Home Laboratory medicine and medical oncology: the tale of two Cinderellas
Article Publicly Available

Laboratory medicine and medical oncology: the tale of two Cinderellas

  • Bohuslav Melichar

    Professor Bohuslav Melichar, MD, PhD was born on March 6, 1965 in Hradec Králové (then Czechoslovakia). After attending, between 1983 and 1989, the Charles University Medical School in Hradec Králové, Czechoslovakia, he graduated in 1989. Between 1989 and 1992 he was a resident in internal medicine at 2nd Department of Internal Medicine, Charles University Medical School and Teaching Hospital in Hradec Králové, Czechoslovakia. In 1992 he entered a Ph.D. program in medical immunology and worked simultaneously as a physician at the 2nd Department of Internal Medicine, Charles University Medical School and Teaching Hospital in Hradec Králové. In 1992 he was a visiting scholar in the University of Innsbruck, Austria, working with Professor Wachter and Professor Fuchs on the pteridines as indicators of immune response. In 1993–1994 he was a visiting scholar in the University of Bordeaux, France, working in the laboratory of Professor Mégraud, focusing on Helicobacter pylori infection. Between 1994 and 1996 he was a Fulbright scholar at the M.D. Anderson Cancer Center, Houston, Texas, United States, working first on antitumor activity of macrophages in experimental colorectal cancer, and subsequently in the laboratory of Professor Ralph S. Freedman on immunology of peritoneal carcinomatosis and intraperitoneal immunotherapy. After coming back from the United States he continued working as a physician in the 2nd Department of Internal Medicine and, later, in the Section of Hematology/Oncology of the Charles University Medical School and Teaching Hospital in Hradec Králové, Czech Republic. Simultaneously, he served as an instructor in internal medicine at the Charles University Medical School. In October 1998 he defended Ph.D. thesis “Increased production of neopterin and other indicators of immune activation in patients with malignant tumors and its biologic significance”. In March 1999 he passed a specialization examination in medical oncology. Since January 2000 he was working as medical oncologist in the Department of Oncology and Radiotherapy of the Charles University Medical School and Teaching Hospital in Hradec Králové. Between March 2001 and August 2001 he was (on a NATO scholarship) visiting scientist in the Department of Gynecologic Oncology of the University of Texas, M.D. Anderson Cancer Center, Houston (working again with Professor Ralph S. Freedman). In 2001 he was appointed associate professor of medicine and in November 2006 full professor. In February 2008 Professor Melichar was appointed the Head of the Department of Oncology, Palacký University Medical School and Teaching Hospital, Olomouc, Czech Republic.

    In January 2009 Professor Melichar was appointed associate editor for oncology of Clinical Chemistry and Laboratory Medicine. Professor Melichar is currently the president-elect of the International Society of Pteridinology and scientific secretary of the Czech Oncology Society. He is a member of Czech Society of Clinical Biochemisty, American Society of Clinical Oncology and European Society of Medical Oncology. The main areas of interest of Professor Melichar are the study of biomarkers of immune response in cancer patients and the study of new medical therapies for solid tumors, including breast cancer, renal cell carcinoma, ovarian cancer and gastrointestinal malignancies. Professor Melichar is the author of 147 papers in journals with the impact factor, including 93 papers as the first author.

    EMAIL logo
Published/Copyright: September 15, 2012

Abstract

Cancer represents a leading cause of death in the developed countries. The past 50 years have witnessed major progress in both laboratory medicine and clinical oncology that has translated into improved prognosis of cancer patients. From the humble beginnings as unrelated specialties, major advances in the understanding of molecular bases of cancer progression led to increased interactions between laboratory medicine and clinical (mostly medical) oncology. Laboratory medicine is now an integral part of the management of cancer patients. The many aspects of the role of laboratory medicine in clinical oncology include the determination of biomarkers that are used in establishing the diagnosis, predicting response to therapy or prognosis, study of the host response to tumor growth, detection of treatment toxicity and determining the concentrations of anticancer drugs.

Introduction

In the developed countries, cancer is currently the second leading cause of death. It is estimated that every third person will be affected with cancer during his or her lifetime, and every fourth person will die of cancer. Historically, cancer as a leading cause of death and a major public health issue is a relatively recent phenomenon. Despite the fact that malignant tumors have been known since antiquity, only a few generations back, cancer was relatively rare. The incidence of most malignant tumors has increased sharply during the 20th century as a result of lifestyle changes and the prolongation of life expectancy.

The history of laboratory medicine also dates back more than thousand years [1, 2]. Fifty years ago the standing of what was then medical oncology and clinical chemistry/laboratory medicine in the medical community was quite similar. Both specialties were at that time mostly outside the perimeter of interest of society, as well as the medical community. Since then, medical oncology and laboratory medicine have undergone a remarkable metamorphosis not unlike that from Cinderella to a princess. Despite the fact that 50 years ago in most institutions departments of oncology or laboratory medicine were hidden somewhere in the basements, today these departments represent the leading edge of the medical profession and occupy extensive space on separate floors or even in separate buildings. Thus, the following review may be more a tale of two Cinderellas and a journal that sprouted in the shadow of the Berlin Wall.

The essential feature of tumor growth is its systemic nature. While the assessment of local tumor extent using imaging or even histological examination may reveal only one aspect of neoplastic growth and progression, the assessment of systemic behavior of cancer is predominantly in the realm of laboratory medicine. The role of laboratory medicine in oncology also increased because of the need to define the changes associated with the tumor growth and progression at the molecular level. The rise of laboratory medicine as an independent specialty is also a relatively recent development. As mentioned above, the developments of medical oncology and laboratory medicine in the past 50 years have much in common. Both specialties rose from humble beginnings to being currently the fastest growing areas of medicine. There were times when the paths of medical oncology and laboratory medicine might have seemed to wind in parallel, but now, with these paths more resembling two busy speedways, there appear to be multiple crossings.

Our understanding of cancer pathogenesis has evolved enormously during the past 50 years. In a seminal paper published in 2000, Hanahan and Weinberg defined the six hallmarks of cancer, defined as functional properties responsible for malignant phenotype that include sustained proliferative signals, evasion of growth suppression, resistance to apoptosis, replicative immortality, induction of angiogenesis or invasion and formation of metastases [3]. The acquisition of these properties is made possible by two characteristics of malignant tissues, i.e., genomic instability of cancer cells and elicitation of the inflammatory response. Both genomic instability and inflammation then promote tumor progression. In a review revisiting the same topic published in 2011, two more hallmarks of cancer were added, including alteration of energy metabolism of cancer cells and evasion of immune response [4]. The importance of the concept of tumor microenvironment encompassing tumor cells, stromal cells and multiple interactions between the numerous cellular populations in the tumor is also being increasingly recognized [4].

For obvious reasons, the introduction of new therapies into the clinical practice has lagged significantly behind the discoveries in the basic research. In fact, in many cancers considerable progress of research of the molecular basis of tumor growth and progression has still failed to translate into the clinic. Laboratory medicine as a specialty has been instrumental in translating the progress in basic science into clinical medicine and has been bridging the gap between basic and clinical research. The role of laboratory medicine in medical, surgical or radiation oncology is by no means limited to the assessment of the tumor burden or dynamics. An at least equally important, although still underestimated aspect of laboratory medicine in oncology, is the detection of toxicity. Today, clinical oncology and laboratory medicine may be regarded as indispensable partners in the care of cancer patients [5].

During the past decades, with the expanding role of laboratory medicine in oncology and the introduction of new medical treatment options, the number of papers dealing with these topics has increased sharply. Oncology has also been increasingly covered in Clinical Chemistry and Laboratory Medicine. The present review aims at highlighting both the currently established and potential contribution of laboratory medicine in oncology, mainly medical oncology. As outlined above, both medical oncology and laboratory medicine are relatively new specialties, and the interaction between the two disciplines is even more recent, therefore the focus in the present review is on more recent papers defining a field that was almost non-existent only 50 years ago.

50 Years of Clinical Chemistry and Laboratory Medicine in the changing world of laboratory medicine and oncology

Numerous parallels may come to the mind when looking back at the past 50 years since the first issue of Clinical Chemistry and Laboratory Medicine. The journal has changed as the world was changing. What began as a German language journal (with some papers in English, and, exceptionally, in French) has been transformed into an international journal publishing exclusively English language papers. The name of the journal changed four times from the original Zeitschrift für Klinische Chemie to Zeitschrift für Klinische Chemie und Klinische Biochemie in 1967, putting the English translation Journal of Clinical Chemistry and Clinical Biochemistry ahead of the German title in 1976, in 1991 changing the title to European Journal of Clinical Chemistry and Clinical Biochemistry and, finally, in 1998 to Clinical Chemistry and Laboratory Medicine. The outside world was changing, too. The journal is still being published in Berlin as it was 50 years ago. The first issue appeared <18 months after the construction of the Berlin Wall, just few hundred meters east from the building that serves as the editorial office for the journal. Similarly to this infamous structure that divided the city, Germany, Europe and, finally, the world into two parts that had limited communication, the topics we encountered in the first issues of the Journal were similarly separated from the practice of then nascent specialty of medical oncology. In fact, only a few papers in the first years were dedicated to topics with any relation to cancer or oncology (one paper each in 1963 and 1964, three papers in 1965 and three papers in 1966). Thus, in the first 3 years of the journal (under the title Zeitschrift für Klinische Chemie) only five papers related to cancer were published, much less than in many single monthly issues in recent years. This can be contrasted, e.g., with nine papers accompanied by an editorial covering oncology topics in the October 2011 issue of Clinical Chemistry and Laboratory Medicine[5–14]. Moreover, the predominant topic of communications in the early issues of the journal, the experimental studies on the Ehrlich mouse ascites tumor had little to say to the physicians practicing clinical oncology at that time [15–17].

The science and practice of medicine changed even more rapidly than society. Like the Berlin Wall, the separation between the worlds of clinical chemistry and laboratory medicine on one side and clinical oncology in the broad sense (encompassing medical, surgical and radiation oncology) has also disappeared, probably less conspicuously, but by no means less permanently. Fortunately, the advances of clinical oncology also resulted in improved prognosis of cancer patients. In one of the papers of relevance to clinical practice, a brief report describing elevations of lactate dehydrogenase published in 1967, the final sentence of the paragraph describing a case of a patient with leukemia is “Tod 14 Tage später” (Death 14 days later) [18]. In contrast, in Clinical Chemistry and Laboratory Medicine, volume 50, we can read a case report of a patient with breast cancer presenting with liver metastases and hyperbilirubinemia who is alive and well on targeted therapy with trastuzumab 7 years after the diagnosis [19].

With time, the number of papers on topics related to oncology in Zeitschrift für Klinische Chemie und Klinische Biochemie, Journal of Clinical Chemistry and Clinical Biochemistry, European Journal of Clinical Chemistry and Clinical Biochemistry and, finally, Clinical Chemistry and Laboratory Medicine increased sharply. The number of papers on topics related to oncology almost tripled in Clinical Chemistry and Laboratory Medicine during the 15 years of the journal bearing this name (Figure 1). Starting from 2009 many issues of the journal include a section “Cancer diagnostics” featuring papers focusing on oncology.

Figure 1 
					Number of papers on oncological topics in Clinical Chemistry and Laboratory Medicine.
					Shown is the number of papers dealing with oncological topics from 1998 (volume 36, first volume under the title of Clinical Chemistry and Laboratory Medicine) to 2012 (volume 50).
Figure 1

Number of papers on oncological topics in Clinical Chemistry and Laboratory Medicine.

Shown is the number of papers dealing with oncological topics from 1998 (volume 36, first volume under the title of Clinical Chemistry and Laboratory Medicine) to 2012 (volume 50).

The current role of laboratory medicine in the management of cancer patient

From its humble beginnings, evidenced by scarce papers in the first years of the journal, the role of laboratory medicine in the management of cancer patients has expanded enormously. Molecular changes associated with the tumor arise in the complex interactions of tumor microenvironment, and subsequently spread systemically. Laboratory methods may be used both to study the tumor microenvironment as well as to study tumor effects on the systemic level. Four major areas of clinical chemistry/laboratory medicine in clinical (mostly medical) oncology may be defined, including tumor biomarkers, detection of the host response to tumor, biomarkers of toxicity and measurement/prediction of anticancer drug concentrations.

Tumor markers – the old and the new

The measurement of tumor markers is a traditional topic that has been providing a link between oncology and laboratory medicine for decades. During that time, the well-established paradigm of using tumor marker detection for early diagnosis of cancer or cancer recurrence has evolved into a more diversified utilization. The traditional concept of tumor marker, a protein or glycoprotein, has been expanded to cover a wide range of molecules [20], and the term biomarker is now preferred to reflect this molecular diversity. Also peripheral blood is no longer the exclusive sample matrix used for assessment of these biomarkers, and other sample matrices including urine, breath, malignant effusions, secretions or tumor tissue are now being increasingly used.

Increased concentrations of circulating protein or carbohydrate biomarkers (“tumor markers”) have been detected across the range of tumors, from common tumors, mainly of epithelial origin (carcinomas) [11, 21–28] to more rare tumors, e.g., meningeoma [29]. Despite the many controversies, the advent of tumor markers has virtually transformed the management of some malignant disorders, including prostate cancer, epithelial ovarian carcinoma and hepatocellular carcinoma. The determination of prostate specific antigen (PSA) plays an indispensable role in the early diagnosis as well as the management of prostate cancer [30]. In fact, the introduction of PSA has transformed the way this tumor is treated and, consequently, may have changed the course of this disease. The evaluation of disease course in patients with prostate cancer is largely based on serial PSA measurements. The detection of a sustained rise of PSA is often the basis for the diagnosis of relapse, termed biochemical relapse, and leads to institution of therapy.

Determination of tumor markers also plays an important role in the management of epithelial ovarian carcinoma. Since surgery is the principal prognostic factor in patients affected with this tumor it is important that the diagnosis is established before the procedure and the patient may be referred to an experienced surgeon [31]. The introduction of human epididymis protein 4 (HE4) has provided another biomarker to improve the preoperative diagnosis of epithelial ovarian carcinoma [21–24]. The simultaneous determination of carbohydrate antigen (CA) 125 and HE4 with imaging represents a reliable estimate of the probability of the presence of epithelial ovarian carcinoma [22, 23].

Determination of α-fetoprotein plays an essential role in the diagnosis of hepatocellular carcinoma. In fact, hepatocellular carcinoma represents an exception in the rule that histological or cytological verification is always required for cancer diagnosis. In patients with hepatocellular carcinoma, the detection of high serum concentrations of α-fetoprotein in association with specific imaging findings can be a substitute for histological diagnosis because the biopsy could be associated with considerable risk in some patients. Measurement of α-fetoprotein is used for early diagnosis of hepatocellular carcinoma in patients at risk, to detect recurrence and to follow the patients during or after the therapy [27]. Des-γ-carboxyprothrombin and fucosylated α-fetoprotein are sometimes used along with α-fetoprotein [27].

The role of tumor markers in most other tumors is far less established compared to prostate cancer, hepatocellular carcinoma or epithelial ovarian carcinoma. Much has been said about misuses of tumor marker determination in clinical practice [32], sometimes culminating in what has been termed as “tumor marker terrorism”. In fact, virtually all tumor markers currently used in routine practice may be increased in a range of benign disorders as recently comprehensively reviewed by Trapé et al. [8]. Increased tumor marker concentrations in subjects with benign disorders represent an important limitation that has to be considered when using tumor marker determination as a diagnostic aid.

While the utility of the determination of “classical” tumor markers may still be, in many instances, controversial, the potential of non-protein molecules as cancer biomarkers is being increasingly recognized. In fact, there has been a constant movement from assessments based on biomarkers only indirectly linked with the pathogenesis of cancer to the detection of biomarkers associated with causal events in the tumor growth and progression. In this respect, the analysis of nucleic acids has been of major importance.

Pathogenesis of cancer involves a sequence of events caused by the alterations in the genome of the cancer cells. Typically, mutations of multiple genes are found that are responsible for the aberrant behavior of the cell. Gene mutations may be associated with a specific tumor phenotype, and, most importantly, the response to therapy. The determination of specific gene mutations has entered routine practice. Some of these mutations are germ-line mutations and indicate high risk for developing a specific tumor, e.g., BRCA-1 mutations associated with breast and ovarian carcinoma [33]. Interestingly, the presence of BRCA-1 mutations is associated with specific tumor phenotype and sensitivity to some agents, e.g., platinum compound, and targeted therapy with poly-(ADP-ribose)-polymerase 1 (PARP1) is currently under investigation for the treatment of these tumors. Other mutations are acquired mutations present only in tumor cells. Among these mutations, Kirsten RAS (KRAS) mutations are used to predict the efficacy of monoclonal antibodies targeting epidermal growth factor receptor (EGFR), cetuximab and panitumumab, in metastatic colorectal carcinoma, BRAF mutations are associated with the response to vemurafenib, a BRAF inhibitor, in malignant melanoma, and EGFR mutations predict the efficacy of low-molecular-weight EGFR inhibitors erlotinib and gefitinib in non-small cell lung cancer [34, 35].

In addition to the detection of DNA mutations, the role of epigenetic aberrations [36], mainly DNA methylation [37–39], but also histone acetylation [40] and microRNAs [6, 41], in the tumor growth and progression has been increasingly recognized in recent years. Although current methods of investigation of epigenetic processes in daily practice have been limited almost entirely to the study of DNA methylation, with the advent of drugs targeting epigenetic pathways, mainly histone deacetylase inhibitors, the applications may spread to the study of histone modifications to follow the efficacy of therapy [42]. Among other classes of nucleic acids, considerable attention has been devoted in the past few years to microRNAs. MicroRNAs are small non-coding RNA molecules that post-transcriptionally modulate gene expression, and are involved in the epigenetic regulation of different biological processes relevant to tumor growth and metastasis, including cell proliferation, apoptosis, differentiation and epithelial-mesenchymal transition [6]. Specifically cell-free circulating microRNAs represent promising new biomarkers, both in aiding the diagnosis and determining the prognosis, in a range of malignant tumors, including lung cancer or epithelial ovarian carcinoma [6, 41, 43].

The role of molecular biomarkers is well-established in predictive diagnostics in relation with targeted therapies. In fact, the utilization of many targeted agents that are active in 5%–15% of patients affected with a particular tumor would not be possible without the use of predictive diagnostics that not only identifies patients likely to benefit from the targeted agents, but also excludes patients in whom there would be no benefit, incurring only cost and toxicity. This concept can be illustrated on the example of human epidermal growth factor receptor (HER)-2-positive breast carcinoma. Historically, in patients with breast carcinoma the over-expression of HER-2 has been associated with poor outcome. The introduction of drugs targeting HER-2 has not only transformed the management of HER-2- positive breast carcinoma, but also served as a paradigm for the introduction of targeted therapy in medical oncology [19]. HER-2 is a 185-kDa transmembrane tyrosine kinase receptor, member of the HER family of receptors that also includes HER-1, HER-3 and HER-4. No ligand has been described for the HER-2 receptor. HER-2 is constitutively active and may undergo ligand-independent dimerization with other HER receptors, leading to tyrosine phosphorylation and activation of downstream signaling [44]. As the over-expression of HER-2 is encountered only in about 15% of patients with breast carcinoma, the determination of tumor HER-2 status is of critical importance. From the methods used to examine HER-2 status of the tumor, immunohistochemistry and fluorescence in situ hybridization (FISH) are used in clinical practice [44]. HER-2-targeted agents are effective only in breast cancer patients with tumors over-expressing HER-2. Given the relatively low prevalence of HER-2 over-expressing tumors, only the identification of a reliable predictive biomarker, in this case the determination of HER-2 over-expression, means that the activity of HER-2 targeted agents could be established. Currently, a number of anti-HER-2 therapies are available or are about to be registered, including humanized monoclonal antibody trastuzumab, low-molecular-weight inhibitor lapatinib, another antibody pertuzumab and an antibody-toxin conjugate trastuzumab emtamsine (T-DM1). Recently, trastuzumab has been introduced into the therapy of HER-2 over-expressing metastatic gastric carcinoma. HER-2 protein may also be measured in serum, but in contrast to the tumor HER-2 status, serum HER-2 determination has so far found little use in clinical practice [45, 46]. Other important predictive biomarkers used in medical oncology recently identified, including the presence KRAS mutations in gastrointestinal carcinomas [47], BRAF mutations [35, 48], and EGFR mutations in non-small cell lung cancer [34], have been mentioned above. Complex assays utilizing multigene expression profiles, e.g., Oncotype Dx, that determine the expression of a panel of genes associated with tumor progression and metastases have been introduced into the clinical practice and are used in therapeutic decisions [9].

The technological advances have also allowed the identification and measurement of circulating cancer cells in patients with solid tumors [7]. While the presence of circulating neoplastic cells has been used for decades in the diagnosis and monitoring of the effect of therapy in patients with leukemia, and it has been known for more than a century that circulating tumor cells are present in patients with solid tumors, the detection of these cells has been hampered by an extremely low frequency with approximately one circulating tumor cells in 106–107 leukocytes [7]. Moreover, the prognostic significance of detection of circulating tumor cells is limited by the phenomenon of metastatic inefficiency, with only one circulating tumor cell in many thousands being able to initiate metastasis [49]. Despite these problems, circulating tumor cells represent an emerging class of cancer biomarkers that allow the most direct assessment of the tumor, close to a “liquid biopsy”. However, a number of studies illustrate the difficulties associated with the detection of circulating tumor cells [50, 51].

Another aspect of laboratory medicine of importance in clinical practice is the detection of infection by oncogenic viruses, e.g., human papilloma viruses that cause carcinoma of the uterine cervix as well as other tumors, including anal cancer and some cases of head and neck carcinomas [52, 53]. Among other biomarkers, the detection of parameters associated with angiogenesis, one of the hallmarks of cancer, is being slowly introduced into the laboratory and the clinic [54]. Serum concentrations of vascular endothelial growth factor (VEGF) and its receptors are increased in patients with advanced cancer [55]. Some data indicate that high circulating VEGF concentrations may predict the response to anti-VEGF therapy, e.g., bevacizumab [56]. Similarly, the determination of matrix metaloproteinases, enzymes that play an important role in tumor invasion and formation of metastases has been introduced [57].

There are many issues in the field of cancer biomarkers that urgently need to be addressed. For example, apart from circulating VEGF there are still no predictive biomarkers for drugs targeting VEGF or VEGF receptors that currently represent the backbone of therapy in some tumors, e.g., metastatic renal cell carcinoma [58], and these therapies are currently prescribed more or less indiscriminately. While most assays used either tumor tissue or peripheral blood (including plasma or serum) as the sample matrix for biomarker determination, methods have been introduced to determine tumor biomarkers in cerebrospinal fluid [59–61], urine [62–64], stool [65], secretions [66] or breath [67–69]. Breath analysis has been in the forefront of research in recent years and volatile organic compounds have been identified as potential tumor biomarkers [67–69]. The presence of volatile compounds associated with tumor growth may also represent an explanation for canine olfactory detection of cancer, a phenomenon that has been well documented [70, 71].

Detection of the host response against tumor

Tumor growth is not only the result of proliferation of populations of malignant cells, but an equally important role may be played by the host response. Until recently, this aspect has been mostly neglected or underestimated. The changing view of the role of the host response can be illustrated by the two reviews by Hanahan and Weinberg that have been already discussed above [3, 4]. It is symptomatic that the earlier review enumerated six hallmarks mostly associated with the properties of tumor cells, while revisiting the topic one decade later the authors included evasion of immune response as an important hallmark and elicitation of inflammatory response as one of the enabling characteristics [4]. It is now evident that tumor growth elicits host response mediated both by specific (immune) and non-specific (inflammatory) signals that can be detected using different laboratory methods. It is being increasingly recognized that immune response in relation to tumor growth is a double-edge sword, and immune and inflammatory phenomena may result in both tumor elimination and progression. Unlike in infectious disorders, the detection of specific immune response (formation of antibodies) is mostly not being utilized diagnostically, but to assess host-tumor interactions. Although the practical significance of the measurement of host response to tumor may be presently viewed as limited, the study of these phenomena is important for the understanding of the pathogenesis of tumor progression and metastatic spread. Moreover, there is cumulative evidence indicating that the host response to neoplasia is an important prognostic factor. With the expanding role of targeted therapies, including agents that modulate immune response, e.g., ipilimumab, in the management of cancer patients, the role of laboratory assessment of immune or inflammatory response is bound to increase.

The specific (immune) and non-specific (inflammatory) phenomena associated with the host response against the tumor may be difficult to separate and many parameters currently used reflect both aspects of the host response. Parameters used to assess the host response include the phenotypic and functional characterization of leukocyte populations, determination of antibodies, cytokines that regulate the host response or small molecules produced or metabolized as the result of host response, and the host response may be studied both at the local and systemic levels. Tumors may, e.g., induce the formation of specific antibodies, such as antibodies against HER-2 in patients with breast cancer [72]. There are reports indicating that the detection of autoantibodies in patients with cancer could be useful diagnostically as demonstrated for anti-survivin antibodies in patients with colorectal cancer [73]. Interestingly, the presence of autoimmune response may be a positive prognostic factor, e.g., the presence of anti-thyroid peroxidase antibodies in patients with breast cancer [74].

The systemic immune and inflammatory phenomena associated with the host response to neoplasia result in marked changes of molecules that may be measured in the laboratory and used to assess the host response. It has been known for many years that cancer progression is associated with acute phase response that results in elevations of some circulating proteins, e.g., C-reactive protein, or the decrease of other proteins, e.g., albumin [75, 76]. Among other laboratory parameters, malignant tumors have also been associated with an increase of concentrations of neopterin or changes of tryptophan metabolism.

Neopterin is a pteridine produced by macrophages activated with interferon-gamma from guanosine triphophate (GTP) in a reaction catalyzed by GTP cyclohydrolase I as a first step in a pathway leading to the production of 5,6,7,8-tetrahydrobiopterin. For a reason that is still not fully clarified activated human macrophages possess only the activity of GTP cyclohydrolase I while lacking the activity of enzymes distal to this pathway leading to the accumulation of the large amounts of neopterin. Neopterin may be determined in the serum or in the urine by immunoassay or by high-performance liquid chromatography [77, 78]. Increased neopterin concentrations are observed across a spectrum of disorders ranging from acute myocardial infarction [79, 80], infectious or autoimmune disorders [81] to malignant tumors [82]. Increased urinary or serum neopterin concentrations in patients are also associated with poor prognosis across the range of primary tumors [82, 83]. Moreover, increased neopterin concentrations are associated with the decrease of circulating CD4+ T-lymphocytes [84] and a decline of lymphocyte function [85]. Neopterin may also serve as a biomarker of surgical stress, e.g., in patients undergoing liver resection for cancer or benign disorders [86]. Neopterin concentrations may also increase after administration of systemic chemotherapy [87].

Another enzyme induced by interferon-γ is indoleamine 2,3-dioxygenase that catalyzes the production of kynurenine from tryptophan [88]. Indoleamine 2,3-dioxygenase may be expressed by both tumor cells and monocytes infiltrating the tumor. Tryptophan depletion results in cell growth suppression, affecting both the tumor cell proliferation [89] as well as the proliferation of lymphocytes [90]. Experimental data indicate that the induction of indoleamine 2,3-dioxygenase may result in the suppression of immune response in the tumor microenvironment. Moreover, kynurenine and its metabolite hydroxykynurenine have antiproliferative activity [91].

Activation of the immune response may also be associated with increased production of nitric oxide, although to lesser extent in humans compared to rodents. The production of nitric oxide may be assessed by measuring serum or urinary nitrate concentrations [92]. Increased nitrate concentrations have been described in patients with disorders associated with the activation of the immune system, e.g., inflammatory bowel disease [93], but, in contrast to neopterin or kynurenine, data on nitrate in cancer patients are scanty.

The utility of the concept of tumor microenvironment is evident in relation to the study of the host response against the tumor [94]. At the local level, the presence of leukocytes may be detected histologically in tumors. It has been demonstrated that of particular interest is the presence of lymphocytes that are termed tumor-infiltrating lymphocytes. Across a spectrum of tumors of different primary locations, the presence of tumor-infiltrating lymphocytes has been associated with prognosis [95], or response to chemotherapy [96]. Populations of leukocytes, including lymphocytes and monocytes/macrophages in the tumor microenvironment may be studied even more easily in malignant effusions, e.g., ascites [97, 98], and these measurements may be used for the determination of response to therapy in the tumor microenvironment [99]. The concentrations of molecules associated with immune and inflammatory response, e.g., neopterin or tryptophan, have also been studied in malignant ascites.

Biomarkers of treatment toxicity

Unless adequately treated cancer is a deadly disease. Therefore, aggressive therapies need to be prescribed for cancer patients. Local treatments, including surgery or radiation therapy, may be very effective in the control of localized tumors, but what kills the patient is usually distant spread resulting in systemic metastases. Systemic disease can usually be treated only by a systemic approach, i.e., drug therapy. Given the catastrophic consequences when systemic treatment fails, anti-cancer drugs are usually administered at maximum tolerated doses. Only in selected cases the problem of systemic toxicity may be circumvented by regional administration of the drug, e.g., hepatic arterial infusion [100]. Due to individual differences in treatment tolerance, the administration of maximum tolerated dose results in serious toxicity in a significant proportion of patients. Therefore, the prediction and early detection of treatment toxicity is of major importance in clinical practice.

The problem of detection of treatment toxicity has been studied for all agents, but is of particular importance for cytotoxic drugs, a class of drugs characterized by low therapeutic index. These drugs damage not only the neoplastic cells, but also normal tissues. The toxicity of cytotoxic drugs may be, in general, divided into toxic effects that are common to most cytotoxic drugs and result from toxicity on rapidly dividing tissues, and to toxic effects that are specific to a given drug. The toxic effects common to most cytotoxic drugs include toxic effects resulting from the suppression of bone marrow and gastrointestinal mucosa.

The myelotoxicity may easily be detected with simple peripheral blood cell count that can be performed in almost any laboratory. The detection of the second most common side effect of chemotherapy, gastrointestinal toxicity, still relies heavily on complaints reported by the patient that may be unreliable. For some agents, e.g., patupilone, gastrointestinal toxicity is the principal adverse event determining treatment tolerance [101]. Currently, there are no laboratory methods used in daily practice that would detect or could be used to follow gastrointestinal disturbances associated with administration of anticancer agents. Promising results have been reported with the measurement of gastrointestinal permeability or determination of serum citrulline concentrations. Testing of intestinal permeability using differential sugar absorption measured as urinary recovery has been established as a useful test in the assessment of disorders of the small intestine, with the results being expressed as a ratio [102]. Changes of intestinal permeability in cancer patients may be detected by determining the disaccharide/monosaccharide (e.g., lactulose/mannitol) ratio [103]. Parameters of intestinal permeability are increased in cancer patients even before the therapy, and it has been demonstrated that the lactulose/mannitol ratio is markedly increased in cancer patients experiencing gastrointestinal toxicity [103, 104]. Gastrointestinal mucosal damage may also be detected by measuring serum citrulline. Low citrulline concentrations have been reported in patients treated with intensive chemotherapy [105]. Gastrointestinal toxicity is also one of the principal manifestations of the graft vs. host disease in patients after allogeneic hematopoietic stem cell transplantation. The results of a pilot study indicate that serum cytokeratin 18 concentrations may represent an early biomarker of gastrointestinal graft vs. host disease [10].

The toxic effects on other organs are less common, but the consequences may still be catastrophic. Prominent among these side effects is cardiotoxicity, a toxicity that typically manifests late after the therapy. Concerns about the devastating consequences of cardiotoxicity of anticancer agents and efforts to identify patients at risk and detect early signs of cardiac dysfunction have led to the constitution of an interdisciplinary field of cardioncology. Early biomarkers of cardiotoxicity have been identified, including troponins, natriuretic peptides and markers of endothelial dysfunction or myocardial ischemia [106]. Cardiotoxicity as a result of anthracycline exposure is relatively rare. Emerging data indicate that a more common problem is the acceleration of atherosclerosis in young patients treated with curative chemotherapy. This phenomenon was first documented in survivors of germ cell tumors. Germ cell tumors typically affect young adults and can be cured even at the metastatic stage. Several studies have noted an increased prevalence of cardiovascular risk factors and a higher incidence of cardiovascular events in survivors of germ cell tumors [107–109]. Laboratory risk factors of atherosclerosis in patients with cancer are, similar to the general population, associated with alterations of parameters of immune and inflammatory response [110]. A negative impact of chemotherapy on the risk factors of atherosclerosis has also been described in patients with breast cancer [111]. Another example of late toxicity with devastating consequences is represented by second primary tumors, including myeloid neoplasms, that may be associated with genetic predisposition [112], e.g., BRCA-1 mutations, that are being increasingly used in testing persons at risk [33, 113].

New therapies are associated with hitherto unknown toxicities. This can be demonstrated in the case of hypomagnesemia induced by the antibodies against EGFR [114]. Hypomagnesemia is, in general, an underestimated problem in clinical medicine [115]. Severe hypomagnesemia that may be observed after anticancer agents, including anti-EGFR antibodies or platinum compounds could be manifested by serious symptoms. Therefore, serial magnesium determination is essential in the management of cancer patients treated with these drugs.

Monitoring or predicting the concentrations of anticancer drugs

For the drugs with a narrow therapeutic index, e.g., cytotoxic agents, the optimal approach for dosing involves measuring of systemic drug concentrations. With the advent of new technological approaches, the possibilities of monitoring concentrations of anticancer drugs have expanded significantly in recent years. While in clinical practice, dosing based on body surface area is usually used, the actual doses obtained vary significantly in individual patients. The technological advances in liquid chromatography, gas chromatography and mass spectrometry made possible the development of therapeutic drug monitoring in patients treated with cytotoxic chemotherapy [116]. Monitoring of serum concentrations may also be useful in cancer patients treated with targeted agents. Highly sensitive methods have been developed for a number of agents, e.g., mammalian targeted of rapamycin inhibitors [117]. However, monitoring of therapeutic drug concentrations has still not entered routine use in medical oncology. A number of factors limit the use of therapeutic drug monitoring, including the complexity of drug-tumor interactions, cumulation of side effects or late toxicity, instability of anticancer drugs, heterogeneity and instability of tumor cells, local bioactivation that is not reflected in systemic drug concentrations or variable tumor blood supply [116]. One of the principal reasons for limited utilization of therapeutic drug monitoring is also that in the daily practice the information on drug concentrations may arrive at a moment when it is too late to adjust the medication dose. From this perspective, the prediction of drug concentrations based on pharmacogenetic information seems to be a better strategy compared to serial drug monitoring.

The marked inter-individual differences in the metabolism of anticancer agents have sparked the fast advancement of pharmacogenomic approaches in medical oncology [9]. Broadly speaking, pharmacogenetic analyses in medical oncology may be distinguished into the determination of the presence of appropriate target in the tumor, or to the study of genetic polymorhisms associated with drug metabolism. Genetic analyses of the tumor were introduced to predict response to targeted agents as outlined above. There is some evidence, that tumors associated with germ-line mutations of some genes, e.g., BRCA-1, may be more responsive to certain agents, such as platinum compounds [118]. Of equal importance is the pharmacogenetic analysis of genes involved in the drug metabolism to predict both treatment efficacy and toxicity [9]. The polymorphisms of genes coding for the cytochrome P450 enzymes have been studied for more than a decade [119]. For example, the significance of polymorphisms of the cytochrome P450 (CYP) 2D6 the enzyme responsible for biotransformation of tamoxifen to its active metabolite, endoxifen, is well characterized [9]. Prediction of treatment toxicity may be even more important. 5-Fluorouracil represents a drug constituting the backbone of chemotherapy regimens in gastrointestinal cancer. 5-Fluorouracil is well-tolerated in the majority of patients, but may rarely induce serious, sometimes even fatal toxicity that is associated with polymorphisms of the gene coding for dihydropyrimidine dehydrogenase, the enzyme responsible for 5-fluorouracil degradation. Genotyping may identify patients who are at risk of serious toxicity [120].

Measurement of drug concentrations may be useful not only in anticancer therapy, but also in symptomatic treatment of patients with advanced cancer. A method allowing for simultaneous measurement of urinary concentrations of 19 drugs or metabolites using liquid chromatography-tandem mass spectrometry has recently been introduced [121].

Conclusions

The role of laboratory medicine in the management of cancer patients is rapidly expanding. In the spectrum of different presentations of malignant disorders we can hardly find a situation in which determination of a diagnostic, predictive or prognostic biomarker, a parameter reflecting the host response and/or treatment toxicity, or a study of pharmacokinetic or pharmacodynamic parameters would not play an indispensable role in patient management. The emancipation of clinical chemistry/laboratory medicine as well as medical oncology has been accomplished simultaneously in mutual symbiosis. Instead of the two Cinderellas of 50 years ago we have now two princesses ready to work together to take the leading role in the most prestigious endeavor, saving human lives. As Brugsch and Schütte, the first publishers of this journal, wrote in an article introducing its first issue “…die Aufgabe des Wissenschaftlers ist, zum Wohle der Mitmenschen Wissen und Werte zu schaffen, denn nur dann wird die Wissenschaft bestehen” (“the role of the scientist is to create knowledge and values for the benefit of the fellow humans because only then it is science”) [122]. This sentence is as valid today as it was 50 years ago. The cooperation of physicians and other scientists of different specialities is the only way forward in the global effort to combat cancer.

Conflict of interest statement

Authors’ conflict of interest disclosure: The authors stated that there are no conflicts of interest regarding the publication of this article.

Research funding: None declared.

Employment or leadership: None declared.

Honorarium: None declared.


Corresponding author: Prof. MUDr. Bohuslav Melichar, PhD, Department of Oncology, Palacký University Medical School and Teaching Hospital, Fakultní nemocnice, I.P. Pavlova 6, 775 20 Olomouc, Czech Republic, Phone: +420 588444288, Fax: +420 588442522

About the author

Bohuslav Melichar

Professor Bohuslav Melichar, MD, PhD was born on March 6, 1965 in Hradec Králové (then Czechoslovakia). After attending, between 1983 and 1989, the Charles University Medical School in Hradec Králové, Czechoslovakia, he graduated in 1989. Between 1989 and 1992 he was a resident in internal medicine at 2nd Department of Internal Medicine, Charles University Medical School and Teaching Hospital in Hradec Králové, Czechoslovakia. In 1992 he entered a Ph.D. program in medical immunology and worked simultaneously as a physician at the 2nd Department of Internal Medicine, Charles University Medical School and Teaching Hospital in Hradec Králové. In 1992 he was a visiting scholar in the University of Innsbruck, Austria, working with Professor Wachter and Professor Fuchs on the pteridines as indicators of immune response. In 1993–1994 he was a visiting scholar in the University of Bordeaux, France, working in the laboratory of Professor Mégraud, focusing on Helicobacter pylori infection. Between 1994 and 1996 he was a Fulbright scholar at the M.D. Anderson Cancer Center, Houston, Texas, United States, working first on antitumor activity of macrophages in experimental colorectal cancer, and subsequently in the laboratory of Professor Ralph S. Freedman on immunology of peritoneal carcinomatosis and intraperitoneal immunotherapy. After coming back from the United States he continued working as a physician in the 2nd Department of Internal Medicine and, later, in the Section of Hematology/Oncology of the Charles University Medical School and Teaching Hospital in Hradec Králové, Czech Republic. Simultaneously, he served as an instructor in internal medicine at the Charles University Medical School. In October 1998 he defended Ph.D. thesis “Increased production of neopterin and other indicators of immune activation in patients with malignant tumors and its biologic significance”. In March 1999 he passed a specialization examination in medical oncology. Since January 2000 he was working as medical oncologist in the Department of Oncology and Radiotherapy of the Charles University Medical School and Teaching Hospital in Hradec Králové. Between March 2001 and August 2001 he was (on a NATO scholarship) visiting scientist in the Department of Gynecologic Oncology of the University of Texas, M.D. Anderson Cancer Center, Houston (working again with Professor Ralph S. Freedman). In 2001 he was appointed associate professor of medicine and in November 2006 full professor. In February 2008 Professor Melichar was appointed the Head of the Department of Oncology, Palacký University Medical School and Teaching Hospital, Olomouc, Czech Republic.

In January 2009 Professor Melichar was appointed associate editor for oncology of Clinical Chemistry and Laboratory Medicine. Professor Melichar is currently the president-elect of the International Society of Pteridinology and scientific secretary of the Czech Oncology Society. He is a member of Czech Society of Clinical Biochemisty, American Society of Clinical Oncology and European Society of Medical Oncology. The main areas of interest of Professor Melichar are the study of biomarkers of immune response in cancer patients and the study of new medical therapies for solid tumors, including breast cancer, renal cell carcinoma, ovarian cancer and gastrointestinal malignancies. Professor Melichar is the author of 147 papers in journals with the impact factor, including 93 papers as the first author.

References

1. Diamandopoulos AA, Goudas PC. The late Greco-Roman and Byzantine contribution to the evolution of laboratory examinations of bodily excrement. Part 1: urine, menses and stools. Clin Chem Lab Med 2003;41:963–9.10.1515/CCLM.2003.147Search in Google Scholar

2. Diamandopoulos A, Goudas PC. The late Greco-Roman and Byzantine contribution towards the evolution of laboratory examinations of bodily excrement. Part 2: sputum, vomit, blood, sweat, autopsies. Clin Chem Lab Med 2005;43:90–6.10.1515/CCLM.2005.014Search in Google Scholar

3. Hanahan D, Weinberg RA. Hallmarks of cancer. Cell 2000;100:57–70.10.1016/S0092-8674(00)81683-9Search in Google Scholar

4. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144:646–74.10.1016/j.cell.2011.02.013Search in Google Scholar PubMed

5. Melichar B, Plebani M. Laboratory medicine: an essential partner in the care of cancer patients. Clin Chem Lab Med 2011;49:1575–8.10.1515/CCLM.2011.244Search in Google Scholar PubMed

6. Markou A, Liang Y, Lianidou E. Prognostic, therapeutic and diagnostic potential of microRNAs in non-small cell lung cancer. Clin Chem Lab Med 2011;49:1591–603.10.1515/CCLM.2011.661Search in Google Scholar PubMed

7. Lianidou ES, Markou A. Circulating tumor cells as emerging tumor biomarkers in breast cancer. Clin Chem Lab Med 2011;49:1579–90.10.1515/CCLM.2011.628Search in Google Scholar

8. Trape J, Filella X, Alsina-Donadeu M, Juan-Pereira L, Bosch-Ferrer A, Rigo-Bonnin R. Increased plasma concentrations of tumour markers in the absence of neoplasia. Clin Chem Lab Med 2011;49:1605–20.10.1515/CCLM.2011.694Search in Google Scholar PubMed

9. Soh TI, Yong WP, Innocenti F. Recent progress and clinical importance on pharmacogenetics in cancer therapy. Clin Chem Lab Med 2011;49:1621–32.10.1515/CCLM.2011.715Search in Google Scholar

10. Waterhouse M, Samek E, Torres M, Bertz H, Finke J. Diagnostic utility of a soluble cytokeratin 18 assay for gastrointestinal graft-vs.-host disease detection. Clin Chem Lab Med 2011;49:1695–7.10.1515/CCLM.2011.644Search in Google Scholar

11. Darago A, Sapota A, Matych J, Nasiadek M, Skrzypinska-Gavrysiak M, Kilanowicz A. The correlation between zinc and insulin-like growth factor 1 (IGF-1), its binding protein (IGBF-3) and prostate-specific antigen (PSA) in prostate cancer. Clin Chem Lab Med 2011;49:1699–705.10.1515/CCLM.2011.651Search in Google Scholar PubMed

12. Albert H, Santos S, Battaglia E, Brito M, Monteiro C, Bagrel D. Differential expression of CDC25 phosphatases splice variants in human breast cancer cells. Clin Chem Lab Med 2011;49:1707–14.10.1515/CCLM.2011.635Search in Google Scholar PubMed

13. Wang F, Chen L, Ding W, Wang J, Luo L, Cong H, et al. Serum APRIL, a potential tumor marker in pancreatic cancer. Clin Chem Lab Med 2011;49:1715–9.10.1515/CCLM.2011.608Search in Google Scholar PubMed

14. Yamada S, Tabata C, Tabata R, Fukuoka K, Nakano T. Clinical significance of pleural effusion mesothelin in malignant pleural mesothelioma. Clin Chem Lab Med 2011;49:1721–6.10.1515/CCLM.2011.242Search in Google Scholar PubMed

15. Warburg O, Gawehn K, Geissler W, Lorenz S. Uber Heilung von Mause-Ascites-Krebs durch D- un L-Glycerinaldehyd. Z Klin Chem 1963;1:175–7.10.1515/cclm.1963.1.6.175Search in Google Scholar

16. Luhrs W, Chrometzka K. Anaerobe Glykolyse bei Ehrlich-Aszites-Tumorzellen. 2. Mitteilung: Beinflussung durch Antidiabetika. Z Klin Chem 1965;3:25–9.10.1515/cclm.1965.3.1.25Search in Google Scholar

17. Gunther T, Wenzel M, Eberhard T. Zum Stoffwechsel des Glukosamins in Ascitestumorzellen. III. Mitteilung: Stoffwechselwirkungen von Glukosaminderivaten. Z Klin Chem 1965;3:76–81.10.1515/cclm.1965.3.3.76Search in Google Scholar

18. Voigt A. Über eine Besonderheit des LDH-Isoenzyms im Serum. Z Klin Chem Klin Biochem 1967;5:146–7.Search in Google Scholar

19. Melichar B, Plebani M. Targeted therapy for HER-2: personalized medicine for her, too. Clin Chem Lab Med 2012;50:1–4.10.1515/cclm.2011.724Search in Google Scholar PubMed

20. Hayes DF, Bast RC, Desch CE, Fritsche H, Kemeny NE, Jessup M, et al. Tumor marker utility grading system: a framework to evaluate clinical utility of tumor markers. J Natl Cancer Inst 1996;88:1456–66.10.1093/jnci/88.20.1456Search in Google Scholar PubMed

21. Kim YM, Whang DH, Park J, Kim SH, Lee SW, Park HA, et al. Evaluation of the accuracy of serum human epididymis protein 4 in combination with CA125 for detecting ovarian cancer: a prospective case-control study in a Korean population. Clin Chem Lab Med 2011;49:527–34.10.1515/CCLM.2011.085Search in Google Scholar PubMed

22. Montagnana ME, Danese E, Ruzzenente O, Bresciani V, Nuzzo T, Gelati G, et al. The ROMA (Risk of Ovarian Malignancy Algorithm) for estimating the risk of epithelial ovarian cancer in women presenting with pelvic mass: is it really useful? Clin Chem Lab Med 2011;49:521–5.10.1515/CCLM.2011.075Search in Google Scholar PubMed

23. Lenhard M, Stieber P, Hertlein L, Kirschenhofer A, Furst S, Mayr D, et al. The diagnostic accuracy of two human epididymis protein 4 (HE4) testing systems in combination with CA125 in the differential diagnosis of ovarian masses. Clin Chem Lab Med 2011;49:2081–8.10.1515/CCLM.2011.709Search in Google Scholar PubMed

24. Plebani M. HE4 in gynecological cancers: report of a European investigators and experts meeting. Clin Chem Lab Med 2012;50:2127–36.10.1515/cclm-2012-0373Search in Google Scholar PubMed

25. Zanotti L, Bignotti E, Calza S, Bandiera E, Ruggeri G, Galli C, et al. Human epididymis protein 4 as a serum marker for diagnosis of endometrial carcinoma and prediction of clinical outcome. Clin Chem Lab Med 2012;50:2189–98.10.1515/cclm-2011-0757Search in Google Scholar PubMed

26. Hertlein L, Stieber P, Kirschenhofer A, Furst S, Mayr D, Hofman K, et al. Human epididymis protein 4 (HE4) in benign and malignant diseases. Clin Chem Lab Med 2012;50:2181–88.10.1515/cclm-2012-0097Search in Google Scholar PubMed

27. Masuda T, Miyoshi E. Cancer biomarkers for hepatocellular carcinoma: from traditional markers to recent topics. Clin Chem Lab Med 2011;49:959–66.10.1515/CCLM.2011.152Search in Google Scholar PubMed

28. Kontos CK, Scorilas A. Kallikrein-related peptidases (KLKs): a gene family of novel cancer biomarkers. Clin Chem Lab Med 2012;50:1877–91.10.1515/cclm-2012-0247Search in Google Scholar PubMed

29. Abeloos L, Lefranc F. Could serum measurments of S100 proteins be reliable markers to predict recurrence in meningiomas? Clin Chem Lab Med 2011;49:971–5.10.1515/CCLM.2011.157Search in Google Scholar PubMed

30. Ulmert D, Vickers AJ, Scher HI, Becker C, Iversen P, Frankel D, et al. Rapid elimination kinetics of free PSA or human kallikrein-related peptidase 2 after initiation of gonadotropin-releasing hormone-antagonist treatment of prostate cancer: potential for rapid monitoring of treatment responses. Clin Chem Lab Med 2012;50:1993–98.10.1515/cclm-2011-0967Search in Google Scholar PubMed PubMed Central

31. Plebani M, Melichar B. ROMA or death: advances in epithelial ovarian cancer diagnosis. Clin Chem Lab Med 2011;49:443–5.10.1515/CCLM.2011.073Search in Google Scholar PubMed

32. Basuyau J-P, Leroy M, Brunelle P. Determination of tumor markers in serum. Pitfalls and good practice. Clin Chem Lab Med 2001;39:1227–33.10.1515/CCLM.2001.197Search in Google Scholar

33. Poumpouridou N, Kroupis C. Hereditary breast cancer: beyond BRCA genetic analysis. Clin Chem Lab Med 2012;50:423–34.10.1515/cclm-2011-0840Search in Google Scholar PubMed

34. Cho MC, Choi CM, Jang S, Jang S, Park CJ, Chi HS, et al. Direct sequencing in cytological specimens as a potentially useful strategy for detecting EGFR mutations as non-small cell lung cancer patients. Clin Chem Lab Med 2012;50:565–72.10.1515/cclm.2011.704Search in Google Scholar PubMed

35. Li G, Luo X, He J, Zhu Z, Yu G, Qin H, et al. A novel liquidchip platform for simultaneous detection of 70 alleles of DNA somatic mutations on EGFR, KRAS, BRAF and PIK3CA from formalin-fixed and parafin-embedded slides containing tumor tissue. Clin Chem Lab Med 2011;49:191–5.10.1515/CCLM.2011.040Search in Google Scholar PubMed

36. Melichar B, Kroupis C. Cancer epigenomics: moving slowly, but at a steady pace from lab bench to clinical practice. Clin Chem Lab Med 2012;50:1699–701.10.1515/cclm-2012-0495Search in Google Scholar PubMed

37. Markopoulou S, Nikolaidis G, Liloglou T. DNA methylation biomarkers in biological fluids for early detection of respiratory tract cancer. Clin Chem Lab Med 2012;50:1723–31.10.1515/cclm-2012-0124Search in Google Scholar PubMed

38. Torano EG, Petrus S, Fernandez AF, Fraga MF. Global DNA hypomethylation in cancer: review of validated methods and clinical significance. Clin Chem Lab Med 2012;50: 1733–42.10.1515/cclm-2011-0902Search in Google Scholar PubMed

39. Heichman KA, Warren JD. DNA methylation biomarkers and their utility for solid cancer diagnostics. Clin Chem Lab Med 2012;50:1707–21.10.1515/cclm-2011-0935Search in Google Scholar

40. Yang Z, Zhou L, Wu LM, Xie HY, Zhang F, Zheng SS. Combination of polymorphisms within the HDAC1 and HDAC3 gene predict tumor recurrence in hepatocellular carcinoma patients that have undergone transplant therapy. Clin Chem Lab Med 2010;48:1785–91.10.1515/CCLM.2010.353Search in Google Scholar PubMed

41. Kuhlmann JD, Rasch J, Wimberger P, Kasimir-Bauer S. microRNA and the pathogenesis of ovarian cancer – a new horizon for molecular diagnostics and treatment? Clin Chem Lab Med 2012;50:601–15.10.1515/cclm-2011-0847Search in Google Scholar PubMed

42. Tate CR, Rhodes LV, Segar HC, Driver JL, Pounder FN, Burow ME, et al. Targeting triple-negative breast cancer cells with the histone deacetylase inhibitor panobinostat. Breast Cancer Res 2012;14:R79.10.1186/bcr3192Search in Google Scholar PubMed PubMed Central

43. Vinci S, Gelmini S, Pratesi N, Conti S, Malentacchi F, Simi L, et al. Genetic variants in miR-146a, miR196a2, miR-499 and their influence on relative expression in lung cancers. Clin Chem Lab Med 2011;49:2073–80.10.1515/CCLM.2011.708Search in Google Scholar PubMed

44. Tafe LJ, Tsongalis GJ. The human epidermal growth factor receptor 2 (HER2). Clin Chem Lab Med 2012;50:23–30.10.1515/cclm.2011.707Search in Google Scholar

45. Sorensen PD, Madsen J, Brandslund I. Serum HER-2/ECD in monitoring breast cancer patients. Clin Chem Lab Med 2012;50:175–6.10.1515/cclm.2011.722Search in Google Scholar

46. Molina R, Escudero JM, Munoz M, Auge JM, Filella X. Circulating levels of HER-2/neu oncoprotein in breast cancer. Clin Chem Lab Med 2012;50:5–21.10.1515/cclm.2011.822Search in Google Scholar PubMed

47. Guo W, Zhang C, Wu J, Song B, Wang B, Zhou Y, et al. Unlabeled-probe high-resolution melting to detect KRAS codon 12 and 13 mutations in pancreatic adenocarcinoma tissues. Clin Chem Lab Med 2012;50:1035–40.10.1515/cclm-2011-0601Search in Google Scholar PubMed

48. Pelizzo MR, Boschin IM, Barollo S, Pennelli G, Toniato A, Zambonin L, et al. BRAF analysis by fine needle aspiration biopsy of thyroid nodules improves preoperative identification of papillary thyroid carcinoma and represents a prognostic factor. A mono-institutional experience. Clin Chem Lab Med 2011;49:325–9.10.1515/CCLM.2011.031Search in Google Scholar

49. Weiss L. Inefficiency of metastasis from colorectal carcinomas. Relationship to local therapy for hepatic metastasis. Cancer Treat Res 1994;69:1–11.10.1007/978-1-4615-2604-9_1Search in Google Scholar

50. Hekimian K, Stein EL, Pachmann U, Pachmann K. Demasking of epithelial cell adhesion molecule (EpCAM) on circulating epithelial tumor cells by Tween20 treatment in breast cancer patients. Clin Chem Lab Med 2012;50:701–8.10.1515/cclm.2011.812Search in Google Scholar PubMed

51. Vendittelli F, Raffaelli M, Fadda G, Carelli-Alinovi C, Paolillo C, Bellantone R, et al. Blood presence of circulating oncofetal fibronectin mRNA, by RT-PCR, does not represent a useful specific marker for the management and follow-up of thyroid cancer patients. Clin Chem Lab Med 2012;50:715–20.10.1515/cclm-2011-0848Search in Google Scholar PubMed

52. Kroupis C, Vourlidis N. Human papilloma virus (HPV) molecular diagnostics. Clin Chem Lab Med 2011;49:1783–99.10.1515/cclm.2011.685Search in Google Scholar

53. Micalessi IM, Boulet GA, Bogers JJ, Benoy IH, Depuydt CE. High-throughput detection, genotyping and quantification of the human papillomavirus using real-time PCR. Clin Chem Lab Med 2012;50:655–61.10.1515/cclm.2011.835Search in Google Scholar PubMed

54. Zimmermann R, Weisbach V, Eckstein R. Choice of sample for the analysis of circulating angiogenic cytokines. Clin Chem Lab Med 2008;46:1328.10.1515/CCLM.2008.255Search in Google Scholar

55. Pezzilli R, Fabbri D, Corsi MM, Imbrogno A, Barassi A, Morselli-Labate AM, et al. Plasma concentrations of angiogenetic factors and angiogenetic inhibitors in patients with ductal pancreatic neoplasms. A pilot study. Clin Chem Lab Med 2011;49:1047–51.10.1515/CCLM.2011.159Search in Google Scholar

56. Van Cutsem E, de Haas S, Kang YK, Ohtsu A, Tebbutt NC, Ming Xu J, et al. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a biomarker evaluation from the AVAGAST randomized phase III trial. J Clin Oncol 2012;30:2119–27.10.1200/JCO.2011.39.9824Search in Google Scholar

57. Emara M, Cheung PY, Grabowski K, Sawicki G, Wozniak M. Serum levels of matrix metalloproteinase-2 and -9 and conventional tumor markers (CEA and CA 19-9) in patients with colorectal and gastric cancers. Clin Chem Lab Med 2009;47:993–1000.10.1515/CCLM.2009.212Search in Google Scholar

58. Buchler T, Klapka R, Melichar B, Brabec P, Dusek L, Vyzula R, et al. Sunitinib followed by sorafenib or vice versa for metastatic renal cell carcinoma – data from the Czech registry. Ann Oncol 2012;23:395–401.10.1093/annonc/mdr065Search in Google Scholar

59. Corsini E, Bernardi G, Gaviani P, Silvani A, de Grazia U, Ciusani E, et al. Intrathecal synthesis of tumor markers is a highly sensitive test in the diagnosis of leptomeningeal metastasis from solid cancers. Clin Chem Lab Med 2009;47:874–9.10.1515/CCLM.2009.183Search in Google Scholar

60. Mroczko B, Groblewska M, Lukaszewicz-Zajac M, Bandurski R, Kedra B, Szmitkowski M. Pre-treatment serum and plasma levels of matrix metalloproteinase 9 (MMP-9) and tissue inhibitor of matrix metalloproteinases 1 (TIMP-1) in gastric cancer patients. Clin Chem Lab Med 2009;47:1133–9.10.1515/CCLM.2009.253Search in Google Scholar

61. Liu BL, Cheng JX, Zhang W, Zhang X, Wang R, Lin H, et al. Quantitative detection of multiple gene promoter hypermethylation in tumor tissue, serum, and cerebrospinal fluid predicts prognosis of malignant gliomas. Neuro Oncol 2010;12:540–8.10.1093/neuonc/nop064Search in Google Scholar

62. Melichar B, Solichová D, Freedman RS. Neopterin as an indicator of immune activation and prognosis in patients with gynecological malignancies. Int J Gynecol Cancer 2006;16:240–52.10.1111/j.1525-1438.2006.00294.xSearch in Google Scholar

63. Solichova D, Melichar B, Blaha V, Klejna M, Vavrova J, Palicka V, et al. Biochemical profile and survival in non-agenarians. Clin Biochem 2001;34:563–9.10.1016/S0009-9120(01)00261-2Search in Google Scholar

64. Melichar B, Urbanek L, Krcmova L, Kalabova H, Svobodova I, Dragounova E, et al. Urinary neopterin in patients with ovarian cancer. Pteridines 2006;17:145–53.10.1515/pteridines.2006.17.4.145Search in Google Scholar

65. Kalimutho M, Blanco Gdel V, Gravina P, Cretella M, Manucci L, Mannisi E, et al. Quantitative denaturing high performance liquid chromatography (Q-dHPLC) detection of APC long DNA in faeces from patients with colorectal cancer. Clin Chem Lab Med 2010;48:1303–11.10.1515/cclm.2010.245Search in Google Scholar

66. Antill YC, Mitchell G, Johnson SA, Devereux L, Milner A, Di lulio J, et al. Gene methylation in breast ductal fluid from BRCA1 and BRCA2 mutation carriers. Cancer Epidemiol Biomarkers Prev 2010;19:265–74.10.1158/1055-9965.EPI-09-0359Search in Google Scholar PubMed

67. Buszewski B, Ulanowska A, Kowalkowski T, Cieslinski K. Lung cancer biomarkers by hyphenated separation techniques and chemometrics. Clin Chem Lab Med 2012;50:573–81.10.1515/cclm.2011.769Search in Google Scholar

68. Ligor M, Ligor T, Bajtarevic A, Ager C, Pienz M, Klieber M, et al. Determination of volatile organic compounds in exhaled breath of patients with lung cancer using solid phase microextraction and gas chromatography mass spectrometry. Clin Chem Lab Med 2009;47:550–60.10.1515/CCLM.2009.133Search in Google Scholar PubMed

69. Badjagbo K. Exhaled breath analysis for early cancer detection: principle and progress in direct mass spectrometry techniques. Clin Chem Lab Med 2012;50:1893–902.10.1515/cclm-2012-0208Search in Google Scholar PubMed

70. Lippi G, Cervellin G. Canine olfactory detection of cancer versus laboratory testing: myth or opportunity? Clin Chem Lab Med 2012;50:435–9.10.1515/cclm.2011.672Search in Google Scholar

71. Melichar B, Plebani M. Cancer diagnosis: from dogs to DNA or from DNA to dogs? Clin Chem Lab Med 2012;50:415–8.10.1515/cclm.2011.786Search in Google Scholar PubMed

72. Lauterlein JJ, Petersen ER, Olsen DA, Ostergaard B, Brandslund I. Quantification of HER2 autoantibodies in the amplification phenomenon of HER2 in breast cancer. Clin Chem Lab Med 2011;49:877–83.10.1515/CCLM.2011.135Search in Google Scholar PubMed

73. Chen JS, Chen KT, Fan WC, Yu JS, Chang YS, Chan EC. Combined analysis of survivin autoantibody and carcinoembryonic antigen biomarkers for improved detection of colorectal cancer. Clin Chem Lab Med 2010;48:719–25.10.1515/CCLM.2010.123Search in Google Scholar PubMed

74. Farahati J, Roggenbuck D, Gilman E, Schutte M, Jagminaite E, Zakavi RS, et al. Anti-thyroid peroxidase antibodies are associated with the absence of distant metastases in patients with newly diagnosed breast cancer. Clin Chem Lab Med 2012;50:709–14.10.1515/cclm.2011.819Search in Google Scholar

75. Groblewska M, Mroczko B, Wereszczynska-Siemiatkowska U, Kedra B, Lukaszewicz M, Baniukiewicz A, et al. Serum interleukin 6 (IL-6) and C-rective protein (CRP) levels in colorectal adenoma and cancer patients. Clin Chem Lab Med 2008;46:1423–8.10.1515/CCLM.2008.278Search in Google Scholar PubMed

76. Krzystek-Korpacka M, Matusiewicz M, Diakowska D, Grabowski K, Blachut K, Kustrzeba-Wojcicka I, et al. Acute-phase response proteins are related to cachexia and accelerated angiogenesis in gastroesophageal cancers. Clin Chem Lab Med 2008;46:359–64.10.1515/CCLM.2008.089Search in Google Scholar PubMed

77. Hammerer-Lercher A, Moser C, Leichtfried V, Schobersberger W, Griesmacher A, Fuchs D. Comparison of a commercial urinary neopterin radioimmunoassay with high performance liquid chromatography. Clin Chem Lab Med 2012;50:1075–8.10.1515/cclm-2011-0861Search in Google Scholar PubMed

78. Moutereau S, Ech Chad N, Devanlay M, Esmilaire L, Loric S. Improved neopterin ELISA kit: a good compromise between HPLC results and clinical practice. Clin Chem Lab Med 2011;49:553–4.10.1515/CCLM.2011.071Search in Google Scholar PubMed

79. Melichar B, Gregor J, Solichova D, Lukes J, Tichy M, Pidrman V. Increased urinary neopterin in acute myocardial infarction. Clin Chem 1994;40:338–9.10.1093/clinchem/40.2.338Search in Google Scholar

80. Montagnana M, Lippi G, Danese E, Salvagno GL, Cervellin G, Guidi GC. Serum concentration of neopterin on admission does not improve the diagnostic performance of highly-sensitive troponin I. Clin Chem Lab Med 2012;50:747–8.10.1515/cclm.2011.829Search in Google Scholar PubMed

81. Plata-Nazar K, Jankowska A. Clinical usefulness of determining the concentration of neopterin. Pteridines 2011;22:77–89.10.1515/pteridines.2011.22.1.77Search in Google Scholar

82. Melichar B, Solichova D, Melicharova K, Malirova E, Cermanova M, Zadak Z. Urinary neopterin in patients with advanced colorectal carcinoma. Int J Biol Markers 2006;21:190–8.10.1177/172460080602100309Search in Google Scholar PubMed

83. Cincibuch J, Cwiertka K, Prochazkova-Studentova H, Zezulova M, Neoral C, Kalabova H, et al. Prognostic significance of serum neopterin in patients with esophageal carcinoma. Pteridines 2012;23:22–6.10.1515/pteridines.2012.23.1.22Search in Google Scholar

84. Melichar B, Touskova M, Solichova D, Kralickova P, Kopecky O. CD4+ T-lymphocytopenia and systemic immune activation in patients with primary and secondary liver tumours. Scand J Clin Lab Inv 2001;61:363–70.10.1080/003655101316911404Search in Google Scholar PubMed

85. Melichar B, Jandik P, Krejsek J, Solichova D, Drahosova M, Skopec F, et al. Mitogen-induced lymphocyte proliferation and systemic immune activation in cancer patients. Tumori 1996;82:218–20.10.1177/030089169608200306Search in Google Scholar

86. Jerin A, Pozar-Lukanovic N, Sojar V, Stanisavljevic D, Paver-Erzen V, Osredkar J. Neopterin-an early marker of surgical stress and hypoxic reperfusion damage during liver surgery. Clin Chem Lab Med 2002;40:663–6.10.1515/CCLM.2002.113Search in Google Scholar PubMed

87. Melichar B, Kalabova H, Krcmova L, Kasparova M, Plisek J, Cerman J, et al. Urinary neopterin in patients with metastatic colon cancer treated with patupilone. Pteridines 2011;22:61–5.10.1515/pteridines.2011.22.1.61Search in Google Scholar

88. Krcmova L, Solichova D, Melichar B, Kasparova M, Plisek J, Sobotka L, et al. Determination of neopterin, kynurenine, tryptophan and creatinine in human serum by high throuput HPLC. Talanta 2011;85:1466–71.10.1016/j.talanta.2011.06.027Search in Google Scholar PubMed

89. Burke F, Knowles RG, East N, Balkwill FR. The role of indoleamine 2,3-dioxygenase in the anti-tumour activity of human interferon-gama in vivo. Int J Cancer 1995;60:115–22.10.1002/ijc.2910600117Search in Google Scholar PubMed

90. Munn DH, Shafizadeh E, Attwood JT, Bondarev I, Pashine A, Mellor AL. Inhibition of T cell proliferation by macrophage tryptophan catabolism. J Exp Med 1999;189:1363–72.10.1084/jem.189.9.1363Search in Google Scholar PubMed PubMed Central

91. Melichar B, Ferrandina G, Verschraegen CF, Loercher A, Abbruzzese JL, Freedman RS. Growth inhibitory effects of aromatic fatty acids on ovarian tumor cell lines. Clin Cancer Res 1998;4:3069–76.Search in Google Scholar

92. Djordjevic VB, Stojanovic I, Cosic V, Zvezdanovic L, Deljanin-Ilic M, Dimic S, et al. Serum neopterin, nitric oxide, inducible nitric oxide synthase and tumor necrosis factor-alpha levels in patients with ischemic heart disease. Clin Chem Lab Med 2008;46:1149–55.10.1515/CCLM.2008.213Search in Google Scholar PubMed

93. Melichar B, Karlicek R, Tichy M. Increased urinary nitrate excretion in inflammatory bowel disease. Eur J Clin Chem Clin Biochem 1994;32:3–4.10.1515/cclm.1994.32.1.3Search in Google Scholar PubMed

94. Rahir G, Moser M. Tumor microenvironment and lymphocyte infiltration. Cancer Immunol Immunother 2012;61:751–9.10.1007/s00262-012-1253-1Search in Google Scholar PubMed

95. Tomsova M, Melichar B, Sedlakova I, Steiner I. Prognostic significance of CD3+ tumor-infiltrating lymphocytes in ovarian carcinoma. Gyn Oncol 2008;108:415–20.10.1016/j.ygyno.2007.10.016Search in Google Scholar PubMed

96. Hornychova H, Melichar B, Tomsova M, Mergancova J, Urminska H, Ryska A. Tumor-infiltrating lymphocytes predict response to neoadjuvant chemotherapy in patients with breast carcinoma. Cancer Invest 2008;26:1024–31.10.1080/07357900802098165Search in Google Scholar PubMed

97. Melichar B, Freedman RS. Immunology of the peritoneal cavity: relevance for host-tumor relation. Int J Gynecol Cancer 2002;12:3–17.10.1046/j.1525-1438.2002.01093.xSearch in Google Scholar PubMed

98. Melichar B, Savary CA, Patenia R, Templin S, Melicharova K, Freedman RS. Phenotype and antitumor activity of ascitic fluid monocytes in patients with ovarian carcinoma. Int J Gynecol Cancer 2003;13:435–43.10.1046/j.1525-1438.2003.13331.xSearch in Google Scholar PubMed

99. Freedman RS, Vadhan-Raj S, Butts C, Savary C, Melichar B, Verschraegen C, et al. Pilot study of Flt3 ligand comparing intraperitoneal with subcutaneous routes on hematologic and, immunologic responses in patients with peritoneal carcinomatosis and mesotheliomas. Clin Cancer Res 2003;9:5228–37.Search in Google Scholar

100. Melichar B. Hepatic arterial infusion in colorectal carcinoma: is anatomical targeting still relevant in an era of molecularly-targeted therapy? Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2012;156:81–92.10.5507/bp.2012.047Search in Google Scholar PubMed

101. Melichar B, Casado E, Bridgewater J, Bennouna J, Campone M, Vitek P, et al. Clinical activity of patupilone in patients with pretreated advanced/metastatic colon cancer: results of a phase I dose escalation trial. Br J Cancer 2011;105:1646–53.10.1038/bjc.2011.438Search in Google Scholar PubMed PubMed Central

102. Hessels J, Eidhof HH, Steggink J, Roeloffzen WW, Wu K, Tan G, et al. Assessment of hypolactasia and site-specific intestinal permeability by differential sugar absorption of raffinose, lactose, sucrose and mannitol. Clin Chem Lab Med 2003;41:1056–63.10.1515/CCLM.2003.163Search in Google Scholar PubMed

103. Melichar B, Dvorak J, Hyspler R, Zadak Z. Intestinal permeability in the assessment of intestinal toxicity of cytotoxic agents. Chemotherapy 2005;51:336–8.10.1159/000088957Search in Google Scholar PubMed

104. Melichar B, Kohout P, Bratova M, Solichova D, Kralickova P, Zadak Z. Intestinal permeability in patients with chemotherapy-induced stomatitis. J Cancer Res Clin Oncol 2001;127:314–8.10.1007/s004320000209Search in Google Scholar PubMed

105. Lutgens LC, Blijlevens NM, Deutz NE, Donnelly JP, Lambin P, de Pauw BE. Monitoring myeloablative therapy-induced small bowel toxicity by serum citrulline concentration. A comparison with sugar permeability tests. Cancer 2005;103:191–9.10.1002/cncr.20733Search in Google Scholar PubMed

106. Cardinale D, Salvatici M, Sandri MT. Role of biomarkers in cardioncology. Clin Chem Lab Med 2011;49:1937–48.10.1515/CCLM.2011.692Search in Google Scholar PubMed

107. Huddart RA, Norman A, Shahidi M, Horwich A, Coward D, Nicholls J, et al. Cardiovascular disease as a long-term complication of treatment for testicular cancer. J Clin Oncol 2003;21:1513–23.10.1200/JCO.2003.04.173Search in Google Scholar PubMed

108. Meinardi MT, Gietema JA, van der Graaf WT, van Veldhuisen DJ, Runne MA, Sluiter WJ, et al. Cardiovascular morbidity in long-term survivors of metastatic testicular cancer. J Clin Oncol 2000;18:1725–32.10.1200/JCO.2000.18.8.1725Search in Google Scholar PubMed

109. Zagars GK, Ballo MT, Lee AK, Strom SS. Mortality after cure of testicular seminoma. J Clin Oncol 2004;22:640–7.10.1200/JCO.2004.05.205Search in Google Scholar PubMed

110. Kralickova P, Kalabova H, Krcmova L, Kasparova M, Plisek J, Ungermann L, et al. Correlation of peripheral blood CD14+16+ monocytes, urinary neopterin and the risk factors of atherosclerosisin patients with breast carcinoma. Pteridines 2011;22:66–72.10.1515/pteridines.2011.22.1.66Search in Google Scholar

111. Kalabova H, Melichar B, Ungermann L, Dolezal J, Krcmova L, Kasparova M, et al. Intima-media thickness, myocardial perfusion and laboratory risk factors of atherosclerosis in patients with breast cancer treated with anthracycline-based chemotherapy. Med Oncol 2011;28:1281–7.10.1007/s12032-010-9593-1Search in Google Scholar PubMed

112. Melichar B, Laco J, Fridrichova P, Simkovic M, Papajik T, Foretova L. Therapy-related myeloid neoplasms in epithelial ovarian cancer patients carrying BRCA1 mutation: report of two cases. Acta Oncol 2012;51:136–8.10.3109/0284186X.2011.594810Search in Google Scholar PubMed

113. Vietri MT, Molinari AM, De Paola ML, Cantile F, Fasano M, Cioffi M. Identification of novel in-frame deletion in BRCA2 and analysis of variants of BRCA 1/2 in Italian patients affected with hereditary breast and ovarian cancer. Clin Chem Lab Med 2012;50:2171–80.10.1515/cclm-2012-0154Search in Google Scholar PubMed

114. Melichar B, Kralickova P, Hyspler R, Kalabova H, Cerman J, Holeckova P, et al. Hypomagnesaemia in patients with metastatic colorectal carcinoma treated with cetuximab. Hepatogastroenterology 2012;59:366–71.Search in Google Scholar

115. Ismail Y, Ismail AA, Ismail AA. The underestimated problem of using serum magnesium measurements to exclude magnesium deficiency in adults, a health warning is needed for normal results. Clin Chem Lab Med 2010;48:323–7.10.1515/CCLM.2010.077Search in Google Scholar PubMed

116. Dumez H, Guetens G, De Boeck G, Highley MS, Maes RA, van Oosterom AT, et al. The relevance of therapeutic drug monitoring in plasma and erythrocytes in anti-cancer drug treatment. Clin Chem Lab Med 2004;42:1219–27.10.1515/CCLM.2004.244Search in Google Scholar PubMed

117. Ivanova M, Artusi C, Polo G, Zaninotto M, Plebani M. High-throughput LC-MS/MS method for monitoring sirolimus and everolimus in the routine clinical laboratory. Clin Chem Lab Med 2011;49:1151–8.10.1515/CCLM.2011.192Search in Google Scholar PubMed

118. Melichar B, Fridrichova P, Tomsova M, Malirova E. Repeat chemosensitivity of epithelial ovarian carcinoma in a BRCA1 mutation carrier to paclitaxel/platinum combination chemotherapy. Eur J Gynaecol Oncol 2009;30:323–5.Search in Google Scholar

119. MacLeod SL, Nowell S, Massengill J, Jazieh A, McClure G, Plaxco J, et al. Cancer therapy and polymorphisms of cytochromes P450. Clin Chem Lab Med 2000;38:883–7.10.1515/CCLM.2000.128Search in Google Scholar PubMed

120. Di Francia R, Berretta M, Catapano O, Canzoniero LM, Formisano L. Molecular diagnostics for pharmacogenomic testing of fluoropyrimidine based-therapy: costs, methods and applications. Clin Chem Lab Med 2011; 49:1105–11.10.1515/CCLM.2011.181Search in Google Scholar PubMed

121. Yuan C, Heideloff C, Kozak M, Wang S. Simultaneous quantification of 19 drugs/metabolites in urine important for pain management by liquid chromatography-tandem mass spectrometry. Clin Chem Lab Med 2012;50:95–103.10.1515/cclm.2011.739Search in Google Scholar

122. Brugsch J, Schutte E. Zum Geleit. Zeitschrift fur Kliniche Chemie 1963;1:1.10.1515/cclm.1963.1.1.1Search in Google Scholar

Received: 2012-07-31
Accepted: 2012-08-01
Published Online: 2012-09-15
Published in Print: 2013-01-01

©2013 by Walter de Gruyter Berlin Boston

Articles in the same Issue

  1. Masthead
  2. Masthead
  3. Editorials
  4. Preface: Happy 50th anniversary!
  5. Clinical Chemistry and Laboratory Medicine: an appreciation
  6. Clinical Chemistry and Laboratory Medicine: progress and new challenges for our 50-year-old journal
  7. History
  8. 50th anniversary of Clinical Chemistry and Laboratory Medicine – a historical overview1)
  9. Reviews
  10. The CCLM contribution to improvements in quality and patient safety
  11. The theory of reference values: an unfinished symphony
  12. A history of HbA1c through Clinical Chemistry and Laboratory Medicine
  13. Contributions of CCLM to advances in quality control
  14. Laboratory diagnostics of myocardial infarction – troponins and beyond
  15. Laboratory hemostasis: milestones in Clinical Chemistry and Laboratory Medicine
  16. Laboratory medicine and medical oncology: the tale of two Cinderellas
  17. Genetics and molecular biology in laboratory medicine, 1963–2013
  18. Laboratory hematology in the history of Clinical Chemistry and Laboratory Medicine
  19. Current state of diagnostic technologies in the autoimmunology laboratory
  20. Genetic defects in folate and cobalamin pathways affecting the brain
  21. Expanded newborn screening and confirmatory follow-up testing for inborn errors of metabolism detected by tandem mass spectrometry
  22. Diabetes as a complication of adipose tissue dysfunction. Is there a role for potential new biomarkers?
  23. Mini Reviews
  24. Towards harmonization of quality indicators in laboratory medicine
  25. Clinical applications of maternal plasma fetal DNA analysis: translating the fruits of 15 years of research
  26. Bioanalytical methods for quantitation of levamisole, a widespread cocaine adulterant
  27. Metal release from hip prostheses: cobalt and chromium toxicity and the role of the clinical laboratory
  28. Opinion Papers
  29. Adding value to laboratory medicine: a professional responsibility
  30. Preanalytical quality improvement: in quality we trust
Downloaded on 13.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/cclm-2012-0496/html
Scroll to top button