4 fragments. The peak intensities of all putative ITIH4 fragments were decreased in breast cancer compared to control. ITIH4, a 120 kDa plasma glycoprotein expressed mainly in the liver, acts as a positive acute phase reactant and is extensively proteolytically processed (27). Plasma kallikrein readily cleaves ITIH4 into an N-terminal 85 kDa and C-terminal 35 kDa fragment, after which the 85 kDa fragment is further cleaved into an N-terminal 57 kDa and a putative 28 kDa fragment. The latter fragment has not been detected in its entirety hitherto, as it is rapidly cleaved into subsequent smaller fragments (27). Changes in the abundance of different fragments have been found associated with various types of cancer (e.g., prostate, breast, ovarian, colorectal and pancreatic cancer) (25-27), indicating cancer-type specific proteolytic processing of ITIH4. Three of the 11 putative ITIH4 fragments (i.e., m/z 2733, m/z 3282, and m/z 4292) have been reported as potential markers for breast cancer (27). Unlike our results, however, this study found increased peak intensities of the three fragments in cancer compared to control (27). 94 Diagnostic serum protein profiles for breast cancer Figure 2 Peak intensity correlation matrix for the 14 peaks found significantly different in expression between breast cancer and healthy control in the training set (for clarity, Pearsons´ correlation coefficients were converted into absolute values). 1 m/z 2733 m/z 3166 0.9 m/z 3282 0.8 m/z 3299 m/z 3691 0.7 m/z 3782 0.6 m/z 3965 0.5 m/z 3980 m/z 3997 0.4 m/z 4292 0.3 m/z 4309 m/z 8940 0.2 m/z 4219 0.1 m/z 11745 m/z 11745 m/z 4219 m/z 8940 m/z 4309 m/z 4292 m/z 3997 m/z 3980 m/z 3965 m/z 3782 m/z 3691 m/z 3299 m/z 3282 m/z 3166 m/z 2733 The m/z 4292 ITIH4 fragment has also been described by Li et al. (9;10). They initially observed a 4.3 kDa ITIH4 fragment to be downregulated in breast cancer (9), but found this peak upregulated upon validation (10). In their original discovery study, the cancer sera were collected during a (non-specified) longer time interval than the control sera, whereas in the validation study, sera of both cases and controls were collected within a two-year time interval. Combined with the postulated instability of the ITIH4 fragment (causing further truncation during prolonged storage), this could indeed explain their discrepant results. Nonetheless, following analysis of prospectively collected sera, Mathelin et al. (11) also observed a decreased expression of the m/z 4292 ITIH4 peak intensity in breast cancer. This decrease was also observed following analysis of a subset of the current study population for validation of the markers reported by Li et al. (21). However, the decrease of m/z 4292 observed in the breast cancer cases of the current study could not be explained by the difference in storage duration between the cancer and control sera, as correction for this parameter by logistic regression analyses did not affect the performance of the m/z 4292 peak. In addition, evidence for the alleged 4.3 kDa ITIH4 fragment instability is only limited. Peak intensities of this fragment were 95 Chapter 3.1 found both in- (28) and decreased by different (pre-) analytical parameters (25;27), though the fragmentation pattern was not altered (25;27). Perhaps the discrepant results of the various studies are caused by differences between the patient populations investigated in the various studies. Other markers Of the 14 peak clusters found significantly different in expression between breast cancer and healthy controls, both m/z 4219 and m/z 11745 were not correlated to any of the other peak clusters. While we previously identified m/z 4219 as a putative fibrinogen fragment, the identity of m/z 11745 peak is yet unknown. The m/z 5350 peak cluster, included in the logistic regression model, was identified earlier as a fibrinogen fragment as well (i.e., fibrinogen alpha-E fragment FGA576-625). The multivariate classification model furthermore designated the m/z 28183 peak cluster as a potential marker, in combination with m/z 4219, 4309, and 5350. Although peak intensities of both m/z 5350 and m/z 28183 were not significantly different in expression between breast cancer and healthy control, combination with other markers evidently improved their diagnostic performance. The m/z 5350 peak cluster, though not structurally identified, has been reported earlier as significantly increased in sera of patients with lung cancer (43) and hypopharyngeal squamous cell carcinoma (46). Based on the observed mass, we hypothesise the m/z 28183 peak cluster to represent apolipoprotein A-I. This protein was previously identified by our group as a potential marker for colorectal cancer by serum SELDI-TOF MS analyses (47). Synthesised both in the liver and small intestine, apolipoprotein A-I constitutes the major component of high-density lipoproteins (48). It is a negative acute phase reactant (49), explaining the decreased expression we observed in cancer vs. healthy control (Table 2, crude odds ratio < 1). Its decreased expression in cancer is confirmed by other studies investigating breast- (48), ovarian- (50), colorectal- (47), and hepatocellular cancer (51). Conclusion In conclusion, using SELDI-TOF MS, we discovered and validated 10 peak clusters that significantly differ in expression between sera of breast cancer patients and healthy controls. These peak clusters were structurally identified as the high abundant C3adesArg anaphylatoxin, and putative ITIH4 and fibrinogen fragments. Logistic regression analyses in the training set yielded a classification model with a performance comparable to those reported in previously performed independent validation studies. As these moderate performances most likely originate from the highly heterogeneous nature of breast cancer, selection of breast cancer subgroups for comparison with healthy controls is expected to improve results of future diagnostic SELDI-TOF MS studies. 96 Diagnostic serum protein profiles for breast cancer Acknowledgement This study was supported by a grant of the Dutch Cancer Society (project NKI 20053421). References (1) Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T et al. Cancer statistics, 2008. CA Cancer J Clin 2008; 58(2):71-96. (2) Ries L, Melbert D, Krapcho M, Stinchcomb D, Howlader N, Horner M et al. SEER Cancer Statistics Review, 1975-2005, National Cancer Institute. Bethesda, MD, http://seer.cancer.gov/csr/1975_2005/ , based on November 2007 SEER data submission, posted on the SEER website. 2008. (3) Stieber P, Molina R, Chan DW, Fritsche HA, Beyrau R, Bonfrer JM et al. Clinical evaluation of the Elecsys CA 15-3 test in breast cancer patients. Clin Lab 2003; 49(1-2):15-24. (4) Banks RE, Dunn MJ, Hochstrasser DF, Sanchez JC, Blackstock W, Pappin DJ et al. Proteomics: new perspectives, new biomedical opportunities. Lancet 2000; 356(9243):1749-1756. (5) Becker S, Cazares LH, Watson P, Lynch H, Semmes OJ, Drake RR et al. Surfaced-enhanced laser desorption/ionization time-of-flight (SELDI-TOF) differentiation of serum protein profiles of BRCA-1 and sporadic breast cancer. Ann Surg Oncol 2004; 11(10):907-914. (6) Belluco C, Petricoin EF, Mammano E, Facchiano F, Ross-Rucker S, Nitti D et al. Serum Proteomic Analysis Identifies a Highly Sensitive and Specific Discriminatory Pattern in Stage 1 Breast Cancer. Ann Surg Oncol 2007; 14(9):2470-2476. (7) Hu Y, Zhang S, Yu J, Liu J, Zheng S. SELDI-TOF-MS: the proteomics and bioinformatics approaches in the diagnosis of breast cancer. Breast 2005; 14(4):250-255. (8) Laronga C, Becker S, Watson P, Gregory B, Cazares L, Lynch H et al. SELDI-TOF serum profiling for prognostic and diagnostic classification of breast cancers. Dis Markers 2003; 19(4-5):229-238. (9) Li J, Zhang Z, Rosenzweig J, Wang YY, Chan DW. Proteomics and bioinformatics approaches for identification of serum biomarkers to detect breast cancer. Clin Chem 2002; 48(8):1296-1304. (10) Li J, Orlandi R, White CN, Rosenzweig J, Zhao J, Seregni E et al. Independent Validation of Candidate Breast Cancer Serum Biomarkers Identified by Mass Spectrometry. Clin Chem 2005; 51(12):2229-2235. (11) Mathelin C, Cromer A, Wendling C, Tomasetto C, Rio MC. Serum biomarkers for detection of breast cancers: a prospective study. Breast Cancer Res Treat 2005;1-8. (12) Vlahou A, Laronga C, Wilson L, Gregory B, Fournier K, McGaughey D et al. A novel approach toward development of a rapid blood test for breast cancer. Clin Breast Cancer 2003; 4(3):203-209. (13) Goncalves A, Esterni B, Bertucci F, Sauvan R, Chabannon C, Cubizolles M et al. Postoperative serum proteomic profiles may predict metastatic relapse in high-risk primary breast cancer patients receiving adjuvant chemotherapy. Oncogene 2006; 25(7):981-989. (14) Pusztai L, Gregory BW, Baggerly KA, Peng B, Koomen J, Kuerer HM et al. Pharmacoproteomic analysis of prechemotherapy and postchemotherapy plasma samples from patients receiving neoadjuvant or adjuvant chemotherapy for breast carcinoma. Cancer 2004; 100(9):1814-1822. (15) Heike Y, Hosokawa M, Osumi S, Fujii D, Aogi K, Takigawa N et al. Identification of serum proteins related to adverse effects induced by docetaxel infusion from protein expression profiles of serum using SELDI ProteinChip system. Anticancer Res 2005; 25(2B):1197-1203. 97 Chapter 3.1 (16) Ransohoff DF. Lessons from controversy: ovarian cancer screening and serum proteomics. J Natl Cancer Inst 2005; 97(4):315-319. (17) Villanueva J, Philip J, Chaparro CA, Li Y, Toledo-Crow R, DeNoyer L et al. Correcting common errors in identifying cancer-specific serum peptide signatures. J Proteome Res 2005; 4(4):1060-1072. (18) Gast MC, Bonfrer JM, van Dulken EJ, de Kock L, Rutgers EJ, Schellens JH et al. SELDI-TOF MS serum protein profiles in breast cancer: assessment of robustness and validity. Cancer Biomark 2006; 2(6):235248. (19) Hu J, Coombes KR, Morris JS, Baggerly KA. The importance of experimental design in proteomic mass spectrometry experiments: some cautionary tales. Brief Funct Genomic Proteomic 2005; 3(4):322-331. (20) Karsan A, Eigl BJ, Flibotte S, Gelmon K, Switzer P, Hassell P et al. Analytical and preanalytical biases in serum proteomic pattern analysis for breast cancer diagnosis. Clin Chem 2005; 51(8):1525-1528. (21) van Winden AWJ, Gast MCW, Beijnen JH, Rutgers EJ, Grobbee DE, Peeters PHM et al. Validation of previously identified serum biomarkers for breast cancer with SELDI-TOF MS: a case control study. BMC Medical Genomics 2008; Accepted for publication. (22) Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A 2001; 98(19):10869-10874. (23) Perou CM, Sorlie T, Eisen MB, van de RM, Jeffrey SS, Rees CA et al. Molecular portraits of human breast tumours. Nature 2000; 406(6797):747-752. (24) Bertucci F, Birnbaum D, Goncalves A. Proteomics of Breast Cancer: Principles and Potential Clinical Applications. Mol Cell Proteomics 2006; 5(10):1772-1786. (25) Fung ET, Yip TT, Lomas L, Wang Z, Yip C, Meng XY et al. Classification of cancer types by measuring variants of host response proteins using SELDI serum assays. Int J Cancer 2005; 115(5):783-789. (26) Villanueva J, Shaffer DR, Philip J, Chaparro CA, Erdjument-Bromage H, Olshen AB et al. Differential exoprotease activities confer tumor-specific serum peptidome patterns. J Clin Invest 2006; 116(1):271284. (27) Song J, Patel M, Rosenzweig CN, Chan-Li Y, Sokoll LJ, Fung ET et al. Quantification of fragments of human serum inter-alpha-trypsin inhibitor heavy chain 4 by a surface-enhanced laser desorption/ionization-based immunoassay. Clin Chem 2006; 52(6):1045-1053. (28) Timms JF, Arslan-Low E, Gentry-Maharaj A, Luo Z, T'Jampens D, Podust VN et al. Preanalytic influence of sample handling on SELDI-TOF serum protein profiles. Clin Chem 2007; 53(4):645-656. (29) Hugli TE. Human anaphylatoxin (C3a) from the third component of complement. Primary structure. J Biol Chem 1975; 250(21):8293-8301. (30) Bohana-Kashtan O, Ziporen L, Donin N, Kraus S, Fishelson Z. Cell signals transduced by complement. Mol Immunol 2004; 41(6-7):583-597. (31) Sahu A, Sunyer JO, Moore WT, Sarrias MR, Soulika AM, Lambris JD. Structure, functions, and evolution of the third complement component and viral molecular mimicry. Immunol Res 1998; 17(1-2):109-121. (32) de Bruijn MH, Fey GH. Human complement component C3: cDNA coding sequence and derived primary structure. Proc Natl Acad Sci U S A 1985; 82(3):708-712. (33) Nettesheim DG, Edalji RP, Mollison KW, Greer J, Zuiderweg ER. Secondary structure of complement component C3a anaphylatoxin in solution as determined by NMR spectroscopy: differences between crystal and solution conformations. Proc Natl Acad Sci U S A 1988; 85(14):5036-5040. (34) Gabay C, Kushner I. Acute-phase proteins and other systemic responses to inflammation. N Engl J Med 1999; 340(6):448-454. (35) Carli M, Bucolo C, Pannunzio MT, Ongaro G, Businaro R, Revoltella R. Fluctuation of serum complement levels in children with neuroblastoma. Cancer 1979; 43(6):2399-2404. 98 Diagnostic serum protein profiles for breast cancer (36) Gminski J, Mykala-Ciesla J, Machalski M, Drozdz M, Najda J. Immunoglobulins and complement components levels in patients with lung cancer. Rom J Intern Med 1992; 30(1):39-44. (37) Maness PF, Orengo A. Serum complement levels in patients with digestive tract carcinomas and other neoplastic diseases. Oncology 1977; 34(2):87-89. (38) Lee IN, Chen CH, Sheu JC, Lee HS, Huang GT, Chen DS et al. Identification of complement C3a as a candidate biomarker in human chronic hepatitis C and HCV-related hepatocellular carcinoma using a proteomics approach. Proteomics 2006; 6(9):2865-2873. (39) Habermann JK, Roblick UJ, Luke BT, Prieto DA, Finlay WJ, Podust VN et al. Increased serum levels of complement C3a anaphylatoxin indicate the presence of colorectal tumors. Gastroenterology 2006; 131(4):1020-1029. (40) Ward DG, Suggett N, Cheng Y, Wei W, Johnson H, Billingham LJ et al. Identification of serum biomarkers for colon cancer by proteomic analysis. Br J Cancer 2006; 94(12):1898-1905. (41) Miguet L, Bogumil R, Decloquement P, Herbrecht R, Potier N, Mauvieux L et al. Discovery and identification of potential biomarkers in a prospective study of chronic lymphoid malignancies using SELDI-TOF-MS. J Proteome Res 2006; 5(9):2258-2269. (42) Shin S, Cazares L, Schneider H, Mitchell S, Laronga C, Semmes OJ et al. Serum biomarkers to differentiate benign and malignant mammographic lesions. J Am Coll Surg 2007; 204(5):1065-1071. (43) Han KQ, Huang G, Gao CF, Wang XL, Ma B, Sun LQ et al. Identification of lung cancer patients by serum protein profiling using surface-enhanced laser desorption/ionization time-of-flight mass spectrometry. Am J Clin Oncol 2008; 31(2):133-139. (44) Hamad OA, Ekdahl K, Lambris JD, Nilsson B. Complement activation triggered by thrombin receptoractivated platelets. Mol.Immunol. 44, 180. 2007. (45) Banks RE, Stanley AJ, Cairns DA, Barrett JH, Clarke P, Thompson D et al. Influences of blood sample processing on low-molecular-weight proteome identified by surface-enhanced laser desorption/ionization mass spectrometry. Clin Chem 2005; 51(9):1637-1649. (46) Zhou L, Cheng L, Tao L, Jia X, Lu Y, Liao P. Detection of hypopharyngeal squamous cell carcinoma using serum proteomics. Acta Otolaryngol 2006; 126(8):853-860. (47) Engwegen JY, Helgason HH, Cats A, Harris N, Bonfrer JM, Schellens JH et al. Identification of serum proteins discriminating colorectal cancer patients and healthy controls using surface-enhanced laser desorption ionisation-time of flight mass spectrometry. World J Gastroenterol 2006; 12(10):1536-1544. (48) Chang SJ, Hou MF, Tsai SM, Wu SH, Hou LA, Ma H et al. The association between lipid profiles and breast cancer among Taiwanese women. Clin Chem Lab Med 2007; 45(9):1219-1223. (49) Van Lenten BJ, Reddy ST, Navab M, Fogelman AM. Understanding changes in high density lipoproteins during the acute phase response. Arterioscler Thromb Vasc Biol 2006; 26(8):1687-1688. (50) Zhang Z, Bast RC, Jr., Yu Y, Li J, Sokoll LJ, Rai AJ et al. Three biomarkers identified from serum proteomic analysis for the detection of early stage ovarian cancer. Cancer Res 2004; 64(16):5882-5890. (51) Steel LF, Shumpert D, Trotter M, Seeholzer SH, Evans AA, London WT et al. A strategy for the comparative analysis of serum proteomes for the discovery of biomarkers for hepatocellular carcinoma. Proteomics 2003; 3(5):601-609. 99 Chapter SELDI-TOF MS serum protein profiles in breast cancer: assessment of robustness and validity Marie-Christine W. Gast Johannes M.G.Bonfrer Eric J. van Dulken Lieve de Kock Emiel J. Th. Rutgers Jan H.M. Schellens Jos H. Beijnen Cancer Biomarkers 2006;2(6):235-48 3.2 Chapter 3.2 Abstract There is an urgent need for new serum markers that can be applied in the early detection of breast cancer. Following detection of new, potential biomarkers, such as those reported by Vlahou et al. (Clin Breast Cancer 2003;4:230-9) and Laronga et al. (Dis Markers 2003;19:229-38), assessment of both their robustness and validity is essential to confirm their clinical applicability. We therefore aimed to determine robustness and validity of biomarkers reported by the authors mentioned, by analysis of an independent sample set (breast cancer: n = 47, normal women: n = 48) in our laboratory, according to the methods described by both authors. Although all markers for the differentiation between breast cancer patients and normal women, discovered in the study of Vlahou et al., were recovered in our validation data set, none had sufficient performance to be applied as a classifier. The markers discovered by Laronga et al. in the differentiation between lymph node positive and -negative breast cancer patients were in part recovered from our validation data set, but were also not applicable as a classifier. In conclusion, although (part of) the proteins discovered and designated as markers by either author could be detected, their validity as biomarkers could not be confirmed by the current study. This finding stresses that, when reporting on a potential biomarker, confirmation of both robustness and validity is essential in obtaining its true clinical applicability. 102 Assessment of robustness and validity Introduction Following the introduction of the proteomic surface-enhanced laser desorption/ionisation time-of-flight mass spectrometry (SELDI-TOF MS) platform, many efforts have been made in the search for new biomarkers applicable in a.o. cancer diagnosis. This has resulted in several publications, reporting the analysis of SELDI-TOF MS protein profiles by sophisticated bioinformatics techniques, yielding potential biomarkers with a high sensitivity and specificity in the detection of different types of cancer, e.g., prostate, ovarian, lung and breast cancer (1-5). Despite these promising results, this technology faces some limitations. Of particular concern has been the reproducibility of the SELDI-based approach. This is illustrated by the comment that for the same cancer type, different biomarkers have been identified by different research groups (6). It may, however, be very unlikely to obtain identical protein profiles when studies of the same cancer type use different study populations, different methods to collect the data (e.g., different ProteinChip Array types, sample handling and assay conditions), and different bioinformatics methods to analyse the data (7). Still, standardisation of all these variables does not guarantee detection of reproducible protein profiles. Discriminative protein profiles could well be explained by chance, due to overfitting of the data. This can easily occur when large numbers of possible predictors are fitted by a multivariable model, as is often the case in proteomic data analysis (8). To confirm the robustness of biomarker protein profiles, and to preclude the possibility of being caused by chance, identification of these profiles across different sample sets and different laboratories is imperative (8). Furthermore, structural identification of the proteins or peptides that form part of discriminative protein profiles is essential in undermining problems with robustness. Results should be validated by analysis of an independent, but similar sample set, that is handled like the sample set in which the biomarkers were discovered. Although validation is essential in obtaining the true clinical applicability of the biomarker, very little attention has been paid to this issue thus far. To date, only biomarkers for ovarian and breast cancer have been validated by the analysis of samples from different institutes (3;9). A second, more elaborate validation study for prostate cancer is currently still ongoing (10;11). In breast cancer, eight studies in which the SELDI-TOF MS platform was applied in the identification of serum markers for diagnosis, prognosis, or monitoring of therapy efficacy or toxicity, have been published until now (5;12-18). Thus far, only one panel of biomarkers discovered by SELDI-TOF MS in breast cancer has been validated yet by both intra- (9) and inter-laboratory (19) analysis of two independent sample sets. The two validation studies, however, reported only partial replication of prior results. Thus, detection of reproducible serum protein profiles in different sample sets and at different laboratories has not been achieved yet. We therefore aimed to assess robustness and validity of two high-performance breast cancer protein profiles published earlier by 103 Chapter 3.2 Vlahou et al. (17) and by Laronga et al. (15), through analysis of our own sample set in our laboratory, using the procedures as described by both authors. Methods Patients Serum samples of 47 female patients with pathologically confirmed primary breast cancer, who had not received any prior treatment at time of sample withdrawal, were included in cohort A of this study. The control samples included in this cohort were randomly collected from 48 female normal volunteers. All sera in cohort A were stored at -20°C. Furthermore, serum samples of 19 female patients with pathologically confirmed primary breast cancer who had not received any prior treatment at time of sample withdrawal were included in cohort B of this study. All sera in this cohort were stored at -80°C, and serum aliquots of 7 patients were simultaneously stored at -20°C. All sera included in this study were obtained after signing an Institutional review board approved Informed Consent. Withdrawal, processing and storage of all serum samples were performed under strictly defined conditions at the Department of Clinical Chemistry of the Institute. Study participant and sample characteristics of cohort A and B are, along with study data of Vlahou et al. (17) and Laronga et al. (15), provided in Table 1 and Table 2. Serum protein profiling Serum protein profiling was performed manually on immobilized metal affinity capture (IMAC30) and strong anion exchange (Q10) arrays, as described by Adam et al. (1), and Vlahou et al. (17), respectively. Both array types consist of an identical chromatography as the array types used in the studies of Vlahou et al. (17) and Laronga et al. (15) (IMAC and SAX arrays, respectively). Samples were randomized for processing. In brief, sample pretreatment consisted of mixing 20 μl serum with 30 μl of 8 M urea / 1% CHAPS in PBS pH 7.4 for 10 min at 4°C, followed by the addition of 100 μl 1 M urea / 0.125% CHAPS. Next, 600 μl of binding buffer (PBS pH 7.4 for the IMAC30 assay, and 20 mM HEPES / 0.1% TritonX-100 for the Q10 assay) was added, and samples were placed on ice. Arrays were assembled into a bioprocessor, a device that holds up to twelve 8-spot arrays and allows for the addition of larger volumes, and was shaken on a platform shaker at 250 rpm throughout the assay. IMAC30 arrays were activated by the application of 20 μl 100 mM CuSO4 for 5 min, followed by 10 rinses with double distilled water. Next, 20 μl 100 mM SodiumAcetate was added for 5 min, again followed by 10 rinses with double distilled water. Both IMAC30 and Q10 arrays were subsequently equilibrated twice for 5 min with 200 μl of their respective binding buffer. Next, pretreated serum samples (50 μl) were randomly applied to the arrays. After a 30 min incubation, arrays were washed three times for 5 min with binding buffer and 104 Assessment of robustness and validity three times with double distilled water. Following air drying of the arrays, sinapinic acid was applied twice to each spot. Since both authors do not clearly specify either EAM composition or volume added per spot, we applied 1μl of a 50% solution of sinapinic acid in 50% ACN / 0.5% TFA to each spot twice, according to manufacturers instructions. Following air-drying, the array was inserted in the PBS IIc ProteinChip Reader (Ciphergen Biosystems Inc., Freemont, CA, USA). Since both studies were performed with this instruments’ predecessor, reported acquisition parameters could only partially be applied. Both laser intensity and detector sensitivity were optimised for the detection of the reported biomarkers. Time-Of-Flight mass spectra were generated for IMAC30 and Q10 arrays by averaging 192 laser shots with intensity 150 and 157, respectively (arbitrary units), and detector sensitivity 6 and 5, respectively (arbitrary units). Spectra were generated with different focus lag times. For IMAC30 arrays, a lag time of 900 ns was maintained, as specified by Adam et al. (1) For Q10 arrays, focus lag time was not specified; following optimisation, we maintained a lag time of 528 ns. For mass accuracy, the instrument was calibrated on the day of measurements using a peptide molecular mass standard (Ciphergen Inc.), containing [Arg8] vasopressin (1084.3 Da), somatostatin (1637.9 Da), dynorphin (2147.5 Da), ACTH (2933.5 Da), insulin β-chain (bovine) (3495.9 Da), insulin (human recombinant) (5807.7 Da) and hirudin (7033.6 Da). Statistics and Bioinformatics All spectra were processed by the ProteinChip Software v3.1 (Ciphergen Inc.). Spectra were compiled in one file, baseline subtracted and normalised for total ion current from 1000 Da (Laronga et al. (15)) or 1500 Da (Vlahou et al.(17)) to the spectrum’s end. Spectra with normalisation factors > 2 or < 0.5 were excluded from further analysis. Next, the Biomarker Wizard (BMW), an application within the ProteinChip Software, was applied for peak detection and clustering. First, peaks detected by Vlahou et al. (17) and Laronga et al. (15) were manually searched for and labelled at their centroid, irrespective of signal-to-noise (S/N) ratio. Second, to investigate whether the spectra contained other discriminative protein peaks, automatic peak detection was performed along the entire spectrum. Software settings applied herein were as specified by Vlahou et al.(17) and Laronga et al. (15). For Vlahou et al.(17), peaks with a S/N ratio > 3, occurring in at least 10% of all spectra were clustered initially, in both IMAC30 and Q10 spectra. Clusters were completed by peaks with S/N > 1.5 in a cluster mass window of 0.3%. For Laronga et al. (15), peaks occurring in at least 5% of all spectra, with S/N > 3, were clustered. Peak clusters were completed by peaks with S/N > 2 in a mass window of 0.2% and 0.3% for IMAC30 and Q10 data, respectively. Following peak detection and clustering, average peak intensities for both groups (i.e., breast cancer vs. normal, and lymph node positive vs. -negative) were calculated. Next, peak expression differences between spectra of both groups were calculated by the Biomarker Wizard, using the non-parametric Mann-Whitney-U test. P values < 0.05 were considered 105 Chapter 3.2 statistically significant. Peak information was subsequently exported as spreadsheet files, and data of both array types were merged in one file. Files were analysed for pattern recognition and sample classification by the Biomarker Patterns Software v.5.0.1 (BPS; Ciphergen Inc.). To investigate the effect of storage time on the expression of the reported markers, peak intensities of spectra in cohort A were plotted against their respective sample storage times. Plots were visually inspected for random distribution of peak intensity across sample age. All statistical analyses, other than those executed by the BMW or BPS, were performed by using SPSS statistical software, v.11.0.1 (SPSS Inc., Chicago, IL, USA). For all analyses, a 2-tailed P value < 0.05 was considered significant. Results Patientss Patient As is depicted by the data presented in Table 1, only minor differences existed in participant and sample characteristics of both studies. Breast cancer patients in cohort A, however, differed in stage of disease. While the study participants of Vlahou et al. (17) had Stage I to IV disease or DCIS, cohort A represented only Stage II and III disease. The median age of breast cancer patients and normal women differed slightly between both samples sets, but the range of participants’ age was similar. Moreover, all sera were withdrawn prior to therapy. Sera of both breast cancer patients and normal women in cohort A were sampled in the same time period. The samples in cohort A were stored at a different temperature (-20°C) than the samples analysed by Vlahou et al. (17) (-80°C). Table 1 Characteristics of study participants (breast cancer vs. normal women) and study samples, in comparison with those reported by Vlahou et al. (17). Characteristic N Normal Cohort A 48 Normal Vlahou et al. 47 Cancer Cohort A 47 Cancer Cohort B 19 Cancer Vlahou et al. 45 Median age [range] (years) 51.0 [21-71] 46.5 [21-78] 61.4 [34-86] 60.5 [27-80] 59.3 [31-91] Stage* n.a. n.a. DCIS 1 2A 2B 3A 4 Sampling period 25 15 7 Apr-’03 Jan-‘05 Dec-’01 May-‘02 (53%) (32%) (15%) Aug-’03 Dec-‘04 5 11 3 (26%) (58%) (16%) Apr-’05 Jan-‘06 8 14 14 (18%) (31%) (31%) 6 3 (13%) (7%) Dec-’01 May-‘02 Storage temp -20°C -80°C -20°C -20°C / -80°C -80°C Abbreviations: DCIS: ductal carcinoma in situ, n.a.: not applicable, temp: temperature. * Pathologically determined stage. 106 Assessment of robustness and validity The sample set of Laronga et al. (15) contained significantly more participants in comparison with cohort A of the current sample set (Table 2). Although the median age of lymph node positives and -negatives differed between both sample sets, participants age was not significantly different between lymph node positives and -negatives in cohort A (independent samples T-test; p = 0.06). Both lymph node positive and negative breast cancer patients were sampled in the same time period. The samples in cohort A were stored at a different temperature (-20°C) than the samples analysed by Laronga et al. (15) (-80°C). Table 2 Characteristics of study participants (lymph node positive vs. lymph node negative) and study samples, in comparison with those reported by Laronga et al. (15). Characteristic N LN neg Cohort A 11 LN neg Cohort B 10 LN neg Laronga 71 LN pos Cohort A 36 LN pos Cohort B 9 LN pos Laronga 27 Mean age [range] (yrs) 68.4 [43-86] 57.6 [40-71] 56 60.0 [34-86] 62.8 [27-80] 58 Diagnosis* IDC ILC 11 9 1 70 1 30 6 8 1 26 1 Sampling period Oct-‘03 Dec-‘04 Apr-‘05 Jan-‘06 Dec-‘01 May-‘02 Aug-‘03 Dec-‘04 May-‘05 Sept-‘05 Dec-‘01 May-‘02 Storage temp -20°C -20°C / -80°C -20°C -20°C / -80°C -80°C -80°C Abbreviations: IDC: invasive ductal carcinoma, ILC: invasive lobular carcinoma, LN neg: lymph node negative, LN pos: lymph node positive, temp: temperature. * Pathologically determined diagnosis. Serum protein profiling Differentiation of breast cancer and normal women; Q10 data Following normalisation of the Q10 data, the spectra of two breast cancer patients and three normal women were excluded due to a normalisation factor > 2, leaving 45 spectra of breast cancer patients and 45 spectra of normal women for further analysis. In search for the peaks identified by Vlahou et al. (17) (2.95 kDa, 3.68 kDa, and 4.27 kDa), we manually identified three peak clusters at m/z 2935, m/z 3689 and m/z 4295 on this array type. The mean expression of these peaks was, however, not significantly different between breast cancer patients and normal women (Mann-Whitney U test; p > 0.5). Moreover, whereas the 3.68 kDa marker was downregulated in breast cancer in the sample set of Vlahou et al. (17), this marker was found upregulated in breast cancer in the current sample set, and vice versa for the 4.27 kDa marker (Table 3). With the data of these three peak clusters solely, the Biomarker Patterns Software constructed a decision tree with a cross validated sensitivity and specificity of 73.3% and. 24.4%, respectively (Figure 1A). Over the entire length of spectra, the Biomarker Wizard detected a total of 145 peak clusters. Of 10 peak clusters, mean peak intensities were significantly different between 107 Chapter 3.2 breast cancer patients and normal women (data not shown). Using all peak cluster data, the Biomarker Patterns Software constructed an optimal decision tree consisting of three nodes (m/z 6307, m/z 5073 and m/z 8238), with a cross validated sensitivity and specificity of 51.1% and 57.8%, respectively (Table 4). Table 3 Summary of the markers identified by Vlahou et al. (17) and their presence and performance in the validation sample set of the current study. Vlahou et al. Array Peak (kDa) Intensity cut off ↑ or ↓ in BC Current study Array Peak (m/z) SAX 2.95 3.68 4.27 ≤ 0.841 ≤ 0.217 ≤ 2.699 Down Down Up Q10 2935 3689 4295 Ave BC peak intensity 2.309 2.950 1.744 Ave N peak intensity 2.221 2.826 1.771 ↑ or ↓ in N IMAC 2.95 3.94 3.97 ≤ 0.751 ≤ 1.192 ≤ 8.901 Down Up Down IMAC30 2961 3964 3979 1.416 25.787 16.161 1.301 29.548 17.343 Up Down Down SAX 4.03 ≤ 0.658 Down Q10 4022 2.912 2.782 Up Down Up Down Abbreviations: BC: breast cancer, N: normal. Differentiation of breast cancer and normal women; Q10 & IMAC30 data Upon normalisation, all IMAC30 spectra had a normalisation factor between 0.5 and 2, and thus, none were excluded from further analysis. We manually identified all discriminative peaks discovered by Vlahou et al. (17) (2.95 kDa, 3.94 kDa, and 3.97 kDa) at m/z 2961, m/z 3964, and m/z 3979. Peak intensity data were subsequently generated by the Biomarker Wizard. In our dataset, none of the three peaks was significantly different in mean peak expression between breast cancer patients and normal women (Mann-Whitney U-test; p > 0.09). Moreover, except for the 3.97 kDa peak, the regulation of protein expression in breast cancer patients in the current dataset was opposite to that observed in the dataset of Vlahou et al. (17) (Table 3). The discriminative 4.03 kDa peak discovered on the SAX array type by Vlahou et al. (17) was manually detected at m/z 4022 in the current Q10 dataset. Intensity data for this peak were generated by the Biomarker Wizard. In our dataset, mean m/z 4022 peak intensity did not differ significantly between spectra of breast cancer patients and normal women (Mann-Whitney U test; p > 0.5). Moreover, in the current dataset, the peak at m/z 4022 was upregulated in breast cancer, opposite to the downregulation observed in the dataset of Vlahou et al. (17) (Table 3). Peak intensity data of the IMAC30 clusters (m/z 2961, m/z 3964, and m/z 3979) and the Q10 cluster (m/z 4022) were merged in one file, and subsequently analysed by the Biomarker Patterns Software. The optimum decision tree consisted of 3 nodes (Figure 1B), with a cross validated sensitivity and specificity of 51.1% and. 44.4%, respectively. Using the software settings applied by Vlahou et al. (17), the Biomarker Wizard detected a total number of 82 peaks in the entire length of IMAC30 spectra, 8 of which 108 Assessment of robustness and validity significantly differed in peak expression between spectra of breast cancer patients and normal women (data not shown). Following combination of peak intensity data from both the Q10 and IMAC30 array type in one file, the Biomarker Patterns Software constructed an optimal decision tree consisting of 6 decision nodes, each containing a different peak. This tree had a sensitivity and specificity of 48.9% and 57.8%, respectively, as determined by cross validation (Table 4). Figure 1 Decision trees for the classification of breast cancer (BC) and normal women (N). A. based on Q10 data solely, and B. based on both Q10 data and IMAC30 data. The decision trees were constructed on the three (A) and four (B) discriminative protein peaks identified by Vlahou et al. (17), as manually detected in cohort A of the current dataset. Spectra that follow the decision rules depicted in each node will proceed to the left descendant node and vice versa. 1B 1A m/z 2935 ≤ 0.607 BC: n = 45 N: n = 45 Cancer BC: n = 4 N: n = 0 m/z 3964 ≤ 29.589 BC: n = 45 N: n = 45 m/z 3689 ≤ 1.857 BC: n = 41 N: n = 45 Normal BC: n = 1 N: n = 10 Cancer BC: n = 40 N: n = 35 m/z 3964 ≤ 7.595 BC: n = 25 N: n = 15 Normal BC: n = 2 N: n = 5 Cancer BC: n = 23 N: n = 10 m/z 4022 ≤ 3.447 BC: n = 20 N: n = 30 Normal BC: n = 14 N: n = 28 Cancer BC: n = 6 N: n = 2 Differentiation of lymph node positive and -negative breast cancer patients; Q10 & IMAC30 data Following normalisation, all IMAC30 spectra had a normalisation factor between 0.5 and 2. Thus, none were excluded from further analysis. A total of 114 clusters were detected on the IMAC30 array type, 7 of which differed significantly in peak expression between lymph node positive and negative breast cancer patients (data not shown). All three discriminating peaks discovered by Laronga et al. (15) on the IMAC surface (1437, 1349, and 1003 Da) were recovered in the current IMAC30 dataset. Mean intensities of none of these peaks was significantly different between lymph node positive and negative breast cancer patients in the current dataset (Mann-Whitney U test; p > 0.1) (Table 5). Following normalisation of the Q10 data, 3 spectra (2 from lymph node positive-, 1 from lymph node negative breast cancer patients) had a normalisation factor > 2. These spectra were excluded from further analysis. In total, 18 peak clusters were recovered from the current dataset, with one peak cluster (m/z 43,462) having a significantly mean expression difference between lymph node positive and -negative breast cancer 109 Chapter 3.2 patients (data not shown). None of the discriminating peaks discovered by Laronga et al. (15) were, however, detected in the current dataset. Table 4 Comparison of decision trees constructed in the study of Vlahou et al. (17), Laronga et al. (15), and in the current study. Fig - Study Vlahou Distinction Distinction BC vs. N Array SAX Inclusion of All BMW detected peaks Classifiers 4.27 kDa 3.68 kDa 2.95 kDa Cut off ≤ 2.699 ≤ 0.217 ≤ 0.841 Sens 82.2 80.0 Spec 85.1 78.7 1A Current BC vs. N Q10 Manually detected peaks m/z 2935 m/z 3689 ≤ 0.607 ≤ 1.857 97.8 73.3 22.2 24.4 - Current BC vs. N Q10 All BMW detected peaks m/z 6307 m/z 5073 m/z 8238 ≤ 1.866 ≤ 2.544 ≤ 1.493 71.1 51.1 88.9 57.8 - Vlahou BC vs. N SAX & IMAC All BMW detected peaks 3.94 kDa 3.97 kDa 4.03 kDa 2.95 kDa ≤ 1.192 ≤ 8.901 ≤ 0.658 ≤ 0.751 90.0 90.0 96.7 93.3 1B Current BC vs. N Q10 & IMAC30 Manually detected peaks m/z 3964 m/z 4022 m/z 3964 ≤ 29.59 ≤ 3.447 ≤ 7.595 64.4 51.1 73.3 44.4 - Current BC vs. N Q10 & IMAC30 All BMW detected peaks m/z 6307 m/z 5073 m/z 23425 m/z 8238 m/z 53987 m/z 18743 ≤ 1.866 ≤ 2.544 ≤ 0.207 ≤ 1.493 ≤ 0.064 ≤ 0.321 95.6 48.9 88.9 57.8 - Laronga LN pos vs. LN neg SAX & IMAC All BMW detected peaks 74144 Da 59065 Da 40277 Da 1437 Da 100 81.0 87.3 77.0 1003 Da ≤ 0.014 ≤ 0.010 ≤ 1.444 ≤ 0.446 / 0.537 ≤ 1.367 / 2.014 ≤ 16.90 m/z 1276 m/z 96409 ≤ 1.249 ≤ 0.018 70.6 50.0 100 30.0 1349 Da - Current LN pos vs. LN neg Q10 & IMAC30 All BMW detected peaks Abbreviations: BC: breast cancer, BMW: Biomarker Wizard software, LN neg: lymph node negative, LN pos: lymph node positive, N: normal, Sens: sensitivity (%), Spec: specificity (%), both obtained by the learning dataset (in regular font) and by cross validation (in bold font). Peak intensity data of the IMAC30 and Q10 array types were merged in one file and submitted for analysis by the Biomarker Patterns Software. None of the trees constructed had a satisfactory performance, since the cross validated sensitivity and specificity of the optimum tree did not exceed 50% (Table 4). 110 Assessment of robustness and validity Table 5 Summary of the markers identified by Laronga et al. (15) and their presence and performance in the validation sample set of the present study. Laronga et al. Array Peak (kDa) Intensity cut off ↑ or ↓ in LN+ Current study Array Peak (m/z) SAX 74144 59065 40277 ≤ 0.014 ≤ 0.010 ≤ 1.444 Up Down Up Q10 n.d. n.d. n.d. Ave LNpeak intensity - Ave LN+ peak intensity - ↑ or ↓ in LN+ IMAC 1437 1437 1349 1349 1003 ≤ 0.446 ≤ 0.537 ≤ 1.367 ≤ 2.014 ≤ 16.90 Down Down Down Up Down IMAC30 1455 1.356 1.135 Down 1352 0.717 0.421 Down 1004 14.354 12.499 Down - Abbreviations: LN-: lymph node negative, LN+: lymph node positive, n.d.: not detected. Influence of sample storage temperature and -time on biomarker expression All discriminating peaks reported by Vlahou et al. (17) on both the IMAC and SAX surface were recovered from the spectra in our -80°C / -20°C dataset. Of markers discovered by Laronga et al. (15), only those reported on the IMAC surface were recovered from our -80°C and -20°C spectra. Peak intensities of recovered markers were not significantly different between samples stored at -80°C and -20°C (Mann-Whitney U test; p > 0.05), except for two peaks discovered by Vlahou et al. (17) and recovered on the Q10 surface (Mann-Whitney U test; m/z 4022, p = 0.026; m/z 4295, p = 0.040). However, 95% confidence intervals (CI) of mean intensities of these two peaks overlapped between -20°C and -80°C spectra (mean (95% CI); m/z 4022 (-20°C): 3.95 (3.37-4.53); m/z 4022 (-80°C): 4.64 (4.29-5.00); m/z 4295 (-20°C): 2.45 (2.26-2.65); m/z 4295 (-80°C): 2.80 (2.62-2.97)). Peak intensity distribution of m/z 4022 and m/z 4295 in -80°C and -20°C spectra are presented in Figure 2, along with the intensities of three other representative peaks (m/z 3689 (Q10; Vlahou et al. (17)), m/z 2961 (IMAC30; Vlahou et al. (17)), and m/z 1455 (IMAC30; Laronga et al. (15)). Of the peaks reported by Laronga et al. (15), and recovered in our -80°C dataset, none were significantly different in mean peak intensities between lymph node positive and negative breast cancer sera (Mann-Whitney U test; p > 0.05). The intensities of all recovered peaks in the current dataset were plotted against their respective sample storage times. Visual inspection of these plots revealed a random distribution of peak intensities across sample age. Figure 3 presents three plots, representative for all peaks recovered in cohort A. 111 Chapter 3.2 Figure 2 Peak intensity distributions of the peaks at m/z 3689, m/z 4022, m/z 4295 (Q10), m/z 2961 (IMAC30) (all four reported by Vlahou et al. (17)), and m/z 1455 (IMAC30; reported by Laronga et al. (15)) (LN-: lymph node negative, LN+: lymph node positive) in cohort B of the current dataset. 7 storage temp peak intensity 6 -20°C -80°C 5 4 3 2 1 0 LN- LN- LN+ m/z 3689 m/z 4022 m/z 4295 m/z 2961 m/z 1455 Discussion Differentiation of breast cancer and normal Breast cancer was the most commonly diagnosed cancer among women in the USA in 2004. (20) While early detection of breast cancer can lead to improved clinical outcomes (21;22), 34% of breast cancer patients in the USA are diagnosed in a late stage (20). Currently used serum tumour markers, such as CA15.3, lack adequate sensitivity (23%) and specificity (69%) to be applicable in cancer detection (23), and are therefore recommended only for use as markers for monitoring therapy or recurrence (24). Even with mammography, being the most widely applied imaging test today, approximately 20% of breast cancers will remain undetected (25). Therefore, new, robust and valid serum biomarkers that can be applied in the (early) detection of breast cancer are urgently needed. In search for these biomarkers, Vlahou et al. (17) reported the application of a number of peaks, detected by SELDI-TOF MS, in the differentiation between sera obtained from breast cancer patients and normal women. Although all reported classifiers were recovered from the current dataset, identified peak mass-to-charge ratio’s differed slightly between the datasets of Vlahou and Laronga and the current dataset. Mass shifts between spectra can have its origin in the low resolution of the SELDI-(linear)TOF MS (26), in its mass accuracy of 0.1%, or in the different calibrations that were applied to both datasets (10;27). Finally, since none of the peaks discovered were structurally identified, it cannot be excluded that corresponding peak masses between both studies 112 Assessment of robustness and validity possibly represent different proteins / peptides, or result from post-translational modifications or slight differences in sample storage and processing, thus explaining the observed mass shift. Figure 3 Sample age vs. intensity of the peaks at A. m/z 3689 (Q10), B. m/z 2961 (IMAC30) (both reported by Vlahou et al.(17)), and C. m/z 1455 (IMAC30; reported by Laronga et al. (15)) in cohort A of the current dataset. m/z 3689 7 BC N 6 Peak intensity m/z 2961 group 5 4 3 2 4 m/z 1455 group 4 group BC N 3 LNLN+ 3 2 2 1 1 0 0 1 0 200 400 600 800 200 400 600 800 200 400 600 800 Sample age (days) The promising recovery of classifiers in our own dataset did, however, not develop further into a satisfactory discrimination between breast cancer and normal by these classifiers. Although SELDI-TOF MS peak intensity data are known to be affected by the different sample collection methods and instrument settings applied in these studies (28;29), peak expression in both datasets was not only different, but even reversed for a number of classifiers (i.e., up- vs. downregulated in breast cancer patients). Moreover, mean peak intensities of the classifiers recovered in our dataset were not significantly different between both groups. An exact parallel between the peak expression differences in both studies is, however, difficult to draw. Peak expression differences in the total study population (as determined in the current study) do not necessarily correspond to peak expression differences observed in a subgroup of the total population (as deduced from the decision trees reported by Vlahou et al. (17)). Still, peak expression differences of the first markers applied in reported decision trees (4.27 kDa and 3.94 kDa), for which aforesaid is not relevant, was reversed between both datasets. As a result, only decision trees with suboptimal performance could be constructed, and validity of classifiers could not be ascertained by our own dataset. Differentiation of lymph node positive and -negative The study performed by Laronga et al. (15) had multiple aims, one of which was the identification of serum biomarkers specific for lymph node involvement in breast cancer patients. Currently, lymph node status is determined by sentinel lymph node biopsy. Although false negative rates of 33% have been reported, sentinel lymph node 113 Chapter 3.2 detection has an overall accuracy of ≥ 96% (30). The procedure has, however, a highly operator dependent character, and in the event of multicentric tumours and lymph nodes with high tumour burden, accuracy can be limited (31). Thus, serum biomarkers for lymph node status, provided their performance is satisfactory, can reduce morbidity associated with biopsy and aid in determining whether dissection of axillary lymph nodes is required. In search for these markers, Laronga et al. (15) reported an eight-node decision tree, constructed out of six features (three of each ProteinChip array type applied), to differentiate between lymph node positive and -negative breast cancer patients with a sensitivity and specificity of 81% and 77%, respectively, as determined by cross validation. With the exception of the 1003 Da peak, intensity cut off values of all peaks used in the discrimination between lymph node positive and -negative breast cancer patients are quite low. As demonstrated by Semmes et al. (10), interlaboratory agreement on peak m/z values is more difficult to achieve when peak intensities decrease. This finding is also reflected by our own observation, since none of the low intensity peaks discovered on the SAX surface were recovered in our dataset. The 1003 Da classifier, reported on the IMAC surface by Laronga et al. (15), had the highest intensity cut off value and was indeed recovered in our own dataset. This peak, however, is located in the very low m/z region of the spectrum, where the matrix noise contribution to the baseline signal is largest (27). Hence, this peak most likely does not represent a functional peptide, but might also be an adduct or artefact of the energyabsorbing-molecules or other chemical contaminants. As such, the biological validity of the peak is open to question (1). Structural identification clearly is a prerequisite to unequivocally determine whether an alleged biomarker is biologically valid (32). Data analysis Both Vlahou et al. (17) and Laronga et al. (15) applied the same bioinformatics software as used in the current study, i.e., the Biomarker Patterns software. This software package, applied in pattern recognition and sample classification, constructs decision trees by means of forward selection. A drawback of this method is that each successive split (‘node’) is less well-founded statistically, since sample size concomitantly decreases with an increasing number of decision nodes (33). Thus, with each successive decision rule, the tree becomes more strongly fitted to the training dataset, thereby reducing the likelihood of generalisation to unseen (test) data. This overfitting of data is more likely to occur when a large number of possible classifiers is applied in the construction of a multivariable model, such as a decision tree, as is often the case in proteomic studies (8). Overfitting of a model can unequivocally be detected by cross validation. While the error rate in the training set tends to decrease with an increasing number of classifiers, the error rate in the test set (as determined by cross validation) will increase. Thus, the performance of decision trees that do not suffer from overfitting will be similar during training and cross validation. Cross validation of the decision tree reported by Laronga 114 Assessment of robustness and validity et al. (15) yielded a sensitivity and specificity of 77% and 81%, respectively, while during training, a sensitivity and specificity of 100% and 87.3%, respectively, was achieved, indicating probable overfitting. Classifiers applied in overfitted trees are seldom robust, since they often represent peculiarities of the data set used for tree construction (33), providing a possible explanation for our inability to recover part of the classifiers detected by Laronga et al. (15). Furthermore, for data analysis procedures, Laronga et al. (15) refer to the publication of Vlahou et al. (17), in which spectra were normalised for total ion current in the 1.5-200 kDa mass range. The biomarkers reported by Laronga et al. (15) on the IMAC surface are, however, all < 1.5 kDa in mass. Since the application of a normalisation factor is only valid in the mass range employed during computation, intensities of the < 1.5kDa peaks applied by Laronga et al. (15) suffer from faulty normalisation. This may well provide an explanation for our inability to validate the markers we recovered from the IMAC30 surface. Sample storage temperature and -time Recovery of the SAX markers detected by Laronga et al. (15) in our validation sample set should be achieved when markers are robust, even though both sample handling and assay procedures presumably were not completely identical between studies. However, of the markers that were proven to be robust, validity could not be ascertained by our sample set. Our inability to recover and validate (part of) the reported markers could result from the difference in storage temperature between the sample sets analysed by Vlahou et al. (17), Laronga et al. (15) (both at -80°C), and the sample set analysed in the current study (-20°C). To investigate dependence of peak expression on storage temperature, we analysed an additional serum sample set from primary breast cancer patients, with identical samples stored at both -80°C and -20°C. Regarding the markers reported by Laronga et al. (15), only markers detected on the IMAC30 surface were recovered from our -80°C / -20°C dataset. Peak intensities of recovered markers were not significantly different between -80°C and -20°C spectra. None of the markers reported on the IMAC surface and recovered in our -80°C sample set was able to differentiate between lymph node positive and -negative breast cancer patients. Thus, expression of these three markers does not seem to be influenced by sample storage temperature (-20°C vs. -80°C). Regarding the markers reported by Vlahou et al. (17), all were recovered from the -80°C / -20°C sample set. No significant difference in peak intensities of recovered markers was observed between -80°C and -20°C spectra, except for m/z 4022 and m/z 4295. However, as the 95% CI of the mean peak intensities overlapped between the -80°C and -20°C spectra, expression of these peaks is most likely to be independent of storage temperature. Yet, since our -80°C sample set consisted solely of breast cancer sera, decisive conclusions with respect to influence of storage temperature on marker 115 Chapter 3.2 expression in sera of normal women, and thus on marker performance, could not be drawn. Although the structural identity of reported markers has not been elucidated, we hypothesize these markers to represent fragments of host response proteins, originating from protease (or protease inhibitor) activity, since these fragments frequently have been identified as potential biomarkers in the proteomic biomarker studies published to date (34-36). Assuming this hypothesis is correct, then protease (inhibitor) activity in breast cancer sera is most likely not affected by storage temperature, since peak intensities were similar in both -80°C and -20°C spectra under the tested conditions. We therefore hypothesize that protease (inhibitor) activity in normal control sera is not influenced by sample storage temperature as well, and, as a consequence, sample storage temperature most likely does not influence biomarker performance in this study. Another parameter that can influence the expression of reported peaks, is storage time. When plotting peak intensity vs. sample storage time, a random distribution of peak intensity across sample age was observed for all peaks. Thus, the expression of reported biomarkers, recovered in our dataset, is most likely not influenced by storage time. Evidently, further research is warranted to draw definitive conclusions. Differentiation by other biomarkers Satisfactory differentiation between sera of either breast cancer patients and normal women or lymph node positive and -negative breast cancer patients could not be achieved by any other (pattern of) features in the protein profiles generated on either IMAC30 or Q10 surfaces. The current approach of using unfractionated sera clearly lacks sensitivity to differentiate between these populations. Different pre-fractionation techniques may, however, provide us with a more in-depth view of especially the lowabundant proteome (37;38), thereby offering a possible means of differentiation. Moreover, since breast cancer is a highly heterogeneous disease, study patients may fall into a wide variety of different subgroups. Then, sample size will likely become too small to detect differences between subclasses of cancer or between cancer and normal, and detection of specific biomarkers may be hampered. Increasing sample size can provide a solution. Conclusion Conclusion In this study, both robustness and validity of the breast cancer biomarkers detected by Vlahou et al. (17) and Laronga et al. (15) were assessed. Following analysis of a different set of breast cancer and normal control sera in a different laboratory by meticulously using the reported assays, all biomarkers reported by Vlahou et al. (17) were recovered in our dataset. However, none of these biomarkers, applied either alone or in combination with each other, could satisfactorily differentiate between breast cancer sera and normal sera. Thus, although robustness of these biomarkers in our dataset was 116 Assessment of robustness and validity proven, their validity could not. The biomarkers Laronga et al. (15) reported to be specific for lymph node involvement in breast cancer patients were partially recovered from our dataset. Since no satisfactory differentiation between lymph node positive and -negative breast cancer sera could be achieved using the recovered markers, validity of these biomarkers could not be ascertained by analysis of our small sample set. In conclusion, this study demonstrates that, although results reported by both Vlahou et al. (17) and Laronga et al. (15) were promising, the validity or serum biomarkers discovered herein could not be ascertained by analysis of our independent sample set. Although structural identification of a potential biomarker is no absolute prerequisite in validation, it can not only undermine problems with robustness, but also determine its biological validity. Our findings stress that, when reporting on potential biomarkers, confirmation of both robustness and validity is pivotal in obtaining their clinical applicability, and structural identification of a potential biomarker, prior to its validation, is strongly recommended. References (1) Adam BL, Qu Y, Davis JW, Ward MD, Clements MA, Cazares LH et al. Serum protein fingerprinting coupled with a pattern-matching algorithm distinguishes prostate cancer from benign prostate hyperplasia and healthy men. Cancer Res 2002; 62(13):3609-3614. (2) Woong-Shick A, Sung-Pil P, Su-Mi B, Joon-Mo L, Sung-Eun N, Gye-Hyun N et al. Identification of hemoglobin-alpha and -beta subunits as potential serum biomarkers for the diagnosis and prognosis of ovarian cancer. Cancer Sci 2005; 96(3):197-201. (3) Zhang Z, Bast RC, Jr., Yu Y, Li J, Sokoll LJ, Rai AJ et al. Three biomarkers identified from serum proteomic analysis for the detection of early stage ovarian cancer. Cancer Res 2004; 64(16):5882-5890. (4) Xiao X, Liu D, Tang Y, Guo F, Xia L, Liu J et al. Development of proteomic patterns for detecting lung cancer. Dis Markers 2003; 19(1):33-39. (5) Li J, Zhang Z, Rosenzweig J, Wang YY, Chan DW. Proteomics and bioinformatics approaches for identification of serum biomarkers to detect breast cancer. Clin Chem 2002; 48(8):1296-1304. (6) Diamandis EP. Point: Proteomic patterns in biological fluids: do they represent the future of cancer diagnostics? Clin Chem 2003; 49(8):1272-1275. (7) Grizzle WE, Meleth S. Clarification in the point/counterpoint discussion related to surface-enhanced laser desorption/ionization time-of-flight mass spectrometric identification of patients with adenocarcinomas of the prostate. Clin Chem 2004; 50(8):1475-1476. (8) Ransohoff DF. Lessons from controversy: ovarian cancer screening and serum proteomics. J Natl Cancer Inst 2005; 97(4):315-319. (9) Li J, Orlandi R, White CN, Rosenzweig J, Zhao J, Seregni E et al. Independent Validation of Candidate Breast Cancer Serum Biomarkers Identified by Mass Spectrometry. Clin Chem 2005; 51(12):2229-2235. (10) Semmes OJ, Feng Z, Adam BL, Banez LL, Bigbee WL, Campos D et al. Evaluation of serum protein profiling by surface-enhanced laser desorption/ionization time-of-flight mass spectrometry for the detection of prostate cancer: I. Assessment of platform reproducibility. Clin Chem 2005; 51(1):102-112. 117 Chapter 3.2 (11) Grizzle WE, Adam BL, Bigbee WL, Conrads TP, Carroll C, Feng Z et al. Serum protein expression profiling for cancer detection: validation of a SELDI-based approach for prostate cancer. Dis Markers 2003; 19(4-5):185-195. (12) Becker S, Cazares LH, Watson P, Lynch H, Semmes OJ, Drake RR et al. Surfaced-enhanced laser desorption/ionization time-of-flight (SELDI-TOF) differentiation of serum protein profiles of BRCA-1 and sporadic breast cancer. Ann Surg Oncol 2004; 11(10):907-914. (13) Heike Y, Hosokawa M, Osumi S, Fujii D, Aogi K, Takigawa N et al. Identification of serum proteins related to adverse effects induced by docetaxel infusion from protein expression profiles of serum using SELDI ProteinChip system. Anticancer Res 2005; 25(2B):1197-1203. (14) Hu Y, Zhang S, Yu J, Liu J, Zheng S. SELDI-TOF-MS: the proteomics and bioinformatics approaches in the diagnosis of breast cancer. Breast 2005; 14(4):250-255. (15) Laronga C, Becker S, Watson P, Gregory B, Cazares L, Lynch H et al. SELDI-TOF serum profiling for prognostic and diagnostic classification of breast cancers. Dis Markers 2003; 19(4-5):229-238. (16) Pusztai L, Gregory BW, Baggerly KA, Peng B, Koomen J, Kuerer HM et al. Pharmacoproteomic analysis of prechemotherapy and postchemotherapy plasma samples from patients receiving neoadjuvant or adjuvant chemotherapy for breast carcinoma. Cancer 2004; 100(9):1814-1822. (17) Vlahou A, Laronga C, Wilson L, Gregory B, Fournier K, McGaughey D et al. A novel approach toward development of a rapid blood test for breast cancer. Clin Breast Cancer 2003; 4(3):203-209. (18) Goncalves A, Esterni B, Bertucci F, Sauvan R, Chabannon C, Cubizolles M et al. Postoperative serum proteomic profiles may predict metastatic relapse in high-risk primary breast cancer patients receiving adjuvant chemotherapy. Oncogene 2006; 25(7):981-989. (19) Mathelin C, Cromer A, Wendling C, Tomasetto C, Rio MC. Serum biomarkers for detection of breast cancers: a prospective study. Breast Cancer Res Treat 2005;1-8. (20) Jemal A, Tiwari RC, Murray T, Ghafoor A, Samuels A, Ward E et al. Cancer statistics, 2004. CA Cancer J Clin 2004; 54(1):8-29. (21) Etzioni R, Urban N, Ramsey S, McIntosh M, Schwartz S, Reid B et al. The case for early detection. Nat Rev Cancer 2003; 3(4):243-252. (22) Smith RA, Cokkinides V, Eyre HJ. American Cancer Society guidelines for the early detection of cancer, 2004. CA Cancer J Clin 2004; 54(1):41-52. (23) Stieber P, Molina R, Chan DW, Fritsche HA, Beyrau R, Bonfrer JM et al. Clinical evaluation of the Elecsys CA 15-3 test in breast cancer patients. Clin Lab 2003; 49(1-2):15-24. (24) Chan DW, Beveridge RA, Muss H, Fritsche HA, Hortobagyi G, Theriault R et al. Use of Truquant BR radioimmunoassay for early detection of breast cancer recurrence in patients with stage II and stage III disease. J Clin Oncol 1997; 15(6):2322-2328. (25) Olsen O, Gotzsche PC. Cochrane review on screening for breast cancer with mammography. Lancet 2001; 358(9290):1340-1342. (26) Petricoin EF, Liotta LA. SELDI-TOF-based serum proteomic pattern diagnostics for early detection of cancer. Curr Opin Biotechnol 2004; 15(1):24-30. (27) Baggerly KA, Morris JS, Coombes KR. Reproducibility of SELDI-TOF protein patterns in serum: comparing datasets from different experiments. Bioinformatics 2004; 20(5):777-785. (28) Rai AJ, Stemmer PM, Zhang Z, Adam BL, Morgan WT, Caffrey RE et al. Analysis of Human Proteome Organization Plasma Proteome Project (HUPO PPP) reference specimens using surface enhanced laser desorption/ionization-time of flight (SELDI-TOF) mass spectrometry: multi-institution correlation of spectra and identification of biomarkers. Proteomics 2005; 5(13):3467-3474. (29) Rai AJ, Gelfand CA, Haywood BC, Warunek DJ, Yi J, Schuchard MD et al. HUPO Plasma Proteome Project specimen collection and handling: towards the standardization of parameters for plasma proteome samples. Proteomics 2005; 5(13):3262-3277. 118 Assessment of robustness and validity (30) Scoggins CR, Chagpar AB, Martin RC, McMasters KM. Should sentinel lymph-node biopsy be used routinely for staging melanoma and breast cancers? Nat Clin Pract Oncol 2005; 2(9):448-455. (31) Tafra L, Lannin DR, Swanson MS, Van Eyk JJ, Verbanac KM, Chua AN et al. Multicenter trial of sentinel node biopsy for breast cancer using both technetium sulfur colloid and isosulfan blue dye. Ann Surg 2001; 233(1):51-59. (32) Gillette MA, Mani DR, Carr SA. Place of pattern in proteomic biomarker discovery. J Proteome Res 2005; 4(4):1143-1154. (33) Wiemer JC, Prokudin A. Bioinformatics in proteomics: application, terminology, and pitfalls. Pathol Res Pract 2004; 200(2):173-178. (34) Fung ET, Yip TT, Lomas L, Wang Z, Yip C, Meng XY et al. Classification of cancer types by measuring variants of host response proteins using SELDI serum assays. Int J Cancer 2005; 115(5):783-789. (35) Li J, White N, Zhang Z, Rosenzweig J, Mangold LA, Partin AW et al. Detection of prostate cancer using serum proteomics pattern in a histologically confirmed population. J Urol 2004; 171(5):1782-1787. (36) Tolson J, Bogumil R, Brunst E, Beck H, Elsner R, Humeny A et al. Serum protein profiling by SELDI mass spectrometry: detection of multiple variants of serum amyloid alpha in renal cancer patients. Lab Invest 2004; 84(7):845-856. (37) Righetti PG, Castagna A, Antonioli P, Boschetti E. Prefractionation techniques in proteome analysis: the mining tools of the third millennium. Electrophoresis 2005; 26(2):297-319. (38) Thulasiraman V, Lin S, Gheorghiu L, Lathrop J, Lomas L, Hammond D et al. Reduction of the concentration difference of proteins in biological liquids using a library of combinatorial ligands. Electrophoresis 2005; 26(18):3561-3571. 119 Chapter Haptoglobin phenotype is not a predictor of recurrence free survival in high-risk primary breast cancer patients Marie-Christine W. Gast Harm van Tinteren Marijke Bontenbal René Q.G.C.M. van Hoesel Marianne A. Nooij Sjoerd Rodenhuis Paul N. Span Vivianne C.G. Tjan-Heijnen Elisabeth G.E. de Vries Nathan Harris Jos W.R. Twisk Jan H.M. Schellens Jos H. Beijnen BMC Cancer 2008;8;389 3.3 Chapter 3.3 Abstract Better breast cancer prognostication may improve selection of patients for adjuvant therapy. We conducted a retrospective follow-up study in which we investigated sera of high-risk primary breast cancer patients, to search for proteins predictive of recurrence free survival. Two sample sets of high-risk primary breast cancer patients participating in a randomised national trial investigating the effectiveness of high-dose chemotherapy were analysed. Sera in set I (n = 63) were analysed by surface-enhanced laser desorption/ionisation time-of-flight mass spectrometry (SELDI-TOF MS) for biomarker finding. Initial results were validated by analysis of sample set II (n = 371), using one-dimensional gel-electrophoresis. In sample set I, the expression of a peak at mass-to-charge ratio 9198 (relative intensity ≤ 20 or > 20), identified as haptoglobin (Hp) alpha-1 chain, was strongly associated with recurrence free survival (global Log-rank test; p = 0.0014). Haptoglobin is present in three distinct phenotypes (Hp 1-1, Hp 2-1, and Hp 2-2), of which only individuals with phenotype Hp 1-1 or Hp 2-1 express the haptoglobin alpha-1 chain. As the expression of the haptoglobin alpha-1 chain, determined by SELDI-TOF MS, corresponds to the phenotype, initial results were validated by haptoglobin phenotyping of the independent sample set II by native one-dimensional gel-electrophoresis. With the Hp 1-1 phenotype as the reference category, the univariate hazard ratio for recurrence was 0.87 (95% CI: 0.56-1.34, p = 0.5221) and 1.03 (95% CI: 0.65-1.64, p = 0.8966) for the Hp 2-1 and Hp 2-2 phenotypes, respectively, in sample set II. In contrast to our initial results, the haptoglobin phenotype was not identified as a predictor of recurrence free survival in high-risk primary breast cancer in our validation set. Our initial observation in the discovery set was probably the result of a type I error (i.e., false positive). This study illustrates the importance of validation in obtaining the true clinical applicability of a potential biomarker. 122 Prognostic serum protein profiles for breast cancer Introduction Following lung cancer, breast cancer currently is the second leading cause of cancer deaths in women (1). A substantial survival benefit is achieved by treatment with adjuvant systemic therapy. The main prognostic factors in breast cancer include clinical (age) and pathological parameters (tumour size, lymph node status, and grade of malignancy), whereas the hormone-receptor and Her2/neu-receptor status are (also) predictive factors (2). However, 30-50% of breast cancer patients will eventually develop metastatic relapse and die, despite locoregional treatment and adjuvant systemic chemotherapy (3), while there is a small percentage that would have survived without adjuvant chemotherapy and hormonal therapy. Clearly, improved breast cancer prognostication is urgently needed to more accurately predict clinical outcome in individual patients and as such reduce both over- and undertreatment of the disease. High-throughput genomic and transcriptomic approaches have recently demonstrated to generate signatures that better predict clinical outcome than conventional prognosis criteria. For example, investigators from our institutes have published gene expression profiles in tumour tissue that outperformed all clinical variables in predicting disease outcome (distant metastases) (4-7). Similarly, a RT-PCR based multigene assay was recently shown to accurately predict both the probability of recurrence and the magnitude of chemotherapy benefit in node-negative, oestrogen-receptor positive breast cancer (8). An alternative and complementary approach is to perform protein expression analysis. As the proteome reflects gene expression as well as protein stability and posttranslational modifications, protein data could, in principle, be used for the same purpose. One of the techniques currently applied in proteomics research of breast cancer is surface-enhanced laser desorption/ionisation time-of-flight mass spectrometry (SELDI-TOF MS). Until now, only two studies have been published in which this platform was applied in the identification of serum markers for prognosis of breast cancer (9;10). Comparing the tumour cytosolic extract of node-negative sporadic breast tumours with or without a recurrence, Ricolleau et al. (10) identified a high level of ubiquitin and / or a low level of ferritin light chain to be associated with a good prognosis in breast cancer (n = 60). Goncalves et al. (9) constructed a multiprotein model, consisting of 40 proteins, that correctly predicted relapse in 67 of the 81 patients of which fractionated sera were investigated. These promising results need to be interpreted cautiously, as in both studies only a limited number of patients was investigated, and results have not been validated yet by analysis of independent study populations. Hence, the aim of the current study is to investigate sera of high-risk primary breast cancer patients to search for proteins predictive of recurrence free survival, and to validate our results by analysis of an independent study population. 123 Chapter 3.3 Materials and Methods Study population From 1993 to 1999, high-risk primary breast cancer patients who had undergone modified radical mastectomy or breast conserving surgery with complete axillary clearance participated in a randomised multicentre phase III trial. This study investigated the benefit of high-dose adjuvant chemotherapy in patients with ≥ 4 axillary lymph node metastases. The design of the study has been described elsewhere (11). Major eligibility criteria were histologically confirmed stage 2A, 2B or 3A breast cancer with at least 4 tumour-positive axillary lymph nodes but no evidence of distant metastases, age under 56 years, and no previous other malignancies. In sample set I, sera of 63 study patients who were treated in the Netherlands Cancer Institute were included. Sera were procured after surgery (7-51 days), but prior to adjuvant chemotherapy (0-45 days). All sera were obtained and stored under strictly defined conditions at the Institutional Serum Bank. In sample set II, serum / plasma samples (procured at any time point in therapy) of 371 study patients treated in the Netherlands Cancer Institute (sera; n = 15, plasma; n = 38), the Erasmus Medical Center - Daniel den Hoed Cancer Center (sera; n = 114), the Radboud University Medical Center Nijmegen (sera; n = 87), the University Medical Center Groningen (sera; n = 69), and the University Medical Center Leiden (sera; n = 48) were included. All samples were obtained with medical-ethics approval and all patients gave informed consent. Chemicals All used chemicals were obtained from Sigma, St. Louis, MO, USA, unless stated otherwise. Biomarker discovery Protein profiling was performed using the ProteinChip SELDI Reader (Bio-Rad Laboratories, Hercules, CA, USA). Several chromatographic array surfaces with suitable binding conditions were screened for discriminative mass-to-charge ratio’s (m/z) between unfractionated sera of breast cancer patients of set I either experiencing a recurrence at a relatively short follow-up (Recurrence Free Survival (RFS) < 16 months, n = 4), or experiencing no recurrence after a long follow-up (> 75 months, n = 4). Optimal discrimination between both groups was obtained by Q10 arrays (strong anion exchange chromatography) with 100 mM Tris-HCl pH 8 / 0.1% TritonX-100 as a binding buffer. This assay was subsequently applied in the analysis of all sera in sample set I (n = 63). In brief, samples were thawed on ice and denatured by 1:10 dilution in 9 M urea / 2% 3[(3-cholamidopropyl)dimethylammonio]-1-propanesulfonate (CHAPS) / 1% dithiotreitol (DTT). Arrays were assembled in a 96-well bioprocessor (Bio-Rad Labs), which was 124 Prognostic serum protein profiles for breast cancer placed on a platform shaker at 350 rpm at all steps of the protocol. Arrays were equilibrated twice with 200 µl of binding buffer for 5 min. Pretreated serum samples were diluted 1:10 in binding buffer and were randomly applied to the arrays. After a 30 min incubation, the arrays were washed twice with binding buffer and twice with 100 mM Tris-HCl pH 8 for 5 min. Following a quick rinse with deionised water (Braun, Emmenbrücke, Germany), arrays were air-dried. A 50% solution of sinapinic acid (BioRad Labs) in 50% acetonitrile (ACN) / 0.5% trifluoroacetic acid (TFA) was applied twice (1.0 µl) to the array as matrix. Following air-drying, the arrays were analysed using the ProteinChip SELDI (PBS IIc) Reader (Bio-Rad Labs). For mass accuracy, the instrument was calibrated on the day of measurements with All-In-One peptide standard (Bio-Rad Labs). Data were collected between 0 and 200 kDa, averaging 65 laser shots with intensity 158, detector sensitivity 5, and a focus lag time of 746 ns. Spectra were baseline subtracted and normalised to the total ion current from 1.5 to 200 kDa. The Biomarker Wizard software package (version 3.1, Bio-Rad Labs) was applied for peak detection. Peaks were auto-detected when occurring in at least 25% of spectra and when having a signal-to-noise ratio of at least 5. Peak clusters were completed with peaks with a signal-to-noise ratio of at least 2 in a cluster mass window of 0.3%. Biomarker characterisation A 500 µl serum sample containing the biomarker of interest marker (i.e., m/z 9198) was denatured in 9 M urea / 2% CHAPS / 1% DTT in 50 mM Tris-HCl pH 9. The sample was subsequently fractionated on Q Ceramic HyperD beads with a strong anion exchange moiety (Biosepra Inc., Marlborough, MA, USA). After binding of denatured sample to the beads, the flow through was collected and bound proteins were subsequently eluted with buffers of pH 9-3. The fraction containing the marker was further purified by size fractionation, using Microcon 50 kDa MW spin concentrators (YM50, Millipore, Billerica, MA, USA) with increasing concentrations of ACN / 0.1% TFA. The filtrate containing the m/z 9198 marker was subsequently de-salted by application on reversed phase RP18 beads (Varian Inc., Palo Alto, CA, USA), followed by elution with increasing concentrations of ACN containing 0.1% TFA. The purification process was monitored by profiling each fraction on Q10 arrays and NP20 arrays (a non-selective, silica chromatographic surface). Eluates containing the m/z 9198 marker were dried and redissolved in loading buffer for SDS-PAGE, which was performed on Novex NuPage gels (18% Tris-Glycine gel; Invitrogen, San Diego, CA, USA). Following Coomassie staining (Simply Blue; Invitrogen), protein bands of interest were excised and collected. The proteins within the excised bands were eluted by washing twice with 30% ACN / 100 mM ammonium bicarbonate, followed by dehydration in 100% ACN. Gel bands were subsequently heated at 50°C for 5 min and eluted with 45% formic acid / 30% ACN / 10% isopropanol under sonification for 30 min. After leaving the eluates overnight at room temperature, they were profiled on NP20 arrays. Eluates were subsequently dried, resuspended in 20 ng/µl trypsin 125 Chapter 3.3 (Promega, Madison, WI, USA) in 10% ACN / 25 mM ammonium bicarbonate, followed by incubation at room temperature for 4 h for protein digestion. For in-gel protein digestion, gel bands were first washed with 40% methanol / 10% acetic acid twice, followed by a 30% ACN / 100 mM ammonium bicarbonate wash. Gel bands were dried by SpeedVac and digested for 12 h by trypsin (20 ng/µl 100 mM ammonium bicarbonate). All tryptic digests were profiled on NP20 chips, using 1 µl 20% alphacyano-4-hydroxy cinnaminic acid solution in 50% ACN / 0.5% TFA as matrix. Peptides in the digests were investigated with the NCBI database using the ProFound search engine at http://prowl.rockefeller.edu/prowl-cgi/profound.exe with the following search parameters: standard cleavage rules for trypsin, 1 missed cleavage allowed. Confirmation of protein identity was provided by sequencing tryptic digest peptides by quadrupole-TOF (Q-TOF) MS (Applied Biosystems / MSD Sciex, Foster City, CA, USA) fitted with a ProteinChip Interface. Fragment ion spectra resulting from Q-TOF analyses were taken to search the SwissProt 44.2 database (Homo Sapiens: 11072 sequences) using the MASCOT search engine at www.matrixscience.com (Matrix Science Ltd., London, UK), with the following search parameters: monoisotopic precursor mass tolerance: 40 ppm, fragment mass tolerance: 0.2 Da, variable modifications: methionine oxidation, and trypsin cleavage site. Throughout the identification experiments, a serum sample lacking the m/z 9198 marker was run concurrently as a negative control. Haptoglobin phenotyping assay The haptoglobin (Hp) phenotype of all samples in set I and II was assessed by native one-dimensional gel electrophoresis, followed by peroxidase staining. One µl of serum or plasma sample was mixed with 19 µl of a 1:100 dilution of haemolysate in phosphate buffered saline. Following incubation for 5 min at room temperature, 10 µl of 3x native sample buffer (30 ml glycerol / 18.8 ml 1 M Tris-HCl pH 6.8 / 1.5 ml 1% (w/v) bromophenol blue, made to 100 ml with water) was added and mixed. Samples were then loaded onto a 3-8% gradient Tris-Acetate NuPAGE precast gel (Invitrogen, Karlsruhe, Germany). Samples were run at a constant 150 V, gradient 18-7 mA for 3 h, using a running buffer of 25 mM Tris / 250 mM glycine, adjusted to pH 8.6. After staining with 1% (w/v) rhodamine 1%, the gel was incubated for 10 min in a 1:1 waterdiluted leucomalachite green peroxidase-development buffer (0.2 g leucomalachite green / 0.02 g EDTA in 25 ml 40% (v/v) acetic acid with 0.06% (v/v) H2O2). The phenotype of each sample was subsequently determined by its specific migration pattern, which appears as black bands in the gel (Figure 1A) (12). Statistical analysis Survival curves were analysed according to the Kaplan-Meier method from the date of randomisation to the time of first recurrence or death, or the date of last follow-up. The curves were compared by log-rank statistics. To investigate the relation of haptoglobin 126 Prognostic serum protein profiles for breast cancer phenotype and other variables with recurrence-free survival time, a Cox proportional hazards model was used. Relations were expressed in terms of hazard ratios with 95% confidence intervals. Possible confounding clinical variables that either have known prognostic or predictive value (i.e., treatment (high dose vs. conventional dose chemotherapy), age (≥ 40 yrs vs. < 40 yrs), number of positive lymph node (0 - 9 vs. ≥ 10), tumour size (< 5 cm vs. ≥ 5 cm), Her2/neu status (negative vs. positive, of note, patients did not yet receive adjuvant trastuzumab), receptor status (oestrogen and / or progesterone receptor (ER/PR) positive vs. negative), and Bloom-Richardson grade (grade I vs. grade II vs. grade III)), or variables that were related to the exposure haptoglobin phenotype (i.e., surgery (breast conserving vs. mastectomy)) were incorporated into the model. Figure 1 Haptoglobin phenotype assessment using a native PAGE system. A. Specific migration pattern of Hp 1-1, Hp 2-1, and Hp 2-2, in a 3-8% gradient Tris-Acetate gel. B. Composition of the three haptoglobin phenotypes Hp 1-1, Hp 2-1, and Hp 2-2 (adapted from (13)). A Hp 1-1 Hp 2-1 Hp 2-2 B Hp 1-1 Hp α-1 Hp 2-1 Hp α-2 Hp 2-2 Hp β The distribution of patient characteristics over the two sample sets were compared using either the Chi-square test or the Fisher’s exact test for categorical variables and the Mann-Whitney U test for continuous variables. All statistical analyses were performed using SPSS statistical software, version 13.0 (SPSS Inc., Chicago, IL, USA) and SAS statistical software, version 9.1.3 (SAS Institute Inc., Cary, NC, USA). Statistical tests were two sided at the 5% level of significance. 127 Chapter 3.3 Results Study population At time of analysis, in sample set I (n = 63), 28 patients had a recurrence or had died and 35 patients were censored at a median follow-up of 6.6 years. In sample set II (n = 371), 149 patients had a recurrence or had died and 222 patients were censored at a median follow-up of 8.0 years. Characteristics of both sample sets are provided in Table 1. All patient characteristics were similarly distributed between sample set I and sample set II, as determined by the Chi-square test or the Mann-Whitney U test. Biomarker discovery Following evaluation of several chromatographic array surfaces with suitable binding conditions, the Q10 array with 100mM Tris-HCl pH 8 / 0.1% TritonX-100 as a binding buffer gave optimal results in our screening population (n = 8). Using this SELDI-TOF MS assay, the spectra of sera from patients experiencing no recurrence (n = 4) could clearly be distinguished by the spectra of sera from patients that recurred at a relatively short follow-up (n = 4) by overexpression of a peak at m/z 9198. The clear dichotomous distribution in the relative m/z 9198 peak intensity was subsequently confirmed in the acquired mass spectra of all 63 sera in sample set I (peak intensity > 20: n = 40, ≤ 20: n = 23). Representative SELDI-TOF MS spectra are presented in Figure 2. The KaplanMeier curve (Figure 3) shows a significant difference in the probability of remaining recurrence free (Log-rank test, p = 0.0014) between high-risk primary breast cancer patients exhibiting a peak at m/z 9198 with a relative intensity > or ≤ than 20. The univariate hazard ratio was 3.22 (95% CI: 1.51-6.85, p = 0.0024). Biomarker characterisation Following anion exchange fractionation, the m/z 9198 marker was eluted in the pH 5 eluate. This fraction was concentrated on YM50 spin concentrators, and the marker was found in the water wash. De-salting of the water wash on RP18 beads resulted in elution of the marker in the 60% ACN / 0.1% TFA eluate, which was subsequently subjected to SDS-PAGE analysis. After staining, a clear band in the 9 kDa region was visible, which was excised. Elution of the proteins within the excised bands was followed by tryptic digestion of the eluate. Profiling of the gel-eluate confirmed the presence of the marker. Peptide mapping of the tryptic digest identified the marker as haptoglobin alpha-1 chain (estimated Z-score 1.49, 48% sequence coverage), which is an 83 amino acid peptide with a theoretical mass of 9192.21 Da and a pI of 5.23. This identity [SwissProt: P00738] was confirmed by amino acid sequencing of 4 peptides in the tryptic digest by tandem MS on a Q-TOF (76% coverage, Figure 4 A). 128 Prognostic serum protein profiles for breast cancer Table 1 Patient and tumour characteristics of sample set I and II. Sample set I N (%) Sample set II N (%) Patient characteristics N 63 371 age Mean [range] < 40 years ≥ 40 years 45.8 [33-55] 10 53 (16%) (84%) 43.9 [26-55] 94 277 (25%) (75%) 49 11 3 (78%) (17%) (5%) 317 40 14 (85%) (11%) (4%) Mastectomy Breast conserving 56 7 (89%) (11%) 291 80 (78%) (22%) Conventional dose High dose 27 36 (43%) (57%) 158 213 (43%) (57%) 4-9 ≥ 10 40 23 (63%) (37%) 241 130 (65%) (35%) T1 (< 2 cm) T2 (2-5 cm) T3 (≥ 5 cm) 9 41 13 (14%) (65%) (21%) 90 225 56 (24%) (61%) (15%) Her2/neu status Negative Positive Unknown 42 16 5 (67%) (25%) (8%) 274 81 16 (74%) (22%) (4%) Oestrogen / Progesterone receptor status ER and PR negative ER and/or PR positive Unknown 10 50 3 (16%) (79%) (5%) 101 250 20 (27%) (67%) (5%) Bloom-Richardson grade Grade I Grade II Grade III Unknown 13 26 20 4 (21%) (41%) (32%) (4%) 62 112 170 27 (17%) (30%) (46%) (7%) Menopausal status Pre Post Unknown Surgery Treatment Tumour characteristics Number Tumour size Haptoglobin occurs in vivo as polymers of an alpha and beta chain complex, interlinked via disulfide bridges. There are two major alpha chains: alpha-1 (83 amino acids, 9.2 kDa) and alpha-2 (142 amino acids, 16 kDa), of which the alpha-2 chain is the product of unequal crossing over between two alpha-1 alleles (14). Due to this genetic variation, haptoglobin occurs in three major (pheno)types: Hp 1-1, Hp 2-1 and Hp 2-2, occurring in 16%, 48%, and 36%, respectively, of the northwestern European population (13). The 129 Chapter 3.3 Hp 1-1 phenotype consists of an [alpha-1 - beta] dimer (86 kDa), whereas Hp 2-1 consist of two [alpha-1 - beta] units flanking a variable length [alpha-2 - beta] polymer (86-300 kDa). Hp 2-2, the largest species, consists of multiple repeats of an [alpha-2 beta] unit (170-900 kDa) (Figure 1B) (15;16). Expression of the Hp alpha-1 chain, as determined by SELDI-TOF MS, will correspond to the actual haptoglobin phenotype, since the haptoglobin alpha-1 chain is only expressed by individuals with the Hp 1-1 or Hp 2-1 phenotype (12). Indeed, following haptoglobin phenotype assessment by native one-dimensional gel-electrophoresis, all patients with m/z 9198 ≤ 20 carried the Hp 2-2 phenotype (n = 23), while patients with m/z 9198 > 20 were shown to have either the Hp 1-1 (n = 14) or Hp 2-1 (n = 26) phenotype (Figure 5). Figure 2 Representative example of serum protein profiles (sample set I) obtained with the optimized SELDI-TOF MS assay, showing the clear dichotomous expression of the m/z 9198 peak (dotted box). 8000 9000 10000 11000 75 Pt 1 0 75 Pt 2 Relative intensity → 0 75 Pt 3 0 75 Pt 4 0 75 Pt 5 0 75 Pt 6 0 75 Pt 7 0 75 Pt 8 0 8000 9000 10000 11000 Mass-to-charge ratio → Following Kaplan-Meier analysis by haptoglobin phenotype in sample set I (n = 63), the Hp 1-1, 2-1 and 2-2 phenotypes were shown to be associated with a good, intermediate and poor prognosis, respectively (global Log-rank test, p = 0.0029) (Figure 6). With Hp 1-1 phenotype as the reference category, the univariate hazard ratio was 3.08 (95% CI: 0.67-14.10, p = 0.1464) for Hp 2-1, and 7.37 (95% CI: 1.69-32.23, p = 0.0079) for Hp 2-2 130 Prognostic serum protein profiles for breast cancer phenotype. In the multivariate Cox regression analysis, haptoglobin phenotype was independently associated with recurrence free survival (Hp 2-2; p = 0.0098), while for receptor status (ER/PR negative; p = 0.0962) and treatment arm (conventional dose; p = 0.0509) a borderline significant association was observed (Table 2). Figure 3 Recurrence free survival in sample set I (n = 63) according to m/z 9198 peak intensity > 20 or ≤ 20, as determined by SELDI-TOF MS. 1.0 m/z 9198 > 20 RFS probability 0.8 0.6 m/z 9198 <= 20 0.4 0.2 0.0 p = 0.0014 (Log-rank test, two-sided) 40 23 0 38 22 12 34 16 32 11 29 8 24 36 48 months from randomisation 29 7 60 24 > 20 6 <= 20 72 Biomarker validation The distribution of the haptoglobin phenotype of patients in the validation sample set II (n = 371) was subsequently assessed for validation purposes. As the haptoglobin phenotype (i.e., genotype) is not influenced by treatment, samples in set II were collected at any time point in therapy. All patient characteristics were similarly distributed between sample set I and sample set II. The Hp 1-1, 2-1, and 2-2 phenotype was determined in 70, 189, and 112 patients, respectively, yielding an allele frequency of 0.44, which is in concordance with previously reported frequencies. (13) The Kaplan-Meier curve, however, did not show a significant difference in the probability of recurrence free survival (global Log-rank test, p = 0.6158) between the high-risk primary breast cancer patients in sample set II having the Hp 1-1, 2-1 or 2-2 phenotype (Figure 7). With the Hp 1-1 phenotype as the reference category, the univariate hazard ratio was 0.87 (95% CI: 0.56-1.34, p = 0.5221) and 1.03 (95% CI: 0.651.64, p = 0.8966) for the Hp 2-1 and Hp 2-2 phenotypes, respectively. This finding was not affected by tumour size, which was found to be the only independently associated variable for recurrence free survival (p = 0.0374). Her2/neu status (p = 0.0589) and 131 Chapter 3.3 treatment arm (p = 0.0809) were only borderline significantly associated with recurrence free survival in set II (Table 2). Table 2 Multivariable proportional-hazards analyses for the risk of recurrence for patients in sample set I and II. Variable Sample set I (n = 63) Sample set II (n = 371) HR (95% CI) p-value HR (95% CI) p-value Haptoglobin phenotype Hp 1-1 Hp 2-1 Hp 2-2 1 4.21 17.76 (0.44-39.83) (2.00-157.44) 0.2105 0.0098 1 0.94 1.26 (0.58-1.52) (0.76-2.08) 0.8059 0.3653 Surgery Breast conserving Mastectomy 1 1.26 (0.24-6.67) 0.7843 1 0.91 (0.59-1.39) 0.6514 Treatment arm High dose Conventional dose 1 0.33 (0.11-1.00) 0.0509 1 1.36 (0.96-1.92) 0.0809 Age ≥ 40 yrs < 40 yrs 1 1.26 (0.32-4.93) 0.7413 1 0.97 (0.65-1.46) 0.8890 No. of positive lymph nodes 4-9 ≥ 10 1 0.72 (0.23-2.29) 0.5785 1 1.00 (0.69-1.45) 0.9964 Tumour size < 5 cm ≥ 5 cm 1 1.35 (0.50-3.65) 0.5497 1 1.63 (1.03-2.60) 0.0374 Her2/neu status Negative Positive 1 1.47 (0.53-4.08) 0.4641 1 1.47 (0.99-2.20) 0.0589 Receptor status ER/PR positive ER/PR negative 1 3.39 (0.80-14.27) 0.0962 1 1.11 (0.73-1.70) 0.6138 Bloom-Richardson grade Grade I Grade II Grade III 1 0.92 1.28 (0.26- 3.23) (0.37-4.46) 0.9016 0.6946 1 0.87 1.34 (0.50-1.50) (0.79-2.26) 0.6187 0.2796 Abbreviations: 95% CI: 95% confidence interval, HR: hazard ratio. Discussion The introduction of high-throughput analytical platforms, such as the genomic/ transcriptomic microarray technology, or the proteomic SELDI-TOF MS technology, has enabled the advent of discovery-based research. Large quantities of data can now be analysed without underlying hypotheses, to search for patterns that discriminate between patients with different diagnosis, prognosis or response to treatment. 132 Prognostic serum protein profiles for breast cancer Assessment of validity, however, is pivotal in this discovery-based ‘-omics’ research, as the meaning of such patterns from a biological perspective often is unknown. Figure 4a Structural identification of the m/z 9198 peak cluster. Peptide mapping of the m/z 9198 marker. MS spectrum of the m/z 9198 tryptic digest in the gel eluate. All peptides were sequenced with tandem MS using Q-TOF for confirmation. Results from the MASCOT search for protein identification include start and end positions of the peptide sequence starting from the amino acid terminal of the whole protein, the observed m/z (Mr(obs)), transformed to its experimental mass (Mr(expt)), the calculated mass (Mr(calc)) from the matched peptide sequence, as well as their mass difference (Delta), the number of missed cleavage sites for trypsin (Miss), and the peptide sequence (in grey: the amino acid sequence determined by Q-TOF MS). 3294.6096 100 1976.8243 3293.5781 2379.0537 1975.8258 3295.5713 2378.0378 1708.7289 2380.0500 3292.6165 1977.8232 1709.7421 % 1743.7511 2090.8633 2091.8340 1590.6274 2465.0471 2466.0205 1573.6035 1000 1200 1400 1600 3276.5515 3067.4463 2467.0144 1563.6398 0 3296.5806 2381.0464 3080.3804 2706.1506 1800 2000 2200 2400 2600 2800 3297.5657 4387.0762 4385.0986 3000 3200 3400 3600 3800 4000 4200 4400 m/z, amu MASCOT peptide mapping results: m/z 9198 haptoglobin alpha-1 chain StartMr Mr Mr Delta Miss Sequence StartEnd (obs) (expt) (calc) 35-49 1590.70 1589.69 1589.79 -0.10 0 K.PPEIAHGYVEHSVR.Y 58-72 1708.80 1708.79 1707.84 -0.05 1 K.LRTEGDGVYTLNNEK.Q 78-94 1743.75 1742.74 1742.87 -0.13 0 K.AVGDKLPECEAVCGKPK.N 19-49 3292.61 3291.60 3291.51 0.09 0 A.VDSGNDVTDIADDGCPKPPEIAHGYVEHSVR.Y Amino acid sequence of m/z 9198 haptoglobin alpha-1 chain (start: 18 - end: 101, 76% sequence coverage): VDSGNDVTDIADDGCPKPPEIAHGYVEHSVRYQCKNYYKLRTEGDGVYTLNNEKQWINKAVGDKLPECEA VCGKPKNPANPVQ Our initial findings were, however, endorsed by the various biological functions of haptoglobin and their phenotype-dependency. The main physiological function of haptoglobin is binding of free haemoglobin. The haptoglobin-haemoglobin complex is too large to be filtered at the kidney glomerulus and is therefore retained. Both iron loss 133 Chapter 3.3 and free-radical mediated damage, caused by the haem-iron mediated generation of free hydroxyl radicals (by means of the Fenton reaction: H2O2 + Fe2+ → Fe3+ + OH- + ·OH) are thus prevented (13). Haptoglobin has also been identified as a strong angiogenic agent, activating endothelial cell growth and differentiation. This function was shown to be phenotype dependent, as the Hp 2-2 phenotype has been found to be more angiogenic than the other phenotypes (17). The poor prognosis of our Hp 2-2 breast cancer patients in our discovery set (n = 63) could be exerted via this haptoglobin function, since angiogenesis is well known to be involved tumour growth, proliferation, and metastasis (18). Figure 4b Structural identification of the m/z 9198 peak cluster. Matched amino acid sequence of the m/z 9198 marker (in grey: amino acid sequence sequenced by Q-TOF MS), haptoglobin alpha-1 chain and the corresponding N-terminus of haptoglobin-related-protein (in grey: amino acid substitutions between haptoglobin and haptoglobin-related-protein) (19). Comparison of amino acid sequences m/z 9198 marker VDSGNDVTDIADDGCPKPPEIAHGYVEHSVRYQCKNYYKLRTEGDGVYTLNNEKQWINKAVGDKLPECEAVCGKPKN PANPVQ Haptoglobin alphaalpha-1 chain (Mw 9192.21, pI 5.23) VDSGNDVTDIADDGCPKPPEIAHGYVEHSVRYQCKNYYKLRTEGDGVYTLNNEKQWINKAVGDKLPECEAVCGKPKN PANPVQ HaptoglobinHaptoglobin-relatedrelated-protein (Mw 9492.64, pI 6.76) LYSGNDVTDISDDRFPKPPEIANGYVEHLFRYQCKNYYRLRTEGDGVYTLNDKKQWINKAVGDKLPECEAVCGKPKN PANPVQ Both haptoglobin and its phenotype have been described previously in relation to various diseases (including cancer), a finding which is not surprising in view of its biology. Both the intact protein and its subunits have been found overexpressed in serum of patients with various solid tumours, for example ovarian and small-cell lung cancer (20-22). The 9.2 kDa haptoglobin alpha-1 chain has been specifically detected by Tolson et al. (12) in sera of renal cell carcinoma patients and healthy controls. Following haptoglobin phenotyping, all patients having the Hp 2-2 phenotype indeed proved to be missing the 9.2 kDa haptoglobin alpha-1 peak in their serum protein profile. Due to its phenotypic distribution, this protein could, however, not be considered as a diagnostic marker. The influence of haptoglobin phenotype on recurrence free survival has not been investigated (12). Using the SELDI-TOF MS platform, the 9.2 kDa alpha-1 chain was also detected by Goncalves et al. (9). Unlike our own observations, they found the 9.2 kDa peak to be overexpressed in sera of high-risk primary breast cancer patients (n = 81) experiencing a relapse versus long-term disease free survivors (9). The absolute intensities of the m/z 9192 peak in SELDI-TOF MS spectra of Goncalves et al. (9) ranged between 0 and 2, and a clear dichotomous peak intensity distribution was not observed. These discrepancies from our initial findings most likely originate in the serum pre-fractionation that was performed in this study. 134 Prognostic serum protein profiles for breast cancer During protein purification, we repeatedly found the 9.2 kDa peak to be predominantly present in the pH 5 fraction. Goncalves et al. (9) however, subjected only the pH 9 / flow through, pH 4, and organic solvent fractions to SELDI-TOF MS analysis, resulting in a suboptimal assay for haptoglobin alpha-1 detection. Peak intensity of the m/z 9198 marker (as determined by SELDI-TOF MS) vs. haptoglobin phenotype (as assessed by 1D gel-electrophoresis) of samples in set I. m/z 9198 peak intensity Figure 5 80 60 40 20 0 Hp 1 1-1 Hp 2 2-1 Hp 3 2-2 Another association between protein expression and recurrence free survival in breast cancer has previously been reported by Kuhajda et al. (23;24). They described a decreased tumour tissue expression of haptoglobin-related-protein, quantitated immunohistochemically, to be associated with a prolonged recurrence free survival in 70 breast cancer patients (23). Their findings differ from our observations by the biological matrix analysed (tumour tissue vs. serum), by the exposure used for prediction of recurrence free survival (protein expression vs. phenotype), and by the identity of the protein used for prognostication (haptoglobin-related-protein vs. haptoglobin). Although coded for by two different genes, both proteins have more than 90% amino acid sequence homology. There are, however, distinct differences between the alpha-1 chain of haptoglobin and haptoglobin-related-protein, due to 8 amino acid substitutions (19). Amino acid sequencing of the m/z 9198 marker, taking 7 out of 8 amino acid substitutions into account, enabled us to unequivocally identify our candidate biomarker as haptoglobin alpha-1 chain (Figure 4 B). The haptoglobin phenotype has not been identified as a predictor of recurrence free survival in breast cancer thus far. The phenotype has nevertheless been associated with clinical outcome of other pathologies, a.o. mortality (25), nephropathy (26), and cardiovascular disease outcome in diabetic patients (27), mortality in HIV infection (28) and mortality in tuberculosis (29). In these studies, the Hp 2-2 phenotype was invariably associated with worse clinical outcome, in contrast though to the study of Depypere et al. (30), who found the Hp 1-1 phenotype associated with more severe hypertension and proteinuria in patients with preeclampsia. 135 Chapter 3.3 Despite the potential biological justifications, our promising initial result of the haptoglobin phenotype being a predictor of recurrence free survival in a limited number (n = 63) of high-risk primary breast cancer patients was not confirmed following validation by analysis of a six-fold larger sample set (n = 371). It is unlikely that our findings result from differences in patient characteristics between our discovery and validation sample set, since all (known) characteristics were similarly distributed between both sample sets. Figure 6 Recurrence free survival in sample set I (n = 63) by haptoglobin phenotype. 1.0 Hp 1-1 RFS probability 0.8 Hp 2-1 0.6 Hp 2-2 0.4 0.2 0.0 p = 0.0029 (logrank test, two-sided) 14 26 23 0 14 24 22 12 13 21 16 13 19 11 12 17 8 24 36 48 months from randomisation 12 17 7 60 11 Hp 1-1 13 Hp 2-1 6 Hp 2-2 72 Despite the potential biological justifications, our promising initial result of the haptoglobin phenotype being a predictor of recurrence free survival in a limited number (n = 63) of high-risk primary breast cancer patients was not confirmed following validation by analysis of a six-fold larger sample set (n = 371). It is unlikely that our findings result from differences in patient characteristics between our discovery and validation sample set, since all (known) characteristics were similarly distributed between both sample sets. The two major threats to validity of discovery-based proteomics research come from chance and bias (31). Sources of bias include differences in sample collection and storage, or in analysis (32). The haptoglobin phenotype however, is not influenced by specimen collection and storage. Besides, the native 1D-gelelectrophoresis method for assessment of haptoglobin phenotype is robust and reproducible (33). Study results are therefore unlikely to have been influenced by bias, but rather result from chance. Due to small sample sizes and the artifice of discovery strategies, many biomarker candidates are prone to be false positive, i.e., be a type I error (erroneous rejection of the null hypothesis). The chance of candidate biomarkers being type I errors is inferred 136 Prognostic serum protein profiles for breast cancer by the fact that most proteomic datasets are subject to both the ‘curse of dimensionality’ (large number of features) and the ‘curse of dataset sparsity’ (limited number of samples) (34). As such, datasets are frequently subjected to multiple testing in search for candidate biomarkers. Yet, even a level of significance for type I errors of 0.01 is no guarantee that false positive findings are debarred, even following correction for multiple testing. Problems caused by chance are best avoided by analysis of an independent validation dataset, in which false positive markers will be ruled out, as they are unique to the discovery sample set (32;35). The above-mentioned hurdles in proteomics research apply equally to all other ‘-omics’ research (e.g., genomics and metabolomics), as in general, these research approaches suffer from a limited number of samples in comparison with the large number of generated features. Figure 7 Recurrence free survival in sample set II (n = 371) by haptoglobin phenotype. 1.0 RFS probability Hp 2-1 Hp 1-1 0.8 0.6 Hp 2-2 0.4 0.2 p = 0.6158 (Log-rank test, two-sided) 70 189 0.0 112 0 63 176 103 12 57 160 90 51 143 80 48 138 73 24 36 48 months from randomisation 43 124 66 60 33 Hp 1-1 107 Hp 2-1 58 Hp 2-2 72 Conclusion In conclusion, although we initially found the haptoglobin phenotype to be a predictor of recurrence free survival in a limited number of high-risk primary breast cancer patients, this was not confirmed following validation by analysis of a similar, but sixfold larger sample set. Clearly, validation of initial results is of pivotal importance in determining the clinical significance of a candidate biomarker. In spite of this, few, if any, related clinical diagnostic tests have yet been validated for clinical use, although the number of papers reporting on candidate protein biomarkers is large and still expanding. This lack of validation can result in chance results and erroneous 137 Chapter 3.3 conclusions, leading to disappointment when results cannot be reproduced (36). Distillation of true positives from the total pool of candidate biomarkers is the single greatest challenge in biomarker development, and should therefore be the emphasis in data-driven proteomics research. Acknowledgement This study was supported by a grant of the Dutch Cancer Society (project NKI 20053421). References (1) Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T et al. Cancer statistics, 2008. CA Cancer J Clin 2008; 58(2):71-96. (2) Goldhirsch A, Wood WC, Gelber RD, Coates AS, Thurlimann B, Senn HJ. Meeting highlights: updated international expert consensus on the primary therapy of early breast cancer. J Clin Oncol 2003; 21(17):3357-3365. (3) Polychemotherapy for early breast cancer: an overview of the randomised trials. Early Breast Cancer Trialists' Collaborative Group. Lancet 1998; 352(9132):930-942. (4) 't Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature 2002; 415(6871):530-536. (5) Foekens JA, Atkins D, Zhang Y, Sweep FC, Harbeck N, Paradiso A et al. Multicenter validation of a gene expression-based prognostic signature in lymph node-negative primary breast cancer. J Clin Oncol 2006; 24(11):1665-1671. (6) van de Vijver MJ, He YD, van't Veer LJ, Dai H, Hart AA, Voskuil DW et al. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med 2002; 347(25):1999-2009. (7) Wang Y, Klijn JG, Zhang Y, Sieuwerts AM, Look MP, Yang F et al. Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet 2005; 365(9460):671-679. (8) Paik S, Tang G, Shak S, Kim C, Baker J, Kim W et al. Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor-positive breast cancer. J Clin Oncol 2006; 24(23):37263734. (9) Goncalves A, Esterni B, Bertucci F, Sauvan R, Chabannon C, Cubizolles M et al. Postoperative serum proteomic profiles may predict metastatic relapse in high-risk primary breast cancer patients receiving adjuvant chemotherapy. Oncogene 2006; 25(7):981-989. (10) Ricolleau G, Charbonnel C, Lode L, Loussouarn D, Joalland MP, Bogumil R et al. Surface-enhanced laser desorption/ionization time of flight mass spectrometry protein profiling identifies ubiquitin and ferritin light chain as prognostic biomarkers in node-negative breast cancer tumors. Proteomics 2006; 6(6):19631975. (11) Rodenhuis S, Bontenbal M, Beex LV, Wagstaff J, Richel DJ, Nooij MA et al. High-dose chemotherapy with hematopoietic stem-cell rescue for high-risk breast cancer. N Engl J Med 2003; 349(1):7-16. (12) Tolson J, Bogumil R, Brunst E, Beck H, Elsner R, Humeny A et al. Serum protein profiling by SELDI mass spectrometry: detection of multiple variants of serum amyloid alpha in renal cancer patients. Lab Invest 2004; 84(7):845-856. (13) Langlois MR, Delanghe JR. Biological and clinical significance of haptoglobin polymorphism in humans. Clin Chem 1996; 42(10):1589-1600. 138 Prognostic serum protein profiles for breast cancer (14) Bowman BH, Kurosky A. Haptoglobin: the evolutionary product of duplication, unequal crossing over, and point mutation. Adv Hum Genet 1982; 12:189-4. (15) Dobryszycka W. Biological functions of haptoglobin--new pieces to an old puzzle. Eur J Clin Chem Clin Biochem 1997; 35(9):647-654. (16) Wassell J. Haptoglobin: function and polymorphism. Clin Lab 2000; 46(11-12):547-552. (17) Cid MC, Grant DS, Hoffman GS, Auerbach R, Fauci AS, Kleinman HK. Identification of haptoglobin as an angiogenic factor in sera from patients with systemic vasculitis. J Clin Invest 1993; 91(3):977-985. (18) de Castro JG, Puglisi F, de Azambuja E, El Saghir NS, Awada A. Angiogenesis and cancer: A cross-talk between basic science and clinical trials (the "do ut des" paradigm). Crit Rev Oncol Hematol 2006; 59(1):40-50. (19) Maeda N. Nucleotide sequence of the haptoglobin and haptoglobin-related gene pair. The haptoglobinrelated gene contains a retrovirus-like element. J Biol Chem 1985; 260(11):6698-6709. (20) Bharti A, Ma PC, Maulik G, Singh R, Khan E, Skarin AT et al. Haptoglobin alpha-subunit and hepatocyte growth factor can potentially serve as serum tumor biomarkers in small cell lung cancer. Anticancer Res 2004; 24(2C):1031-1038. (21) Rai AJ, Gelfand CA, Haywood BC, Warunek DJ, Yi J, Schuchard MD et al. HUPO Plasma Proteome Project specimen collection and handling: towards the standardization of parameters for plasma proteome samples. Proteomics 2005; 5(13):3262-3277. (22) Ye B, Cramer DW, Skates SJ, Gygi SP, Pratomo V, Fu L et al. Haptoglobin-alpha subunit as potential serum biomarker in ovarian cancer: identification and characterization using proteomic profiling and mass spectrometry. Clin Cancer Res 2003; 9(8):2904-2911. (23) Kuhajda FP, Piantadosi S, Pasternack GR. Haptoglobin-related protein (Hpr) epitopes in breast cancer as a predictor of recurrence of the disease. N Engl J Med 1989; 321(10):636-641. (24) Kuhajda FP, Katumuluwa AI, Pasternack GR. Expression of haptoglobin-related protein and its potential role as a tumor antigen. Proc Natl Acad Sci U S A 1989; 86(4):1188-1192. (25) Suleiman M, Aronson D, Asleh R, Kapeliovich MR, Roguin A, Meisel SR et al. Haptoglobin polymorphism predicts 30-day mortality and heart failure in patients with diabetes and acute myocardial infarction. Diabetes 2005; 54(9):2802-2806. (26) Nakhoul FM, Zoabi R, Kanter Y, Zoabi M, Skorecki K, Hochberg I et al. Haptoglobin phenotype and diabetic nephropathy. Diabetologia 2001; 44(5):602-604. (27) Levy AP, Hochberg I, Jablonski K, Resnick HE, Lee ET, Best L et al. Haptoglobin phenotype is an independent risk factor for cardiovascular disease in individuals with diabetes: The Strong Heart Study. J Am Coll Cardiol 2002; 40(11):1984-1990. (28) Delanghe JR, Langlois MR, Boelaert JR, Van Acker J, Van Wanzeele F, van der GG et al. Haptoglobin polymorphism, iron metabolism and mortality in HIV infection. AIDS 1998; 12(9):1027-1032. (29) Kasvosve I, Gomo ZA, Mvundura E, Moyo VM, Saungweme T, Khumalo H et al. Haptoglobin polymorphism and mortality in patients with tuberculosis. Int J Tuberc Lung Dis 2000; 4(8):771-775. (30) Depypere HT, Langlois MR, Delanghe JR, Temmerman M, Dhont M. Haptoglobin polymorphism in patients with preeclampsia. Clin Chem Lab Med 2006; 44(8):924-928. (31) Ransohoff DF. Bias as a threat to the validity of cancer molecular-marker research. Nat Rev Cancer 2005; 5(2):142-149. (32) Ransohoff DF. Lessons from controversy: ovarian cancer screening and serum proteomics. J Natl Cancer Inst 2005; 97(4):315-319. (33) Linke RP. Typing and subtyping of haptoglobin from native serum using disc gel electrophoresis in alkaline buffer: application to routine screening. Anal Biochem 1984; 141(1):55-61. 139 Chapter 3.3 (34) Somorjai RL, Dolenko B, Baumgartner R. Class prediction and discovery using gene microarray and proteomics mass spectroscopy data: curses, caveats, cautions. Bioinformatics 2003; 19(12):1484-1491. (35) Belluco C, Petricoin EF, Mammano E, Facchiano F, Ross-Rucker S, Nitti D et al. Serum Proteomic Analysis Identifies a Highly Sensitive and Specific Discriminatory Pattern in Stage 1 Breast Cancer. Ann Surg Oncol 2007; 14(9):2470-2476. (36) Ransohoff DF. Discovery-based research and fishing. Gastroenterology 2003; 125(2):290. 140 Chapter Post-operative serum proteomic profiles may predict recurrence free survival in high-risk primary breast cancer Marie-Christine W. Gast Marc Zapatka Jan H.M. Schellens Jos H. Beijnen Interim analysis 3.4 Chapter 3.4 Abstract Better breast cancer prognostication may improve selection of patients for adjuvant therapy. We conducted a retrospective follow-up study in which we investigated sera of high-risk primary breast cancer patients, to search for proteins predictive of recurrence free survival. Sera of 82 breast cancer patients procured after surgery, but prior to the administration of adjuvant therapy, were fractionated using anion-exchange chromatography, to facilitate detection of the low abundant serum proteome. Selected fractions were subsequently analysed by surface-enhanced laser desorption/ionisation time-of-flight mass spectrometry (SELDI-TOF MS), and resulting protein profiles were searched for prognostic markers by appropriate bioinformatics tools. Four peak clusters (i.e., m/z 3073, m/z 3274, m/z 4405, and m/z 7973) were found to bear significant prognostic value (p ≤ 0.01). The m/z 3274 candidate marker was structurally identified as inter-alpha-trypsin inhibitor heavy chain 4 fragment658-688 in serum. Except for the m/z 7973 peak cluster, these peaks remained independently associated to recurrence free survival upon multivariate Cox regression analysis, including clinical parameters of known prognostic value in this study population. Hence, investigation of the postoperative serum proteome by e.g., anion-exchange fractionation followed by SELDITOF MS analysis, is promising for the detection of novel prognostic factors. However, regarding the rather limited study population, validation of these results by analysis of independent study populations is warranted to assess the true clinical applicability of discovered prognostic markers. In addition, structural identification of the other markers will aid in elucidation of their role in breast cancer prognosis, as well as enable development of absolute quantitative assays. 144 Prognostic serum protein profiles for breast cancer Introduction Breast cancer is at present the most commonly diagnosed neoplasm among women in the USA (1). In addition, despite the substantial progress made in cancer therapy, breast cancer is the second leading cause of female cancer deaths, following lung cancer (1). The main prognostic factors currently used to determine eligibility for administration of adjuvant therapy include both clinical and pathological parameters, e.g., patient’s age at diagnosis, tumour size, lymph node status, grade of malignancy, and hormone-receptor and Her2/neu receptor status (the latter two being predictive factors as well) (2). However, despite appropriate locoregional treatment and adjuvant therapy, 30-50% of breast cancer patients will develop metastatic relapse and die (3), while there is a small percentage of patients that would have survived without adjuvant chemo- and hormonal therapy. Evidently, currently applied prognostic markers do not suffice for precise risk-group determination in breast cancer. This failure most likely originates in the high molecular heterogeneity of breast cancer pathogenesis and progression, which the currently used prognostic parameters clearly cannot fully address. Improved prognostic markers that might help to reduce both over- and undertreatment of the disease are thus urgently needed. In search for these markers, investigators from our institutes have published gene expression profiles in tumour tissue that outperformed all prognostic parameters in predicting disease outcome (i.e., distant metastases) (4-7). Nonetheless, it is currently understood that the functional “end-unit” of the genome, i.e., the proteome, might have greater ability in reflecting the molecular complexity of (breast) cancer. Covering posttranslational and post-transcriptional modifications, the proteome reflects both the intrinsic genetic programme of the cell and the impact of its immediate environment, providing a highly dynamic and accurate view of a biological status (8), and hence, a rich and complementary source of potential biomarkers. One of the proteomic technologies used extensively in the search for novel markers is surface-enhanced laser desorption/ionisation time-of-flight mass spectrometry (SELDITOF MS) (9). Combining retention chromatography with laser desorption/ionisation MS instrumentation, this platform has enabled high-throughput mass profiling of highly complex biological samples, such as tissue lysates and serum. Thus far, only two studies have reported the use of SELDI-TOF MS for discovery of prognostic breast cancer markers (10;11). Ricolleau et al. (11) investigated tumour cytosolic extracts of 60 breast cancer patients, and identified ubiquitin and ferritin light chain to be associated with prognosis. Goncalves at al. (10), on the other hand, investigated serum, being an easier accessible biological matrix that provides a good reflection of the human proteome as it perfuses all tissues of the body. Following SELDI-TOF MS analysis of fractionated sera, they constructed a multiprotein model consisting of 40 proteins, correctly predicting relapse in 67 of 81 patients (10). Our research group has previously performed a prognostic SELDI-TOF MS study in serum as well (12). Although we 145 Chapter 3.4 initially discovered the haptoglobin phenotype to be a strong, independent, prognostic parameter in high-risk primary breast cancer (n = 63), this result most likely was false positive, as it was not confirmed following analysis of our validation sample set (n = 371) (12). In contrast to the study of Goncalves et al. (10), we investigated raw, unfractionated sera in our previous study. While only 22 proteins comprise more than 99% of the human serum proteome, the low abundant proteins make up for the remaining < 1% (13). This large dynamic range of proteins in crude serum hampers detection of the allegedly high-informative low abundant serum proteins. Serum fractionation, however, is likely to facilitate detection of the low abundant proteins through reduction of this dynamic range (14). We therefore aimed to specifically explore the low abundant serum proteome for the presence of markers that can be applied in the prognostication of breast cancer. To this end, sera of 82 breast cancer patients procured after surgery, but prior to the administration of adjuvant therapy, were fractionated using anion-exchange chromatography. Selected fractions were subsequently analysed by SELDI-TOF MS, and resulting protein profiles were searched for prognostic markers by appropriate bioinformatics tools. Materials and Methods Study population From 1993 to 1999, high-risk primary breast cancer patients who had undergone modified radical mastectomy or breast conserving surgery with complete axillary clearance participated in a randomised multicentre phase III trial. This study investigated the benefit of high-dose adjuvant chemotherapy in patients with ≥ 4 axillary lymph node metastases. The design of the study has been described elsewhere (15). Major eligibility criteria were histologically confirmed stage 2A, 2B or 3A breast cancer with at least 4 tumour-positive axillary lymph nodes, but no evidence of distant metastases, age under 56 years, and no previous other malignancies. In the current study, sera of 82 study patients who were treated in the Erasmus Medical Center - Daniel den Hoed Cancer Center (Erasmus: n = 24), or in the Radboud University Medical Center Nijmegen (Radboud: n = 58) were included. Sera were procured after surgery (13-55 days), but prior to the administration of adjuvant chemotherapy (0-41 days), and all sera were stored at -80°C. All serum samples were obtained with medical-ethics approval and all patients gave informed consent. Chemicals All used chemicals were obtained from Sigma, St. Louis, MO, USA, unless stated otherwise. 146 Prognostic serum protein profiles for breast cancer Serum fractionation Sera were fractionated manually using a strong anion exchange Q ceramic resin (BioRad Labs, Hercules, CA, USA), according to manufacturers’ protocol. Briefly, sera (20 µl) were denatured in 9 M urea / 2% 3[(3-cholamidepropyl)-dimethylammonio]propane sulfonate (CHAPS), after which they were randomly allocated in duplicate to two 96-well ProteinChip Q filtration plates, prefilled with Q ceramic HyperD F resin (Bio-Rad Labs). In addition, one serum sample was randomly assigned to 12 different wells of each fractionation plate for quality control purposes. Following incubation (30 min), the flow through was collected using a vacuum manifold (Millipore, Billerica, MA, USA). Bound proteins were subsequently eluted with a stepwise pH gradient using wash buffers ranging from pH 9 to pH 3, followed by an organic buffer for elution of remaining proteins. As a result, 6 serum fractions (F) were obtained, i.e., F1 (flowthrough plus pH 9), F2 (pH 7), F3 (pH 5), F4 (pH 4), F5 (pH 3), and F6 (organic buffer). Prior to protein profiling, fractions were stored overnight at +4°C. SELDISELDI-TOF MS protein profiling Protein profiling of serum fractions was performed using the ProteinChip SELDI (PCS 4000) Reader (Bio-Rad Labs). Various array chemistries and fractions were initially evaluated to determine which combination provided the best protein profiles in terms of number and resolution of proteins. Following assay optimisation, we selected Immobilized Metal Affinity Capture (IMAC30) arrays for analysis of F3 and F4, and weak cation exchange (CM10) arrays for analysis of F5 and F6. Throughout the manual assay, arrays were assembled in a 96-well bioprocessor, which was shaken on a MicroMix 5 platform shaker (DPC Cirrus Inc., Los Angeles, CA, USA) at setting 20/7. IMAC30 arrays were charged with 50 µl 100 mM copper sulphate (Merck, Darmstadt, Germany) for 10 min, followed by neutralisation (5 min) with 200 µl 100 mM sodium acetate buffer pH 4. Next, both IMAC30 and CM10 arrays were equilibrated twice for 5 min with 200 µl of their respective binding buffers (IMAC30: 0.01 M phosphate buffered saline pH 7.4 / 0.5 M sodium chloride (Merck), CM10: 20 mM sodium acetate pH 4). Arrays were subsequently loaded with 85 µl binding buffer and 15 µl of the fractionated sample. After incubation (30 min), arrays were washed three times with 200 µl binding buffer, and following a quick rinse with MilliQ water (Millipore), arrays were air-dried. A 50% sinapinic acid (Bio-Rad Labs) solution in 50% acetonitrile (Labscan Ltd., Dublin, Ireland) / 0.5% trifluoroacetic acid (Merck) was applied twice (1.0 µl) to the arrays as the matrix. Following air-drying, the arrays were analysed using the ProteinChip SELDI (PCS 4000) Reader. Data were collected between 0 and 300 kDa, averaging 530 laser shots with 3500 nJ intensity, at focus mass 7.5 kDa and matrix attenuation 1000 Da. For mass accuracy, the instrument was calibrated on the day of measurements with All-in-One protein standard (Bio-Rad Labs). 147 Chapter 3.4 Statistics and bioinformatics Mass spectrometry data were processed using the tbimass R-package (www.rproject.org, publication in preparation). After pre-processing (resampling, baseline correction, normalization, alignment correction), peaks were recognised using PROcess (www.bioconductor.org) on the mean spectra of each experimental group (fraction/ ProteinChip array type). For discovery of peak clusters with significant prognostic value, a subpopulation (n = 68) containing patients diagnosed with a recurrence within 36 months of follow-up (n = 32) and patients experiencing no recurrence after a followup of at least 48 months (n = 36) were extracted from the total study population. Investigating this subpopulation using Cox proportional hazards analysis, the peak clusters associated with recurrence were identified within all peaks of the combined data of all fractions/ProteinChip array types. For selection, a stepwise method was applied (i.e., stepBIC), an algorithm sequentially searching through all possible Cox proportional hazard models for the one that minimises the Bayesian Information Criterion (BIC). Recurrence free survival was calculated from the date of randomisation to the time of first recurrence or death, or the date of last follow-up. Clinical parameters were selected on known impact on recurrence in the total study population, to prevent overfitting of the data to the model. To this end, a Cox proportional hazards analysis was performed including the known clinical parameters presented in Table 1, based on forward entry (p < 0.05). In addition, the clinical parameter ‘treatment’ was selected to correct for the different treatment arms of the original clinical trial. A Cox proportional hazards model was subsequently build on the total study population, by inclusion of the relevant clinical parameters only. To investigate whether the relationship between peak intensities and recurrence free survival could be explained by any of the relevant clinical parameters, the hazard ratios were adjusted for these clinical parameters by construction of a Cox proportional hazards model on the total study population, incorporating the selected peak clusters and the relevant clinical parameters. Since our study population originated from two different hospitals that allegedly used different sample collection protocols, our results could have been influenced by various pre-analytical factors. The influence of the different collection protocols on the SELDITOF MS protein profiles was investigated by multidimensional scaling of the SELDITOF MS spectra. Herewith, the degree of similarity or dissimilarity between the samples withdrawn at the two different hospitals is graphically expressed: points representing similarity tend to cluster together, while points representing dissimilarity tend to be far apart. The influence of collection center on the protein profile was furthermore investigated by Cox proportional hazards analysis for each peak cluster separately, incorporating one peak cluster, relevant clinical parameters and collection center. 148 Prognostic serum protein profiles for breast cancer The reproducibility of the assay was assessed by analysis of one quality control serum sample, fractionated 24 times by random assignment to 12 different wells of each of the 2 fractionation plates. Within the quality control spectra, all peaks with a signal-tonoise ratio (S/N) ≥ 2 were detected, after which the coefficient of variation was calculated on the corresponding peak intensities. All statistical tests were two-sided, and p < 0.05 was considered statistically significant. Peptide identification For identification purposes, peptides of interest were extracted from serum(fractions) by reversed-phase C18 magnetic beads (Dynabeads RPC18, Invitrogen, Breda, The Netherlands) using a Kingfisher 96 pipetting robot (Thermo Fisher Scientific, Waltham, MA, USA), according to the optimized protocol described in (16). Briefly, sera were diluted in TFA 0.1%, after which the peptide content was bound to the beads. The beads were subsequently washed with 0.1% TFA, and eluted with 50% ACN. Eluate (1 µl) was mixed with α-cyano-4-hydroxy-cinnamic acid matrix (2 µl), after which the mixture was spotted (0.7 µl) on a MALDI target plate. Analyses were performed on a 4800 MALDI-TOF/TOF mass spectrometer (Applied Biosystems, Forster City, CA, USA). Fragment ion spectra were taken to search the NCBI 20081128 database (Homo sapiens: 216937 sequences) using the MASCOT search engine at http://www.matrixscience.com (Matrix Science Ltd., London, UK), with the following search parameters: monoisotopic precursor mass tolerance: 18 ppm, fragment mass tolerance: 1 Da, variable modifications: methionine oxidation, and no specified protease cleavage site. Results Study population At time of analysis, 45 patients (Erasmus: 19 pts, Radboud: 26 pts) had a recurrence or had died and 37 patients (Erasmus: 5 pts, Radboud: 32 pts) were censored at a median follow-up of 6.5 years (Erasmus: 7.8 years, Radboud: 6.3 years). Patient characteristics are provided in Table 1. All patient characteristics were similarly distributed between the samples obtained from the Erasmus Medical Center and the Radboud University Medical Center, as determined by the Chi-square test or the Mann-Whitney U test. SELDI--TOF MS protein profiling SELDI After selection of the peak clusters with stepBIC, a Cox proportional hazards model on combined peak clusters identified in all measured fractions/ProteinChip arrays was build. Out of the 400 peak clusters tested for inclusion into the model, four peak clusters (m/z 3073 (F4/IMAC30), m/z 3273 (F4/IMAC30), m/z 4404 (F6/CM10), m/z 149 Chapter 3.4 7973 (F5/CM10)) were selected due to their significant association with recurrence free survival in the subpopulation (Table 2). Table 1 Patient and tumour characteristics of the study population. Erasmus (n = 24) n (%) Radboud (n = 58) n (%) Total (n = 82) n (%) Age, mean [range] < 40 years ≥ 40 years 43.5 [26-54] 6 (25%) 18 (75%) 43.2 [28-54] 15 (26%) 43 (74%) 43.3 [26-54] 21 (26%) 61 (74%) Menopausal status Premenopausal Postmenopausal Unknown 21 3 0 (88%) (12%) (0%) 52 5 1 (90%) (9%) (1%) 73 8 1 (89%) (10%) (1%) Mastectomy Breast conserving 15 9 (63%) (37%) 40 18 (69%) (31%) 55 27 (67%) (33%) Conventional dose High dose 13 11 (54%) (46%) 30 28 (52%) (48%) 43 39 (52%) (48%) 15 9 (63%) (37%) 38 20 (66%) (34%) 53 29 (65%) (35%) T1 (< 2 cm) T2 (2-5 cm) T3 (≥ 5 cm) 7 13 4 (29%) (54%) (17%) 17 34 7 (29%) (59%) (12%) 24 47 11 (29%) (57%) (14%) Negative Positive Unknown 13 10 1 (54%) (42%) (4%) 37 19 2 (64%) (33%) (3%) 50 29 3 (61%) (35%) (4%) Oestrogen receptor status ER negative ER positive Unknown 9 14 1 (38%) (58%) (4%) 23 35 0 (40%) (60%) (0%) 32 49 1 (39%) (60%) (1%) Progesterone receptor status PR negative PR positive Unknown 10 13 1 (42%) (54%) (4%) 24 34 0 (41%) (59%) (0%) 34 47 1 (42%) (57%) (1%) Bloom-Richardson grade Grade I Grade II Grade III Unknown 0 6 17 1 (0%) (25%) (71%) (4%) 7 18 32 1 (12%) (31%) (55%) (2%) 7 24 49 2 (9%) (29%) (60%) (2%) Patient characteristics Surgery Treatment Tumour characteristics Number of positive lymph nodes 4-9 ≥ 10 Tumour size Her2/neu status 150 Prognostic serum protein profiles for breast cancer Cox proportional hazards analysis on all known clinical parameters based on forward entry revealed the parameters ‘age’, ‘number of positive lymph nodes’, and ‘progesterone receptor status’ to be significantly associated with recurrence free survival in the total population. In addition, the parameter ‘treatment’ was included to correct for the different treatment arms of the original clinical trial. In the Cox proportional hazards model constructed on the total population by inclusion of the relevant clinical parameters, age (≥ 40 years, p = 0.021), number of positive lymph nodes (≥ 10, p = 0.012), and progesterone receptor status (positive, p = 0.003) were found significantly associated with recurrence free survival (Table 2). Table 2 Multivariate proportional hazards analyses for the risk of recurrence on selected peak clusters, before (model 1, subpopulation) and after (model 3, total study population) adjustment for relevant clinical parameters, and on relevant clinical parameters solely (model 2, total study population). Parameter Peak cluster m/z 3073 m/z 3273 m/z 4405 m/z 7973 Treatment CONV HD Age < 40 yrs ≥ 40 yrs No. of LN+ ≥ 10 4-9 PR status PR(-) PR(+) Model 1 - peak clusters Model 2 - clinical parameters Model 3 - combined HR (95% CI) p-value HR (95% CI) p-value HR (95% CI) p-value 3.17 10.18 0.02 0.05 (2.03-4.96) (1.99-52.01) (0.01-0.25) (0.01-0.48) < 0.001 0.005 0.003 0.010 2.48 11.71 0.01 0.24 (1.78-3.48) (2.05-66.90) (0.01-0.17) (0.03-1.78) < 0.001 0.006 0.001 0.160 1 1.59 (0.86-2.95) 0.140 1 2.48 (1.24-4.98) 0.011 1 0.44 (0.22-0.88) 0.021 1 0.35 (0.16-0.74) 0.006 1 0.44 (0.23-0.84) 0.012 1 0.37 (0.19-0.72) 0.003 1 0.40 (0.22-0.73) 0.003 1 0.28 0.14-0.55) < 0.001 Abbreviations: BR grade: Bloom-Richarson grade, CONV.: conventional dose arm, HD: high dose arm, LN+: number of positive lymph nodes, PR: progesterone receptor status positive (+) and negative (-). To investigate whether the prognostic value of the four peak clusters was independent from confounding effects due to differences in the clinical parameters, a combined Cox proportional hazards model including the four selected peaks and all relevant clinical parameters was build on the total study population. Three of the four selected peak clusters (i.e., m/z 3072, m/z 3273, and m/z 4404) remained significantly associated with recurrence free survival in combination with the clinical variables (Table 2). Furthermore, using multidimensional scaling, we investigated the influence of the different collection protocols (allegedly used by the two hospitals) on the SELDI-TOF MS serum protein profiles. As depicted in Figure 1 for F4/CM10, spectra of the sera collected in the Erasmus Medical Center and the Radboud University Medical Center are randomly distributed, indicating no structural differences in the SELDI-TOF MS 151 Chapter 3.4 serum protein profiles of both hospitals. In addition, following Cox proportional hazards analysis including one peak cluster, relevant clinical parameters and collection center, all peak clusters except m/z 7973 remained (borderline) significant (i.e., m/z 3073: HR = 3.44, p = 0.046, m/z 3274: HR = 2.39, p = 0.051, m/z 4405: HR = 0.107, p < 0.001, and m/z 7973: HR = 0.35, p = 0.160). Figure 1 MDS plot of Fraction 4/IMAC30 data (i.e., duplicate spectra) on Center of withdrawal (R: Erasmus, N: Radboud, O: quality control sample). Lastly, the reproducibility of the assay was investigated by calculation of the coefficient of variation of all peak clusters with S/N > 2 detected in the quality control spectra (n = 24 per fraction/ProteinChip array type) (Figure 2). The median coefficient of variation of the peak intensities following fractionation and SELDI-TOF MS analysis ranged from 13.4% to 24.2% for the different fractions/ProteinChip arrays investigated, with an overall average CV of 20.2%. Peptide identification The MALDI serum / serum fraction peptide profiles obtained using C18 magnetic beads were searched for the presence of prognostic SELDI peaks based on mass matching. Due to the different chemistries used for peptide capture for SELDI-TOF MS (IMAC30 Cu) and MALDI-TOF MS (C18), and to the mass limitations for direct fragmentation, we were able to elucidate the identity of one of the four candidate prognostic peak clusters in the spectra of whole serum. The SELDI-TOF MS peak cluster at m/z 3274 was detected by MALDI-TOF/TOF MS as MH+ ions at m/z 3271.69 (default calibration), and identified by MALDI-TOF/TOF MS/MS (Figure 3) in conjunction with database searching as a fragment of inter-alpha-trypsin inhibitor heavy chain 4 (ITIH4658-688), with a MASCOT score of 69 (expect: 0.0025). 152 Prognostic serum protein profiles for breast cancer Figure 2 Coefficient of variation (y-axis) of the peak cluster identified in the quality control sample (x-axis) fractionated on fractionation plate 1 (black) and plate 2 (grey). CM10 F5 CM10 F6 peaks peaks IMAC30 F3 IMAC30 F4 peaks peaks Discussion In the current study, we investigated sera of 82 breast cancer patients procured after surgery, but prior to the administration of adjuvant therapy, in search for novel prognostic biomarkers. To facilitate detection of the low-abundant serum proteome, sera were fractionated using anion-exchange chromatography, after which selected fractions were analysed by SELDI-TOF MS. Resulting protein profiles were searched for prognostic markers by appropriate bioinformatics tools. Considering solely the peak clusters detected in the SELDI-TOF MS protein profiles, four peak clusters (i.e., m/z 3073, m/z 3274, m/z 4405, and m/z 7973) were found to bear significant prognostic value. The m/z 3274 candidate marker was structurally identified as ITIH4658-688 in serum. Moreover, except for the m/z 7973 peak cluster, these peaks remained independently associated to recurrence free survival upon multivariate Cox regression analysis, including clinical parameters of known prognostic value in this study population. Hence, investigation of the post-operative serum proteome by e.g., anionexchange fractionation followed by SELDI-TOF MS analysis, is promising for the detection of novel prognostic factors. However, regarding the rather limited study population, validation of our results by analysis of similar, prospectively collected, independent, study populations is warranted to assess the true clinical applicability of identified prognostic markers. In addition, structural identification of the other markers will aid in elucidation of their role in breast cancer prognosis, as well as enable development of absolute quantitative assays (e.g., (17)). 153 Chapter 3.4 Figure 3 Annotated MALDI-TOF/TOF MS/MS spectrum of m/z 3271.69. 100 90 119.5 70.0738 b14 2120.0483 80 b23 2431.2012 70 % Intensity b20-NH3 60 50 b23-NH3 2103.0208 110.0566 2414.2258 155.0333 245.9986 b2 40 z22 c19 30 c22 2022.0994 120.0500 84.0851 129.0643 b9 y9 20 c9 1054.1283 b5 1019.1696 y11 1810.9154 1616.4342 2342.2510 y20 2184.1443 1245.4258 MH-NH3 3256.5027 2333.2383 y20 2724.3333 10 0 9.0 698.4 1387.8 Mass (m/z) 2077.2 2766.6 3456.0 Metastases are thought to arise from clinically undetectable residual or micrometastatic disease, activated by a.o. stroma-generated growth factors, early impediment of immune surveillance and enhancement of angiogenesis (18-21). These early post-surgical host reponse processes are potentially affected by surgical extirpation of the tumour, as this disrupts the intricate interactions between malignant cells and physiological tumourcontrol mechanisms (22;23). Hence, the early post-operative serum proteome can bear prognostic information, since it reflects the host response processes that can play a key role in metastatic progression. The candidate prognostic markers detected in the current study therefore most likely correlate with this post-operative host response. In addition, since all study participants were treated with adjuvant chemotherapy, these differentially expressed proteins may also relate to the tumour phenotype and its chemosensitivity. Nonetheless, the four candidate markers could also arise directly from residual or micrometastatic disease. Considering the nadir in tumour burden following surgery, however, serum concentrations of tumour-secreted proteins most likely are well below the detection limit of the SELDI-TOF MS platform, even following serum fractionation. Lastly, the four candidate prognostic markers can also result from tumour-secreted proteases that process host-response proteins upon their exposure to the tumour microenvironment (24;25). Since these modified host response proteins generally are present at substantially higher circulatory concentrations than the 154 Prognostic serum protein profiles for breast cancer enzymes that process them upon their exposure to the tumour microenvironment, they can be detected in blood by SELDI-TOF MS. This latter hypothesis is in fact endorsed by the structural identity of the candidate m/z 3274 marker, i.e., the inter-alpha-trypsin inhibitor heavy chain 4658-688 fragment, identified in serum. We previously found serum levels of this fragment decreased in breast cancer compared to control (Gast et al., submitted). Other studies have detected this fragment in serum as well, reporting either a lack of discriminative value (24;25), or an increase in breast cancer compared to control (26). Most likely, these contradictory findings originate from the heterogeneity of the different study populations investigated, or from the postulated instability of ITIH4 fragments (24;26;27). In addition, changes in the abundance of the m/z 3274 ITIH4 fragment have been found associated to various types of cancer (e.g., prostate, breast, ovarian, colorectal, and pancreatic cancer) (24-26). This evident lack of specificity does not hamper its use as prognostic marker, however. The various serum ITIH4 fragments are currently hypothesised to result from tumour-secreted proteases that process host response proteins upon their exposure to the tumour microenvironment (24;26;28). Hence, in the current study, the m/z 3274 marker could well originate from proteolytic activity associated with residual (micrometastatic) disease. According to this hypothesis, this candidate prognostic marker might possibly be applied in other malignancies as well, as the protease activity has been shown to be cancer-type specific (24-26). Hence, future validation studies should also include other types of malignancies. Structural identification is imperative to investigate origin and function of the other three candidate biomarkers. In addition, concerning the rather limited study population, results must be validated by analysis of an independent, similar, sample set. Such validation sets may prove difficult to obtain, however, regarding the extended follow-up window needed to reliably investigate breast cancer prognosis. Fortunately, cross validation can already offer some indication of the generalisibility of the classification model. For instance, the performance of the multiprotein index constructed by Goncalves et al. (10) declined from 83% during training, to 72% after cross-validation. This drop in performance indicates probable overfitting of the data, which most likely is caused by the high number of proteins used for classification (n = 40) compared to the limited study population (n = 81). Conversely, we included only 4 protein peaks (and the 4 clinical parameters that best predicted recurrence free survival) in our model to purposely preclude overfitting of the data to the model. Hence, despite the lack of an independent validation set, we hypothesise this model to be generalisible to similar, new, study populations. While serum is generated by coagulation, its proteome is prone to the proteases involved in this cascade, as well as to those involved in the complement cascade, activated upon clotting. Various pre-analytical parameters, such as sampling device, clotting temperature, and storage time, can thus all exert a distinct influence on the serum proteome. Since our study populations originated from two different hospitals 155 Chapter 3.4 that allegedly used different sample collection protocols, our results could have been influenced by the various pre-analytical factors. However, as depicted in Figure 2, we did not observe such an influence on the protein profiles, indicating that the investigated serum proteome most likely is rather robust to (small) differences in collection protocols. Moreover, despite the different characteristics of the two study groups, all peak clusters except m/z 7973 remained (borderline) significant after inclusion of the collection center in the Cox proportional hazards model. The three peaks are therefore of additional prognostic value, even if the different collection centers are taken into account. The reliability of our results is furthermore endorsed by the reproducibility of the assay (average CV: 20.2%), which is well in agreement to previous reports (10;29). Conclusion In conclusion, using serum anion-exchange fractionation in combination with SELDITOF MS analysis, we discovered 4 peak clusters, one of which identified as serum ITIH4658-688, with significant prognostic value in a study population of 82 high-risk primary breast cancer patients. Three peak clusters (including ITIH4658-688) remained significantly associated to recurrence free survival following inclusion of clinical parameters. These results are promising, as the prognostic profile identified in the current study could eventually improve therapeutic accuracy. However, the rather limited study population requires extension and re-assessment in other, similar, study populations, to confirm the performance of identified prognostic peak clusters. In addition, the precise roles of the candidate markers in breast cancer remain to be elucidated, as this could help in the identification of new molecular targets. Hence, enabling development of absolute quantitative assays as well, structural identification of the candidate markers is warranted. Lastly, profiling of other serum fractions on different types of ProteinChip arrays might reveal more protein peaks with prognostic value. Acknowledgement This study was supported by a grant of the Dutch Cancer Society (project NKI 20053421). We gratefully acknowledge Annemieke van Winden for help with serum fractionation. 156 Prognostic serum protein profiles for breast cancer References (1) Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T et al. Cancer statistics, 2008. CA Cancer J Clin 2008; 58(2):71-96. (2) Goldhirsch A, Wood WC, Gelber RD, Coates AS, Thurlimann B, Senn HJ. Meeting highlights: updated international expert consensus on the primary therapy of early breast cancer. J Clin Oncol 2003; 21(17):3357-3365. (3) Polychemotherapy for early breast cancer: an overview of the randomised trials. Early Breast Cancer Trialists' Collaborative Group. Lancet 1998; 352(9132):930-942. (4) 't Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature 2002; 415(6871):530-536. (5) Foekens JA, Atkins D, Zhang Y, Sweep FC, Harbeck N, Paradiso A et al. Multicenter validation of a gene expression-based prognostic signature in lymph node-negative primary breast cancer. J Clin Oncol 2006; 24(11):1665-1671. (6) van de Vijver MJ, He YD, van't Veer LJ, Dai H, Hart AA, Voskuil DW et al. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med 2002; 347(25):1999-2009. (7) Wang Y, Klijn JG, Zhang Y, Sieuwerts AM, Look MP, Yang F et al. Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet 2005; 365(9460):671-679. (8) Banks RE, Dunn MJ, Hochstrasser DF, Sanchez JC, Blackstock W, Pappin DJ et al. Proteomics: new perspectives, new biomedical opportunities. Lancet 2000; 356(9243):1749-1756. (9) Hutchens TW, Yip TT. New desorption strategies for the mass spectrometric analysis of macromolecules. Rapid Commun Mass Spectrom 1993; 7:576-580. (10) Goncalves A, Esterni B, Bertucci F, Sauvan R, Chabannon C, Cubizolles M et al. Postoperative serum proteomic profiles may predict metastatic relapse in high-risk primary breast cancer patients receiving adjuvant chemotherapy. Oncogene 2006; 25(7):981-989. (11) Ricolleau G, Charbonnel C, Lode L, Loussouarn D, Joalland MP, Bogumil R et al. Surface-enhanced laser desorption/ionization time of flight mass spectrometry protein profiling identifies ubiquitin and ferritin light chain as prognostic biomarkers in node-negative breast cancer tumors. Proteomics 2006; 6(6):19631975. (12) Gast MC, van Tinteren H, Bontenbal M, van Hoesel QC, Nooij MA, Rodenhuis S et al. Haptoglobin phenotype is not a predictor of recurrence free survival in high-risk primary breast cancer patients. BMC Cancer 2009; 8:389. (13) Anderson NL, Anderson NG. The human plasma proteome: history, character, and diagnostic prospects. Mol Cell Proteomics 2002; 1(11):845-867. (14) Hoffman SA, Joo WA, Echan LA, Speicher DW. Higher dimensional (Hi-D) separation strategies dramatically improve the potential for cancer biomarker detection in serum and plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2007; 849(1-2):43-52. (15) Rodenhuis S, Bontenbal M, Beex LV, Wagstaff J, Richel DJ, Nooij MA et al. High-dose chemotherapy with hematopoietic stem-cell rescue for high-risk breast cancer. N Engl J Med 2003; 349(1):7-16. (16) Jimenez CR, El Filali Z, Knol JC, Hoekman K, Kruyt FAE, Giaccone G et al. Automated serum peptide profiling using novel magnetic C18 beads off-line coupled to MALDI-TOF-MS. Proteomics Clin Appl 2007; 1(6):598-604. (17) Broek van den I, Sparidans RW, Schellens JH, Beijnen JH. Liquid chromatography/tandem mass spectrometric method for the quantification of eight proteolytic fragments of ITIH4 with biomarker potential in human plasma and serum. Rapid Commun Mass Spectrom 2008; 22(18):2915-2928. (18) Demicheli R, Retsky MW, Swartzendruber DE, Bonadonna G. Proposal for a new model of breast cancer metastatic development. Ann Oncol 1997; 8(11):1075-1080. 157 Chapter 3.4 (19) Demicheli R, Retsky MW, Hrushesky WJ, Baum M, Gukas ID. The effects of surgery on tumor growth: a century of investigations. Ann Oncol 2008; 19(11):1821-1828. (20) Heimann R, Hellman S. Individual characterisation of the metastatic capacity of human breast carcinoma. Eur J Cancer 2000; 36(13 Spec No):1631-1639. (21) Pupa SM, Menard S, Forti S, Tagliabue E. New insights into the role of extracellular matrix during tumor onset and progression. J Cell Physiol 2002; 192(3):259-267. (22) Fisher B, Gunduz N, Coyle J, Rudock C, Saffer E. Presence of a growth-stimulating factor in serum following primary tumor removal in mice. Cancer Res 1989; 49(8):1996-2001. (23) Tagliabue E, Agresti R, Carcangiu ML, Ghirelli C, Morelli D, Campiglio M et al. Role of HER2 in woundinduced breast carcinoma proliferation. Lancet 2003; 362(9383):527-533. (24) Fung ET, Yip TT, Lomas L, Wang Z, Yip C, Meng XY et al. Classification of cancer types by measuring variants of host response proteins using SELDI serum assays. Int J Cancer 2005; 115(5):783-789. (25) Villanueva J, Shaffer DR, Philip J, Chaparro CA, Erdjument-Bromage H, Olshen AB et al. Differential exoprotease activities confer tumor-specific serum peptidome patterns. J Clin Invest 2006; 116(1):271284. (26) Song J, Patel M, Rosenzweig CN, Chan-Li Y, Sokoll LJ, Fung ET et al. Quantification of fragments of human serum inter-alpha-trypsin inhibitor heavy chain 4 by a surface-enhanced laser desorption/ionization-based immunoassay. Clin Chem 2006; 52(6):1045-1053. (27) Timms JF, Arslan-Low E, Gentry-Maharaj A, Luo Z, T'Jampens D, Podust VN et al. Preanalytic influence of sample handling on SELDI-TOF serum protein profiles. Clin Chem 2007; 53(4):645-656. (28) Villanueva J, Philip J, Chaparro CA, Li Y, Toledo-Crow R, DeNoyer L et al. Correcting common errors in identifying cancer-specific serum peptide signatures. J Proteome Res 2005; 4(4):1060-1072. (29) Albrethsen J. Reproducibility in protein profiling by MALDI-TOF mass spectrometry. Clin Chem 2007; 53(5):852-858. 158 Chapter Protein profiling of tissue 4 Chapter Detection of breast cancer by SELDI-TOF MS tissue and serum protein profiling Marie-Christine W. Gast Eric J. van Dulken Thea K.G. van Loenen Florine Kingma-Vegter Johan Westerga Claudie C. Flohil Jaco C. Knol Connie R. Jimenez Carla H. van Gils Lodewijk F.A. Wessels Jan H.M. Schellens Jos H. Beijnen Submitted for publication 4.1 Chapter 4.1 Abstract Breast cancer is estimated to be the second leading cause of female cancer deaths in the USA in 2008. Despite the advances made in cancer therapy, early detection remains the best route to decrease overall (breast) cancer mortality. However, current modalities (e.g., mammography) lack adequate performance to be applicable in early detection, and new, improved, markers are urgently needed. In the past decade, novel markers were extensively searched for in the proteome, using a.o. the surface-enhanced laser desorption/ionisation time-of-flight mass spectrometry (SELDI-TOF MS) technology. The majority of SELDI-TOF MS studies have thus far investigated samples originating from biorepositories, which are likely to suffer from variable adherence to collection protocols, thereby hampering biomarker discovery. We therefore investigated breast cancer (n = 75) and control (n = 26) serum and tissue samples, collected prospectively by rigorous adherence to a strictly defined protocol, to discover novel breast cancer associated markers. Sera were collected pre- and post-operatively, and both serum and tissue samples were analysed by SELDI-TOF MS using two different array-types (IMAC30 Ni and Q10 pH 8). Following serum analyses, one Q10 peak cluster (m/z 3939) was found significantly increased in breast cancer compared to control, while two IMAC30 peak clusters (m/z 4292 and m/z 4301) were significantly decreased in expression following surgery of the breast cancer patients. Conversely, proteome analyses at the tumour level yielded 27 peak clusters, discriminative between breast cancer and control tissue. In addition, several peak clusters gradually in- or decreased in intensity from healthy to benign to cancer, or with increasing cancer stage, apparently visualising disease progression. Constructed classification trees had a 10-fold cross validated performance of 67% to 87%. Two tissue peak clusters were identified as N-terminal albumin fragments. These fragments are likely to have been generated by (breast) cancer specific proteolytic activity in the tumour microenvironment. As such, they can potentially provide insight into the pathophysiological mechanisms associated with, or underlying, breast cancer, and aid in improving breast cancer diagnosis. 164 Diagnostic tissue and serum protein profiles for breast cancer Introduction Accounting for 26% of all new cancer cases, breast cancer is estimated to be the most commonly diagnosed neoplasm among women in the USA in 2008 (1). Following lung cancer, it is the second leading cause of USA cancer deaths in the prognosis for 2008 (1). Despite the substantial progress made in cancer therapy, the best route to decrease overall mortality from (breast) cancer is through early detection, as cancer survival is inversely proportional to disease stage at presentation (2). Unfortunately, due to a lack of adequate detection methods, only 63% of breast cancers are confined to the breast at the time of diagnosis (1). Although mammography currently is the most widely applied imaging test, it has only limited predictive value in women with dense breast tissue and small lesions. In addition, established serum tumour markers (e.g., Cancer Antigen 15.3) lack adequate performance to be applicable in early detection, and are thus applied only in monitoring therapy of advanced breast cancer or recurrence (3). Evidently, new biomarkers for reliable detection of breast cancer, either individually or in conjunction with existing modalities, are urgently needed. With cancer being a genetic disease, these markers were initially searched for by the investigation of the cancer genome and transcriptome. It is expected, however, that the proteome will have complementary use in the detection of novel cancer markers (4). Covering post-transcriptional as well as post-translational modifications, the proteome provides a more dynamic and accurate reflection of a biological status, as it mirrors both the intrinsic genetic programme of the cell and the impact of its immediate environment (5). In search for novel cancer markers, the proteome has been investigated by various techniques, including surface-enhanced laser desorption/ionisation time-of-flight mass spectrometry (SELDI-TOF MS) (6). As this platform enables analysis of highly complex biological matrices (e.g., serum, tissue lysates) in a high-throughput fashion, it has been used extensively for discovery of novel serum markers for e.g., breast (7;8), colorectal- (9;10), and renal cell carcinoma (11;12). The majority of SELDI-TOF MS studies published thus far in breast cancer have investigated sera originating from biorepositories (7;8;13-15), allowing timely study progression, since prospective sample collection usually takes years. Biorepositories may, however, vary in their adherence to consistent sample processing protocols over time, potentially affecting SELDI-TOF MS serum protein profiles (16). Although this renders biorepository samples representative of routinely collected “real world” samples, it may also seriously hamper biomarker detection, as breast cancer is a highly complex disease, thereby complicating biomarker discovery in its own right. Hence, in the current study, we aimed to discover novel high-performance SELDI-TOF MS markers for breast cancer detection through comparison of breast cancer and control sera, collected prospectively by rigorous adherence to a strictly defined protocol. Moreover, we also investigated serum proteome transitions occurring after surgery, 165 Chapter 4.1 since monitoring protein expression dynamics in response to surgery may help to better understand breast cancer pathogenesis. Lastly, collection of tissue specimens from each participant during surgery enabled us to perform biomarker discovery directly at the tumour level. Identification of cancer associated proteins at the tumour level might provide us more insight into the pathophysiological mechanisms associated with, or underlying, breast cancer. Materials and methods Study population Women above 18 years old presenting with an indication for (reductive) breast surgery at the Department of (Plastic) Surgery of the Slotervaart Hospital (Amsterdam) were asked for participation in this study. Of the 111 women asked for study participation, 84 were diagnosed with breast cancer (BC). The control group (CON) consisted of 14 women diagnosed with benign breast disease (BBD), and 13 women with healthy breast tissue (healthy control; HC). Ten participants were found ineligible according to the inclusion criteria: 8 participants (BC: n = 7, BBD: n = 1) had prior malignancies, and in 2 participants (BC), sample collection failed. Serum was collected prior to surgery and at least 2 weeks after surgery (but prior to the eventual administration of adjuvant therapy). Of 6 participants, no pre-surgery serum sample was collected (BC: n = 3, BBD: n = 1, HC: n = 2). Post-surgery samples were not available of 10 participants (BC: n = 8, BBD: n = 2), either because collection failed, or collection took place following initiation of adjuvant therapy. Tissue sample collection was not successful in 14 participants (BC: n = 12, HC: n = 2). Serum and tissue collection was done following a strict procedure. According to manufacturers’ protocol, blood samples were collected in 9.5 ml BD Vacutainer tubes (Beckton-Dickinson, Breda, The Netherlands) and allowed to clot for exactly 30 min at room temperature, after which they were centrifuged at 1500 g for 15 min at room temperature. Sera were then immediately aliquotted and stored at -70°C. Tissue was collected dry and tissue sections were snap frozen in liquid nitrogen immediately after collection at the Department of Pathology, and stored in liquid nitrogen until analysis. All serum and tissue samples were collected between April 2005 and December 2007, after approval by the local medical ethics committee, with individuals’ written informed consent. Chemicals All used chemicals were obtained from Sigma, St. Louis, MO, USA, unless stated otherwise. 166 Diagnostic tissue and serum protein profiles for breast cancer Preparation of tissue lysates Snap frozen (whole) tissue sections were disintegrated in deep frozen state by pulverisation with a Mikro-dismembrator II (Sartorius AG, Göttingen, Germany). First, tissues were cut into smaller blocks, placed into a pre-cooled shaking flask with a stainless steel ball and then pulverised in three rounds of shaking (55 sec) and cooling in liquid nitrogen (3 min). About 10 mg of the resulting frozen tissue powder was then added to 100 μl of each of two denaturation buffers (buffer 1: 9 M urea and 2% 3-[(3cholamidopropyl)dimethylammonio-]-1-propanesulfonic acid (CHAPS), and buffer 2: 9 M urea, 2% CHAPS, and 1% dithiotreitol (DTT)). Lysates were stored at -70°C until analysis, for which they were thawed on ice and centrifuged at 15000 rpm for 5 min. The protein concentration of each supernatant was subsequently determined using the 2D-Quant Kit (GE Healthcare, Diegem, Belgium), following manufacturers’ instructions. Serum and tissue protein profiling Serum protein profiling was performed using the ProteinChip SELDI (PBSIIc) Reader (Bio-Rad Labs, Hercules, CA, USA). Various chip chemistries, binding- and washingprocedures and sample pretreatments were initially evaluated to determine which procedure provided the best serum profiles in terms of number and resolution of proteins. Both strong anion exchange (Q10) and Immobilized Metal Affinity Capture (IMAC30) arrays were selected for further analysis. Throughout the assay, arrays were assembled in a 96-well bioprocessor, which was shaken on a platform shaker at 250 rpm. Sample processing was manual, and all serum samples were randomly attributed to one of 3 measurement series (per array type) before analysis. Each series was measured in duplicate on one day and samples were allocated randomly to the arrays. Tissue lysates were analysed in duplicate in one separate series, using the same procedures as for serum. For each sample, the amount of lysate applied to the arrays was adjusted to its protein concentration. For the IMAC30 assay, arrays were charged twice with 50 µl 100 mM nickel sulphate (Merck, Darmstadt, Germany) for 15 min, followed by three rinses with deionised water (Braun, Emmenbrücke, Germany) and two equilibrations with 200 µl Phosphate Buffered Saline (PBS; 0.01 M) pH 7.4 / 0.5 M sodium chloride / 0.1% TritonX-100 (binding buffer; sodium chloride from Merck) for 5 min. For the Q10 assay, arrays were equilibrated twice with 200 µl 20 mM Tris-HCl buffer pH 8.0 / 0.1% TritonX-100 (binding buffer). Unfractionated serum samples were thawed on ice and denatured twice; once for the IMAC30 assay (by 1:10 dilution in 9 M urea / 2% CHAPS), and once for the Q10 assay (by 1:10 dilution in 9 M urea / 2% CHAPS / 1% DTT). Pretreated samples were diluted 1:10 in binding buffer and randomly applied to the arrays. After a 30 min incubation, the arrays were washed twice with binding buffer and twice with PBS pH 7.4 / 0.5 M sodium chloride (IMAC30) or 20 mM Tris-HCl buffer pH 8 (Q10) 167 Chapter 4.1 for 5 min. Following a quick rinse with deionised water, arrays were air-dried. A 50% solution of sinapinic acid (Bio-Rad Labs) in 50% acetonitrile (ACN; Biosolve, Valkenswaard, The Netherlands) / 0.5% trifluoroacetic acid (TFA; Merck) was applied twice (1.0 µl) to the arrays as the matrix. Following air-drying, the array was inserted in a ProteinChip SELDI (PBS IIc) Reader. Using the ProteinChip Software v3.1 (Bio-Rad Labs), data were collected between 0 and 100 kDa, averaging 80 laser shots with intensity 147 (IMAC30) / 140 (Q10), detector sensitivity 5, and focus lag time of 746 ns. Settings for tissue analysis were optimised independently, resulting in an average of 80 laser shots per spectrum at intensity 147 (IMAC30) / 144 (Q10), detector sensitivity 5, and focus lag time of 746 ns. For mass accuracy, the instrument was calibrated on each day of measurements with All-in-One peptide standard (Bio-Rad Labs). Statistics and bioinformatics Spectra of serum samples were processed per measurement series by the ProteinChip Software v3.1 (Bio-Rad Labs). Following baseline subtraction, spectra were normalised to the total ion current. Spectra with normalisation factors < 0.5 or > 2 were excluded from further analysis. Next, the spectra of the three measurement series were merged in one experiment file, and the Biomarker Wizard (BMW) software package was applied for peak detection. Peaks were auto-detected when occurring in at least 15% of spectra and when having a signal-to-noise (S/N) ≥ 4. Peak clusters were completed with peaks with a S/N ≥ 1.5 in a cluster mass window of 0.4%. Peak information was subsequently exported as spreadsheet files, and peak intensities from the duplicate analyses were averaged. The sera were analysed on three consecutive days, a parameter known to influence spectral data (17;18). As such, merging peak intensity data of the three measurement series could lead to spurious results. To this end, peak intensities were log transformed to obtain normal distributions. Per measurement series, the log transformed peak intensities were converted to standard Z-values by subtracting the mean and dividing by the standard deviation. The log-Z transformed data of the three series were subsequently merged in one file. The T-test was then applied in the comparison of the mean log-Z peak intensities between BC and CON (i.e., BBD and HC) per time-point (i.e., pre- and post-surgery). Mean log-Z peak intensities of the different time points (pre- vs. post-surgery) were compared by the paired T-test in each group (BC and CON) separately, to preclude bias by group. All tissue samples were analysed within one measurement series per array type. Spectra were pre-processed as described above, after which the BMW software package was applied for peak detection. Peaks were auto-detected when occurring in at least 10% of spectra and when having a S/N ≥ 3. Peak clusters were completed with peaks with S/N ≥ 1 in a cluster mass window of 0.45%. Peak information was subsequently exported as spreadsheet files, and peak intensities from the duplicate analyses were averaged. Median peak intensities between BC and CON (i.e., BBD and HC) were compared using the non-parametric Mann-Whitney U test (MWU). All p-values were corrected for 168 Diagnostic tissue and serum protein profiles for breast cancer multiple testing by the Bonferroni method, by multiplying p-values with the number of peak clusters detected and tested. The classification performance of both serum and tissue protein profiles was assessed by building classification trees with the Biomarker Patterns Software v.5.0.1. (BPS; BioRad Labs), inputting all peaks detected by the BMW. A ten-fold cross-validation was performed to estimate the sensitivity and specificity of each tree. The breast cancer samples were collected during a longer time interval than the control samples (Table 1), which, despite storage at -70°C (serum) and -196°C (tissue), potentially can introduce bias (19-21). Therefore, all analyses were also performed in subsets of the total study population, containing only breast cancer samples that were matched to the control samples for sample storage duration. Table 1 Patient and sample characteristics of diagnostic groups evaluable for serum protein profiling. Breast cancer (total) Breast cancer (subgroup†) Benign Healthy N 75 26 13 13 Age (years), median [IQR] 57.4 [48.4-69.9] 55.1 [45.2-67.1] 40.8 [30.6-48.9] 43.4 [43.0-48.7] n.a. n.a. 3 34 21 / 10 3/3 1 2 9 6/5 2/2 0 n.a. n.a. Stage ‡ 0 1 2A / 2B 3A / 3C Unknown Benign diagnosis Mastopathy Periductitis Fibroadenoma Sample storage duration (months), median [IQR] Pre-surgery sera Post-surgery sera n.a. 7 1 5 17.0 [10.7-23.2] 14.8 [8.8-20.4] 8.4 [5.1-12.3] 7.4 [4.1-10.9] 8.0 [3.1-9.6] 5.2 [2.1-8.2] 9.1 [5.5-12.0] 7.5 [4.5-10.6] Abbreviations: IQR: interquartile range, n.a.: not applicable. † Subgroup: subgroup of breast cancer patients matched to the control patients for sample storage duration. ‡ Stage: pathologically determined stage. Finally, we also investigated whether the relationship between peak intensity and breast cancer / surgery status was influenced by patients’ age or stage of disease. To this end, breast cancer samples were split according to tertiles of patients’ age, or to stage of disease (Stage 1, and 2). Peak intensity differences between the categories were subsequently investigated by ANOVA and T-test statistics (pre- and post-surgery serum), and the Kruskall-Wallis and Mann-Whitney U (MWU) test (tissue). For breast cancer/surgery status-associated peak clusters found significantly related to patients’ age and or stage of disease, the relationship between peak intensity and breast 169 Chapter 4.1 cancer/surgery status was investigated in subgroups of age (i.e., < and > median age) and stage (i.e., Stage 1, and 2). identification Peptide identificati on For identification purposes, peptides of interest were extracted from tissue lysates by reversed-phase C18 magnetic beads (Dynabeads RPC18, Invitrogen, Breda, The Netherlands) using a Kingfisher 96 pipetting robot (Thermo Fisher Scientific, Waltham, MA, USA), according to the optimized protocol described in (22). Briefly, tissue lysates were diluted in TFA 0.1%, after which the peptide content was bound to the beads. The beads were subsequently washed with 0.1% TFA, and eluted with 50% ACN. Eluate (1.5 µl) was mixed with α-cyano-4-hydroxy-cinnamic acid matrix (1.5 µl), after which the mixture was spotted (0.7 µl) on a MALDI target plate. Analyses were performed on a 4800 MALDI-TOF/TOF mass spectrometer (Applied Biosystems, Forster City, CA, USA). Fragment ion spectra resulting from TOF/TOF analyses were searched manually for b- and y-ion (related) peaks. Results Study population The characteristics of all assessable study participants are described in Table 1 (serum) and 2 (tissue). At the time of inclusion, the breast cancer patients were significantly older than the controls in both the total and matched study population (MWU; p < 0.001). The majority of breast cancer patients was diagnosed with Stage 1 (45%) or Stage 2 (41%) disease. The cancer samples were collected during a longer time interval than the control samples, resulting in significantly different sample storage times. Storage durations were not significantly different between both groups in the matched study population. Serum protein profiling Representative serum and tissue SELDI-TOF MS spectra are presented in Figure 1 and 2. Following serum IMAC30 analyses, the Biomarker Wizard detected 51 peak clusters. None of the detected peak clusters were found significantly different in expression between breast cancer and control in either the total study population or the matched subgroup. Comparing pre- and post-surgery samples, significant differences were observed in the breast cancer group, as intensities of the m/z 4285 peak cluster and its oxidised form at m/z 4301 significantly decreased following surgery (MWU; Bonferroni corrected p = 0.042, and 0.001, respectively, Figure 3). The Biomarker Wizard detected 54 peak clusters following Q10 serum analyses. In the pre-surgery samples, intensities of the m/z 3939 peak cluster were found significantly increased in breast cancer compared to control in the total study and matched 170 Diagnostic tissue and serum protein profiles for breast cancer population (T-test; Bonferroni corrected p = 0.003, and 0.022, respectively). Postsurgery m/z 3939 peak intensities were similar between cancer and control (Figure 3), as were all other peak clusters. None of the peak clusters were found significantly different in intensity between the pre- and post-surgery samples in either one of the diagnostic groups. Table 2 Patient and sample characteristics of diagnostic groups evaluable for tissue protein profiling. Breast cancer (total) Breast cancer (subgroup†) Benign Healthy N 63 22 13 11 Age (years), median [IQR] 59.1 [48.7-70.6] 56.2 [45.6-76.3] 41.2 [30.9-49.3] 36.6 [43.1-48.3] n.a. n.a. 2 28 17 / 10 3/3 2 8 5/3 2/2 n.a. n.a. Stage ‡ 0 1 2A / 2B 3A / 3C Benign diagnosis n.a. 7 1 5 Sample storage duration (months), median [IQR] 17.5 [10.1-25.4] 10.3 [7.4-13.8] 9.3 [4.8-11.1] 8.8 [6.4-13.4] Abbreviations: IQR: interquartile range, n.a.: not applicable. † Subgroup: subgroup of breast cancer patients matched to the control patients for sample storage duration. ‡ Stage: pathologically determined stage. None of the three significant peak clusters (i.e., m/z 4285, 4301, and 3939) were found related to patients’ age (ANOVA; p > 0.05) and stage of disease (T-test; p > 0.05) in either the pre- or post-surgery samples. In addition, no suitable classification trees were obtained in either the IMAC30 or Q10 serum data (10-fold cross validated performance < 66%, data not shown). Tissue protein profiling Of the 114 peak clusters detected by the Biomarker Wizard in the IMAC30 spectra, 20 were found significantly different in intensity between breast cancer and control. None of these 20 peak clusters were found related to patients’ age (Kruskall-Wallis test; p > 0.05), while 10 peak clusters were found significantly discriminative in the matched sample set as well (Table 3). Some of these peaks showed a gradual increase (i.e., m/z 6833) or decrease (i.e., m/z 4505) in peak intensity from HC to BBD to BC (Figure 4). We also observed some clusters (i.e., m/z 9517) to gradually change in intensity with increasing cancer stage (Figure 4), though none of the peak clusters were found related to stage of disease (MWU test; p > 0.05). 171 Chapter 4.1 Figure 1 Representative example of serum IMAC30 (pre- vs. post-surgery) and Q10 (pre-surgery BC vs. HC) protein profiles. 4000 40 20 0 40 20 0 40 20 0 40 20 0 6000 8000 4285.4+H 4301.8+H BC pre-surgery (IMAC30) BC post-surgery (IMAC30) HC pre-surgery (Q10) 3938.5+H 4000 BC pre-surgery (Q10) 6000 8000 In the Q10 spectra, 113 peak clusters were detected by the Biomarker Wizard. Of the 27 peak clusters found significantly different in intensity between breast cancer and control in the total sample set, 17 were also significantly different in the matched sample set (Table 4). None of the significant peak clusters were found related to patient’s age (Kruskall-Wallis test; p > 0.05). Similar to the IMAC30 data, some peaks showed a gradual increase or decrease (i.e., m/z 7286) in peak intensity going from BC to BBD to HC (Figure 5). Again, some clusters were found to gradually increase (i.e., m/z 9745) or decrease (i.e., m/z 2612) in intensity with increasing cancer stage (Figure 5). However, intensities of none of the significant peak clusters were found related to stage of disease (MWU test; p > 0.05). Optimal discrimination between breast cancer and control was achieved by one-node classification trees, applying either the m/z 5430 (IMAC30) or the m/z 19899 (Q10) peak cluster in both the total and the matched study population. Ten-fold cross-validated performance of classification trees ranged from 67% to 87% (data not shown). Peptide identification The MALDI tissue peptide profiles obtained using C18 magnetic beads were searched for the presence of discriminative SELDI peaks based on mass matching. Due to the different chemistries used for peptide capture for SELDI-TOF MS (Q10 and IMAC30 Ni) and MALDI-TOF MS (C18), and to the mass limitations for direct fragmentation, we were able to elucidate the identity of only two Q10 tissue lysate peak clusters found significantly different in expression between cancer and control. The SELDI-TOF MS peak clusters at m/z 3090 and m/z 4169 were detected by MALDI-TOF/TOF MS as MH+ 172 Diagnostic tissue and serum protein profiles for breast cancer ions at m/z 3084.80 and m/z 4163.04 (default calibration), respectively. Using one a-ion peak, eleven b-ion peaks, one c-ion peak, and three y-ion peaks detected by MALDITOF/TOF MS/MS (Figure 6), m/z 3084.80 was identified manually as the albumin25-51 fragment DAHKSEVAHRFKDLGEENFKALVLIAF (theoretical monoisotopic mass 3083.62 Da, pI 6.04). Six of the b-ion peaks were also identified in the MALDITOF/TOF MS/MS spectrum of m/z 4163.04, explaining the major peaks in the spectrum. The m/z 4169/4163.04 peptide corresponds to the N-terminal albumin25-60 fragment (sequence DAHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPF, theoretical monoisotopic mass 4162.11 Da, and pI 6.04). On the SELDI platform, the peak intensities of the discriminative m/z 3549, 3563, and 3711 peak clusters were found highly correlated to the peak intensities of the N-terminal m/z 3090 and 4369 albumin fragments (Spearman’s R ≥ 0.75), suggesting structural homology between these peak clusters, or cleavage by the same protease acting on a different substrate. Figure 2 Representative example of tissue IMAC30 and Q10 protein profiles (BC vs. HC). 4000 6000 8000 4504.6+H 40 20 0 HC (IMAC30) 40 20 0 40 20 0 BC (IMAC30) 2610.5+H HC (Q10) 40 20 0 BC (Q10) 4000 6000 8000 Discussion In the current study, we analysed pre- and post-surgery serum and tissue samples of breast cancer patients (n = 75) and controls (n = 26) using the SELDI-TOF MS technology. All samples were collected prospectively by rigorous adherence to a strictly defined protocol. Following serum analyses, one Q10 peak cluster (m/z 3939) was found significantly increased in breast cancer compared to control, while IMAC30 analyses revealed two peak clusters (m/z 4292 and m/z 4301) that significantly decreased in expression following surgery of the breast cancer patients. In contrast, tissue analyses yielded 10 IMAC30 and 17 Q10 peak clusters with a significantly different expression 173 Chapter 4.1 between cancer and control. A number of peak clusters gradually increased or decreased in intensity with increasing cancer stage, or from healthy to benign to cancer (Figure 4 and 5). Ten-fold cross validation performances of the various classification trees ranged from 67% to 87%. Figure 3 Intensities of the significantly different peak clusters detected in the IMAC30 (m/z 4285 and m/z 4301) and Q10 (m/z 3939) serum analysis (y-axis: log-Z transformed peak intensity, x-axis: group CON (control), and BC (breast cancer)). m/z 4285 4 m/z 4301 p = 0.042 4 m/z 3939 p = 0.001 4 2 2 2 0 0 0 -2 -2 -2 CON BC CON BC p = 0.003 CON BC Serum analyses Although one peak cluster (m/z 3939) differed significantly in expression between groups (BC vs. CON), obtained serum protein profiles could not be applied in a satisfactory classification of samples. The detection of discriminating peak clusters might have been hampered by our limited study population, as well as by the composition of the diagnostic groups investigated. Since our control group contains benign breast disease patients, and our cancer group contains predominantly early stage disease, the diagnostic groups are less divergent, which is likely to have impeded group distinction. Nonetheless, comparison of breast cancer to solely benign breast disease or healthy control did not yield significantly different peak clusters (data not shown), a finding most likely caused by the very limited samples sizes of the respective control groups (BBD: n = 13, HC: n = 11). Since monitoring protein expression dynamics in response to surgery may help to better understand breast cancer pathogenesis, we also investigated serum proteome transitions occurring after surgery. To this end, protein profiles of sera procured prior to and following surgery were compared. Using paired statistics, we aimed to detect intraindividual differences, which might otherwise be masked by inter-individual variation (23). Both m/z 4285 and m/z 4301 peak clusters were observed to significantly decrease following surgery. Since this effect was detected solely in the breast cancer group (n = 75), we hypothesise both peak clusters to be cancer-associated. 174 Diagnostic tissue and serum protein profiles for breast cancer Table 3 Significantly different peak clusters detected in the IMAC30 tissue protein profiles. Peak cluster (m/z) Total study population Matched study population p (MWU) Peak ratio† p (MWU) Peak ratio† 3347 3545 3600 3897 4207 4505 4876 5225 5278 5431 6833 7950 9518 10938 15296 15895 16076 31199 31818 66656 0.001 0.008 0.001 0.028 0.020 0.001 < 0.001 0.003 < 0.001 < 0.001 0.002 0.009 < 0.001 < 0.001 0.002 0.006 < 0.001 < 0.022 0.026 0.004 0.30 1.86 18.62 4.24 3.47 0.30 5.96 4.81 0.23 0.31 4.87 0.46 2.49 7.00 0.44 0.50 0.47 0.45 0.42 0.24 n.s. n.s. n.s. 0.021 0.002 0.006 n.s. n.s. 0.027 0.027 0.027 n.s. 0.009 0.014 n.s. n.s. 0.049 n.s. n.s. 0.039 0.41 1.46 17.38 5.21 4.75 0.26 5.49 4.97 0.26 0.36 5.64 0.47 2.91 4.80 0.48 0.53 0.49 0.48 0.47 0.24 Abbreviations: MWU: Mann-Whitney U test BC vs. CON, Bonferroni corrected p-value, n.s.: not significant. † Peak ratio: average peak intensity in breast cancer spectra divided by the average intensity in control spectra. The m/z 4285 and m/z 4301 peak clusters have both been reported previously as diagnostic markers for breast cancer (8;15;24-26). Identified as a putative ITIH4 fragment (15;25) and its oxidised form, previous studies have described either a decreased (8;24;26) or increased (15;25) serum m/z 4285 ITIH4 expression in breast cancer. Most likely, these contradictory findings originate from the postulated instability of ITIH4 fragments (25;27;28), as well as the heterogeneity of the different study populations investigated. Despite these considerations, ITIH4 fragments are currently understood to contain cancer-type specific marker utility, as they are hypothesised to be generated by proteases contributed by cancer cells (25;27;29). Tissue analysis In tissue, we detected several peak clusters to differ significantly in expression between cancer and control. Of the 47 discriminative peak clusters observed in the total study population, 27 were also found significantly different in the matched subgroup. This discrepancy between the total and the matched study population is most likely due to the limited sample size, as effect estimates (i.e., peak ratio’s) are similar between both populations (see Table 3 and 4). Regarding the 67 to 87% performance of constructed 175 Chapter 4.1 classification trees, the tissue protein profiles had improved diagnostic utility compared to the serum protein profiles. Figure 4 Intensities of the significantly different peak clusters detected in the IMAC30 tissue analysis (yaxis: peak intensity, x-axis: group HC (healthy control), BBD (benign breast disease), and BC 1 – 3 (breast cancer stage 1-3)). m/z 6833 m/z 4505 m/z 9518 8 50 6 6 40 30 4 4 20 10 0 HC Figure 5 BBD 2 2 0 0 HC BC BBD BC HC BBD BC 1 BC 2 BC 3 Intensities of the significantly different peak clusters detected in the Q10 tissue analysis (y-axis: peak intensity, x-axis: group HC (healthy control), BBD (benign breast disease), and BC 1-3 (breast cancer stage 1-3)). m/z 7286 m/z 2612 30 20 10 m/z 9745 12 8 9 6 6 4 3 2 0 0 HC BBD BC 1 BC 2 BC 3 0 HC BBD BC HC BBD BC 1 BC 2 BC 3 By analogy to the linear model of the development of colon cancer (i.e., the adenomacarcinoma sequence), breast lesions are believed to progress in a linear fashion through the sequential stages of normal epithelium, to usual hyperplasia (without atypia), to atypical hyperplasia, to carcinoma in situ, and, ultimately, invasive breast cancer (30;31). The progression to malignant breast disease is associated with accumulation of an increasing number of genetic mutations (32), as well as changes in the expression of cell cycle-related and apoptosis-related proteins (33). This continuum of breast alterations appears to be visualised by the peak clusters that gradually increase or 176 Diagnostic tissue and serum protein profiles for breast cancer decrease in intensity from healthy to benign to cancer, and with increasing cancer stage. Table 4 Significantly different peak clusters detected in the Q10 tissue protein profiles. Peak cluster (m/z) Total study population Matched study population p (MWU) Peak ratio† p (MWU) Peak ratio† 1871 2021 2074 2089 2504 2612 2959 2976 3091 3201 3298 3326 3549 3563 3711 3987 4169 4857 7286 7339 9745 9958 12173 12636 16804 19899 35988 0.048 0.011 0.005 0.005 < 0.001 < 0.001 0.018 0.021 < 0.001 0.002 < 0.001 < 0.001 < 0.001 < 0.001 < 0.001 0.424 0.006 < 0.001 < 0.001 0.049 < 0.001 0.002 < 0.001 < 0.001 0.012 < 0.001 < 0.001 0.62 2.93 0.38 0.36 0.39 0.15 2.68 2.19 0.38 2.38 0.32 0.36 0.33 0.28 0.34 0.41 0.33 0.36 0.29 0.54 3.87 2.14 8.53 2.58 1.80 4.17 1.81 n.s. n.s. n.s. n.s. 0.006 0.020 n.s. n.s. 0.022 n.s. 0.018 0.022 0.006 0.006 0.015 n.s. 0.038 0.029 0.042 n.s. 0.002 0.024 0.003 0.010 n.s. < 0.001 0.029 0.62 2.31 0.44 0.38 0.37 0.24 2.64 1.84 0.35 2.30 0.30 0.34 0.26 0.25 0.32 0.45 0.27 0.36 0.47 0.55 4.20 2.26 4.57 2.89 1.93 4.57 2.04 Abbreviations: MWU: Mann-Whitney U test BC vs. CON, Bonferroni corrected p-value, n.s.: not significant. † Peak ratio: average peak intensity in breast cancer spectra divided by the average intensity in control spectra. Of the discriminative peak clusters detected in the Q10 tissue lysate analyses, m/z 3090 and m/z 4169 were identified as differentially truncated N-terminal albumin fragments. Three other discriminative peaks (m/z 3549, 3563, and 3711) were found highly correlated to these peaks, indicating structural similarity or cleavage by the same protease. Their diagnostic value could, however, possibly be compromised by the continuous aspecific proteolytic (albumin) degradation known to occur during prolonged storage at -30°C. For example, we previously found the expression of the m/z 3090 albumin25-51 fragment to be significantly associated with storage duration of breast 177 Chapter 4.1 cancer sera at -30°C. Peak intensities were found to increase up to approximately 5 years of storage, after which they gradually decreased. A similar association to sample storage duration was observed in an other study by our group, in which we investigated sera of colorectal cancer patients stored for 1.4 years at -30°C (19). In the current study, however, the discriminative N-terminal albumin fragments were detected in tissue lysate, rather than in serum. All tissue specimens were snap frozen immediately after surgical excision, after which they were stored in liquid nitrogen (-196°C), further limiting in vitro proteolytic activity compared to -30°C. Indeed, the albumin fragments were found significantly discriminative in both the total and matched study population, indicating a lack of association to sample storage duration. This finding was confirmed by the observation that the albumin fragments were discriminative to the same extent in both the total study population and the sub-population matched for sample storage duration. Hence, the albumin fragments observed in the current study most likely are (breast) cancer specific. Figure 6 100 Annotated MALDI-TOF/TOF MS/MS spectrum of m/z 3084.80. 110.0737 522.4 90 80 % Intensity 70 60 50 b13 40 1521.8853 70.0805 30 b5 20 539.2104 b3 b4 10 b6 452.1948 668.1923 b7 767.2963 0 9.0 b12 b8 838.4666 324.0803 658.6 b10 y26 1406.8763 1131.5055 a11 b16 c12 b17 2969.8669 y19 y21 1821.0933 2247.5828 1250.7838 1423.8265 1950.1238 2417.8674 1308.2 1957.8 2607.4 MH+ 3084.6667 3257.0 Mass (m/z) Although the N-terminal albumin fragments have not been described in breast cancer hitherto, it is currently understood that proteolytic fragments of high-abundant serum proteins can bear cancer(type)-specificity (27;34). Tumourigenesis is associated with changes in the balance between proteases and protease inhibitors that are secreted by 178 Diagnostic tissue and serum protein profiles for breast cancer the tumour in its microenvironment (35-37). These enzymes can not yet be applied in cancer detection, as they generally do not reach detectable levels in the circulation (27). However, since they enzymatically process high-abundant host-response proteins, specific proteolytic fragments thereof can serve as surrogate biomarkers for the presence and action of underlying proteases. Indeed, many cancer-specific proteolytic fragments of high-abundant host-response proteins (e.g., ITIH4, fibrinogen, apolipoproteins, and complement components) have been detected in serum (25;27;34;38), similar to the discriminative serum ITIH4 fragment detected in the current study. As the breast tumour microenvironment is known to exhibit various changes in the amount and activity of proteolytic enzymes (35;39), the differential albumin fragments discovered in the current study could well result from such specific proteolytic activity. Protease levels have been found proportional to tumour size, and hence, tumour stage (40). In the current study, we investigated predominantly early stage disease, suggesting limited protease activity in the tumour microenvironment, which might explain the lack of detection of these albumin fragments in serum. Yet, these fragments might well be detected in blood by other, more sensitive and specific analytical methods. However, upon translation into a diagnostic serum assay, care should be taken to prevent bias by pre-analytical parameters. Nevertheless, provided that these albumin fragments are validated in independent study populations, they can potentially offer further insight into the pathophysiological mechanisms associated with, or underlying, breast cancer. Similarly, structural identification of the other discriminative peak clusters observed in tissue is warranted to assess their potential role in breast cancer. Lastly, these discriminative proteins can potentially aid in improving accurate diagnosis of breast cancer. Conclusion In conclusion, though we detected some discriminative peak clusters following serum analyses, constructed classification models had moderate performances. Likely hampered by the highly complex nature of breast cancer (41), the current approach appears not sensitive enough to reliably detect the cancer-specific markers that are allegedly present in the low-abundant serum proteome (42;43). Analysis at the tumour level, however, yielded several peak clusters with a significantly different expression between breast cancer and control. Two discriminative peak clusters were identified as N-terminal albumin fragments. Presumably generated by (breast) cancer-specific proteolytic activity in the tumour microenvironment, these albumin fragments can potentially offer further insight into the pathophysiological mechanisms associated with, or underlying, breast cancer, and improve accurate breast cancer diagnosis, provided that these fragments are validated in independent study populations. 179 Chapter 4.1 Acknowledgement We gratefully acknowledge Marian van der Linde (mammacare nurse), the Department of Clinical Chemistry, and the Department of Anaesthesiology for help with collection, preparation and storage of serum and tissue samples. References References (1) Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T et al. Cancer statistics, 2008. CA Cancer J Clin 2008; 58(2):71-96. (2) Ries LA, Melbert D, Krapcho M, Mariotto A, Miller BA, Feuer EJ et al. SEER Cancer Statistics Review, 1975-2004. http://seer.cancer.gov/csr/1975_2004/ . 2008. (3) Stieber P, Molina R, Chan DW, Fritsche HA, Beyrau R, Bonfrer JM et al. Clinical evaluation of the Elecsys CA 15-3 test in breast cancer patients. Clin Lab 2003; 49(1-2):15-24. (4) Engwegen JY, Gast MC, Schellens JH, Beijnen JH. Clinical proteomics: searching for better tumour markers with SELDI-TOF mass spectrometry. Trends Pharmacol Sci 2006; 27(5):251-259. (5) Banks RE, Dunn MJ, Hochstrasser DF, Sanchez JC, Blackstock W, Pappin DJ et al. Proteomics: new perspectives, new biomedical opportunities. Lancet 2000; 356(9243):1749-1756. (6) Hutchens TW, Yip TT. New desorption strategies for the mass spectrometric analysis of macromolecules. Rapid Commun Mass Spectrom 1993; 7:576-580. (7) Belluco C, Petricoin EF, Mammano E, Facchiano F, Ross-Rucker S, Nitti D et al. Serum Proteomic Analysis Identifies a Highly Sensitive and Specific Discriminatory Pattern in Stage 1 Breast Cancer. Ann Surg Oncol 2007; 14(9):2470-2476. (8) Li J, Zhang Z, Rosenzweig J, Wang YY, Chan DW. Proteomics and bioinformatics approaches for identification of serum biomarkers to detect breast cancer. Clin Chem 2002; 48(8):1296-1304. (9) Engwegen JY, Helgason HH, Cats A, Harris N, Bonfrer JM, Schellens JH et al. Identification of serum proteins discriminating colorectal cancer patients and healthy controls using surface-enhanced laser desorption ionisation-time of flight mass spectrometry. World J Gastroenterol 2006; 12(10):1536-1544. (10) Habermann JK, Roblick UJ, Luke BT, Prieto DA, Finlay WJ, Podust VN et al. Increased serum levels of complement C3a anaphylatoxin indicate the presence of colorectal tumors. Gastroenterology 2006; 131(4):1020-1029. (11) Engwegen JY, Mehra N, Haanen JB, Bonfrer JM, Schellens JH, Voest EE et al. Validation of SELDI-TOF MS serum protein profiles for renal cell carcinoma in new populations. Lab Invest 2007; 87(2):161-172. (12) Tolson J, Bogumil R, Brunst E, Beck H, Elsner R, Humeny A et al. Serum protein profiling by SELDI mass spectrometry: detection of multiple variants of serum amyloid alpha in renal cancer patients. Lab Invest 2004; 84(7):845-856. (13) Becker S, Cazares LH, Watson P, Lynch H, Semmes OJ, Drake RR et al. Surfaced-enhanced laser desorption/ionization time-of-flight (SELDI-TOF) differentiation of serum protein profiles of BRCA-1 and sporadic breast cancer. Ann Surg Oncol 2004; 11(10):907-914. (14) Hu Y, Zhang S, Yu J, Liu J, Zheng S. SELDI-TOF-MS: the proteomics and bioinformatics approaches in the diagnosis of breast cancer. Breast 2005; 14(4):250-255. (15) Li J, Orlandi R, White CN, Rosenzweig J, Zhao J, Seregni E et al. Independent Validation of Candidate Breast Cancer Serum Biomarkers Identified by Mass Spectrometry. Clin Chem 2005; 51(12):2229-2235. 180 Diagnostic tissue and serum protein profiles for breast cancer (16) Banks RE. Preanalytical influences in clinical proteomic studies: raising awareness of fundamental issues in sample banking. Clin Chem 2008; 54(1):6-7. (17) Karsan A, Eigl BJ, Flibotte S, Gelmon K, Switzer P, Hassell P et al. Analytical and preanalytical biases in serum proteomic pattern analysis for breast cancer diagnosis. Clin Chem 2005; 51(8):1525-1528. (18) Hu J, Coombes KR, Morris JS, Baggerly KA. The importance of experimental design in proteomic mass spectrometry experiments: some cautionary tales. Brief Funct Genomic Proteomic 2005; 3(4):322-331. (19) Engwegen JY, alberts M, knol JC, Jimenez C.R., Depla ACTM, Tuynman H et al. Influence of variations in sample handling on SELDI-TOF MS serum protein profiles for colorectal cancer. Proteomics Clin Appl 2008; 2(6):936-945. (20) Hsieh SY, Chen RK, Pan YH, Lee HL. Systematical evaluation of the effects of sample collection procedures on low-molecular-weight serum/plasma proteome profiling. Proteomics 2006; 6(10):31893198. (21) Rai AJ, Gelfand CA, Haywood BC, Warunek DJ, Yi J, Schuchard MD et al. HUPO Plasma Proteome Project specimen collection and handling: towards the standardization of parameters for plasma proteome samples. Proteomics 2005; 5(13):3262-3277. (22) Jimenez CR, El Filali Z, Knol JC, Hoekman K, Kruyt FAE, Giaccone G et al. Automated serum peptide profiling using novel magnetic C18 beads off-line coupled to MALDI-TOF-MS. Proteomics Clin Appl 2007; 1(6):598-604. (23) Kasthuri RS, Verneris MR, Ibrahim HN, Jilma B, Nelsestuen GL. Studying multiple protein profiles over time to assess biomarker validity. Expert Rev Proteomics 2006; 3(4):455-464. (24) Mathelin C, Cromer A, Wendling C, Tomasetto C, Rio MC. Serum biomarkers for detection of breast cancers: a prospective study. Breast Cancer Res Treat 2005;1-8. (25) Song J, Patel M, Rosenzweig CN, Chan-Li Y, Sokoll LJ, Fung ET et al. Quantification of fragments of human serum inter-alpha-trypsin inhibitor heavy chain 4 by a surface-enhanced laser desorption/ionization-based immunoassay. Clin Chem 2006; 52(6):1045-1053. (26) van Winden AWJ, Gast MCW, Beijnen JH, Rutgers EJ, Grobbee DE, Peeters PHM et al. Validation of previously identified serum biomarkers for breast cancer with SELDI-TOF MS: a case control study. BMC Medical Genomics 2008; Accepted for publication. (27) Fung ET, Yip TT, Lomas L, Wang Z, Yip C, Meng XY et al. Classification of cancer types by measuring variants of host response proteins using SELDI serum assays. Int J Cancer 2005; 115(5):783-789. (28) Timms JF, Arslan-Low E, Gentry-Maharaj A, Luo Z, T'Jampens D, Podust VN et al. Preanalytic influence of sample handling on SELDI-TOF serum protein profiles. Clin Chem 2007; 53(4):645-656. (29) Villanueva J, Philip J, Chaparro CA, Li Y, Toledo-Crow R, DeNoyer L et al. Correcting common errors in identifying cancer-specific serum peptide signatures. J Proteome Res 2005; 4(4):1060-1072. (30) Arpino G, Laucirica R, Elledge RM. Premalignant and in situ breast disease: biology and clinical implications. Ann Intern Med 2005; 143(6):446-457. (31) Santen RJ, Mansel R. Benign breast disorders. N Engl J Med 2005; 353(3):275-285. (32) Reis-Filho JS, Lakhani SR. The diagnosis and management of pre-invasive breast disease: genetic alterations in pre-invasive lesions. Breast Cancer Res 2003; 5(6):313-319. (33) Mommers EC, van Diest PJ, Leonhart AM, Meijer CJ, Baak JP. Balance of cell proliferation and apoptosis in breast carcinogenesis. Breast Cancer Res Treat 1999; 58(2):163-169. (34) Villanueva J, Shaffer DR, Philip J, Chaparro CA, Erdjument-Bromage H, Olshen AB et al. Differential exoprotease activities confer tumor-specific serum peptidome patterns. J Clin Invest 2006; 116(1):271284. (35) Garbett EA, Reed MW, Stephenson TJ, Brown NJ. Proteolysis in human breast cancer. Mol Pathol 2000; 53(2):99-106. 181 Chapter 4.1 (36) Blasi F. Proteolysis, cell adhesion, chemotaxis, and invasiveness are regulated by the u-PA-u-PAR-PAI-1 system. Thromb Haemost 1999; 82(2):298-304. (37) Bank U, Kruger S, Langner J, Roessner A. Review: peptidases and peptidase inhibitors in the pathogenesis of diseases. Disturbances in the ubiquitin-mediated proteolytic system. Proteaseantiprotease imbalance in inflammatory reactions. Role of cathepsins in tumour progression. Adv Exp Med Biol 2000; 477:349-378. (38) Villanueva J, Martorella AJ, Lawlor K, Philip J, Fleisher M, Robbins RJ et al. Serum peptidome patterns that distinguish metastatic thyroid carcinoma from cancer-free controls are unbiased by gender and age. Mol Cell Proteomics 2006; 5(10):1840-1852. (39) Martinez JM, Prieto I, Ramirez MJ, Cueva C, Alba F, Ramirez M. Aminopeptidase activities in breast cancer tissue. Clin Chem 1999; 45(10):1797-1802. (40) Ho CH, Yuan CC, Liu SM. Diagnostic and prognostic values of plasma levels of fibrinolytic markers in ovarian cancer. Gynecol Oncol 1999; 75(3):397-400. (41) Bertucci F, Birnbaum D, Goncalves A. Proteomics of Breast Cancer: Principles and Potential Clinical Applications. Mol Cell Proteomics 2006; 5(10):1772-1786. (42) Hoffman SA, Joo WA, Echan LA, Speicher DW. Higher dimensional (Hi-D) separation strategies dramatically improve the potential for cancer biomarker detection in serum and plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2007; 849(1-2):43-52. (43) Anderson NL, Anderson NG. The human plasma proteome: history, character, and diagnostic prospects. Mol Cell Proteomics 2002; 1(11):845-867. 182 Conclusions and perspectives Conclusions and perspectives Conclusions and Perspectives Recent advances in mass spectrometry, such as surface-enhanced laser desorption/ionisation time-of-flight mass spectrometry (SELDI-TOF MS) technology, have enabled the simultaneous detection of a large part of the proteome in a highthroughput fashion. The relative simplicity of sample preparation, high analytical sensitivity, and speed of data acquisition renders SELDI-TOF MS a promising technology for detection of novel biomarkers in complex biological matrices, such as serum and tissue. In the present thesis, technical aspects of the SELDI-TOF MS technology (e.g., technical improvements, sample handling issues) are described. Subsequently, the use of SELDI-TOF MS for protein profiling of serum and breast tissue in search of novel diagnostic and prognostic biomarkers for breast cancer is described. In a retrospective study performed in serum, we discovered and structurally identified several discriminating proteins. The diagnostic performance of the constructed classification model was only moderate though. In addition, upon analysis of an independent sample set, we could not confirm the diagnostic performance of previously published candidate biomarkers in serum. Furthermore, we investigated the potential of SELDI-TOF MS serum protein profiles in improving breast cancer prognostication. Although in crude serum, we could not detect prognostic protein profiles, analysis of anion-exchange fractionated serum revealed three protein peaks, intensities of which were independently associated to recurrence free survival. In addition, investigation of prospectively collected breast tissues revealed several highly discriminative proteins, two of which were structurally identified as N-terminal albumin fragments. Most probably generated by tumour-specific proteolytic activity, these fragments might provide further insight into the pathophysiological mechanisms associated with, or underlying, breast cancer. These results highlight the potential of tissue SELDI-TOF MS analysis in clinical proteomics research. Yet, its successful application requires further consideration of key issues such as sample handling procedures, reproducibility and external validation of results. SELDI--TOF MS Breast cancer biomarker discovery by SELDI Detection of breast cancer at an early stage, when it is still curable by current treatment modalities, could be greatly facilitated by the application of blood-borne biomarkers. In search for such markers, we performed a retrospective study, investigating sera from breast cancer patients and healthy controls (Chapter 3.1). To obtain reliable estimates of classification model performance, we applied a split-sample approach, dividing the samples into a training and a test set. Several discriminative serum proteins were discovered and structurally identified as acute phase reactants (a.o. C3a des-arginine anaphylatoxin and inter-alpha-trypsin inhibitor heavy chain 4 (ITIH4) fragments). However, the constructed classification model had a moderate performance, 187 Conclusions and perspectives comparable to those reported by previously performed SELDI-TOF MS validation studies. The apparent intricacy associated with biomarker detection in crude serum was confirmed by a second study, in which we aimed to assess the reproducibility of previously reported breast cancer biomarkers (Chapter 3.2). Following analysis of an independent sample set by meticulous use of reported analytical assays, only part of the previously reported candidate biomarkers was recovered, while none of the previously published expression differences could be confirmed. Better breast cancer prognostication may improve selection of patients whom would benefit from adjuvant therapy. Hence, in search for novel prognostic serum markers, sera of high-risk primary breast cancer patients (Chapter 3.3) were investigated and a strong association between haptoglobin phenotype and recurrence free survival was discovered. This result could, however, not be confirmed following validation by analysis of a similar, but six-fold larger sample set, rendering our initial observation most likely false positive. These results emphasize the importance of validation in a sufficiently sized population, as even the most thorough statistical methodology cannot preclude chance results and, hence, erroneous conclusions. Subsequently, to specifically explore the allegedly high-informative low abundant serum proteome, we fractionated a subset of the previous validation set (n = 82) and analysed part of the resulting fractions by SELDI-TOF MS (Chapter 3.4). Four protein peaks, one of which identified as a serum ITIH4 fragment, were found to contain significant prognostic value. Three peak clusters (including the ITIH4 fragment) remained significantly associated to recurrence free survival following inclusion of clinical parameters of known prognostic value. Hence, investigation of the postoperative, anion-exchange fractionated, serum proteome by e.g., SELDI-TOF MS, is promising for the detection of novel prognostic factors. Provided that the (other three) discovered candidate markers are structurally identified and validated by independent study populations, they can eventually be applied to improve breast cancer prognostication. Lastly, we investigated tissue specimens collected prospectively from breast cancer patients, benign breast disease patients and healthy controls (Chapter 4.1). Contrary to the crude serum analyses, proteome analyses at the tumour level yielded multiple SELDI-TOF MS peak clusters with discriminative value between malignant and healthy/benign tissue. Two proteins were structurally identified as N-terminal albumin fragments. Most probably originating from tumour-specific protease activity, following their validation these proteins might provide further insight into the pathophysiological mechanisms associated with, or underlying, breast cancer. Similarly, structural identification of the other discriminative peak clusters observed in tissue is warranted to assess their potential role in breast cancer. Lastly, upon validation, these discriminative proteins can potentially aid in a more accurate diagnosis of breast cancer. The apparent difficulty in the detection of general, high-performance biomarkers in crude serum for diagnosis or prognosis of breast cancer by SELDI-TOF MS probably is 188 Conclusions and perspectives inherent to the highly complex nature of breast cancer, as well as to its high heterogeneity. Selection of breast cancer subgroups is therefore likely to facilitate future biomarker detection in crude serum. Furthermore, the detection of valid biomarkers is hampered by the type of research applied. Most proteomic datasets are subject to both the ‘curse of dimensionality’ (large number of protein peaks) and the ‘curse of dataset sparsity’ (limited number of samples). As such, datasets are frequently subjected to multiple testing. Consequently, classification models are easily over-trained (i.e., overfitted), resulting in many candidate biomarkers prone to be false positive. Crossvalidation can offer some indication of the generalisability of the classification model, provided that potential bias is not hard-wired into the data. Hence, a more reliable estimate of general performance is obtained by analysis of an independent, but similar, sample set. Detection of candidate biomarkers might also have been hampered by the biological matrix commonly investigated (i.e., crude serum). While only 22 proteins comprise more than 99% of the human serum proteome, the low abundant proteins make up for the remaining < 1%. This large dynamic range of proteins in crude serum (~10 orders of magnitude) hampers detection of the allegedly high-informative low abundant serum proteins, since the currently applied proteomic technologies are limited to a dynamic range of 2-4 orders of magnitude. Serum fractionation, however, is likely to facilitate detection of the low abundant proteins through reduction of this dynamic range, as demonstrated in Chapter 3.4. In addition, while serum is generated by coagulation, its proteome is prone to the proteases involved in this cascade, as well as to those involved in the complement cascade, activated upon clotting. Various pre-analytical parameters, such as sampling device, clotting temperature, and storage time, can thus all exert a distinct influence on the serum proteome, as illustrated by Chapter 2.2. Investigating breast cancer sera stored for 1 to 11 years at -30°C, we identified several proteins with a significant (non-) linear association to storage duration. These proteins have, however, also been described previously as potential cancer markers, rendering them specific to both disease and sample handling issues. Hence, to prevent experimental variation from being interpreted erroneously as disease associated variation, assessment of potential (non-)linear confounding effects by pre-analytical parameters is a prerequisite in biomarker discovery studies. Moreover, to ascertain reproducibility and prevent systematic bias and overfitting of data, proteomic studies aiming at the discovery of biomarkers preferably should include two distinct and complementary steps: a discovery phase and a validation phase. The above-mentioned hurdles in serum biomarker detection apply to tissue analyses as well, since the complexity and heterogeneity of breast cancer or the type of research is not affected by the biological matrix investigated. Likewise, the tissue proteome is susceptible to proteolytic activity induced by tumour resection. Nonetheless, analysis of tissue specimens, collected following a strict protocol to prevent bias by pre-analytical 189 Conclusions and perspectives parameters, yielded many more discriminate proteins compared to serum analyses (Chapter 4). As the concentration of potential biomarkers will be highest in the tumour and its immediate microenvironment, the discrepancy in results between tissue and serum is most likely caused by dilution (or degradation) of tissue proteins upon entrance of the circulatory system. Though assessment of these tumour-derived markers in blood would facilitate their clinical application, the discriminative tissue proteins were not detected in serum collected from the same patients by a rigorous sample collection protocol, using the same SELDI-TOF MS assay that was applied in tissue analysis. Yet, these fragments might well be detected in blood by other, more sensitive and specific analytical methods. However, upon translation into a diagnostic serum assay, care should be taken to prevent bias by pre-analytical parameters, as one albumin fragment was previously found to be associated with sample storage duration of serum at -30°C (Chapter 2.2). Nevertheless, provided that these albumin fragments are validated in independent study populations, they can potentially offer further insight into the pathophysiological mechanisms associated with, or underlying, breast cancer, as well as aid in a more accurate diagnosis of breast cancer. Similarly, structural identification and validation of the other discriminative peak clusters observed in tissue is warranted to assess their potential role in breast cancer. Perspectives The ultimate aim of proteomic protein profiling studies has been the identification of novel, specific oncoproteins, to augment knowledge of the molecular mechanisms underlying breast carcinogenesis and to improve breast cancer care. Still, the candidate biomarkers identified thus far constitute of normal cellular proteins and highly abundant proteins normally present in the blood compartment, which were identified in various cancer types. As such, specific oncoproteins have yet not been detected using the SELDI-TOF MS approach. The term ‘oncoprotein’ can, however, be defined in various ways. For instance, the currently approved oncoproteins (e.g., Cancer Antigen 15.3 and 27.29) are proteins for which the genetic blueprint is present in every cell. Thus, in theory, the expression of these oncoproteins is not limited to malignant cells. The resulting lack of specificity is reflected by the moderate performance of these markers. True oncoproteins, on the other hand, are highly tumour-specific, since their genetic blueprint is confined to malignant cells. These oncoproteins are the result of the specific genetic mutations that underlie malignant transformation, leading to aberrant amino acid sequences and/or post-translational modifications. However, as cancer cells are deranged host cells, and most cancers of epithelial origin share similar molecular features, it may be hard to find true cancer-specific proteins, expressed exclusively by one type of malignant cells. Alternatively, since these true oncoproteins are expected to be among the least abundant proteins, they could simply have eluded detection thus far, due to the limited dynamic range of the current protein profiling technologies. The detection of low 190 Conclusions and perspectives abundant proteins can be facilitated by reducing the dynamic range of their biological matrix prior to analysis. Although this can be achieved by either (immuno)depletion of the most abundant proteins, or enrichment of the low abundant proteins by, for instance, anion-exchange fractionation, these approaches all suffer from inherent limitations (e.g., co-depletion of low abundant proteins, loss of reproducibility). As such, they have not yet resulted in identification of true oncoproteins. Alternatively, less complex biological matrices, such as (media from) cell lines could be investigated. Although this simultaneously reduces the biological heterogeneity that is characteristic of human specimens, cells grown in vivo and in vitro are not identical due to adaptation to cell culture conditions. Reduction of heterogeneity can also be accomplished by analysis of mouse models, though mouse and human specimens are likely to be of similar complexity. Furthermore, the identification of true oncoproteins can be hampered by the very same genetic blueprint that defines them as true oncoproteins. Detection in protein profiling studies requires these oncoproteins to sufficiently differ from native proteins in mass and/or physicochemical properties. This will be easily achieved for oncoproteins that are the result of genetic mutations leading to aberrant post-translational modifications. However, the oncoproteins that are defined by aberrant amino acid sequences might prove more difficult to detect, as changes in amino acids do not necessarily alter the isoelectric point or mass of proteins. For instance, apolipoprotein A-I (28 kDa, described in Chapter 3.1) has a mass error of 1.4-15 Da upon detection by high-throughput protein profiling platforms (of 30 to 500 ppm mass accuracy). Hence, its D127N variant, in which aspartic acid (115.09 Da) is substituted by asparagine (114.10 Da), may not be detected. Likewise, substitution of multiple amino acids might not cause a net mass difference. Detection of oncoproteins can also be precluded when multiple genetic mutations result in loss of protein expression. Identification of true oncoproteins is, however, not a prerequisite for improving breast cancer care, as better breast cancer diagnosis and prognosis can also be accomplished by surrogate biomarkers of disease. A class of proteins currently recognised for their surrogate biomarker potential, are the (proteolytic fragments of) high-abundant circulatory proteins. These fragments are hypothesised to either be generated by cancer type-specific exoprotease activity, superimposed on the ex vivo coagulation and complement degradation proteolytic pathways, or result from tumour-secreted proteases that process host-response proteins upon their exposure to the tumour microenvironment. Although in breast cancer, this concept has been investigated for a.o. serum ITIH4, the various studies have reported contradictory results, most likely caused by the susceptibility of the serum proteome to various pre-analytical parameters. Hence, the concept of cancer type-specific (host response) protein fragments generated by tumour-specific proteases still awaits confirmation by validation studies that apply rigorous sample handling protocols. The latter requirement could prove to be a 191 Conclusions and perspectives bottleneck in the clinical use of these markers, as a strict adherence to collection protocols might not be feasible in the long run. Another bottleneck in the clinical application of protein biomarkers lies in the poor reproducibility of the semi-quantitative laser desorption/ionisation (LDI) technologies, such as SELDI-TOF MS. The reproducibility can be improved by standardisation of preanalytical parameters, automation of the experimental work-flow, the use of replicate measurements, and a frequent performance check of the SELDI-TOF MS instrument. However, as the matrix co-crystallisation and desorption/ionisation steps central to the LDI technology are highly complex processes involving different thermodynamic and physicochemical phenomena, which are currently not well understood, the LDI process in itself is essentially uncontrollable. The use of internal standards (a commonly used method to correct for analytical variability), is hampered by competition for array surface binding and ionisation, which play prominent roles in the complex mixtures generally investigated (e.g., serum, tissue lysates). Hence, the variability between measurements over time can not completely be eliminated by analytical quality control procedures, and efforts to develop statistical and/or bioinformatical methods to correct for this variation should be made. As these reproducibility issues hamper the future use of classification algorithms based on relative (SELDI-TOF MS) peak intensities, quantitative measurements of candidate biomarkers are currently still preferable for clinical use. However, quantitative methods such as ELISA will not always suffice. An antibody based assay cannot distinguish the parent protein from its cleaved fragments (the latter of which could possess the greatest diagnostic potential) since the antibody recognises its cognate epitope in both the parent and fragment proteins, thereby precluding correct quantitation. Hence, high-throughput, multiplex immuno mass spectrometry technologies that can discriminate between the different antibody-bound proteins should be developed. In conclusion, mass spectrometry based profiling techniques should be considered as a means of screening the proteome to identify protein patterns indicative of cancer, and biomarkers candidates should be validated in new study populations using other, quantitative, methods. Following their validation, candidate biomarkers must be investigated for their utility as breast cancer biomarkers in larger, prospective, clinical settings, also including different disease types (e.g., benign breast diseases) to ascertain specificity. This move from the discovery phase to the pre-clinical and subsequent clinical validation phase is mandatory, as the sole purpose of a biomarker lies in its application. Overseeing the results of all SELDI-TOF MS protein profiling studies in breast cancer up to now, this platform holds promise as a high-throughput screening tool for discovery of novel breast cancer markers. Provided that these studies are performed with adequate statistical power and analytical rigour, they could eventually fulfil the great promise that protein biomarkers have for improving cancer patient outcome. 192 Summary Samenvatting Summary Summary Breast cancer imposes a significant healthcare burden on women worldwide, as it is estimated to be the most commonly diagnosed neoplasm in women. In addition, preceded only by lung cancer, breast cancer is at present the second leading cause of cancer deaths. Despite the substantial progress made in cancer therapy, the five-year survival rate of breast cancer still is inversely proportional to its stage at the time of diagnosis. Hence, short of prevention, detection of breast cancer at an early, still curable, stage would offer the best route to decrease its mortality rates. However, since many patients present with advanced disease, the current diagnostic screening tools (e.g., mammography) obviously do not suffice for adequate breast cancer diagnosis. In addition, despite the survival benefit achieved by locoregional treatment and adjuvant systemic therapy, many breast cancer patients will eventually develop metastatic relapse and die, while a small percentage of patients would have survived without these treatment modalities. Evidently, the currently applied prognostic and predictive markers (e.g., age, hormone receptor status) lack adequate performance as well. Hence, better markers for early diagnosis, accurate prognosis and treatment prediction, applied either individually or in conjunction with existing modalities, are warranted to improve breast cancer care. As proteins reflect the actual state of an organism and are readily measurable in biological matrices such as blood and tissue, they hold promise as potential biomarkers for cancer. Recent advances in analytical technologies, such as protein microarrays and mass spectrometry (MS), have enabled large-scale proteomic analyses. Two MS based technologies in particular, i.e., matrix-enhanced laser desorption/ionisation time-offlight (MALDI-TOF) MS and its variant surface-enhance laser desorption/ionisation (SELDI-) TOF MS have been widely deployed for cancer biomarker discovery, cue to the relative simplicity of sample preparation, high analytical sensitivity and speed of data acquisition. In Chapter 1.1, a comprehensive overview of the protein profiling studies performed in breast cancer by these two LDI platforms is provided. Many biomarker candidates have been detected. However, structural identification, validation, and investigation in large, prospective clinical trials are obligatory prior to their eventual application in clinical patient care. Nonetheless, the two platforms hold promise as a high-throughput screening tools for discovery of breast cancer biomarkers, provided that studies are performed with adequate statistical power and analytical rigour. In Chapter 2, technical and pre-analytical aspects related to protein profiling research is investigated. In Chapter 2.1, the performance of the first and second generation SELDITOF MS apparatus is compared. No differences between the instruments were observed in the number of peaks detected in whole serum, the biomarker potential of the 197 Summary detected peaks, and the reproducibility of the analyses. However, the second generation SELDI-TOF MS had a superior performance in the analysis of anion-exchange fractionated serum, since up to twice as many peaks were detected compared to the first generation apparatus. It is increasingly recognised that pre-analytical variables, such as sample collection, processing, and storage temperature, can exert profound effects on the serum proteome. However, although the majority of clinical studies investigate samples originating from sample banks, only little is known about the possible effects of the pre-analytical variable ‘sample storage duration’. We therefore investigated the effects of extended storage duration (1 to 11 years) on the SELDI-TOF MS serum protein profile (Chapter 2.2). Several protein peaks, structurally identified as C3a des-Arginine anaphylatoxin and multiple fragments of albumin and fibrinogen were found significantly associated to sample storage duration, following five (non-)linear patterns. Of note, these proteins have also been described as potential cancer markers, rendering them specific to both disease and sample handling issues. Hence, to prevent experimental variation from being interpreted erroneously as disease associated variation, assessment of potential (non-)linear confounding by pre-analytical parameters should be an integral component of biomarker discovery and validation studies. We applied the SELDI-TOF MS technology in the search for novel candidate biomarkers that can be used in diagnosis (Chapter 3.1 and 3.2) or prognosis (Chapter 3.3 and 3.4) in breast cancer. In Chapter 3.1, the identification of serum proteins by which breast cancer patients could be discerned from healthy controls is described. Ten peaks, structurally identified as C3a des-Arginine anaphylatoxin, (tentative) inter-alphatrypsin inhibitor heavy chain (ITIH4) fragments, and a (tentative) fibrinogen fragment, were found significantly discriminative in both the training and the test set. None of these peaks were influenced by clinical (subjects’ age) and pre-analytical (sample storage duration) parameters. Nonetheless, the constructed classification model had an only moderate performance, most likely originating from the highly heterogeneous nature of breast cancer. Hence, selection of breast cancer subgroups for comparison with healthy controls is expected to improve results of future diagnostic SELDI-TOF MS studies. In Chapter 3.2, we describe the validation of diagnostic SELDI-TOF MS serum protein profiles for breast cancer discovered by other research groups, by investigation of an independent study population in our laboratory. Although (part of) the reported markers were recovered from our study population, none had sufficient performance to be applied as a marker, exemplifying analytical (i.e., reproducibility of the SELDI-TOF MS assay) and statistical (e.g., data overfitting) problems associated with this type of research. Confirmation of validity therefore is essential in obtaining the true clinical applicability of candidate biomarkers. 198 Summary Besides diagnostic serum protein profiles, profiles for prognosis may also aid breast cancer management. Better breast cancer prognostication may improve selection of patients whom would benefit from adjuvant therapy, thereby reducing both over- and undertreatment of the disease. In Chapter 3.3, a retrospective follow-up study in which sera of high-risk primary breast cancer patients were investigated in search for proteins predictive of recurrence free survival is described. Although we initially found the haptoglobin phenotype to be a strong predictor of recurrence free survival in our discovery study population (n = 63), this was not confirmed following analysis of a similar, but six-fold larger, validation sample set (n = 371), rendering our initial observation most likely false positive. These results emphasise the importance of validation in a sufficiently sized population, as even the most thorough statistical methodology can not preclude chance results, and hence, erroneous conclusions. Subsequently, to specifically explore the allegedly high-informative low abundant serum proteome, we fractionated a subset of the previous validation set by anionexchange chromatography, and analysed part of the resulting fractions by SELDI-TOF MS (Chapter 3.4). Four protein peaks, one of which identified as a serum ITIH4 fragment, were found to contain significant prognostic value. Three peak clusters (including the ITIH4 fragment) remained significantly associated to recurrence free survival following inclusion of clinical parameters of known prognostic value. Provided that the (other three) discovered candidate biomarkers are structurally identified and validated by independent study populations, they can eventually be applied in improving breast cancer prognostication. In Chapter 4, the analysis of both serum and breast tissue, collected prospectively from breast cancer patients, benign breast disease patients, and female healthy controls, is described. Sera were collected pre- and post-operatively, to assess the applicability of serum protein profiles for follow-up after surgery. Contrary to the analysis of crude serum, proteome analyses at the tumour level yielded multiple SELDI-TOF MS peak clusters with discriminative value between malignant and healthy / benign tissue. Two discriminative proteins were structurally identified as N-terminal albumin fragments. Most probably originating from tumour-specific protease activity, following their validation, these proteins might provide further insight into the pathophysiological mechanisms associated with, or underlying, breast cancer. Similarly, structural identification of the other discriminative tissue peak clusters is warranted to assess their potential role in breast cancer. Lastly, upon validation, these proteins can potentially aid in a more accurate diagnosis of breast cancer. In conclusion, the development of high-throughput mass spectrometric protein profiling approaches such as SELDI-TOF MS has enabled the simultaneous detection of part of the proteome in clinical samples in a high-throughput fashion. Overseeing the results of all SELDI-TOF MS protein profiling studies in breast cancer up to now, this 199 Summary platform holds promise as a high-throughput screening tool for discovery of novel breast cancer markers. Yet, its successful application requires further consideration of key issues such as sample handling procedures, enhancement of the dynamic range, reproducibility, and external validation of results. Provided that these studies are performed with adequate statistical power and analytical rigour, they could eventually fulfil the great promise that protein biomarkers have for improving breast cancer care. 200 Samenvatting Samenvatting Borstkanker is naar schatting de meest gediagnosticeerde vorm van kanker bij vrouwen en vormt daarmee wereldwijd een significante gezondheidsbelasting. Daarnaast heeft borstkanker momenteel (op longkanker na) de hoogste kankergerelateerde mortaliteit. Ondanks de aanzienlijke vooruitgang die op therapiegebied is geboekt, is de vijf-jaars overleving van borstkanker nog steeds omgekeerd evenredig met het stadium ten tijde van diagnose. Op preventie na biedt detectie van borstkanker in een vroeg, geneeslijk, stadium daarom de beste kans op reductie van de mortaliteit. Echter, de huidige detectiemethoden (zoals mammografie) blijken niet optimaal voor vroege detectie, aangezien veel patiënten pas in een laat stadium worden gediagnosticeerd. Bovendien zullen veel patiënten ondanks locoregionale behandeling en adjuvante systemische therapie uiteindelijk overlijden na terugkeer van de ziekte, terwijl een klein aantal patiënten ook zonder therapie langdurig in leven zou zijn gebleven. Duidelijk is dat ook de huidige prognostische en predictieve markers (bijvoorbeeld leeftijd, hormoon receptor status) onvoldoende presteren. Er is daarom behoefte aan verbeterde markers voor vroege diagnose, accurate prognose en predictie van therapie-effectiviteit, die alleen of in combinatie met bestaande methoden de borstkankerzorg kunnen verbeteren. Omdat eiwitten (zowel in hoeveelheid als soort) een goede afspiegeling geven van de staat waarin een organisme verkeert, en omdat eiwitten eenvoudig meetbaar zijn in biologische monsters zoals bloed en weefsel, kunnen zij uitstekende biomarkers voor borstkanker zijn. Recente ontwikkelingen in analytische technologieën, zoals eiwit-microarrays en massaspectrometrie (MS), hebben grootschalige eiwitanalyse mogelijk gemaakt. Gezien hun relatief eenvoudige monstervoorbewerking, grote analytische sensitiviteit en analysesnelheid, zijn twee MS technieken in het bijzonder, dat wil zeggen “matrixenhanced laser desorption/ionisation time-of-flight” (MALDI-TOF) MS en zijn variant “surface-enhanced laser desorption/ionisation” (SELDI-)TOF MS, veelvuldig toegepast in de zoektocht naar nieuwe biomarkers voor kanker. In Hoofdstuk 1.1 wordt een uitgebreid overzicht gegeven van de proteomicsstudies die tot dusver met behulp van deze twee technieken binnen borstkanker zijn uitgevoerd. Er zijn zeer veel potentiële biomarkers gedetecteerd, echter, structurele identificatie, validatie en onderzoek in grote, prospectieve klinische studies zijn noodzakelijk alvorens deze markers binnen de kliniek toegepast kunnen worden. Desalniettemin zijn beide technieken veelbelovend als snelle screeningsmethode voor het vinden van nieuwe borstkankermarkers, mits deze studies met voldoende statische power en analytische nauwkeurigheid worden uitgevoerd. In Hoofdstuk 2 wordt het onderzoek naar de technische en preanalytische aspecten van de proteomicsstudies beschreven. In Hoofdstuk 2.1 wordt de werking van de eerste en 201 Samenvatting tweede generatie SELDI-TOF MS apparaten vergeleken. Er werden geen verschillen gezien tussen het aantal pieken dat werd gedetecteerd in serum, de potentie van de pieken als biomarker en de reproduceerbaarheid van de analyse. Echter, wanneer gefractioneerd serum werd geanalyseerd, werden met de nieuwe generatie SELDI-TOF MS tot tweemaal toe zoveel pieken gedetecteerd als met het apparaat van de oude generatie. Het wordt steeds meer onderkend dat preanalytische variabelen, zoals de manier waarop de monsters zijn afgenomen en verzameld, alsook de temperatuur waarop de monsters worden opgeslagen, van grote invloed kunnen zijn op het eiwitprofiel van biologische monsters. Het merendeel van de klinische proteomicsstudies is retrospectief, waardoor monsters vaak gedurende langere tijd zijn opgeslagen. Desalniettemin is er slechts zeer weinig bekend over de mogelijke invloed van opslagduur op het SELDI-TOF MS eiwitprofiel. In Hoofdstuk 2.2 hebben wij daarom het effect van langdurige opslag (1 tot 11 jaar) op het SELDI-TOF MS serum eiwitprofiel onderzocht. Van verschillende eiwitten bleek de expressie significant geassocieerd te zijn met opslagduur, volgens vijf verschillende, (niet-) lineaire patronen. Geïdentificeerd als C3a des-Arginine anafylatoxine en meerdere fragmenten van albumine en fibrinogeen, bleken deze eiwitten echter tevens beschreven in de literatuur als potentiële markers voor kanker, waardoor ze specifiek zijn voor zowel kanker als preanalytische variabelen. Om uit te sluiten dat experimentele variatie foutief geïnterpreteerd wordt als variatie die aan de ziekte (kanker) is gerelateerd, zou het onderzoek naar potentiële (niet-)lineaire effecten van preanalytische variabelen een vast onderdeel moeten zijn van proteomicsstudies waarin biomarkers worden gezocht cq gevalideerd. De SELDI-TOF MS technologie is vervolgens gebruikt voor de detectie van nieuwe, potentiële, biomarkers die toegepast kunnen worden in de diagnose (Hoofdstuk 3.1 en 3.2), of prognose (Hoofdstuk 3.3 en 3.4) van borstkanker. In Hoofdstuk 3.1 wordt de identificatie beschreven van serumeiwitten waarmee borstkanker patiënten van gezonde vrouwelijke controles onderscheiden kunnen worden. De intensiteiten van tien pieken, geïdentificeerd als C3a des-Arginine anafylatoxine, en (tentatieve) fragmenten van inter-alpha-trypsin inhibitor heavy chain 4 (ITIH4) en fibrinogeen, waren significant onderscheidend tussen borstkanker en controle in zowel de trainingals de test-set. De intensiteit van geen van de pieken werd beïnvloed door klinische (leeftijd) dan wel preanalytische (opslagduur) parameters. Desalniettemin had het geconstrueerde classificatiemodel een slechts bescheiden sensitiviteit en specificiteit, die hoogstwaarschijnlijk veroorzaakt worden door de grote heterogeniciteit van borstkanker. Selectie van borstkanker subgroepen voor vergelijking met gezonde vrijwilligers zal de resultaten van toekomstige SELDI-TOF MS studies naar alle waarschijnlijkheid verbeteren. 202 Samenvatting In Hoofdstuk 3.2 wordt de validatie beschreven van diagnostische SELDI-TOF MS serumeiwitprofielen voor borstkanker ontdekt door andere onderzoeksgroepen, door analyse van een onafhankelijke studiepopulatie in ons laboratorium. Hoewel de gerapporteerde biomarkers (deels) werden gedetecteerd in onze studiepopulatie, was geen van de kandidaat markers voldoende sensitief en specifiek om als marker toegepast te worden. Deze resultaten illustreren de analytische (de reproduceerbaarheid van de SELDI-TOF MS analyse) en statistische (data ‘overfitting’) problemen die inherent zijn aan dit type onderzoek. Bevestiging van de validiteit is daarom essentieel voor het vaststellen van de mogelijke klinische toepasbaarheid van kandidaat-biomarkers. Naast diagnostische eiwitprofielen kunnen ook prognostische eiwitprofielen van belang zijn binnen de borstkankerzorg. Een verbeterde prognostische evaluatie kan helpen bij een meer accurate selectie van patiënten die baat hebben bij therapie, waardoor zowel over- als onderbehandeling gereduceerd kan worden. In Hoofdstuk 3.3 wordt een retrospectieve follow-up studie beschreven, waarin sera van hoogrisico patiënten met primaire borstkanker onderzocht zijn op eiwitten die predictief zijn voor ziektevrije overleving. In eerste instantie bleek het haptoglobine fenotype zeer sterk geassocieerd met ziektevrije overleving in de training-set (n = 63). Echter, dit resultaat was zeer waarschijnlijk vals positief, aangezien de gevonden associatie niet werd bevestigd na analyse van een vergelijkbare, maar zesmaal grotere, validatie-set (n = 371). Deze resultaten onderschrijven het belang van validatie in een voldoende grote test-set, aangezien zelfs de meest rigoureuze statistische methodologie de kans op vals positieve resultaten, en daarmee op onjuiste conclusies, niet kan uitsluiten. Vervolgens is een deel van de sera (n = 82) uit de hierboven beschreven validatie-set gefractioneerd middels anion-exchange chromatografie om zo de vermoedelijk zeer informatieve laagabundante serum eiwitten te onderzoeken op aanwezigheid van prognostische markers (Hoofdstuk 3.4). Na analyse van geselecteerde serum fracties met SELDI-TOF MS bleken vier eiwitpieken (waarvan een geidentificeerd als een serum ITIH4 fragment) significante prognostische informatie te bevatten. Drie van deze eiwitten (inclusief het ITIH4 fragment) bleken ook na inclusie van klinische parameters met bekende prognostische waarde significant geassocieerd met ziektevrije overleving. Deze eiwitten kunnen mogelijk op termijn ingezet worden voor verbetering van borstkankerzorg, mits ze verder worden geïdentificeerd en gevalideerd in onafhankelijke studie populaties. Hoofdstuk 4.1 beschrijft de analyse van zowel serum als borstweefsel, prospectief verzameld van borstkankerpatiënten, patiënten met een goedaardige borstaandoening en gezonde, vrouwelijke, vrijwilligers. De sera werden zowel pre- als postoperatief verzameld om te onderzoeken in hoeverre de serumeiwitprofielen toepasbaar zijn binnen de follow-up na chirurgie. In tegenstelling tot de analyse van de (ongefractioneerde) sera, leverde de analyse van eiwitten op het tumorniveau meerdere SELDI-TOF MS eiwitpieken op met onderscheidende waarde tussen maligne en gezond/benigne weefsel. Twee van deze onderscheidende eiwitten werden 203 Samenvatting geïdentificeerd als albumine fragmenten. Mits gevalideerd, kunnen deze eiwitten, die waarschijnlijk zijn gegenereerd door tumorspecifieke protease activiteit, mogelijk meer inzicht verschaffen in de pathofysiologische mechanismen die ten grondslag liggen of samengaan met de ontwikkeling van borstkanker. Evenzo is de identificatie van de overige onderscheidende eiwitten vereist om hun potentiële rol in borstkanker vast te stellen. Na validatie kunnen deze eiwitten mogelijk helpen in het stellen van een meer accurate diagnose van borstkanker. Concluderend kan worden gesteld dat de ontwikkeling van snelle, massaspectrometrische, proteomics technieken, zoals SELDI-TOF MS, de gelijktijdige detectie van een deel van de eiwitsamenstelling van klinische monsters mogelijk heeft gemaakt. Wanneer de resultaten van de SELDI-TOF MS studies, die tot dusver binnen borstkanker zijn uitgevoerd, in ogenschouw worden genomen, kan worden geconcludeerd dat deze technologie een veelbelovende, snelle, screeningsmethode is, die uitstekend toegepast kan worden in de zoektocht naar nieuwe markers voor borstkanker. Echter, voor een succesvolle toepassing van de technologie moet aandacht geschonken worden aan essentiële zaken, zoals procedures voor monsterafname, verwerking en -opslag, verbetering van de dynamische range, reproduceerbaarheid en externe validatie van resultaten. Mits deze proteomicsstudies uitgevoerd worden met voldoende statistische power en analytische nauwkeurigheid, kunnen zij op termijn de grote belofte die eiwitbiomarkers voor verbetering van de borstkankerzorg hebben. 204 Dankwoord Curriculum vitae List of publications Dankwoord Dankwoord Dit proefschrift is tot stand gekomen met hulp van velen. Een aantal personen wil ik hierbij graag in het bijzonder bedanken. Een eerste woord van dank gaat uit naar alle vrouwen, die bereid zijn geweest om bloed- en weefselmonsters af te staan voor onderzoek naar borstkanker. Zonder hun medewerking had het in dit proefschrift beschreven onderzoek niet uitgevoerd kunnen worden. Daarnaast wil ik mijn beide promotores, prof. dr Jos Beijnen en prof. dr Jan Schellens, danken voor de gelegenheid die ze mij hebben geboden om het in dit proefschrift beschreven onderzoek uit te voeren, en voor de begeleiding hierbij. Beste Jos, ik heb groot respect voor jouw onuitputtelijke ideeën, motivatie en enthousiasme voor de wetenschap, alsook voor de manier waarop jij farmacie, wetenschap en (ziekenhuis)management zo pragmatisch weet te combineren. Ik wil je danken voor je vertrouwen en de vrijheid die je mij geboden hebt, je immer positieve kijk op de onderzoeksresultaten (met name op momenten dat die mij even ontbrak), en het feit dat je deur letterlijk en figuurlijk te alle tijden voor mij heeft opengestaan. Beste Jan, veel dank voor je waardevolle input op klinisch gebied en de kritische blik op mijn manuscripten; ik heb veel van jou geleerd. Carla van Gils en Lodewijk Wessels hebben een belangrijke rol gespeeld bij de totstandkoming van dit proefschrift. Beste Carla en Lodewijk, hoewel jullie commentaar me soms tot wanhoop dreef, ben ik jullie er altijd dankbaar voor geweest; de artikelen zijn er veel beter van geworden. Veel dank voor jullie constructieve begeleiding, ik heb heel veel van jullie geleerd. Marc Zapatka, dear Marc, I have appreciated our numerous informative and pleasant discussions. Thank you for your help with the ´appropriate bioinformatics tools´ (described in Chapter 3.4). Special thanks also to Nathan Harris. Dear Nathan, I have much enjoyed the highly informative days I have spend under your supervision at the Ciphergen Lab in Guildford, and I am very thankful for your help with the protein identifications. Hans Bonfrer, Tiny Korse, Dorothé Linders en Olaf van Tellingen van het Klinisch Chemisch Laboratorium van het AvL hebben een belangrijke rol gespeeld bij de monsterverzameling. Veel dank voor het ontsluiten van de serumbank, alsook voor het gebruik van de mikrodismembrator. Olaf, dank voor jouw waardevolle inbreng tijdens het maandagochtendoverleg. Daarnaast ben ik de chirurgen Eric van Dulken en Lieve de Kock, en de plastisch chirurgen Florine Kingma-Vegter en Thea van Loenen, zeer erkentelijk voor hun onmisbare bijdrage aan het slagen van de klinische studie (beschreven in hoofdstuk 4.1). Dank voor de leerzame en plezierige samenwerking. Ook Marian van der Linde, mammacare verpleegkundige van het Slotervaartziekenhuis, wil ik in dit opzicht niet 209 Dankwoord onbenoemd laten. Beste Marian, zonder jouw inbreng was de klinische studie niet zo soepel verlopen; veel dank hiervoor. De afdeling Klinische Chemie van het Academisch Ziekenhuis Maastricht, in het bijzonder Etiënne Michielsen en Judith Bons, wil ik danken voor de organisatie van de jaarlijkse ‘SELDI-gebruikers dag’, en voor het gebruik van een aantal softwareprogramma’s. Ik heb goede herinneringen aan de dagen bij jullie in Maastricht, met name aan de hoge computerdichtheid bij jullie op de kamer (simultane data-analyse gaat nu eenmaal sneller!). Ook Leo Kruijt van de Animal Science Group te Lelystad wil ik bij dezen danken voor de hartelijke ontvangst en de plezierige werkomgeving; stroomstoringen, SELDI-‘jams’ en gecrashte harde schijven hebben de productiviteit en het plezier van de reis naar Lelystad niet kunnen drukken! Helgi Helgason, onze ‘proteomics’ wegen hebben elkaar regelmatig gekruist; dank voor de zeer plezierige samenwerking. Ik wens je veel succes met de voortzetting van jouw onderzoek in IJsland, en kom je graag een keer opzoeken. Wouter Meuleman wil ik bij dezen graag danken voor zijn hulp bij de data-analyse. Wouter, hoeveel ‘das Experiment’-en hebben we nu uiteindelijk uitgevoerd? Jouw gevoel voor humor (‘aim: world domination’) heb ik altijd bijzonder gewaardeerd. Veel succes met jouw verdere onderzoek. De lab-apotheek analisten wil ik graag bedanken voor het wegwijs maken in deze strakgeregelde afdeling, en de gezellige dagen op het lab. Ik wil mijn collega-OIO’s uit het Slotervaart en het AvL bedanken voor de leuke tijd en de positieve werksfeer, eerst in de Onderwereld, later in de Zonnetempel. De vele SLZlunches en -diners, koffiepauzes, vrijdagmiddagborrels in ‘die Rooie’, schoen-zet-acties met Sinterklaas, OIO-weekendjes, en -uitjes vormen dierbare herinneringen. Natalie en Judith, zoals Jos al eerder opmerkte, waren wij samen op eiwitgebied ‘de drie musketiers’ van het Slotervaartziekenhuis, waarbij Annemieke de rol van D’Artagnan vervulde ;). Natalie, jij als nestrix wist altijd wel raad als wij ergens mee vastliepen. Judith, als ‘de proteomics-dames’ hebben wij jarenlang lief en leed gedeeld; ik kijk er met plezier op terug. Dank voor de vele discussies met synergistisch resultaat. Annemieke, veel dank voor de vele (statistiek-)discussies, maar zeker ook voor de hulp bij de laatste experimenten in Lelystad (fràctionation ;)). Veel succes met het afronden van je onderzoek. Daarnaast wil ik ook in het bijzonder Ly (het orakel), Rob (de shaker is het nèt niet geworden!) en Joost (I love …) danken voor de vele hilarische momenten, het meejuichen in majeure tijden en het luisterend oor in mineure tijden. Veel succes met de laatste loodjes! Jolanda, dank voor de geruisloze en betrouwbare overname van de monsterverzameling als ik er niet was. Elke, mijn oud-kamergenoot, jij weet als geen ander hoe ik jouw positiviteit, eerlijkheid, rust en vertrouwen waardeer. Ik ben heel blij dan je mijn paranimf wilt zijn. Lieve Sylvia en Paul (PotPaPaul), ik prijs mijzelf gelukkig met jullie als potentiële schoonouders! Al blijft het wat surreëel dat ik jullie niets hoef uit te leggen over (mijn) 210 Dankwoord onderzoek. Dank voor het warme onthaal in jullie familie, de gezellige skivakanties, de interesse in mijn onderzoek, de (mentale) ondersteuning en de kritische blik op mijn manuscript. Lieve Gerben en Wendy, dank voor jullie interesse in mijn doen en laten. Gerben, veel succes met je onderzoek; het is bij jou in goede handen. Wendy, wat is het leven zonder kunst? Dank voor jouw alpha-inbreng in mijn bèta-leven ;). Lieve papa en mama, veel dank voor jullie vertrouwen, de (mentale) support en de veilige thuishaven waar ik altijd belangeloos op terug kon vallen; het zijn belangrijke bijdrages aan dit proefschrift geweest. Lieve Bonma, uw scherpe oog en verstand ontgaan niets. Dank u voor de interesse in mijn onderzoek en de wijze adviezen. Elseline en Ruud en de kinderen, wat is het fijn om bij jullie te zijn! Dank voor de ontspanning die jullie mij bieden. Gerrie-Cor en Niels, dank voor jullie belangstelling voor mijn onderzoek, en uiteraard ook voor de gezellige avondjes in de kroeg ;). GerrieCor, bijzonder leuk dat ook jij het promotietraject in bent gegaan. Jij bent een feest der herkenning; ik ben heel blij dat jij mijn paranimf wilt zijn. Lieve Jasper, met jou in mijn leven schijnt iedere dag de zon. Jij bent mijn solide, doch flexibele, thuisbasis. Dank voor jouw liefde, rustig vertrouwen, positiviteit en humor. Ik heb je lief! Marie-Christine Den Haag, december 2008 211 Curriculum vitae Curriculum vitae Marie-Christine Gast werd geboren op 23 februari 1978 te Woerden. In 1996 behaalde zij het atheneumdiploma aan de Minkema Scholengemeenschap te Woerden, waarna ze begon met de studie farmacie aan de Universiteit Utrecht. De doctoraalopleiding werd afgerond met een wetenschappelijke stage aan het Nederlands Forensisch Instituut te Rijswijk. In 2002 behaalde zij het apothekersdiploma. In datzelfde jaar begon ze als projectapotheker binnen de Apotheek van het Slotervaartziekenhuis, waar ze het farmaceutisch toezicht op de GGD Amsterdam heeft opgezet en uitgevoerd. Onder voortzetting van dit toezicht werd aansluitend op dezelfde werkplek gestart met het in dit proefschrift beschreven onderzoek, onder leiding van promotores prof. dr J.H. Beijnen en prof. dr J.H.M. Schellens. 213 List of publications List of publications related to this thesis Gast MCW, Bonfrer JMG, Rutgers EJTh, Schellens JHM, Beijnen JH. Proteomics in breast cancer. EORTC PAMM 2004;25:Abstract 4.03 Bouwman K, Gast MCW, Bonfrer JMG, Schellens JHM, Beijnen JH. Proteomics in de oncologie: eiwitten analyseren om kanker op te sporen. Pharm Weekbl 2004;139(25):879-84 Gast MCW, Bonfrer JMG, Rutgers EJTh, Schellens JHM, Beijnen JH. New discriminatory protein profiles in breast cancer patients. Proceedings of ASCO 2004;23:Abstract 574 Gast MCW, Bonfrer JMG, Rutgers EJTh, Schellens JHM, Beijnen JH. Proteomics in patients with breast cancer: unique profile discriminates patients from healthy controls. Br J Clin Pharmacol 2005;59(1):130 (Abstract) Engwegen JYMN, Gast MCW, Schellens JHM, Beijnen JH. Clinical proteomics: searching for better tumour markers with SELDI-TOF MS mass spectrometry. Trends Pharmacol Sci. 2006;27(5):251-9 Gast MCW, Bonfrer JM, van Dulken EJ, de Kock L, Rutgers EJTh, Schellens JHM, Beijnen JH. SELDI-TOF MS serum protein profiles in breast cancer: Assessment of robustness and validity. Cancer Biomarkers 2006;2(6):235-48 Gast MCW, Engwegen JYMN, Helgason HH, Schellens JHM, Beijnen JH. “Clinical proteomics” in de oncologie. NtvO 2007:4(4);140-52 Gast MCW, Engwegen JYMN, Schellens JHM, Beijnen JH. Comparing the old and new generation SELDI-TOF MS: implications for serum protein profiling. BMC Med Genomics 2008;1:4 Meuleman W, Engwegen JYMN, Gast MCW, Beijnen JH, Reinders MJ, Wessels LFA. Comparison of normalisation methods for surface-enhanced laser desorption and ionisation (SELDI) time-of-flight (TOF) mass spectrometry data. BMC Bioinformatics 2008;9:88 Meuleman W, Engwegen JYMN, Gast MCW, Wessels LFA, Reinders MJ. Analysis of mass spectrometry data using sub-spectra. BMC Bioinformatics 2008; in press. 215 List of publications Gast MCW, van Tinteren H, Bontenbal M, van Hoesel QGCM, Nooij MA, Rodenhuis S, Span PN, Tjan-Heijnen VCG, de Vries EGE, Harris N, Twisk JWR, Schellens JHM, Beijnen JH. Haptoglobin phenotype is not a predictor of recurrence free survival in high-risk primary breast cancer patients. BMC Cancer 2008;8:389 van Winden AWJ, Gast MCW, Beijnen JH, Rutgers EJTh, Grobbee DE, Peeters PHM, van Gils CH. Validation of previously identified serum biomarkers for breast cancer with SELDI-TOF MS: a case control study. BMC Medical Genomics 2009; accepted for publication. Gast MCW, Schellens JHM, Beijnen JH. Clinical proteomics in breast cancer: a review. Breast Cancer Res Treatm 2008; in press. Gast MCW, van Gils CH, Wessels LFA, Harris N, Bonfrer JMG, Rutgers EJTh, Schellens JHM, Beijnen JH. Serum protein profiling using SELDI-TOF MS: Influence of sample storage duration. Submitted for publication. Gast MCW, van Gils CH, Wessels LFA, Harris N, Bonfrer JMG, Rutgers EJTh, Schellens JHM, Beijnen JH. Serum protein profiling for diagnosis of breast cancer using SELDITOF MS. Submitted for publication. Gast MCW, van Dulken EJ, van Loenen TKG, Kingma-Vegter F, Westerga J, Flohil CC, Knol J, Jimenez CR, van Gils C, Wessels LFA, Schellens JHM, Beijnen JH. Detection of breast cancer by SELDI-TOF MS tissue and serum protein profiling. Submitted for publication. 216
© Copyright 2024