Physical Address
304 North Cardinal St.
Dorchester Center, MA 02124
The analysis of circulating tumor cells (CTCs) and tumor cell products (DNA, RNA, extracellular vesicles) released into the blood may provide clinically relevant information as a “liquid biopsy” and provide new insights into tumor biology.
CTCs are complementary to other liquid biopsy biomarkers such as circulating cell-free DNA (ctDNA), circulating microRNAs, extracellular vessels, and tumor-educated platelets. Validation of liquid biopsy assays is essential and has been performed by the EU/IMI consortium CANCER-ID ( www.cancer-id.eu ), an activity sustained now by the European Liquid Biopsy Society (ELBS) consortium ( www.elbs.eu ). Liquid biopsy analyses with validated platforms provide information on early detection of cancer, identification of cancer patients at risk to develop relapse (prognosis), and it may serve to track tumor evolution, therapeutic targets, or mechanisms of resistance on metastatic cells. Metastatic cells might have unique characteristics that can differ from the bulk of cancer cells in the primary tumor currently used for stratification of patients to systemic therapy. Moreover, monitoring of blood samples in the context of therapies might provide unique information for the future clinical management of the individual cancer patient and might serve as surrogate markers for response to therapy. Liquid biopsy analysis can be used to improve the management of individual cancer patients and contribute to personalized medicine.
Tissue biopsies, the current “gold standard” in cancer diagnostics, have some limitations which may be overcome by liquid biopsies ( Figs. 71.1 and 71.2 ). The term liquid biopsy refers to testing of body fluids (e.g., blood, urine, saliva, cerebrospinal fluid [CSF]) , derived mainly from circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), circulating miRNAs, and extracellular vesicles (EVs) (see Fig. 71.1 ). Liquid biopsy offers a minimally invasive insight into a patient’s cancer. Currently the most promising role for liquid biopsy is the profiling of CTCs or ctDNA as a way to monitor patients in the course of therapy—particularly by using novel technologies for a better and earlier indication of either response or emerging resistance to a particular treatment ( Fig. 71.3 ). The next logical step is to better understand the mechanisms of evolving resistance and hopefully to guide treatment strategies to overcome resistance. The utility of liquid biopsy is not just limited to being a mirror of tissue biopsy, but it is a potential tool that can detect unique and impactful information about a patient’s cancer that tissue testing cannot. Just a few years ago, the liquid biopsy approach was limited to research studies, but it is now entering prospective clinical trials. It can be used for patients whose tumors are hard to access by biopsy or when the site of the primary tumor is unknown. With respect to the clinical laboratory, the development of targeted molecular assays as companion diagnostics, for disease monitoring, and even for early cancer detection are all potential possibilities at various stages of development. It may in the future enable decisions for targeted therapies in patients who have failed treatment on a particular drug regimen.
Potentially, liquid biopsy may aid in the investigation of the evolution of subclonal cancer cell populations. Liquid biopsy may be a minimally invasive method for determining dominant clones to direct targeted therapies against. There is hope that this approach can illuminate strategies to combine drugs that affect the dominant mutated populations and also inhibit other subclonal populations from expanding. This approach may impact the definition of minimal residual disease , because it can change the clinician’s ability to predict the risk of recurrence in early-stage cancer patients whose tumors have been surgically removed.
Liquid biopsy as a diagnostic, prognostic, and theranostic tool is appealing because it is minimally invasive and easily performed in a serial manner. However, there are several barriers to the routine clinical use of liquid biopsy. Numerous technologies are available for the detection and molecular characterization of CTCs and ctDNA (and other circulating cancer biomarkers), resulting in different test results even if the same blood samples are analyzed, which points to the urgent need for standardization of the preanalytical and analytical phase of the applied tests to obtain robust and reproducible results. Moreover, well-designed comparison studies on large patient cohorts comparing the clinical relevance of liquid biopsy (CTCs and/or ctDNA) and tissue biopsy with defined clinical endpoints (e.g., progression-free or overall survival [OS]) are still needed to demonstrate clinical utility. Liquid biopsy, tissue biopsy, and imaging may provide complementary information, which could lead to the establishment of a composite diagnostic panel with the highest accuracy and benefit for cancer patients. Thus the present over-competition between researchers working in different biomarker fields (e.g., ctDNA versus CTCs) appears to be counterproductive.
The liquid biopsy approach extracts molecular information from the tumor by detailed analysis of circulating tumor-derived genetic material in the bloodstream. The sources of this material are circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), circulating miRNAs, and extracellular vesicles (EVs).
Liquid biopsy can provide detailed information on tumor genome and transcriptome evolution over time through conventional peripheral blood sampling that can be used for serial monitoring of a patient.
The presence of CTCs was first reported in 1869 by Thomas Ashworth ( Fig. 71.4 ). In 2005, the clinical importance of disseminated tumor cells (DTCs) in the bone marrow of breast cancer patients was shown. However, analysis of DTCs in bone marrow is invasive and thus difficult to repeat. CTCs are rare cells that originate from primary and metastatic tumors that have managed to get into the circulation and that may extravasate to different organs ( Fig. 71.5 ). Despite the assumption that only a small fraction of CTCs will develop into metastasis, the CTC counts at initial diagnosis and during the postsurgical follow-up period are tightly correlated to the risk of relapse in breast cancer and other solid tumors. For example, in breast cancer the recurrence risk was increased more than six times in patients with 5 or more CTCs per 7.5 mL blood before receiving neoadjuvant therapy, and the prognostic value was independent from the response of the primary tumor to neoadjuvant therapy. Cancer metastasis is the main cause of cancer-related death, and dissemination of tumor cells through the blood circulation is an important intermediate step that also exemplifies the switch from localized to systemic disease. Detection of CTCs has also been proposed as a companion diagnostic to identify glioblastoma multiforme patients with extracranial tumor cell spread, in order to exclude these patients as organ donors. CTCs are therefore major players in the liquid biopsy approach and may provide important insights into the biology behind metastatic progression and real-time information on a patient’s disease status.
Many advances have been made in the detection and molecular characterization of CTCs. The presence of CTCs in peripheral blood has been linked to worse prognosis and early relapse in various types of solid cancers. The FDA has cleared the CellSearch system for breast (2004), colorectal (2008), and prostate (2008) cancer based on the critical role that CTCs play in the metastatic spread of carcinomas. Detection of CTCs is correlated with decreased progression-free survival (PFS) and OS in both operable breast cancer and metastatic breast cancer (MBC), and various studies on other tumor types have confirmed this prognostic relevance. This has led to the introduction of a new stage, cM0(i+), characterized by the absence of clinical overt metastases (cM0) but the detection of isolated tumor cells (i+) in blood or other compartments, into the 2018 AJCC classification for breast cancer. Thus CTCs may qualify as a new tool to enrich for high-risk M0 patients, which may be used in future clinical studies assessing the clinical value of adjuvant therapies. Focusing on high-risk patients can speed up time-consuming and costly clinical studies in the adjuvant setting. Nevertheless, enumeration of CTCs might not be sufficient and further downstream analysis will provide more information on the biology of CTCs and their metastasis-initiating potential to extravasate and colonize distant sites. This information might further increase the diagnostic accuracy of CTC analyses. For example, in the analysis of the prognostic value of Bidard and colleagues approximately 25% of breast cancer patients with detectable CTCs had no signs of relapse within the observation period of 70 months. On the other hand, approximately 20% of patients without detectable CTCs had experienced relapse, which points to the need of increasing the sensitivity of CTC analysis, for example, by the introduction of new markers targeting CTCs with nonepithelial phenotype or increasing the blood volume analyzed.
CTCs are targets for understanding tumor biology and tumor cell dissemination in humans. Their molecular characterization offers an exciting approach to understanding resistance to established therapies and elucidating the complex biology of metastasis. Further research on the molecular characterization of CTCs should contribute to a better understanding of the biology of metastatic development in cancer patients and the identification of novel therapeutic targets, especially after elucidating the relationship of CTCs to cancer stem cells (CSCs). This approach may provide individualized targeted treatments and spare cancer patients unnecessary and ineffective therapies.
CTCs are rare, and the amount of available sample is limited, presenting formidable analytical and technical challenges. Recent technical advancements in CTC detection and characterization include multiplex reverse transcription quantitative polymerase chain reaction (PCR) (RT-qPCR) methods, image-based approaches, and microfilter and microchip devices for their isolation. However, direct comparison of different methods for detecting CTCs in blood from patients with breast cancer has revealed a substantial variation in the detection rates. , There is a lack of standardization in reference material, which hampers the implementation of CTC measurement in clinical routine practice. Thus while the potential of CTC analysis is now widely recognized, many challenges remain.
The enrichment and detection technologies for highly pure CTCs are challenging as these cells are extremely rare in the peripheral blood. Indeed, CTCs occur at very low concentrations in the bloodstream, ranging between 1 and 10 cells per 10 mL in most cancer patients. Usually, CTCs are co-isolated with normal peripheral blood mononuclear cells (e.g., leukocytes). Thus efficient enrichment of CTCs can be achieved by approaches that exploit differences between tumor cells and leukocytes, including the differential expression of cell surface proteins or distinct physical properties of the cells.
The combination of high-throughput and automated CTC isolation technologies with validated downstream detection assays are necessary for the routine use of CTC-based diagnostics in the clinical management of cancer patients. CTC analysis includes isolation/enrichment, detection, enumeration, and characterization. The main analytical systems used are described below, and an outline is presented in Fig. 71.6 .
CTCs can be infrequent, depending on the cancer stage. One CTC may be present among 10 6 to 10 8 peripheral blood cells. CTC isolation and enrichment from peripheral blood are thus extremely challenging and very demanding. It is not only that these cells circulate at very low numbers, but they are heterogeneous even within the same patient. Highly standardized and robust isolation protocols are necessary for downstream CTC analysis and molecular characterization. Toward this goal, a lot of effort has focused on developing novel technologies for the isolation and enrichment of CTCs from peripheral blood.
A large spectrum of technologies is available to enrich CTCs from surrounding normal hematopoietic cells (see Fig. 71.6 ). These enrichment methods rely on different properties of CTCs that can be positively or negatively enriched on (i) label-dependent systems based on biological properties (e.g., surface protein expression) and (ii) label-independent systems based on physical properties (e.g., size, density, electric charges, and deformability). In vivo methods for increasing CTC yield have also been developed. Moreover, CTC recovery from blood samples collected close to the tumor (i.e., in vessels located in the drainage area of the primary tumor or metastases) can considerably increase the chance to collect more CTCs ( Fig. 71.7 ). , In colorectal cancer (CRC), the comparative analysis of CTCs obtained from the mesenteric and peripheral blood demonstrated a cascade of genomic events related to metastatic progression ( Fig. 71.8 ).
The most important systems for the isolation/enrichment of CTCs are discussed in the following text. In many cases, these technologies are complementary to each other because they target different properties of CTCs and may define different CTC populations.
Biological properties are used in immunologic procedures with antibodies against either tumor-associated antigens (positive selection) or common leukocytes antigen CD45 (negative selection).
Positive enrichment should reach high cell purity, depending on the antibody specificity used in the assay. Among the current positive systems, most of the technologies target the epithelial cell adhesion molecule (EpCAM) antigen (e.g., FDA-cleared CellSearch system, the current gold standard). However, capturing CTCs that are not expressing EpCAM has pushed researchers to use panels of antibodies against various other epithelial cell surface antigens (such as epidermal growth factor receptor [EGFR], MUC1), tissue-specific antigens (such as PSA, human epidermal growth factor receptor 2 [HER2]), or mesenchymal/stem-cell antigens (such as Snail, ALDH1). Positive selection of CTCs is possible because the phenotype of CTCs is well known. However, many CTCs are heterogenous. Any bias can be avoided by negative selection where CTCs are not targeted directly but the unwanted cells, such as leukocytes, are depleted from the samples. Indeed, negative enrichment targets and removes surrounding normal cells, using antibodies against CD45 (not expressed on cancer cells) and other leukocyte antigens (e.g., CD14, CD8, CD19). The advantages of negative enrichment are: (i) CTCs are not tagged with a difficult-to-remove antibody, (ii) CTCs are not activated or modified via an antibody-protein interaction, and (iii) antibody selection does not bias the subpopulation of CTCs captured. The leukocyte-depletion procedure has a high recovery rate; however, the samples are less pure than positive selection procedures; indeed, more remaining leukocytes are present.
Positive selection is the most widely used CTC isolation/enrichment system. This approach captures CTCs through specific monoclonal antibodies against epithelial cell surface markers that are expressed on CTCs but are absent from normal leukocytes. CTCs can be tagged using antibody-conjugated magnetic microbeads (diameter: 0.5 to 5 μm) or nanoparticles (diameter: 50 to 250 nm) that bind to a specific surface antigen. Intracellular antigens like cytokeratins can also be used as targets. Immunomagnetic assays require a short incubation (∼30 minutes) for antigen/antibody binding that couples the cells to magnetic beads, followed by isolation of the cells using a magnetic field.
Various antigens have been exploited for the positive immunomagnetic isolation of CTCs. Among these, EpCAM is the most common. This approach is well established in terms of proven clinical significance of the captured cells. However, capture with EpCAM has the disadvantage of missing some cells that are undergoing epithelial-mesenchymal transition (EMT; Fig. 71.9 ). We also now know that CTCs are highly heterogeneous, but one approach to partially overcome this issue is to use a “cocktail” of antibodies that targets multiple antigens. Along these lines, several organ- or tumor-specific markers, such as carcinoembryonic antigen (CEA), EGFR, prostate-specific antigen, HER-2, cell surface–associated mucin 1 (MUC-1), ephrin receptor B4 (EphB4), insulin-like growth factor 1 receptor (IGF-1R), cadherin-11 (CAD11), and tumor-associated glycoprotein 72 (TAG-72), have been used to isolate CTCs. The expression of vimentin by CTCs may also be a good marker for epithelial cancers undergoing EMT. By using a specific monoclonal antibody against vimentin, EMT-CTCs were detected in patients undergoing postsurgery adjuvant chemotherapy for metastatic colon cancer. These isolated EMT-CTCs were characterized further using EMT-specific markers, fluorescent in situ hybridization, and single-cell mutation analysis. This antibody exhibited high specificity and sensitivity toward different epithelial cancer cells and was used to detect and enumerate EMT-CTCs from patients. The number of EMT-CTCs detected correlated with the therapeutic outcome of the disease. According to these results, cell surface vimentin is a promising marker for the isolation of EMT-CTCs from a wide variety of tumor types.
Another example of positive selection is the MagSweeper, an immunomagnetic cell separator. In this device, magnetic beads are coated with an antibody targeting epithelial cell surface markers. These immunomagnetic beads are added into blood samples, and cancer cells are attached to the beads. This device gently enriches target cells and eliminates cells that are not bound to magnetic particles by using centrifugal forces. The isolated cells are easily accessible and can be extracted individually based on their physical characteristics to deplete any cells nonspecifically bound to beads. The same group has recently developed the magnetic sifter, a miniature microfluidic chip with a dense array of magnetic pores; tumor cells are labeled with magnetic nanoparticles and are captured from whole blood with high efficiency. The use of isolation technologies that take advantage of magnetic fields may lead the way to routine preparation and characterization of liquid biopsies from cancer patients.
This isolation approach is completely independent of the phenotype of CTCs and is based on the depletion of noncancerous peripheral blood cells. A first step is often the lysis of red blood cells (RBCs) and the second step may use specific markers for white blood cells (WBCs) like CD45 or CD61 to magnetically remove them from the sample. , Another variation of CTC enrichment by negative depletion is the commercially available RosetteSep system (StemCell technologies, Canada): tetrameric antibody complexes target unwanted cells for removal. This approach is independent of the expression of EpCAM on CTCs and has better recoveries than the conventional density gradient method for the isolation of CTCs from normal leukocytes in blood.
Physical properties are another alternative to enrich CTCs from blood samples. During the last decade, numerous marker-independent techniques have been developed for CTC isolation. Label-free enrichment processes based on physical properties, such as density, size, deformability, and electric charges, avoid molecular biases induced by variability of cell biomarker expression associated with tumor heterogeneity. Among physical properties, the size of CTCs is the main characteristic used to enrich them. Indeed, CTCs generally exhibit a larger morphology than leukocytes (8 to 10 μm) and filtration membranes and microfluidic devices using inertial focusing to separate CTCs from blood have been developed.
Density gradient centrifugation using commercially available reagents (e.g., Ficoll, GE Healthcare Life Sciences, Pittsburgh, PA) is one of the most widely used approaches for CTC enrichment. It is based on the lower density of mononuclear cells (including CTCs) compared to RBCs and polymorphonuclear leukocytes. To enrich correctly, the idea is to further combine it to immunomagnetic enrichment (positive or negative selection). Another alternative is to enrich directly the CTCs with a well-defined Ficoll gradient. The OncoQuick system (Greiner Bio-One, Germany) is an improved version of this approach that uses a porous membrane placed on top of the gradient media to prevent mixing. Experiments performed in cell lines have shown CTC recovery rates of 70 to 90%. Although simple and inexpensive, the OncoQuick system has a relatively low yield and enrichment compared to the CellSearch system because in the same group of 61 patients, at least one CTC was detected in 23% with OncoQuick and in 54% with CellSearch system.
Size-based isolation systems are independent of tumor markers and separate CTCs (that are usually larger) from smaller leukocytes. Several different size–based systems have been developed and include membrane filters, microfluidic chips, and hydrodynamic methods. The first CTC filtration device was described by Vona and colleagues in 2000. Since then, filtration devices have been improved, and many downstream applications have been developed. Filters used for CTC isolation/enrichment are often disposable porous membranes (usually polycarbonate) containing numerous randomly distributed 7- to 8-μm-diameter holes that allow blood constituents to cross but capture the larger CTCs. Specific microfabrication techniques have been applied to build microfilters with controlled pore distribution, size, and geometry and different materials like polycarbonate, , , parylene C, nickel, and silicon have been used in the fabrication of these membranes.
Polycarbonate membrane filters are usually accompanied by specific syringes or pumps so the pressure on the filter is optimized to keep fragile CTCs intact. Commercially available filtering devices for CTC isolation include the ISET system (isolation by size of epithelial tumor cells, Rarecells, France), ScreenCell (ScreenCell Inc., France), and the CellSieve system (CREATV MicroTech, Potomac, MD).
CTC isolation by size filtration is simple and reliable; filtration and staining are easy to perform, and the method is rapid. CTCs can be identified using classic cytopathologic criteria. No complex instrumentation or specific training is needed. However, false-negative results may be obtained by using filtration devices where small CTCs are missed. Moreover, endothelial cells or rare hematologic cells such as megakaryocytes may also be present on the filter. For this reason, downstream cytomorphologic, immunocytochemical, and molecular characterization are important to rule out cells that are not CTCs.
CTCs are different from peripheral blood mononuclear cells (PBMCs) with respect to morphology and dielectric properties. Dielectrophoresis (DEP) is a technology for CTC isolation based on electrical properties of cancer cells. Dielectric properties (polarizability) of cells are dependent on cell diameter, membrane area, density, conductivity, and volume. Depending on their phenotype and morphology, different cells have different dielectric properties, which is the principle employed for electro-kinetic isolation of CTCs.
The main advantages of microfluidic systems for CTC isolation/enrichment are their simplicity and the potential to be fully automated, unlike traditional affinity-based CTC isolation techniques. The main disadvantage is the long time needed to run each sample and the low capacity in terms of the volume of peripheral blood that can be processed. These systems are based on a combination of precisely defined topography of microstructures (traps) with laminar flow in microchannels.
With microfluidic-based CTC technology, the chambers are made out of transparent materials that enhance high-resolution imaging, including the use of transmitted light microscopy. Most microfluidic devices are reliant on three-dimensional structures that limit the characterization of cells on the chip. This limitation can be overcome by using functionalized graphene oxide nanosheets on a patterned gold surface. In recent years, various sophisticated microfluidic devices have been developed, including in situ capture and CTC culture in a three-dimensional co-culture model simulating a tumor microenvironment.
The recent development of in vivo systems for CTC isolation from whole blood adds another dimension to the field of CTC isolation. These systems aim to overcome the limitation of small blood sample volumes inherent to the ex vivo CTC isolation techniques previously described.
The first in vivo CTC isolation system used in cancer patients was the CellCollector (CC, Gilupi GmbH, Germany). CellCollector is a nanowire that is coated at its tip (2 cm of length) with pure gold, to which chimeric antibodies directed to EpCAM are covalently attached. The CellCollector is placed into the antecubital vein of a cancer patient for in vivo binding of CTCs. Several studies in patients with lung and prostate cancer have demonstrated technical validity for CTC detection and molecular characterization; however, the clinical relevance of CC-detected CTCs is still under investigation. Recently, Dizdar and colleagues evaluated this technology for CTC detection in CRC patients. While CTC detection in M 0 CRC patients was significantly increased with the CC, the clinical relevance of these CTCs appears inferior to the cells identified by the CellSearch system.
Transdermal photoacoustic flow cytometry can also detect CTCs in vivo. A high–pulse repetition rate diode laser shines light through the skin into a vessel that is up to 3 mm deep to detect acoustic vibrations that result from the absorption of laser light by target nanoparticles. By using this technology, circulating melanoma cells were detected in blood. More recently, Galanzha and colleagues reported a completely new Cytophone platform for photoacoustic detection of CTCs in patients with melanoma. The Cytophone could detect individual CTCs at a concentration of ≥1 CTC/mL in 20 seconds and could also identify clots and CTC-clot emboli, indicating the potential of in vivo blood testing with the Cytophone in melanoma patients.
To analyze a high volume of blood, an alternative to in vivo CTC detection is analysis by leukapheresis. Indeed, leukapheresis is a laboratory procedure in which WBCs or peripheral blood stem cells are separated from blood. During leukapheresis, a patient’s blood is passed through a machine that removes the WBCs or peripheral blood stem cells and then returns the balance of the blood back to the patient. Eifler and colleagues first showed that isolation of CTCs via leukapheresis was feasible. Screening leukapheresis products generated from up to 2.5 L of processed blood per patient demonstrated that CTCs can be detected in more than 90% of nonmetastatic breast cancer patients. Label-free enrichment and molecular characterization of viable CTCs can be performed from diagnostic leukapheresis (DLA) products, and DLA has enabled transcriptomic profiling of single CTCs demonstrating intercellular heterogeneity of endocrine resistance in estrogen receptor (ER)-positive breast cancer.
After the enrichment step, the samples still contain a substantial number of contaminating leukocytes; thus CTCs need to be specifically identified at the single-cell level by a robust reproducible method that can distinguish them from normal blood cells. CTC detection is achieved by (i) immunofluorescence, (ii) molecular nucleic acid analyses (multiplex RT-qPCR), and (iii) functional assays (EPISPOT and EPIDROP). An outline of the main approaches for CTC detection is presented in Fig. 71.6 .
Immunologic technologies are the most frequent methods employed for CTC detection, and they use a combination of membrane and/or intracytoplasmic anti-epithelial, anti-mesenchymal, and anti–tissue-specific markers and anti–tumor-associated antibodies. Over the past 20 years, keratins as constituents of the cytoskeleton of epithelial cells have been established as detection markers for CTCs in patients with various kinds of carcinomas. Many CTC assays follow principles that are similar to the FDA-cleared CellSearch system, which has been the “gold standard” for many years and will be therefore discussed in more detail. After EpCAM–based immunomagnetic enrichment of CTCs, cells are fluorescently stained for epithelial keratins (CK8, 18, and 19) as markers for CTCs, the common leukocyte antigen CD45 as an exclusion marker, and a nuclear dye (4′,6-diamidino-2-phenylindole [DAPI]) to access cellular integrity ( Fig. 71.10 ), and suspicious events are listed in a photo gallery using automated digital microscopy. The main advantages of the CellSearch system are its high reproducibility and robustness including defined preanalytical steps of blood sampling that ensure stability for 96 hours at room temperature. , Most importantly, there is a wealth of data from large-scale clinical studies demonstrating the correlation of CellSearch-based CTC counts with clinical outcome in breast cancer and may other solid tumors. However, limitations to this system are that the enrichment and detection steps depend entirely on the expression of the epithelial markers EpCAM and keratins. Thus CTCs that have undergone a complete EMT and do not express EpCAM and keratins (see below) cannot be detected. Therefore there is an ongoing search for additional markers that detect EMT CTCs and are not expressed on contaminating blood cells to ensure both high sensitivity and specificity of CTC detection.
Molecular techniques identify specific tumor DNA or mRNA in lysates of blood cells to indirectly demonstrate the presence of CTCs. Detection involves designing specific primers for transcripts supposedly associated with CTC-specific genes. These genes either code for tissue-, organ-, or tumor-specific proteins or, more specifically, contain known mutations, translocations, or methylation patterns found in cancer cells. RNA-based methods have the highest sensitivity but can lack specificity, owing to the potential of capturing noncancerous cells that generate false-positive signals, thus decreasing the overall accuracy. Considering the heterogeneity of CTCs, multiplex RT-PCR assays could overcome this limitation. ,
Functional assays that exploit aspects of cellular CTC activity can identify “metastasis-competent cells” among the entire pool of CTCs (see Figs. 71.6 and 71.11 ). The functional epithelial immunospot (EPISPOT, a specific type of ELISPOT or Enzyme-LInked immunoSPOT) assay was introduced for in vitro CTC detection and focuses only on viable CTCs. This technology assesses the presence of CTCs based on secretion, shedding, or release of specific proteins during 24 to 48 hours of short-term culture. This assay has been used in many types of solid cancers, such as breast, colorectal, prostate, and others. The new EPIDROP assay (EPIspot in a DROP) is currently under optimization for the detection of single viable CTCs. It allows the enumeration and characterization of all CTCs, and gives information on their viability and functionality at the single cell level and also on their drug resistance profile in a few hours. More recently, Tang and colleagues described a high-throughput assay for rapid detection of rare metabolically active tumor cells in pleural effusions and the peripheral blood of lung cancer patients.
The establishment of in vitro cultures and permanent lines from CTCs has become a challenging task ( Fig. 71.12 ). Recently, CTC lines have been used to identify key proteins and new pathways involved in cancer stemness and dissemination and to test new drugs to inhibit metastasis-initiator CTCs. Ex vivo CTC cultures have been established for breast, , prostate, lung, colon, and head and neck cancers. Permanent CTC lines from circulating colon cancer cells have been established both before (CTC-MCC-41) , and after the initiation of the anticancer treatment. Important information can be obtained by transplantation of patient-derived CTCs into immunodeficient mice; tumors that grow after xenotransplantation of enriched CTCs have the characteristics of metastasis-initiator cells.
Serial CTC measurement can follow the evolution of tumor subclones during treatment and disease progression and hold the key to understand the biology of the metastatic cascade. Improvements in technologies to yield purer CTC populations improve cellular and molecular investigations. Characterization of single CTCs allows better insight into tumor heterogeneity within assays, including immunofluorescence, array comparative genomic hybridization (CGH), massively parallel sequencing (MPS) of both DNA and RNA, and fluorescence in situ hybridization (FISH).
Immunologic detection and characterization allow isolation of stained CTCs for subsequent molecular characterization. While manual isolation by micromanipulation of CTCs is possible, it is rather arduous and time-consuming. An alternative automated single-cell selection device is the DEPArray, which is based on DEP that traps single cells in DEP cages and is designed for single-cell recovery of CTCs. Multiple clinical studies have used this technology to detect and isolate single CTCs for subsequent genetic analyses.
Another approach is FISH analysis of single CTCs identified by immunocytochemistry. Recently, padlock probe technology, which enables in situ analysis of AR-V7 in CTCs, showed that 71% (22 of 31) of castration-resistant prostate cancer (CRPC) patients had detectable AR-V7 expression ranging from low to high expression. Patients with AR-V7-positive CTCs respond better to taxane-based chemotherapy than novel hormonal therapies, indicating a treatment-selection biomarker.
In 1889, in the very first issue of Lancet , Steve Paget described “the seed and soil hypothesis,” in which “metastasis depends on the cross talk between selected cancer cells (the seed) and specific organ microenvironments (the soil),” a hypothesis revisited many years later by Fidler. Detailing the mechanism of metastasis remains a very hot topic in cancer research today ( Fig. 71.13 ). , Analysis of disease course, tumor growth rates, autopsy studies, clinical trials, and molecular genetic analyses of primary and DTCs all contribute to our understanding of systemic cancer. , Molecular characterization of CTCs provides a level of detail not previously possible and may be the key to our further understanding of metastatic progression.
CTC biology can be viewed as a “window to metastasis” because CTCs play a critical role in the metastatic spread of carcinomas (see Fig. 71.13 ). , If CTCs are effectively targeted or kept in a dormant state, the cancer may be prevented from progressing to metastatic disease. Molecular characterization of CTCs from patients may be the shortest path to determine which and when patients might relapse and to identify specific mechanisms to target these cells. Dormancy gene signatures that identify individuals with dormant disease have also been explored. , In contrast, CTCs with stemness and EMT features display enhanced malignant and metastatic potential. The role of CTCs in treatment failure and disease progression is likely explained by their biological processes, such as EMT, stemness features, dormancy, and heterogeneity ( Fig. 71.14 ).
Mentioning the evolution of dispersal, it is of utmost interest to evaluate whether similar ecologic and evolutionary principles can be applied to metastasis, and how these processes may shape the spatiotemporal dynamics of CTCs. Indeed, CTCs have themselves been observed to disperse alone and in groups of up to around 100 cells, called clusters or microemboli, and cancer cells reproduce asexually, so it is uncertain which of the aforementioned evolutionary outcomes is the most likely to explain a role of kin selection in metastasis ( Fig. 71.15 ).
EMT is an essential process in the metastatic cascade. This biological process is highly associated with an invasive phenotype and enables detachment of tumor cells from the primary site and migration ( Fig. 71.16 ). The reverse process of mesenchymal epithelial transition (MET) might play a crucial role in the further steps of metastasis when CTCs seed distant organs and establish metastasis. The mechanisms and the interplay of EMT and MET have been intensively studied, and current data suggest the existence of the EMT process in CTCs. , It is now clear that CTCs from MBC patients exhibit heterogeneous epithelial and mesenchymal phenotypes and display higher frequencies of partial or full-blown mesenchymal phenotype than carcinoma cells within primary tumors. Mesenchymal-like CTCs are also elevated in patients who are refractory to therapy.
Currently, most systems that detect CTCs, including the CellSearch system, are based on the expression of the epithelial marker EpCAM and do not specifically identify CTC subtypes with EMT. Over the past years, several EMT-related markers have been applied in CTC studies. Three EMT markers ( TWIST1, AKT2, and PI3K α) and the stem cell marker ALDH1 were evaluated in CTCs from 502 primary breast cancer patients by a multiplex RT-PCR assay. A subset of CTCs showed EMT and stem cell characteristics. The expression levels of EMT-inducing transcription factors ( TWIST1, SNAIL1, SLUG, ZEB1, and FOXC2 ) were also determined in CTCs from primary breast cancer patients. In another study, rare primary tumor cells simultaneously expressed mesenchymal and epithelial markers, but mesenchymal cells were highly enriched in CTCs, and serial monitoring suggested an association of mesenchymal CTCs with disease progression. Mesenchymal CTCs occurred as both single cells and multicellular clusters, expressed known EMT regulators, including transforming growth factor (TGF)-β pathway components and the FOXC1 transcription factor. These data support a role for EMT in the blood-borne dissemination of human breast cancer. When the EMT phenotype of CTCs was studied through the expression of two important EMT-connected genes—namely, VIM and SNAIL —using cytokeratin-negative CTCs from nonmetastatic breast cancer patients, the simultaneous detection of both EGFR and EMT markers may improve prognostic or predictive information. A differential expression pattern of ALDH1 (a stemness marker) and TWIST (an EMT marker) on single CTCs was observed both in early and MBC. CTCs expressing high ALDH1 along with nuclear TWIST were more frequently found in patients with MBC, suggesting that CTCs undergoing EMT may prevail during disease progression.
Counter to expectation, Gorges and colleagues reported low CTC numbers in some patients with late metastatic cancers. These results prompted the search for new markers, including those for mesenchymal-like subpopulations. Plastin-3 is an EMT marker in CTCs in CRC. Aberrant expression of plastin-3 was associated with increased CTCs and poor prognosis in CRC and may be involved in the regulation of EMT. Cell-surface vimentin is specifically expressed on the surface of CTCs from epithelial cancers undergoing EMT, and the number of CTCs undergoing EMT correlates with disease outcome.
Cytokeratins are widely used for the identification of CTCs by immunocytochemistry, but even these established markers might be modulated during EMT. Breast cancer cells display a complex pattern of cytokeratin expression with potential biological relevance. Individual cytokeratin antibodies may recognize only certain cytokeratins, and important subsets of biologically relevant CTCs in cancer patients may be missed.
EMT and MET transitions are central to the metastatic potential of CTCs; the elucidation of CTC plasticity on the clinical outcome of cancer patients may shed more light on this important topic. Thus far, a large number of prognostic studies in patients with breast cancer and other solid tumors and recent experimental studies on CTCs , indicate that CTCs with epithelial attributes are relevant for the onset of metastasis. Thus one might postulate that EMT is important for the release of CTCs from tumor tissues, while MET and the resulting epithelial phenotype of CTCs might allow them to colonize distant organs. , However, the most important feature might be the plasticity of CTCs to change their phenotype depending on the actual requirements of their changing microenvironments (i.e., blood or diverse organ sites such as bone, liver, or brain). This plasticity is not easily assessable by snapshot analysis but requires longitudinal analysis or experimental models such as cell lines or xenografts established from CTCs. , ,
Become a Clinical Tree membership for Full access and enjoy Unlimited articles
If you are a member. Log in here