Cancer biomarker

Last updated
Questions that can be answered by biomarkers Cancer biomarker figure.png
Questions that can be answered by biomarkers

A cancer biomarker refers to a substance or process that is indicative of the presence of cancer in the body. A biomarker may be a molecule secreted by a tumor or a specific response of the body to the presence of cancer. Genetic, [1] epigenetic, [2] proteomic, [3] glycomic, [4] and imaging biomarkers can be used for cancer diagnosis, prognosis, and epidemiology. Ideally, such biomarkers can be assayed in non-invasively collected biofluids like blood or serum. [5]

Contents

Cancer is a disease that affects society at a world-wide level. By testing for biomarkers, early diagnosis can be given to prevent deaths. Cancer deaths by type, 2, OWID.svg
Cancer is a disease that affects society at a world-wide level. By testing for biomarkers, early diagnosis can be given to prevent deaths.

While numerous challenges exist in translating biomarker research into the clinical space; a number of gene and protein based biomarkers have already been used at some point in patient care; including, AFP (liver cancer), BCR-ABL (chronic myeloid leukemia), BRCA1 / BRCA2 (breast/ovarian cancer), BRAF V600E (melanoma/colorectal cancer), CA-125 (ovarian cancer), CA19.9 (pancreatic cancer), CEA (colorectal cancer), EGFR (Non-small-cell lung carcinoma), HER-2 (Breast Cancer), KIT (gastrointestinal stromal tumor), PSA (prostate specific antigen) (prostate cancer), S100 (melanoma), and many others. [6] [7] [8] [9] [10] [11] [12] [13] [14] [15] Mutant proteins themselves detected by selected reaction monitoring (SRM) have been reported to be the most specific biomarkers for cancers because they can only come from an existing tumor. [16] About 40% of cancers can be cured if detected early through examinations. [17]

Definitions of cancer biomarkers

Organizations and publications vary in their definition of biomarker. In many areas of medicine, biomarkers are limited to proteins identifiable or measurable in the blood or urine. However, the term is often used to cover any molecular, biochemical, physiological, or anatomical property that can be quantified or measured.

The National Cancer Institute (NCI), in particular, defines biomarker as a: “A biological molecule found in blood, other body fluids, or tissues that is a sign of a normal or abnormal process, or of a condition or disease. A biomarker may be used to see how well the body responds to a treatment for a disease or condition. Also called molecular marker and signature molecule." [18]

In cancer research and medicine, biomarkers are used in three primary ways: [19]

  1. To help diagnose conditions, as in the case of identifying early stage cancers (diagnostic)
  2. To forecast how aggressive a condition is, as in the case of determining a patient's ability to fare in the absence of treatment (prognostic)
  3. To predict how well a patient will respond to treatment (predictive)

Role of biomarkers in cancer research and medicine

Uses of biomarkers in cancer medicine

Risk assessment

Cancer biomarkers, particular those associated with genetic mutations or epigenetic alterations, often offer a quantitative way to determine when individuals are predisposed to particular types of cancers. Notable examples of potentially predictive cancer biomarkers include mutations on genes KRAS, p53, EGFR, erbB2 for colorectal, esophageal, liver, and pancreatic cancer; mutations of genes BRCA1 and BRCA2 for breast and ovarian cancer; abnormal methylation of tumor suppressor genes p16, CDKN2B, and p14ARF for brain cancer; hypermethylation of MYOD1, CDH1, and CDH13 for cervical cancer; and hypermethylation of p16, p14, and RB1, for oral cancer. [20]

Diagnosis

Cancer biomarkers can also be useful in establishing a specific diagnosis. This is particularly the case when there is a need to determine whether tumors are of primary or metastatic origin. To make this distinction, researchers can screen the chromosomal alterations found on cells located in the primary tumor site against those found in the secondary site. If the alterations match, the secondary tumor can be identified as metastatic; whereas if the alterations differ, the secondary tumor can be identified as a distinct primary tumor. [21] For example, people with tumors have high levels of circulating tumor DNA (ctDNA) due to tumor cells that have gone through apoptosis. [22] This tumor marker can be detected in the blood, saliva, or urine. [17] The possibility of identifying an effective biomarker for early cancer diagnosis has recently been questioned, in light of the high molecular heterogeneity of tumors observed by next-generation sequencing studies. [23]

Prognosis and treatment predictions

Another use of biomarkers in cancer medicine is for disease prognosis, which take place after an individual has been diagnosed with cancer. Here biomarkers can be useful in determining the aggressiveness of an identified cancer as well as its likelihood of responding to a given treatment. In part, this is because tumors exhibiting particular biomarkers may be responsive to treatments tied to that biomarker's expression or presence. Examples of such prognostic biomarkers include elevated levels of metallopeptidase inhibitor 1 (TIMP1), a marker associated with more aggressive forms of multiple myeloma, [24] elevated estrogen receptor (ER) and/or progesterone receptor (PR) expression, markers associated with better overall survival in patients with breast cancer; [25] [26] HER2/neu gene amplification, a marker indicating a breast cancer will likely respond to trastuzumab treatment; [27] [28] a mutation in exon 11 of the proto-oncogene c-KIT, a marker indicating a gastrointestinal stromal tumor (GIST) will likely respond to imatinib treatment; [29] [30] and mutations in the tyrosine kinase domain of EGFR1, a marker indicating a patient's non-small-cell lung carcinoma (NSCLC) will likely respond to gefitinib or erlotinib treatment. [31] [32]

Pharmacodynamics and pharmacokinetics

Cancer biomarkers can also be used to determine the most effective treatment regime for a particular person's cancer. [33] Because of differences in each person's genetic makeup, some people metabolize or change the chemical structure of drugs differently. In some cases, decreased metabolism of certain drugs can create dangerous conditions in which high levels of the drug accumulate in the body. As such, drug dosing decisions in particular cancer treatments can benefit from screening for such biomarkers. An example is the gene encoding the enzyme thiopurine methyl-transferase (TPMPT). [34] Individuals with mutations in the TPMT gene are unable to metabolize large amounts of the leukemia drug, mercaptopurine, which potentially causes a fatal drop in white blood count for such patients. Patients with TPMT mutations are thus recommended to be given a lower dose of mercaptopurine for safety considerations. [35]

Monitoring treatment response

Cancer biomarkers have also shown utility in monitoring how well a treatment is working over time. Much research is going into this particular area, since successful biomarkers have the potential of providing significant cost reduction in patient care, as the current image-based tests such as CT and MRI for monitoring tumor status are highly costly. [36]

One notable biomarker garnering significant attention is the protein biomarker S100-beta in monitoring the response of malignant melanoma. In such melanomas, melanocytes, the cells that make pigment in our skin, produce the protein S100-beta in high concentrations dependent on the number of cancer cells. Response to treatment is thus associated with reduced levels of S100-beta in the blood of such individuals. [37] [38]

Similarly, additional laboratory research has shown that tumor cells undergoing apoptosis can release cellular components such as cytochrome c, nucleosomes, cleaved cytokeratin-18, and E-cadherin. Studies have found that these macromolecules and others can be found in circulation during cancer therapy, providing a potential source of clinical metrics for monitoring treatment. [36]

Recurrence

Cancer biomarkers can also offer value in predicting or monitoring cancer recurrence. The Oncotype DX® breast cancer assay is one such test used to predict the likelihood of breast cancer recurrence. This test is intended for women with early-stage (Stage I or II), node-negative, estrogen receptor-positive (ER+) invasive breast cancer who will be treated with hormone therapy. Oncotype DX looks at a panel of 21 genes in cells taken during tumor biopsy. The results of the test are given in the form of a recurrence score that indicates likelihood of recurrence at 10 years. [39] [40]

Uses of biomarkers in cancer research

Developing drug targets

In addition to their use in cancer medicine, biomarkers are often used throughout the cancer drug discovery process. For instance, in the 1960s, researchers discovered the majority of patients with chronic myelogenous leukemia possessed a particular genetic abnormality on chromosomes 9 and 22 dubbed the Philadelphia chromosome. When these two chromosomes combine they create a cancer-causing gene known as BCR-ABL. In such patients, this gene acts as the principle initial point in all of the physiological manifestations of the leukemia. For many years, the BCR-ABL was simply used as a biomarker to stratify a certain subtype of leukemia. However, drug developers were eventually able to develop imatinib, a powerful drug that effectively inhibited this protein and significantly decreased production of cells containing the Philadelphia chromosome. [41] [42]

Surrogate endpoints

Another promising area of biomarker application is in the area of surrogate endpoints. In this application, biomarkers act as stand-ins for the effects of a drug on cancer progression and survival. Ideally, the use of validated biomarkers would prevent patients from having to undergo tumor biopsies and lengthy clinical trials to determine if a new drug worked. In the current standard of care, the metric for determining a drug's effectiveness is to check if it has decreased cancer progression in humans and ultimately whether it prolongs survival. However, successful biomarker surrogates could save substantial time, effort, and money if failing drugs could be eliminated from the development pipeline before being brought to clinical trials.

Some ideal characteristics of surrogate endpoint biomarkers include: [43] [44]

  • Biomarker should be involved in process that causes the cancer
  • Changes in biomarker should correlate with changes in the disease
  • Levels of biomarkers should be high enough that they can be measured easily and reliably
  • Levels or presence of biomarker should readily distinguish between normal, cancerous, and precancerous tissue
  • Effective treatment of the cancer should change the level of the biomarker
  • Level of the biomarker should not change spontaneously or in response to other factors not related to the successful treatment of the cancer

Two areas in particular that are receiving attention as surrogate markers include circulating tumor cells (CTCs) [45] [46] and circulating miRNAs. [47] [48] Both these markers are associated with the number of tumor cells present in the blood, and as such, are hoped to provide a surrogate for tumor progression and metastasis. However, significant barriers to their adoption include the difficulty of enriching, identifying, and measuring CTC and miRNA levels in blood. New technologies and research are likely necessary for their translation into clinical care. [49] [50] [51]

Types of cancer biomarkers

Molecular cancer biomarkers

Tumor typeBiomarker
Breast ER/PR (estrogen receptor/progesteron receptor) [52] [53]
HER-2/neu [52] [53]
Colorectal EGFR [52] [53]
KRAS [52] [54]
UGT1A1 [52] [54]
GastricHER-2/neu [52]
GIST c-KIT [52] [55]
Leukemia/lymphoma CD20 [52] [56]
CD30 [52] [57]
FIP1L1-PDGFRalpha [52] [58]
PDGFR [52] [59]
Philadelphia chromosome (BCR/ABL) [52] [60] [61]
PML/RAR-alpha [52] [62]
TPMT [52] [63]
UGT1A1 [52] [64]
Lung EML4/ALK [52] [65] [66]
EGFR [52] [53]
KRAS [52] [53]
Melanoma BRAF [52] [66]
PancreasElevated levels of leucine, isoleucine and valine [67]
Ovaries CA-125 [68]

Other examples of biomarkers:

Cancer biomarkers without specificity

Not all cancer biomarkers have to be specific to types of cancer. Some biomarkers found in the circulatory system can be used to determine an abnormal growth of cells present in the body. All these types of biomarkers can be identified through diagnostic blood tests, which is one of the main reasons to get regularly health tested. By getting regularly tested, many health issues such as cancer can be discovered at an early stage, preventing many deaths.

The neutrophil-to-lymphocyte ratio has been shown to be a non-specific determinant for many cancers. This ratio focuses on the activity of two components of the immune system that are involved in inflammatory response which is shown to be higher in presence of malignant tumors. [71] Additionally, basic fibroblast growth factor (bFGF) is a protein that is involved in the proliferation of cells. Unfortunately, it has been shown that in the presence of tumors it is highly active which has led to the conclusion that it may help malignant cells reproduce at faster rates. [72] Research has shown that anti-bFGF antibodies can be used to help treat tumors from many origins. [72] Moreover, insulin-like growth factor (IGF-R) is involved in cell proliferation and growth. It has is possible that it is involved in inhibiting apoptosis, programmed cell death due to some defect. [73] Due to this, the levels of IGF-R can be increased when cancer such as breast, prostate, lung, and colorectum is present. [74]

BiomarkerDescriptionBiosensor used
NLR (neutrophil-to-lymphocyte ratio)Elevates with inflammation caused by cancer [75] No
Basic Fibroblast Growth Factor (bFGF)This level increases when a tumor is present, helps with the fast reproduction of tumor cells [76] Electrochemical [77]
Insulin-like Growth Factor (IGF-R)High activity in cancer cells, help reproduction [78] Electrochemical Impedance Spectroscopy Sensor[ citation needed ]

See also

Related Research Articles

<span class="mw-page-title-main">Gastrointestinal stromal tumor</span> Human disease (cancer)

Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal neoplasms of the gastrointestinal tract. GISTs arise in the smooth muscle pacemaker interstitial cell of Cajal, or similar cells. They are defined as tumors whose behavior is driven by mutations in the KIT gene (85%), PDGFRA gene (10%), or BRAF kinase (rare). 95% of GISTs stain positively for KIT (CD117). Most (66%) occur in the stomach and gastric GISTs have a lower malignant potential than tumors found elsewhere in the GI tract.

<span class="mw-page-title-main">Melanoma</span> Cancer originating in melanocytes

Melanoma is the most dangerous type of skin cancer; it develops from the melanin-producing cells known as melanocytes. It typically occurs in the skin, but may rarely occur in the mouth, intestines, or eye. In women, melanomas most commonly occur on the legs; while in men, on the back. Melanoma is frequently referred to as malignant melanoma. However, the medical community stresses that there is no such thing as a 'benign melanoma' and recommends that the term 'malignant melanoma' should be avoided as redundant.

<span class="mw-page-title-main">Cancer immunotherapy</span> Artificial stimulation of the immune system to treat cancer

Cancer immunotherapy (immuno-oncotherapy) is the stimulation of the immune system to treat cancer, improving the immune system's natural ability to fight the disease. It is an application of the fundamental research of cancer immunology and a growing subspecialty of oncology.

<span class="mw-page-title-main">Targeted therapy</span> Type of therapy

Targeted therapy or molecularly targeted therapy is one of the major modalities of medical treatment (pharmacotherapy) for cancer, others being hormonal therapy and cytotoxic chemotherapy. As a form of molecular medicine, targeted therapy blocks the growth of cancer cells by interfering with specific targeted molecules needed for carcinogenesis and tumor growth, rather than by simply interfering with all rapidly dividing cells. Because most agents for targeted therapy are biopharmaceuticals, the term biologic therapy is sometimes synonymous with targeted therapy when used in the context of cancer therapy. However, the modalities can be combined; antibody-drug conjugates combine biologic and cytotoxic mechanisms into one targeted therapy.

<span class="mw-page-title-main">Sunitinib</span> Cancer medication

Sunitinib, sold under the brand name Sutent, is an anti-cancer medication. It is a small-molecule, multi-targeted receptor tyrosine kinase (RTK) inhibitor that was approved by the FDA for the treatment of renal cell carcinoma (RCC) and imatinib-resistant gastrointestinal stromal tumor (GIST) in January 2006. Sunitinib was the first cancer drug simultaneously approved for two different indications.

In biomedical contexts, a biomarker, or biological marker, is a measurable indicator of some biological state or condition. Biomarkers are often measured and evaluated using blood, urine, or soft tissues to examine normal biological processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention. Biomarkers are used in many scientific fields.

In medicine, a biomarker is a measurable indicator of the severity or presence of some disease state. It may be defined as a "cellular, biochemical or molecular alteration in cells, tissues or fluids that can be measured and evaluated to indicate normal biological processes, pathogenic processes, or pharmacological responses to a therapeutic intervention." More generally a biomarker is anything that can be used as an indicator of a particular disease state or some other physiological state of an organism. According to the WHO, the indicator may be chemical, physical, or biological in nature - and the measurement may be functional, physiological, biochemical, cellular, or molecular.

<span class="mw-page-title-main">Oncogenomics</span> Sub-field of genomics

Oncogenomics is a sub-field of genomics that characterizes cancer-associated genes. It focuses on genomic, epigenomic and transcript alterations in cancer.

Triple-negative breast cancer (TNBC) is any breast cancer that either lacks or shows low levels of estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) overexpression and/or gene amplification. Triple-negative is sometimes used as a surrogate term for basal-like.

Minimal residual disease (MRD), also known as Molecular residual disease, is the name given to small numbers of cancer cells that remain in a person either during or after treatment when the patient is in remission. Sensitive molecular tests are either in development or available to test for MRD. These can measure minute levels of cancer cells in tissue samples, sometimes as low as one cancer cell in a million normal cells, either using DNA, RNA or proteins.

A circulating tumor cell (CTC) is a cell that has shed into the vasculature or lymphatics from a primary tumor and is carried around the body in the blood circulation. CTCs can extravasate and become seeds for the subsequent growth of additional tumors (metastases) in distant organs, a mechanism that is responsible for the vast majority of cancer-related deaths. The detection and analysis of CTCs can assist early patient prognoses and determine appropriate tailored treatments. Currently, there is one FDA-approved method for CTC detection, CellSearch, which is used to diagnose breast, colorectal and prostate cancer.

A gene signature or gene expression signature is a single or combined group of genes in a cell with a uniquely characteristic pattern of gene expression that occurs as a result of an altered or unaltered biological process or pathogenic medical condition. This is not to be confused with the concept of gene expression profiling. Activating pathways in a regular physiological process or a physiological response to a stimulus results in a cascade of signal transduction and interactions that elicit altered levels of gene expression, which is classified as the gene signature of that physiological process or response. The clinical applications of gene signatures breakdown into prognostic, diagnostic and predictive signatures. The phenotypes that may theoretically be defined by a gene expression signature range from those that predict the survival or prognosis of an individual with a disease, those that are used to differentiate between different subtypes of a disease, to those that predict activation of a particular pathway. Ideally, gene signatures can be used to select a group of patients for whom a particular treatment will be effective.

<span class="mw-page-title-main">Carlo Gambacorti-Passerini</span> Italian oncologist and hematologist

Carlo Gambacorti-Passerini is an Italian oncologist and hematologist known for his contributions to cancer research.

<span class="mw-page-title-main">Vemurafenib</span> Targeted cancer drug

Vemurafenib (INN), sold under the brand name Zelboraf, is a medication used for the treatment of late-stage melanoma. It is an inhibitor of the B-Raf enzyme and was developed by Plexxikon.

Antineoplastic resistance, often used interchangeably with chemotherapy resistance, is the resistance of neoplastic (cancerous) cells, or the ability of cancer cells to survive and grow despite anti-cancer therapies. In some cases, cancers can evolve resistance to multiple drugs, called multiple drug resistance.

<span class="mw-page-title-main">Circulating tumor DNA</span> Tumor-derived fragmented DNA in the bloodstream

Circulating tumor DNA (ctDNA) is tumor-derived fragmented DNA in the bloodstream that is not associated with cells. ctDNA should not be confused with cell-free DNA (cfDNA), a broader term which describes DNA that is freely circulating in the bloodstream, but is not necessarily of tumor origin. Because ctDNA may reflect the entire tumor genome, it has gained traction for its potential clinical utility; "liquid biopsies" in the form of blood draws may be taken at various time points to monitor tumor progression throughout the treatment regimen.

<span class="mw-page-title-main">Vasculogenic mimicry</span>

Vasculogenic mimicry (VM) is a strategy used by tumors to ensure sufficient blood supply is brought to its cells through establishing new tumor vascularization. This process is similar to tumor angiogenesis; on the other hand vascular mimicry is unique in that this process occurs independent of endothelial cells. Vasculature is instead developed de novo by cancer cells, which under stress conditions such as hypoxia, express similar properties to stem cells, capable of differentiating to mimic the function of endothelial cells and form vasculature-like structures. The ability of tumors to develop and harness nearby vasculature is considered one of the hallmarks of cancer disease development and is thought to be closely linked to tumor invasion and metastasis. Vascular mimicry has been observed predominantly in aggressive and metastatic cancers and has been associated with negative tumor characteristics such as increased metastasis, increased tissue invasion, and overall poor outcomes for patient survival. Vascular mimicry poses a serious problem for current therapeutic strategies due to its ability to function in the presence of Anti-angiogenic therapeutic agents. In fact, such therapeutics have been found to actually drive VM formation in tumors, causing more aggressive and difficult to treat tumors to develop.

Prognostic markers are biomarkers used to measure the progress of a disease in the patient sample. Prognostic markers are useful to stratify the patients into groups, guiding towards precise medicine discovery. The widely used prognostic markers in cancers include stage, size, grade, node and metastasis. In addition to these common markers, there are prognostic markers specific to different cancer types. For example estrogen level, progesterone and HER2 are markers specific to breast cancer patients. There is evidence showing that genes behaving as tumor suppressors or carcinogens could act as prognostic markers due to altered gene expression or mutation. Besides genetic biomarkers, there are also biomarkers that are detected in plasma or body fluid which can be metabolic or protein biomarkers.

Personalized onco-genomics (POG) is the field of oncology and genomics that is focused on using whole genome analysis to make personalized clinical treatment decisions. The program was devised at British Columbia's BC Cancer Agency and is currently being led by Marco Marra and Janessa Laskin. Genome instability has been identified as one of the underlying hallmarks of cancer. The genetic diversity of cancer cells promotes multiple other cancer hallmark functions that help them survive in their microenvironment and eventually metastasise. The pronounced genomic heterogeneity of tumours has led researchers to develop an approach that assesses each individual's cancer to identify targeted therapies that can halt cancer growth. Identification of these "drivers" and corresponding medications used to possibly halt these pathways are important in cancer treatment.

<span class="mw-page-title-main">Tumor mutational burden</span>

Tumour mutational burden is a genetic characteristic of tumorous tissue that can be informative to cancer research and treatment. It is defined as the number of non-inherited mutations per million bases (Mb) of investigated genomic sequence, and its measurement has been enabled by next generation sequencing. High TMB and DNA damage repair mutations were discovered to be associated with superior clinical benefit from immune checkpoint blockade therapy by Timothy Chan and colleagues at the Memorial Sloan Kettering Cancer Center.

References

  1. Calzone KA (May 2012). "Genetic biomarkers of cancer risk". Seminars in Oncology Nursing. 28 (2): 122–128. doi:10.1016/j.soncn.2012.03.007. PMC   10433658 . PMID   22542320.
  2. Herceg Z, Hainaut P (June 2007). "Genetic and epigenetic alterations as biomarkers for cancer detection, diagnosis and prognosis". Molecular Oncology. 1 (1): 26–41. doi:10.1016/j.molonc.2007.01.004. PMC   5543860 . PMID   19383285.
  3. Li D, Chan DW (April 2014). "Proteomic cancer biomarkers from discovery to approval: it's worth the effort". Expert Review of Proteomics. 11 (2): 135–136. doi:10.1586/14789450.2014.897614. PMC   4079106 . PMID   24646122.
  4. Aizpurua-Olaizola O, Toraño JS, Falcon-Perez JM, Williams C, Reichardt N, Boons GJ (2018). "Mass spectrometry for glycan biomarker discovery". TrAC Trends in Analytical Chemistry. 100: 7–14. doi:10.1016/j.trac.2017.12.015.
  5. Mishra A, Verma M (March 2010). "Cancer biomarkers: are we ready for the prime time?". Cancers. 2 (1): 190–208. doi: 10.3390/cancers2010190 . PMC   3827599 . PMID   24281040.
  6. Rhea J, Molinaro RJ (March 2011). "Cancer Biomarkers: Surviving the journey from bench to bedside". Medical Laboratory Observer. Archived from the original on 14 October 2013. Retrieved 26 April 2013.
  7. Behne T, Copur MS (1 January 2012). "Biomarkers for hepatocellular carcinoma". International Journal of Hepatology. 2012: 859076. doi: 10.1155/2012/859076 . PMC   3357951 . PMID   22655201.
  8. Musolino A, Bella MA, Bortesi B, Michiara M, Naldi N, Zanelli P, et al. (June 2007). "BRCA mutations, molecular markers, and clinical variables in early-onset breast cancer: a population-based study". Breast. 16 (3): 280–292. doi:10.1016/j.breast.2006.12.003. hdl: 11381/1629553 . PMID   17257844.
  9. Dienstmann R, Tabernero J (March 2011). "BRAF as a target for cancer therapy". Anti-Cancer Agents in Medicinal Chemistry. 11 (3): 285–295. doi:10.2174/187152011795347469. PMID   21426297.
  10. Lamparella N, Barochia A, Almokadem S (2013). "Impact of Genetic Markers on Treatment of Non-small Cell Lung Cancer". Impact of Genetic Targets on Cancer Therapy. Advances in Experimental Medicine and Biology. Vol. 779. pp. 145–164. doi:10.1007/978-1-4614-6176-0_6. ISBN   978-1-4614-6175-3. PMID   23288638.
  11. Orphanos G, Kountourakis P (2012). "Targeting the HER2 receptor in metastatic breast cancer". Hematology/Oncology and Stem Cell Therapy. 5 (3): 127–137. doi: 10.5144/1658-3876.2012.127 . PMID   23095788.
  12. Deprimo SE, Huang X, Blackstein ME, Garrett CR, Harmon CS, Schöffski P, et al. (September 2009). "Circulating levels of soluble KIT serve as a biomarker for clinical outcome in gastrointestinal stromal tumor patients receiving sunitinib following imatinib failure". Clinical Cancer Research. 15 (18): 5869–5877. doi:10.1158/1078-0432.CCR-08-2480. PMC   3500590 . PMID   19737953.
  13. Bantis A, Grammaticos P (Sep–Dec 2012). "Prostatic specific antigen and bone scan in the diagnosis and follow-up of prostate cancer. Can diagnostic significance of PSA be increased?". Hellenic Journal of Nuclear Medicine. 15 (3): 241–246. PMID   23227460.
  14. Kruijff S, Hoekstra HJ (April 2012). "The current status of S-100B as a biomarker in melanoma". European Journal of Surgical Oncology. 38 (4): 281–285. doi:10.1016/j.ejso.2011.12.005. PMID   22240030.
  15. Ludwig JA, Weinstein JN (November 2005). "Biomarkers in cancer staging, prognosis and treatment selection". Nature Reviews. Cancer. 5 (11): 845–856. doi:10.1038/nrc1739. PMID   16239904. S2CID   25540232.
  16. Wang Q, Chaerkady R, Wu J, Hwang HJ, Papadopoulos N, Kopelovich L, et al. (February 2011). "Mutant proteins as cancer-specific biomarkers". Proceedings of the National Academy of Sciences of the United States of America. 108 (6): 2444–2449. Bibcode:2011PNAS..108.2444W. doi: 10.1073/pnas.1019203108 . PMC   3038743 . PMID   21248225.
  17. 1 2 3 Li X, Ye M, Zhang W, Tan D, Jaffrezic-Renault N, Yang X, Guo Z (February 2019). "Liquid biopsy of circulating tumor DNA and biosensor applications". Biosensors & Bioelectronics. 126: 596–607. doi:10.1016/j.bios.2018.11.037. PMID   30502682. S2CID   56479882.
  18. "biomarker". NCI Dictionary of Cancer Terms. National Cancer Institute. 2011-02-02.
  19. "Biomarkers in Cancer: An Introductory Guide for Advocates" (PDF). Research Advocacy Network. 2010. Archived from the original (PDF) on 2013-10-29. Retrieved 26 April 2013.
  20. Verma M, Manne U (October 2006). "Genetic and epigenetic biomarkers in cancer diagnosis and identifying high risk populations". Critical Reviews in Oncology/Hematology. 60 (1): 9–18. doi:10.1016/j.critrevonc.2006.04.002. PMID   16829121.
  21. Leong PP, Rezai B, Koch WM, Reed A, Eisele D, Lee DJ, et al. (July 1998). "Distinguishing second primary tumors from lung metastases in patients with head and neck squamous cell carcinoma". Journal of the National Cancer Institute. 90 (13): 972–977. doi: 10.1093/jnci/90.13.972 . PMID   9665144.
  22. Lapin M, Oltedal S, Tjensvoll K, Buhl T, Smaaland R, Garresori H, et al. (November 2018). "Fragment size and level of cell-free DNA provide prognostic information in patients with advanced pancreatic cancer". Journal of Translational Medicine. 16 (1): 300. doi: 10.1186/s12967-018-1677-2 . PMC   6218961 . PMID   30400802.
  23. Dragani TA, Matarese V, Colombo F (April 2020). "Biomarkers for Early Cancer Diagnosis: Prospects for Success through the Lens of Tumor Genetics". BioEssays. 42 (4): e1900122. doi:10.1002/bies.201900122. PMID   32128843. S2CID   212406467.
  24. Terpos E, Dimopoulos MA, Shrivastava V, Leitzel K, Christoulas D, Migkou M, et al. (March 2010). "High levels of serum TIMP-1 correlate with advanced disease and predict for poor survival in patients with multiple myeloma treated with novel agents". Leukemia Research. 34 (3): 399–402. doi:10.1016/j.leukres.2009.08.035. PMID   19781774.
  25. Kuukasjärvi T, Kononen J, Helin H, Holli K, Isola J (September 1996). "Loss of estrogen receptor in recurrent breast cancer is associated with poor response to endocrine therapy". Journal of Clinical Oncology. 14 (9): 2584–2589. doi:10.1200/jco.1996.14.9.2584. PMID   8823339.
  26. Harris L, Fritsche H, Mennel R, Norton L, Ravdin P, Taube S, et al. (November 2007). "American Society of Clinical Oncology 2007 update of recommendations for the use of tumor markers in breast cancer". Journal of Clinical Oncology. 25 (33): 5287–5312. doi:10.1200/JCO.2007.14.2364. PMID   17954709.
  27. Kröger N, Milde-Langosch K, Riethdorf S, Schmoor C, Schumacher M, Zander AR, Löning T (January 2006). "Prognostic and predictive effects of immunohistochemical factors in high-risk primary breast cancer patients". Clinical Cancer Research. 12 (1): 159–168. doi: 10.1158/1078-0432.CCR-05-1340 . PMID   16397038.
  28. Vrbic S, Pejcic I, Filipovic S, Kocic B, Vrbic M (Jan–Mar 2013). "Current and future anti-HER2 therapy in breast cancer". Journal of B.U.On. 18 (1): 4–16. PMID   23613383.
  29. Yoo C, Ryu MH, Ryoo BY, Beck MY, Kang YK (October 2013). "Efficacy, safety, and pharmacokinetics of imatinib dose escalation to 800 mg/day in patients with advanced gastrointestinal stromal tumors". Investigational New Drugs. 31 (5): 1367–1374. doi:10.1007/s10637-013-9961-8. PMID   23591629. S2CID   29477955.
  30. Demetri GD, van Oosterom AT, Garrett CR, Blackstein ME, Shah MH, Verweij J, et al. (October 2006). "Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial". Lancet. 368 (9544): 1329–1338. doi:10.1016/S0140-6736(06)69446-4. PMID   17046465. S2CID   25931515.
  31. Herbst RS, Prager D, Hermann R, Fehrenbacher L, Johnson BE, Sandler A, et al. (September 2005). "TRIBUTE: a phase III trial of erlotinib hydrochloride (OSI-774) combined with carboplatin and paclitaxel chemotherapy in advanced non-small-cell lung cancer". Journal of Clinical Oncology. 23 (25): 5892–5899. doi: 10.1200/JCO.2005.02.840 . PMID   16043829.
  32. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, et al. (May 2004). "Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib" (PDF). The New England Journal of Medicine. 350 (21): 2129–2139. doi:10.1056/NEJMoa040938. PMID   15118073.
  33. Sawyers CL (April 2008). "The cancer biomarker problem". Nature. 452 (7187): 548–552. Bibcode:2008Natur.452..548S. doi:10.1038/nature06913. PMID   18385728. S2CID   205213083.
  34. Karas-Kuzelicki N, Mlinaric-Rascan I (August 2009). "Individualization of thiopurine therapy: thiopurine S-methyltransferase and beyond". Pharmacogenomics. 10 (8): 1309–1322. doi:10.2217/pgs.09.78. PMID   19663675.
  35. Relling MV, Hancock ML, Rivera GK, Sandlund JT, Ribeiro RC, Krynetski EY, et al. (December 1999). "Mercaptopurine therapy intolerance and heterozygosity at the thiopurine S-methyltransferase gene locus". Journal of the National Cancer Institute. 91 (23): 2001–2008. doi: 10.1093/jnci/91.23.2001 . PMID   10580024.
  36. 1 2 Schneider JE, Sidhu MK, Doucet C, Kiss N, Ohsfeldt RL, Chalfin D (November 2012). "Economics of cancer biomarkers". Personalized Medicine. 9 (8): 829–837. doi:10.2217/pme.12.87. PMID   29776231.
  37. Henze G, Dummer R, Joller-Jemelka HI, Böni R, Burg G (1997). "Serum S100--a marker for disease monitoring in metastatic melanoma". Dermatology. 194 (3): 208–212. doi:10.1159/000246103. PMID   9187834.
  38. Harpio R, Einarsson R (July 2004). "S100 proteins as cancer biomarkers with focus on S100B in malignant melanoma". Clinical Biochemistry. 37 (7): 512–518. doi:10.1016/j.clinbiochem.2004.05.012. PMID   15234232.
  39. Lamond NW, Skedgel C, Younis T (April 2013). "Is the 21-gene recurrence score a cost-effective assay in endocrine-sensitive node-negative breast cancer?". Expert Review of Pharmacoeconomics & Outcomes Research. 13 (2): 243–250. doi:10.1586/erp.13.4. PMID   23570435. S2CID   33661439.
  40. Biroschak JR, Schwartz GF, Palazzo JP, Toll AD, Brill KL, Jaslow RJ, Lee SY (May 2013). "Impact of Oncotype DX on treatment decisions in ER-positive, node-negative breast cancer with histologic correlation". The Breast Journal. 19 (3): 269–275. doi: 10.1111/tbj.12099 . PMID   23614365. S2CID   30895945.
  41. Moen MD, McKeage K, Plosker GL, Siddiqui MA (2007). "Imatinib: a review of its use in chronic myeloid leukaemia". Drugs. 67 (2): 299–320. doi:10.2165/00003495-200767020-00010. PMID   17284091.
  42. Lemonick M, Park A (May 28, 2001). "New Hope for Cancer". Time . Archived from the original on October 15, 2007. Retrieved 26 April 2013.
  43. Price C, McDonnell D (February 1991). "Effects of niobium filtration and constant potential on the sensitometric responses of dental radiographic films". Dento Maxillo Facial Radiology. 20 (1): 11–16. doi:10.1259/dmfr.20.1.1884846. PMID   1884846.
  44. Cohen V, Khuri FR (2003). "Progress in lung cancer chemoprevention". Cancer Control. 10 (4): 315–324. doi: 10.1177/107327480301000406 . PMID   12915810.
  45. Lu CY, Tsai HL, Uen YH, Hu HM, Chen CW, Cheng TL, et al. (March 2013). "Circulating tumor cells as a surrogate marker for determining clinical outcome to mFOLFOX chemotherapy in patients with stage III colon cancer". British Journal of Cancer. 108 (4): 791–797. doi:10.1038/bjc.2012.595. PMC   3590657 . PMID   23422758.
  46. Balic M, Williams A, Lin H, Datar R, Cote RJ (2013). "Circulating tumor cells: from bench to bedside". Annual Review of Medicine. 64: 31–44. doi:10.1146/annurev-med-050311-163404. PMC   3809995 . PMID   23092385.
  47. Madhavan D, Zucknick M, Wallwiener M, Cuk K, Modugno C, Scharpff M, et al. (November 2012). "Circulating miRNAs as surrogate markers for circulating tumor cells and prognostic markers in metastatic breast cancer". Clinical Cancer Research. 18 (21): 5972–5982. doi: 10.1158/1078-0432.CCR-12-1407 . PMID   22952344.
  48. Redova M, Sana J, Slaby O (March 2013). "Circulating miRNAs as new blood-based biomarkers for solid cancers". Future Oncology. 9 (3): 387–402. doi:10.2217/fon.12.192. PMID   23469974.
  49. Joosse SA, Pantel K (January 2013). "Biologic challenges in the detection of circulating tumor cells". Cancer Research. 73 (1): 8–11. doi: 10.1158/0008-5472.CAN-12-3422 . PMID   23271724.
  50. Hou HW, Warkiani ME, Khoo BL, Li ZR, Soo RA, Tan DS, et al. (2013). "Isolation and retrieval of circulating tumor cells using centrifugal forces". Scientific Reports. 3: 1259. Bibcode:2013NatSR...3E1259H. doi:10.1038/srep01259. PMC   3569917 . PMID   23405273.
  51. Dhondt B, De Bleser E, Claeys T, Buelens S, Lumen N, Vandesompele J, et al. (December 2019). "Discovery and validation of a serum microRNA signature to characterize oligo- and polymetastatic prostate cancer: not ready for prime time". World Journal of Urology. 37 (12): 2557–2564. doi:10.1007/s00345-018-2609-8. hdl:1854/LU-8586484. PMID   30578441. S2CID   58594673.
  52. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 "Table of Pharmacogenomic Biomarkers in Drug Labels". U.S. Food and Drug Administration.
  53. 1 2 3 4 5 "Tumor Markers Fact Sheet" (PDF). American Cancer Society.
  54. 1 2 Heinz-Josef Lenz (2012-09-18). Biomarkers in Oncology: Prediction and Prognosis. Springer Science & Business Media. p. 263. ISBN   978-1-4419-9754-8.
  55. Gonzalez RS, Carlson G, Page AJ, Cohen C (July 2011). "Gastrointestinal stromal tumor markers in cutaneous melanomas: relationship to prognostic factors and outcome". American Journal of Clinical Pathology. 136 (1): 74–80. doi: 10.1309/AJCP9KHD7DCHWLMO . PMID   21685034.
  56. Tam CS, Otero-Palacios J, Abruzzo LV, Jorgensen JL, Ferrajoli A, Wierda WG, et al. (April 2008). "Chronic lymphocytic leukaemia CD20 expression is dependent on the genetic subtype: a study of quantitative flow cytometry and fluorescent in-situ hybridization in 510 patients". British Journal of Haematology. 141 (1): 36–40. doi: 10.1111/j.1365-2141.2008.07012.x . PMID   18324964.
  57. Zhang M, Yao Z, Patel H, Garmestani K, Zhang Z, Talanov VS, et al. (May 2007). "Effective therapy of murine models of human leukemia and lymphoma with radiolabeled anti-CD30 antibody, HeFi-1". Proceedings of the National Academy of Sciences of the United States of America. 104 (20): 8444–8448. Bibcode:2007PNAS..104.8444Z. doi: 10.1073/pnas.0702496104 . PMC   1895969 . PMID   17488826.
  58. Yamada Y, Sanchez-Aguilera A, Brandt EB, McBride M, Al-Moamen NJ, Finkelman FD, et al. (September 2008). "FIP1L1/PDGFRalpha synergizes with SCF to induce systemic mastocytosis in a murine model of chronic eosinophilic leukemia/hypereosinophilic syndrome". Blood. 112 (6): 2500–2507. doi: 10.1182/blood-2007-11-126268 . PMID   18539901.
  59. Nimer SD (May 2008). "Myelodysplastic syndromes". Blood. 111 (10): 4841–4851. doi: 10.1182/blood-2007-08-078139 . PMID   18467609. S2CID   6802096.
  60. Ottmann O, Dombret H, Martinelli G, Simonsson B, Guilhot F, Larson RA, et al. (October 2007). "Dasatinib induces rapid hematologic and cytogenetic responses in adult patients with Philadelphia chromosome positive acute lymphoblastic leukemia with resistance or intolerance to imatinib: interim results of a phase 2 study". Blood. 110 (7): 2309–2315. doi: 10.1182/blood-2007-02-073528 . PMID   17496201.
  61. Boulos N, Mulder HL, Calabrese CR, Morrison JB, Rehg JE, Relling MV, et al. (March 2011). "Chemotherapeutic agents circumvent emergence of dasatinib-resistant BCR-ABL kinase mutations in a precise mouse model of Philadelphia chromosome-positive acute lymphoblastic leukemia". Blood. 117 (13): 3585–3595. doi:10.1182/blood-2010-08-301267. PMC   3072880 . PMID   21263154.
  62. O'Connell PA, Madureira PA, Berman JN, Liwski RS, Waisman DM (April 2011). "Regulation of S100A10 by the PML-RAR-α oncoprotein". Blood. 117 (15): 4095–4105. doi: 10.1182/blood-2010-07-298851 . PMID   21310922.
  63. Duffy MJ, Crown J (November 2008). "A personalized approach to cancer treatment: how biomarkers can help". Clinical Chemistry. 54 (11): 1770–1779. doi: 10.1373/clinchem.2008.110056 . PMID   18801934.
  64. Ribrag V, Koscielny S, Casasnovas O, Cazeneuve C, Brice P, Morschhauser F, et al. (April 2009). "Pharmacogenetic study in Hodgkin lymphomas reveals the impact of UGT1A1 polymorphisms on patient prognosis". Blood. 113 (14): 3307–3313. doi: 10.1182/blood-2008-03-148874 . PMID   18768784.
  65. Li Y, Ye X, Liu J, Zha J, Pei L (January 2011). "Evaluation of EML4-ALK fusion proteins in non-small cell lung cancer using small molecule inhibitors". Neoplasia. 13 (1): 1–11. doi:10.1593/neo.101120. PMC   3022423 . PMID   21245935.
  66. 1 2 Pao W, Girard N (February 2011). "New driver mutations in non-small-cell lung cancer". The Lancet. Oncology. 12 (2): 175–180. doi:10.1016/S1470-2045(10)70087-5. PMID   21277552.
  67. Hewes A (October 2, 2014). "Promising Method for Detecting Pancreatic Cancer Years Before Traditional Diagnosis". Singularity HUB. Retrieved 2016-04-22.
  68. Gupta D, Lis CG (October 2009). "Role of CA125 in predicting ovarian cancer survival - a review of the epidemiological literature". Journal of Ovarian Research. 2 (1): 13. doi: 10.1186/1757-2215-2-13 . PMC   2764643 . PMID   19818123.
  69. Bartels CL, Tsongalis GJ (April 2009). "MicroRNAs: novel biomarkers for human cancer". Clinical Chemistry. 55 (4): 623–631. doi: 10.1373/clinchem.2008.112805 . PMID   19246618.
  70. Paulson KG, Lewis CW, Redman MW, Simonson WT, Lisberg A, Ritter D, et al. (April 2017). "Viral oncoprotein antibodies as a marker for recurrence of Merkel cell carcinoma: A prospective validation study". Cancer. 123 (8): 1464–1474. doi:10.1002/cncr.30475. PMC   5384867 . PMID   27925665.
  71. Proctor MJ, McMillan DC, Morrison DS, Fletcher CD, Horgan PG, Clarke SJ (August 2012). "A derived neutrophil to lymphocyte ratio predicts survival in patients with cancer". British Journal of Cancer. 107 (4): 695–699. doi: 10.1038/bjc.2012.292 . PMC   3419948 . PMID   22828611.
  72. 1 2 Liu M, Xing LQ (August 2017). "Basic fibroblast growth factor as a potential biomarker for diagnosing malignant tumor metastasis in women". Oncology Letters. 14 (2): 1561–1567. doi: 10.3892/ol.2017.6335 . PMC   5529833 . PMID   28789380.
  73. Fürstenberger G, Senn HJ (May 2002). "Insulin-like growth factors and cancer". The Lancet. Oncology. 3 (5): 298–302. doi:10.1016/s1470-2045(02)00731-3. PMID   12067807.
  74. Yu H, Rohan T (September 2000). "Role of the insulin-like growth factor family in cancer development and progression". Journal of the National Cancer Institute. 92 (18): 1472–1489. doi: 10.1093/jnci/92.18.1472 . PMID   10995803.
  75. Vano YA, Oudard S, By MA, Têtu P, Thibault C, Aboudagga H, et al. (2018-04-06). "Optimal cut-off for neutrophil-to-lymphocyte ratio: Fact or Fantasy? A prospective cohort study in metastatic cancer patients". PLOS ONE. 13 (4): e0195042. Bibcode:2018PLoSO..1395042V. doi: 10.1371/journal.pone.0195042 . PMC   5889159 . PMID   29624591.
  76. Liu M, Xing LQ (August 2017). "Basic fibroblast growth factor as a potential biomarker for diagnosing malignant tumor metastasis in women". Oncology Letters. 14 (2): 1561–1567. doi: 10.3892/ol.2017.6335 . PMC   5529833 . PMID   28789380.
  77. Torrente-Rodríguez RM, Ruiz-Valdepeñas Montiel V, Campuzano S, Pedrero M, Farchado M, Vargas E, et al. (2017-04-04). "Electrochemical sensor for rapid determination of fibroblast growth factor receptor 4 in raw cancer cell lysates". PLOS ONE. 12 (4): e0175056. Bibcode:2017PLoSO..1275056T. doi: 10.1371/journal.pone.0175056 . PMC   5380347 . PMID   28376106.
  78. Denduluri SK, Idowu O, Wang Z, Liao Z, Yan Z, Mohammed MK, et al. (March 2015). "Insulin-like growth factor (IGF) signaling in tumorigenesis and the development of cancer drug resistance". Genes & Diseases. 2 (1): 13–25. doi: 10.1016/j.gendis.2014.10.004 . PMC   4431759 . PMID   25984556.