The prognosis of colorectal cancer is closely linked to the occurrence of distant metastases. Systemic dissemination is most likely caused by circulating tumor cells (CTC). Despite the fundamental role of CTC within the metastatic cascade, technical obstacles have so far prevented detailed genomic and, in particular, phenotypic analyses of CTC, which may provide molecular targets to delay or prevent distant metastases. We show here a detailed genomic analysis of single colorectal cancer–derived CTC by array comparative genomic hybridization (aCGH), mutational profiling, and microsatellite instability (MSI) analysis. Furthermore, we report the first gene expression analysis of manually selected colorectal cancer–derived CTC by quantitative real-time PCR (qRT-PCR) to investigate transcriptional changes, enabling CTC to survive in circulation and form distant metastases. aCGH confirmed the tumor cell identity of CellSearch-isolated colorectal cancer–derived CTC. Mutational and MSI analyses revealed mutational profiles of CTC to be similar, but not identical to the corresponding tumor tissue. Several CTC exhibited mutations in key genes such as KRAS or TP53 that could not be detected in the tumor. Gene expression analyses revealed both a pronounced upregulation of CD47 as a potential immune-escape mechanism and a significant downregulation of several other pathways, suggesting a dormant state of viable CTC. Our results suggest mutational heterogeneity between tumor tissue and CTC that should be considered in future trials on targeted therapy and monitoring of response. The finding of upregulated immune-escape pathways, which may be responsible for survival of CTC in circulation, could provide a promising target to disrupt the metastatic cascade in colorectal cancer. Cancer Res; 74(6); 1694–704. ©2014 AACR.

Colorectal cancer ranks third among cancer-related deaths in Western countries (1). As the primary tumor can often be controlled by surgical resection, the survival of patients with colorectal cancer is closely linked to the incidence of distant metastases, mostly in the liver (2). It is widely accepted that the development of metastatic disease is mediated by circulating tumor cells (CTC), which are shed into the blood stream by the primary tumor, survive within the circulation, and home to distant organs to establish metastases (3). The prognostic relevance CTC detection in the blood of patients with colorectal cancer has been shown by several studies and a meta-analysis (4–6). Also, we showed that considerably more CTC are detectable in the mesenteric/portal venous blood as compared with central venous blood of patients with primary colon cancer, indicating that the route of hematogenous tumor cell dissemination determines, at least in part, the occurrence of metastatic lesions (7).

Despite the apparent relevance of CTC within the process of metastasis, little is known about the biology of CTC, i.e., their genotypic and phenotypic characteristics. Genomic and chromosomal profiling approaches of CTC differ widely and transcriptomic analyses are still missing mainly due to technical issues and the abundant background of blood cells (8). As a consequence, molecular markers that allow selective targeting of CTC and thus potentially interrupt the metastatic cascade have not been developed yet.

The dramatic phenotypic changes tumor cells need to undergo to become functional and tumorigenic CTC are no longer present in outgrown metastatic tumors and must therefore be reversible (9–11). This reversible phenotype is probably based on temporary differential regulation of metastasis-related genes. As such changes may present a therapeutic target to inhibit metastatic activity, this study also aimed to evaluate the transcriptomic changes that distinguish circulating from parenchymal tumor cells and for the first time provides data about the transcriptomic characteristics in colorectal cancer–derived CTC.

The present study therefore presents both a comprehensive genomic and a transcriptional analysis of CTC derived from patients with colorectal cancer. Our findings reveal discrepancies in driving mutations between CTC and the corresponding tumor tissue. Furthermore, the results of gene expression analyses of isolated CTC provide evidence for immune-escape mechanisms involved in CTC survival in circulation, which may offer promising targets for future therapeutic approaches to delay or prevent distant metastases in colorectal cancer.

Cell culture

The colorectal cancer cell line HT-29 was purchased from the Leibnitz Institute DSMZ–German Collection of Microorganisms and Cell Cultures (www.dsmz.de) and maintained in RPMI (PAA) supplemented with 10% fetal calf serum (FCS; PAA), 100 U/mL penicillin (PAA) and 100 μg/mL streptomycin (PAA) in a humidified atmosphere of 5% CO2 at 37°C. The authenticity of the cell line was regularly confirmed by DSMZ.

Patient material

The collection and analysis of solid and liquid patient samples as well as clinicopathologic information for that study has been approved by the Medical Ethics Committee of University of Heidelberg (No. 323/2004). Written informed consent was obtained from each patient before surgery. Detailed patient characteristics are given in Supplementary Tables S5–S7.

Blood samples

Blood samples were obtained from patients undergoing surgical resection at the Department of Surgery, University of Heidelberg. After induction of general anesthesia, blood was drawn through a central venous catheter, or intraoperatively from the mesenteric (superior mesenteric vein or inferior mesenteric vein, depending on the location of the tumor) or the metastasis-draining hepatic vein by direct puncture. After collection, blood samples were immediately processed in the laboratory.

Cellsearch CTC enumeration (Veridex)

7.5 mL of blood, collected in CellSave Tubes (Veridex), were used for CTC analysis according to the manufacturer's instructions. Results were evaluated by two independent and Veridex-certified operators; differences were resolved by discussion.

Genomic DNA amplification of single cells

We performed single-cell whole-genome amplification (WGA) using an adapter–linker PCR as originally described by Klein and colleagues (12). Single EPCAM+CK(8/18/19)+ cells as detected by the CellSearch System were manually transferred into PCR tubes by a micromanipulator. One microliter of Proteinase K mix [1× OnePhorAll Buffer (OPA: 100 mmol/L Tris, 100 mmol/L MgAcetate, 500 mmol/L KAcetate), 0.65% Tween, 0.65% Igepal, 1.3 mg/mL Proteinase K] was added. Cells were digested for 10 hours at 42°C in a thermocycler (Eppendorf) followed by an inactivation step of 10 minutes at 80°C. Then, 0.4 μL of MseI enzyme (Roche) was added. After incubation over 3 hours at 37°C, the enzyme was inactivated by heating to 65°C for 5 minutes. LIB1 (LIB1: 5′-AGTGGGATTCCTGCTGTCAGT-3′) and ddMse11 (ddMSE11: 5′-TAACTGACAGCdd-3′) primers were annealed in a reaction mix of 5 μL 10× OPA, 5 μL LIB1 100 μmol/L, 5 μL ddMse11 100 μmol/L, and 15 μL H2O in a water bath. For ligation, 6 μL of preannealed adapters combined with 2 μL of 10 mmol/L ATP (New England Biolabs) together with 2 μL T4 DNA ligase (Roche) were added to the digested cell. Ligation occurred overnight at 15°C. Genomic DNA was amplified with the Expand Long Template PCR System (Roche) according to the manufacturer's instructions. Amplification was performed in a thermocycler under the following conditions: 1× (68°C 3′); 14× (94°C 40″, 57°C 30″, 68°C 90″+1″/cycle); 8× (64°C 40″, 57°C 30″+ 1°C/cycle, 68°C 1′ 45″+ 1″/cycle); 22× (94°C 40″, 65°C 30″, 68°C 1′53″+1′/cycle); 1× (68°C 3′40″). The PCR products were subsequently purified with the GenElute PCR Clean-up Kit (Sigma) according to the manufacturer's instructions.

Sequencing

Amplified PCR products generated by sequence-specific primers (Supplementary Table s12) were primarily preamplified by AmpliTaq Gold DNA Polymerase (Applied Biosystems) in a thermocycler under the following conditions: 1× (95°C 10′); 38× (95°C 45″, 58°C 10″, 72°C 40″); 1× (72°C 10′). Sequence analysis of preamplified PCR products was performed by a sequencing laboratory (GATC Biotech). For that purpose, newly designed primers are shown in Supplementary Table S13.

Tissue preparation and DNA extraction

Fresh frozen tumor tissue samples were used for sectioning. Sections of 10 μm were cut in a cryostat at −20°C and adhered to object slides. One section of each sample was stained with hematoxylin and eosin and examined by light microscopy to discriminate tumor tissue from stroma and healthy tissue. Between 10 and 20 sections, depending on the amount of tumor tissue, were collected for genomic DNA (gDNA) extraction with the Qiagen DNeasy Tissue Kit according to the manufacturer's instructions. The estimated amount of tumor tissue within the dissected area was, in every case, at least 70%.

Microsatellite instability analysis

Amplified gDNA of CTC and tumor tissue samples was tested for microsatellite instability (MSI) using the markers NR21, NR24, and BAT 25 from the standard NCI/ICG-HNPCC marker panel and analyzed as previously described (13–15). These markers were selected because respective amplicons did not contain MseI restriction sites (Supplementary Table S14). In case of defining a CTC as MSI+, a multiplex PCR was subsequently conducted as previously described, targeting the most prominent gene candidates for MSI in their coding sequences (15).

Single-cell array comparative genomic hybridization

Array comparative genomic hybridization (aCGH) for single cells was performed following the protocol developed by Möhlendick and colleagues (16). In case of single CTC analysis, the SurePrint G3 human CGH 4 × 180K Oligo Microarray Kit (Agilent) was used according to the manufacturer's instructions. Regarding the control experiment to prove the sensitivity of aberration detection, male and female leukocyte gDNA was hybridized against each other. Single CTC analysis was conducted with matched leukocytes. aCGH analysis of microdissected snap-frozen tumor tissue probed against matched healthy tissue was carried out with the SurePrint G3 human CGH 8 × 60K, Oligo Microarray Kit (Agilent). Data preprocessing was performed with Scan Control (V 7.0.3); Feature Extraction (V 10.1.1.1; Agilent). Data analysis and visualisation were performed by Genomic Workbench (V 5.0.14; Agilent; parameters: Genome build: hg18; Evaluation algorithm ADM-2, threshold 6; aberration filter: Minimal number of probes in area = 3, minimal mean log2 ratio = 0.25) and subsequently by the R-based package “GLAD” (17) with its defaults (except breaksFdrQ = 0.01) and using the haarseg (18) algorithm and “aCGH” (ref. 19; aberration filter: Minimal number of probes in area = 3, minimal mean log2 ratio = 0.2).

Mononuclear cell enrichment from blood samples

25 to 30 mL of blood was layered over LSM 1077 lymphocyte gradient (PAA) and the peripheral blood mononuclear cell (PBMC) fraction was enriched according to the manufacturer's instructions.

CTC enrichment and staining

PBMC were resuspended in 1.5 mL of beads buffer (PBS, 2 mmol/L EDTA, 0.1% AB serum). Then, 450 μL anti-CD45 magnetic beads (Dynal) were added to the mononuclear cells and incubated in an overhead shaker for 20 minutes at 4°C, followed by magnetic cell separation. The cell pellet was stained with Alexa-Fluor488-anti-EpCAM antibody (Biolegend). EpCAM-positive cells were identified under a fluorescence microscope (Leica, please see Supplementary Fig. S5), manually isolated with a micromanipulator (Eppendorf) and immediately transferred into RNA lysis buffer (PicoPure RNA isolation kit; Invitrogen). To obtain more valid expression data, 2 to 15 CTC from each patient were pooled. EPCAM+ cells isolated from the primary tumor or hepatic metastases served as comparison for CTC profiles; separately isolated CD45+ leukocytes served as controls. One sample exhibited a CD45 expression level comparable with leukocytes and was excluded from the analysis due to putative leukocyte contamination; CD45 expression levels in all other samples were, in average, 10.000× lower than those of leukocyte control samples (ΔΔCt 16.6). To rule out technical problems, primer pairs in which more than 60% of the CTC samples yielded Ct values >35 were excluded from the analysis.

Tumor tissue processing

Tissue specimens were minced through a 40-μm cell strainer (BD Biosciences) and collected in PBS. The resulting dissociated cells were stained with Alexa-Fluor488-anti-EpCAM (Biolegend), manually isolated with a micromanipulator, and immediately transferred into RNA lysis buffer.

RNA extraction and amplification

Total RNA was extracted by the Arcturus PicoPure RNA Isolation Kit (Invitrogen) according to the manufacturer's instructions. Amplification of total RNA was with the WT-Ovation RNA Amplification Kit (Nugen) according to the manufacturer's instructions.

RT-PCR and statistical methods

Before CTC transcriptome analysis, we confirmed the technical feasibility of single-cell qPCR by a pilot study performed with decreasing numbers of HT-29 colorectal cancer tumor cells (Supplementary Figs. S5 and S6).

SYBR Green RT-PCR was performed on a LightCycler qPCR cycler (Roche) according to the manual with an annealing temperature of 60°C and 55 amplification cycles. Amplification plots were analyzed using the second derivative maximum method. All Primers (Supplementary Table S11) were designed with Primer3Plus software and span large intronic regions to exclude amplification of gDNA. Ct values >35 were considered unspecific. CTC expression data were normalized to the corresponding tumor's gene expression by calculating the ΔΔCt values with the following formula: ΔΔCt = ΔCt tumor − ΔCtCTC = [Ct gene of interest (tumor)Cthousekeeper (tumor)] − [Ct gene of interest (CTC)Ct housekeeper (CTC)].

Statistical analysis of CTC expression profiles was achieved by Wilcoxon matched-pairs signed rank test, comparing the ΔCt values of CTC to the ΔCt values of their corresponding tumor tissue. P < 0.05 was considered statistically significant. Statistical outliers were identified and excluded with the ROUT method. (20) Only genes that were expressed in either tumor or CTC in at least 30% of the samples were analyzed. GraphPad Prism 6.01 (GraphPad Software Inc.) was used for statistical analyses and data plotting.

Expression results from a few genes [CK18 (KRT18), CK20 (KRT20), CXCR4, Ki-67 (MKI67), β-catenin (CTTNB1)] in a subgroup of CTC samples (4 of 11 samples) with metastatic disease to the liver have been used in a different publication (Rahbari and colleagues, submitted for publication) to demonstrate the metastatic potential of CTC derived from colorectal liver metastases.

Single-cell aCGH confirms the genomic tumor cell identity of CTC detected by the CellSearch System

To genomically confirm the tumor cell identity of CTC detected by the CellSearch System [CD45/EPCAM+/CK(8,18, 19)+], we performed single-cell aCGH analysis (cf, Fig. 1).

Figure 1.

Flowchart of the experimental set up used in this work.

Figure 1.

Flowchart of the experimental set up used in this work.

Close modal

Eight single CTC from 8 individual patients and the corresponding microdissected tumor tissue were analyzed (Supplementary Table S1), revealing almost identical chromosomal aberrations in CTC and matched tumor tissue along the genome. The penetrance plot diagram showed a typical colorectal cancer pattern of recurrent chromosomal aberrations (gains at 1q, 7, 8q, 13, and 20 and losses at 1p, 4, 8p, 17p, and 18; Fig. 2A; ref. 11). By dissecting the pooled data set into individual ratio plots, cases with rather similar or punctually different profiles could be identified (Supplementary Fig. S1). Two cases are representatively shown in Fig. 2B. An alternative analytic algorithm (haarseq, see Materials and Methods) confirmed the identified alterations of the single CTC (Supplementary Fig. S3).

Figure 2.

Genomic analysis of single colorectal cancer–derived CTC. A, aCGH data of eight CTC and the matched tumor tissue are illustrated as a penetrance plot mapping the frequency of detected aberrations along the human set of chromosomes (1–22, X, Y). Gains are shown in red and losses in green. B, two CTC/tumor tissue ratio profile pairs are representatively displayed. Fairly similar profiles can be observed for patient HD-2095, punctual differences for patient HD-2116. C, amplified length polymorphism analyses exemplified shown for CTC and control leukocytes of patients HD-2215 and HD-2341 for NR21 (blue) and NR24 (green). Asterisks indicate the unchanged microsatellite length as detected in control leukocytes. Arrows point out an altered microsatellite length in single CTC or tumor specimen.

Figure 2.

Genomic analysis of single colorectal cancer–derived CTC. A, aCGH data of eight CTC and the matched tumor tissue are illustrated as a penetrance plot mapping the frequency of detected aberrations along the human set of chromosomes (1–22, X, Y). Gains are shown in red and losses in green. B, two CTC/tumor tissue ratio profile pairs are representatively displayed. Fairly similar profiles can be observed for patient HD-2095, punctual differences for patient HD-2116. C, amplified length polymorphism analyses exemplified shown for CTC and control leukocytes of patients HD-2215 and HD-2341 for NR21 (blue) and NR24 (green). Asterisks indicate the unchanged microsatellite length as detected in control leukocytes. Arrows point out an altered microsatellite length in single CTC or tumor specimen.

Close modal

These data underline that CTC detected by the CellSearch System are genomically true colorectal tumor cells, which, in turn, validates the following in-depth analyses of single CTC.

Genetic characterization of colorectal cancer–derived single CTC reveals disparities in key mutations to the primary tumor

We next performed a comprehensive genetic characterization of single CTC via genomic sequencing (cf, Fig. 1).

Initially, CTC were screened for common colorectal cancer–related mutations (KRAS, BRAF, and TP53). In total, we examined 126 CTC isolated from 31 individual patients (Supplementary Tables S2 and S3). Mutations in the KRAS gene in either CTC or tumor tissue were detected in 11 (35.5%), the BRAF V600E mutation in 3 (9.7%), and TP53 mutations in 12 patients (38.7%), which is in the range reported for colorectal cancer mutation rates (21).

MSI at one or more markers (Supplementary Table S2) was detected in CTC from 2 patients (of 25 with complete MSI data sets; 7.7%, Fig. 2C). In 1 patient, two of 11 tested CTC were MSI despite a microsatellite stable (MSS) tumor (Table 1).

Table 1.

Genomic analysis demonstrated heterogeneity between tissue and CTC and between individual CTC, representatively shown for patient HD-2215

SampleMicrosatellite statusKRAS C12/13BRAF V600ETP53 E5/6TP53 E7TP53 E8
PT MSS G12S WT WT WT WT 
LM MSS G12S WT WT WT WT 
HV CTC 1 MSS G12S WT n.d. n.d. WT 
HV CTC 2 MSS n.d. n.d. n.d. n.d. WT 
MV CTC 1 MSS n.d. n.d. n.d. WT n.d. 
MV CTC 2 MSS G12S WT n.d. WT WT 
MV CTC 3 MSI WT n.d. n.d. n.d. n.d. 
MV CTC 4 MSS G12S WT n.d. n.d. n.d. 
MV CTC 5 MSS WT WT WT WT WT 
MV CTC 6 MSS n.d. n.d. n.d. n.d. n.d. 
MV CTC 7 MSI n.d. WT n.d. n.d. n.d. 
SampleMicrosatellite statusKRAS C12/13BRAF V600ETP53 E5/6TP53 E7TP53 E8
PT MSS G12S WT WT WT WT 
LM MSS G12S WT WT WT WT 
HV CTC 1 MSS G12S WT n.d. n.d. WT 
HV CTC 2 MSS n.d. n.d. n.d. n.d. WT 
MV CTC 1 MSS n.d. n.d. n.d. WT n.d. 
MV CTC 2 MSS G12S WT n.d. WT WT 
MV CTC 3 MSI WT n.d. n.d. n.d. n.d. 
MV CTC 4 MSS G12S WT n.d. n.d. n.d. 
MV CTC 5 MSS WT WT WT WT WT 
MV CTC 6 MSS n.d. n.d. n.d. n.d. n.d. 
MV CTC 7 MSI n.d. WT n.d. n.d. n.d. 

NOTE: Numbers (e.g., CTC 1, CTC 2) indicate different single CTC from the same patient. Pathologic results are indicated by bold text to show that the CTCs are heterogeneous and differ from the PT and the LM.

Abbreviations: HV, hepatic venous; MV, mesenteric venous, WT, wild-type; PT, primary tumor; LM, liver metastasis; n.d., not detected.

Of note, 17 of 31 (54.8%) patients exhibited mutational disparity between CTC and primary tumor in any of the tested mutations; in 3 of these 17 patients (17.6%) at least one of the CTC was mutated, whereas the tumor was wild-type (1× MSI, 1× KRAS, 1× TP53 E8); in 13 of 17 patients (76.5%) the CTC were wild-type despite a mutated tumor. In case of the patient with an MSS tumor with MSI CTC (patient HD-2215), we performed immunohistochemical expression analysis of DNA mismatch repair proteins MLH1, MSH2, MSH6, and PMS2 in sections obtained from the patient's primary tumor and metastatic lesions to rule out contaminating stromal cells as the cause of the MSS genotype. In none of the sections, lack or reduction of nuclear MMR protein expression was observed.

Furthermore, to make sure that contaminating normal cells did not obscure MSI, we reanalyzed the MSI status of HD-2215 in all sections that were also stained for MMR protein expression (one primary tumor, two metastatic lesions). To achieve maximal sensitivity, microdissection was performed from three or more regions of each specimen, and MSI was analyzed using a set of four highly sensitive mononucleotide microsatellite markers (BAT25, BAT26, CAT25, and BAT40). None of the 10 analyzed regions (with estimated tumor cell content of >85%) displayed any alleles indicative of MMR deficiency or MSI.

In 18 of 31 patients (58.1%), we were able to analyze more than one CTC, of which 6 (33.3%) showed cytogenetically heterogeneous CTC populations. One patient is representatively shown in Table 1. The heterogeneity of CTC in terms of their microsatellite stability is representatively shown in Table 2.

Table 2.

CTC with signs of MSI were also examined for MSI within coding regions (cMSI)

SampleNR-21NR-24BAXDD5FLT3LGELAVLMSI classification
MV CTC 1 MSI — — MSS MSS MSI 
MV CTC 2 MSI MSI MSS MSS MSS MSI 
MV CTC 3 MSI MSS MSS MSS MSS MSI 
MV PBMC MSS MSS — MSS MSS MSS — 
SampleNR-21NR-24BAXDD5FLT3LGELAVLMSI classification
MV CTC 1 MSI — — MSS MSS MSI 
MV CTC 2 MSI MSI MSS MSS MSS MSI 
MV CTC 3 MSI MSS MSS MSS MSS MSI 
MV PBMC MSS MSS — MSS MSS MSS — 

NOTE: Results of patient HD-2341 are representatively depicted.Three single CTC were classified as MSI-high level (MSI-H) and showed a mutation in the coding region of the ELAVL gene. A corresponding PBMC from patient HD-2341 did not show any signs of MSI. MSI events are shown in bold text to stress the fact that different markers have shown different results in these CTC.

Abbreviation: MV, mesenteric venous.

In 11 of the 18 patients (61.1%) from which more than one CTC was analysed, we found mutational disparity between tumor and CTC; in contrast, we detected such disparity in only 3 of 13 patients (23.3%) with only one analyzed CTC. This indicates that increasing numbers of analysed CTC consequently increase the sensitivity of CTC genotyping in detecting new mutations not detected during tumor genotyping.

In summary, genomic analysis of single colorectal cancer–derived CTC demonstrated typical key mutations of colorectal cancer in expected frequencies. Interestingly, clinically relevant mutations were found in a subgroup of CTC that were not detected in the primary tumor.

Transcriptome analysis of colorectal cancer–derived CTC reveals a downregulation of epithelial markers, CTC dormancy, and an immune-escape phenotype.

To perform qPCR on CTC, we isolated viable CD45/EPCAM+ CTC from blood samples of patients with colorectal cancer by manual isolation with a micromanipulator (cf, Fig. 1; ref. 22).

To increase the likelihood of CTC detection, we isolated CTC from blood drawn from the tumor-draining vein (mesenteric venous blood in stage III tumors, hepatic venous blood in stage IV disease with hepatic metastases) and compared the isolated CTC with the respective tumor tissue (PT or LM; ref. 7). Blood samples from 72 patients with colorectal cancer were enrolled (Supplementary Tables S4 and S5). To independently control the applied CTC enrichment and isolation protocol, we also analyzed corresponding blood samples on the CellSearch System (Supplementary Table S6).

We succeeded in isolating CTC in nine of 72 cases (14%). Five samples from 3 patients were isolated from the mesenteric vein and compared with tissue from the primary tumor; 6 samples from 5 patients were isolated from the hepatic vein and compared with tissue from liver metastases. A panel of 48 genes (Supplementary Table S8) covering biomarkers for leukocytes, colorectal cancer, EMT (epithelial–mesenchymal transition), cancer, and stemness was chosen.

CD45 expression in CTC did not differ significantly from the corresponding tumor tissue. The epithelial origin of the isolated CTC was confirmed by the expression of EPCAM, CK18 (KRT18), CK19 (KRT19), and CEA (CEACAM7) and the absence of those markers in control leukocyte samples. Generally, gene expression in the CTC samples was on a lower level compared with the corresponding tumor tissue (Fig. 3A and B). Together with the relative downregulation of Ki-67 (MKI67), which was expressed in only 3 of 11 CTC samples (n.s.), and c-Myc (MYC, P = 0.008) as compared with the corresponding tumor tissue, these data indicate a reduced proliferative activity of CTC, which may indicate a state of dormancy in these cells. Interestingly, β-catenin (CTTNB1) is also downregulated (n.s.), indicating reduced activity in the Wnt signaling cascade.

Figure 3.

Transcriptomic analysis of colorectal cancer–derived CTC via qRT-PCR. A, differences in gene expression of selected genes in CTC versus corresponding tumor tissue. Negative values indicate a reduced gene expression in CTC as compared with the tumor; positive values indicate increased expression in CTC. Filled bars indicate significantly downregulated (green) or significantly upregulated (red) genes. Depicted are ΔΔCt values (25–75 percentiles with medians, whiskers indicate min-max). B, heat map depicting the individual ΔΔCt values of CTC samples, normalized to the corresponding tumor tissue. Green squares, downregulated genes in CTC; red squares, upregulated genes; blank fields, missing data.

Figure 3.

Transcriptomic analysis of colorectal cancer–derived CTC via qRT-PCR. A, differences in gene expression of selected genes in CTC versus corresponding tumor tissue. Negative values indicate a reduced gene expression in CTC as compared with the tumor; positive values indicate increased expression in CTC. Filled bars indicate significantly downregulated (green) or significantly upregulated (red) genes. Depicted are ΔΔCt values (25–75 percentiles with medians, whiskers indicate min-max). B, heat map depicting the individual ΔΔCt values of CTC samples, normalized to the corresponding tumor tissue. Green squares, downregulated genes in CTC; red squares, upregulated genes; blank fields, missing data.

Close modal

As expected, most epithelial markers were strongly expressed in the tumor tissue and downregulated in CTC, indicating a potential loss of epithelial phenotype in CTC; the epithelial markers EPCAM (P = 0.027), CK19 (P = 0.012) and CEA (P = 0.002) were significantly downregulated in CTC. However, we did not observe increased expression of the mesenchymal marker vimentin (VIM, P = 0.765).

CTC do not seem to overexpress markers generally associated with “stemness” such as CD166 (ALCAM), CD26 (DPP4), or CD44s. CD133 (PROM1) was not expressed in any of the CTC examined.

Interestingly, the only gene that was overexpressed in CTC compared with the corresponding tumor tissue was CD47 (P = 0.039). CD47 is generally considered as an antiphagocytotic “don't eat me” signal expressed on cancer cells to prevent macrophages and dendritic cells from attacking tumor cells (23). The counterpart of this antiphagocytotic mechanism, the prophagocytic calreticulin (CALR), was significantly downregulated in CTC (P = 0.001).

Migration- and motility-associated genes were generally downregulated, some of which reached or were close to statistical significance [fibronectin (FN1), P = 0.047; CD44v6, P = 0.027; CD151, P = 0.062; TSPAN8, P = 0.064).

In summary, we succeeded in identifying a CTC-specific expression profile as well as disparities to the matched tumor tissue. Tumor cells circulating in the blood stream seem to be in a state of dormancy, indicated by the downregulation of most major signaling pathways, including those responsible for cell migration and motility. The only gene we found significantly upregulated was CD47, which is associated with tumor cell immune escape.

The prognosis of colorectal cancer is closely linked to the occurrence of distant metastases (2). Although CTC play a pivotal role in the metastatic cascade (3) and their prognostic impact has been repeatedly demonstrated (4, 5, 24–27), little is known about their genomic and transcriptomic hallmarks due to the technical obstacles involved in single-cell analysis. The identification of CTC-specific traits would facilitate the development of compounds directly targeting CTC, which could dramatically improve the prognosis of localized disease by preventing metachronous metastases. The aim of the present work was therefore to further characterize CTC and identify genetic and phenotypic hallmarks of CTC, enabling them to survive in the blood stream and ultimately form tumors distant from the primary lesion.

On the basis of the initial proof of genomic tumor cell identity of CTC, we performed a broader mutational characterization of single colorectal cancer–derived CTC. To this end, we aimed to isolate CTC and compare them to the tumor bulk they most likely originated from. We therefore isolated CTC intraoperatively from the appropriate tumor draining vein (e.g., the hepatic vein in case of hepatic metastases, or the mesenteric vein in case of a primary tumor) and compared the isolated CTC with tumor cells isolated from the corresponding tumor tissue. As all blood samples were obtained intraoperatively during tumor resection, we were able to obtain complete sets of blood samples and corresponding tissue samples.

For mutational profiling, we used a robust adapter–linker PCR for WGA that allows comprehensive genomic profiling of fixed and unfixed single cells (12, 16, 28–32). As expected, our data show that CTC are a profoundly heterogeneous cell population. We were able to identify several CTC with mutations that were not present or detected in the matched tumor tissue. This may represent newly acquired mutations in CTC; however, it is more likely that the tumor cell subclone responsible for shedding the mutated CTC into the blood stream was not contained in the biopsy used for genomic analysis of the tumor tissue. Other groups recently observed a similar phenomenon and were subsequently able to detect the mutations found in the CTC at subclonal levels by deep sequencing of the primary tumor tissue (33). As our data strongly support the recent findings of Heitzer and colleagues and other groups (34, 35), especially in the case of KRAS mutations, it is conceivable that many patients receive anti-EGFR treatment although the metastatic subclone within the tumor has already a constitutively active EGF receptor (EGFR) signaling cascade. Testing CTC for KRAS mutations (“liquid biopsy”) may identify such patients and could therefore be a valuable addendum to traditional tumor genotyping; it may detect genetic changes of small regions or CTC-dispatching tumor cell subpopulations in the primary tumor that are not necessarily contained in the tissue sample used for tumor genotyping. Also, as tumor tissue genotyping can only provide a current snapshot and the tumor genotype is subject to constant change, sequential genotyping of CTC may be reasonable to early detect new mutations that may have further therapeutic implications. This hypothesis is well in line with recent findings that circulating KRAS-mutant DNA can be an early predictor of future resistance to anti-EGFR therapy (36). In summary, the current approach using the solid tumor as the sole source of information for therapeutic decision may be substantially improved by additional analysis of CTC or circulating tumor DNA.

Most distant metastases genotypically and phenotypically resemble their primary tumor (11). This implicates that the changes enabling CTC to perform the necessary steps to cause distant metastases must be reversible and, thus, on a transcriptomic rather than a genomic level. Because of the volatile nature of RNA and the miniscule amounts available for analysis, transcriptomic profiling of CTC is technically challenging. We succeeded in isolating viable CTC by manual isolation from the PBMC fraction and subsequently analyzed the transcriptome by qPCR. We demonstrated reduced expression of most tested genes, indicating a state of dormancy of CTC. The downregulation of c-Myc and Ki-67 supports this assumption and is well in line with other reports (37).

Surprisingly, most genes related to cell migration and invasion such as TSPAN8 (38, 39) and CD44v6 were significantly downregulated as well. This, again, may indicate the short-lived nature of the phenotypic changes in CTC: once in circulation, CTC do not require migratory capabilities anymore and may have already lost the mesenchymal phenotype at the time of analysis. Consequently, the tetraspanin CD151, which is involved in cell–cell contacts and which has been shown to be downregulated in metastatic colorectal cancer as compared with nonmetastatic colorectal cancer (40, 41), was significantly downregulated, indicating reduced cell–cell adhesion capabilities of CTC.

The blood stream is a hostile environment for tumor cells; although a tumor presumably sheds millions of tumor cells into the blood stream every day (42), only few can be detected at any given time. This may be due to the rapid clearance of CTC from the blood stream, most likely mediated by natural killer (NK) cells, monocytes/macrophages, and neutrophils. CTC are therefore under evolutionary pressure to develop a phenotype that can protect or hide them from the immune system. In line with this hypothesis, we found CTC to exhibit a distinct nonimmunogenic phenotype by overexpressing CD47, a protein inhibiting the cytotoxic and phagocytic activity of activated immune cells (43–45). CD47 binds to SIRPa (46), an inhibitory receptor on macrophages and T cells (47–49) and is notably the only gene we found to be overexpressed in otherwise mostly dormant CTC. Previous studies on patients with breast cancer indicated the presence of CD47-positive CTC in the bone marrow, which may be responsible for tumor relapse (50). Other groups have shown that constitutive CD47 upregulation is a pivotal requirement for non-Hodgkin lymphoma immunotolerance and dissemination (51, 52). In concordance with the upregulation of CD47 in CTC is the marked downregulation of calreticulin, a chaperone protein acting as a homing beacon, inducing a strong cytotoxic and phagocytic reaction in NK cells and other immune effector cells (53, 54).

The presented data provide new insight into the biology of CTC as it becomes clear that colorectal cancer–derived CTC are mostly dormant and disguised from the immune system. To our knowledge, this is the first study demonstrating both a comprehensive genomic and transcriptomic profiling of colorectal cancer–derived CTC. The data also demonstrate the distinct heterogeneity among CTC, which may explain the relatively low number of metastases compared with the number of CTC present in the blood stream (42), and suggest that only a small fraction of CTC is actually able to form new lesions. A recent work by Baccelli and colleagues showed that EPCAM+CD44+CD47+MET+ CTC but not EPCAM+ CTC isolated from patients with breast cancer are able to form tumors in mice when injected in sufficient numbers (45). This tumor cell population seems to include metastasis-inducing cells; however, the fact that >1,000 of these cells were required to induce tumors suggests that the phenotype needs to be narrowed down even more to define the actual metastasis-inducing cells.

Another unanswered question remains the role of EPCAM CTC. Our expression data show a significant downregulation of EPCAM on CTC, indicating a loss of epithelial phenotype during EMT. Unfortunately, most current methods of CTC detection and isolation, including the methods used in this work, are based on the assumption that CTC constitutively express EPCAM on their surface. CTC which have lost EPCAM expression during EMT remain undetected, which raises the question of their biologic significance. EPCAM CTC may be more capable of inducing distant metastasis as they have undergone EMT to a further extent compared with EPCAM+ CTC. This question needs to be addressed in the future once standardized and reliable markers or physical properties allowing EPCAM-independent detection of CTC have been established. To date, neither the existence of EPCAM-negative colorectal cancer–derived CTC nor their biologic relevance have been reported and the biologic relevance of EPCAM+ CTC in colorectal cancer is well established. Hence, EPCAM still remains the best marker for CTC detection and isolation available today.

In conclusion, our study confirms the genetic heterogeneity of colorectal cancer–derived CTC and demonstrates mutational disparities to the primary tumor. Transcriptome analyses suggest a dormant state of CTC and, for the first time, provide evidence of immune-escape mechanisms of CTC in humans. These data further support the concept of CTC as predictive biomarkers in colorectal cancer. Furthermore, they identified immune-escape mechanisms as potential targets to disrupt the metastatic cascade in colorectal cancer.

No potential conflicts of interest were disclosed.

Conception and design: G. Steinert, S. Schölch, T. Niemietz, U. Bork, M.W. Büchler, J. Weitz, M. Koch

Development of methodology: G. Steinert, S. Schölch, T. Niemietz, U. Bork, M.W. Büchler

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S. Schölch, T. Niemietz, N. Iwata, B. Behrens, A. Voigt, M. Kloor, U. Bork, N.N. Rahbari, J. Weitz

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): G. Steinert, S. Schölch, T. Niemietz, M. Kloor, A. Benner, U. Bork, N.N. Rahbari, M.W. Büchler, N.H. Stoecklein, J. Weitz, M. Koch

Writing, review, and/or revision of the manuscript: G. Steinert, S. Schölch, T. Niemietz, M. Kloor, U. Bork, N.N. Rahbari, M.W. Büchler, N.H. Stoecklein, J. Weitz, M. Koch

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): G. Steinert, S. Schölch, T. Niemietz, S.A. García, B. Behrens, A. Voigt, M.W. Büchler, J. Weitz

Study supervision: G. Steinert, S. Schölch, M.W. Büchler, J. Weitz, M. Koch

The authors thank Melanie Bernhard, Maria Thomalla-Starzl, and Marzena Knyssok-Sypniewski for excellent technical assistance.

This work was supported by the KFO 227 program (Clinical Research Group 227: Colorectal cancer: from primary tumor progression towards metastases (WE 3548/4-1/2) of Deutsche Forschungsgemeinschaft (DFG).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Siegel
R
,
Naishadham
D
,
Jemal
A
. 
Cancer statistics, 2013
.
CA Cancer J Clin
2013
;
63
:
11
30
.
2.
Weitz
J
,
Koch
M
,
Debus
J
,
Höhler
T
,
Galle
PR
,
Büchler
MW
. 
Colorectal cancer
.
Lancet
2005
;
365
:
153
65
.
3.
Pantel
K
,
Alix-Panabières
C
,
Riethdorf
S
. 
Cancer micrometastases
.
Nat Rev Clin Oncol
2009
;
6
:
339
51
.
4.
Rahbari
NN
,
Aigner
M
,
Thorlund
K
,
Mollberg
N
,
Motschall
E
,
Jensen
K
, et al
Meta-analysis shows that detection of circulating tumor cells indicates poor prognosis in patients with colorectal cancer
.
Gastroenterology
2010
;
138
:
1714
26
.
5.
Cohen
SJ
,
Punt
CJA
,
Iannotti
N
,
Saidman
BH
,
Sabbath
KD
,
Gabrail
NY
, et al
Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer
.
J Clin Oncol
2008
;
26
:
3213
21
.
6.
Cohen
SJ
,
Punt
CJA
,
Iannotti
N
,
Saidman
BH
,
Sabbath
KD
,
Gabrail
NY
, et al
Prognostic significance of circulating tumor cells in patients with metastatic colorectal cancer
.
Ann Oncol
2009
;
20
:
1223
9
.
7.
Rahbari
NN
,
Bork
U
,
Kircher
A
,
Nimitz
T
,
Schölch
S
,
Kahlert
C
, et al
Compartmental differences of circulating tumor cells in colorectal cancer
.
Ann Surg Oncol
2012
;
19
:
2195
202
.
8.
Steinert
G
,
Schölch
S
,
Koch
M
,
Weitz
J
. 
Biology and significance of circulating and disseminated tumour cells in colorectal cancer
.
Langenbecks Arch Surg
2012
;
397
:
535
42
.
9.
Vakiani
E
,
Janakiraman
M
,
Shen
R
,
Sinha
R
,
Zeng
Z
,
Shia
J
, et al
Comparative genomic analysis of primary versus metastatic colorectal carcinomas
.
J Clin Oncol
2012
;
30
:
2956
62
.
10.
Vermaat
JS
,
Nijman
IJ
,
Koudijs
MJ
,
Gerritse
FL
,
Scherer
SJ
,
Mokry
M
, et al
Primary colorectal cancers and their subsequent hepatic metastases are genetically different: implications for selection of patients for targeted treatment
.
Clin Cancer Res
2012
;
18
:
688
99
.
11.
Stange
DE
,
Engel
F
,
Longerich
T
,
Koo
BK
,
Koch
M
,
Delhomme
N
, et al
Expression of an ASCL2 related stem cell signature and IGF2 in colorectal cancer liver metastases with 11p15.5 gain
.
Gut
2010
;
59
:
1236
44
.
12.
Klein
CA
,
Schmidt-Kittler
O
,
Schardt
JA
,
Pantel
K
,
Speicher
MR
,
Riethmüller
G
. 
Comparative genomic hybridization, loss of heterozygosity, and DNA sequence analysis of single cells
.
Proc Natl Acad Sci U S A
1999
;
96
:
4494
9
.
13.
Boland
CR
,
Thibodeau
SN
,
Hamilton
SR
,
Sidransky
D
,
Eshleman
JR
,
Burt
RW
, et al
A National Cancer Institute Workshop on Microsatellite Instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer
.
Cancer Res
1998
;
58
:
5248
57
.
14.
Bacher
JW
,
Flanagan
LA
,
Smalley
RL
,
Nassif
NA
,
Burgart
LJ
,
Halberg
RB
, et al
Development of a fluorescent multiplex assay for detection of MSI-High tumors
.
Dis Markers
2004
;
20
:
237
50
.
15.
Findeisen
P
,
Kloor
M
,
Merx
S
,
Sutter
C
,
Woerner
SM
,
Dostmann
N
, et al
T25 repeat in the 3′ untranslated region of the CASP2 gene: a sensitive and specific marker for microsatellite instability in colorectal cancer
.
Cancer Res
2005
;
65
:
8072
8
.
16.
Möhlendick
B
,
Bartenhagen
C
,
Behrens
B
,
Honisch
E
,
Raba
K
,
Knoefel
WT
, et al
A robust method to analyze copy number alterations of less than 100 kb in single cells using oligonucleotide array CGH
.
PLoS ONE
2013
;
8
:
e67031
.
17.
Hupé
P
,
Stransky
N
,
Thiery
J-P
,
Radvanyi
F
,
Barillot
E
. 
Analysis of array CGH data: from signal ratio to gain and loss of DNA regions
.
Bioinformatics
2004
;
20
:
3413
22
.
18.
Ben-Yaacov
E
,
Eldar
YC
. 
A fast and flexible method for the segmentation of aCGH data
.
Bioinformatics
2008
;
24
:
i139
145
.
19.
Dimitrov
P
. 
Classes and functions for array comparative genomic hybridization data.
R package version 1.12.0;
2007
.
20.
Motulsky
HJ
,
Brown
RE
. 
Detecting outliers when fitting data with nonlinear regression - a new method based on robust nonlinear regression and the false discovery rate
.
BMC Bioinformatics
2006
;
7
:
123
.
21.
Cancer Genome Atlas Network
. 
Comprehensive molecular characterization of human colon and rectal cancer
.
Nature
2012
;
487
:
330
7
.
22.
Klein
CA
,
Seidl
S
,
Petat-Dutter
K
,
Offner
S
,
Geigl
JB
,
Schmidt-Kittler
O
, et al
Combined transcriptome and genome analysis of single micrometastatic cells
.
Nat Biotechnol
2002
;
20
:
387
92
.
23.
Jaiswal
S
,
Jamieson
CHM
,
Pang
WW
,
Park
CY
,
Chao
MP
,
Majeti
R
, et al
CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis
.
Cell
2009
;
138
:
271
85
.
24.
Koch
M
,
Kienle
P
,
Kastrati
D
,
Antolovic
D
,
Schmidt
J
,
Herfarth
C
, et al
Prognostic impact of hematogenous tumor cell dissemination in patients with stage II colorectal cancer
.
Int J Cancer
2006
;
118
:
3072
7
.
25.
Weitz
J
,
Kienle
P
,
Magener
A
,
Koch
M
,
Schrödel
A
,
Willeke
F
, et al
Detection of disseminated colorectal cancer cells in lymph nodes, blood and bone marrow
.
Clin Cancer Res
1999
;
5
:
1830
6
.
26.
Weitz
J
,
Kienle
P
,
Lacroix
J
,
Willeke
F
,
Benner
A
,
Lehnert
T
, et al
Dissemination of tumor cells in patients undergoing surgery for colorectal cancer
.
Clin Cancer Res
1998
;
4
:
343
8
.
27.
Weitz
J
,
Koch
M
,
Kienle
P
,
Schrödel
A
,
Willeke
F
,
Benner
A
, et al
Detection of hematogenic tumor cell dissemination in patients undergoing resection of liver metastases of colorectal cancer
.
Ann Surg
2000
;
232
:
66
72
.
28.
Schmidt-Kittler
O
,
Ragg
T
,
Daskalakis
A
,
Granzow
M
,
Ahr
A
,
Blankenstein
TJF
, et al
From latent disseminated cells to overt metastasis: genetic analysis of systemic breast cancer progression
.
Proc Natl Acad Sci U S A
2003
;
100
:
7737
42
.
29.
Gangnus
R
,
Langer
S
,
Breit
E
,
Pantel
K
,
Speicher
MR
. 
Genomic profiling of viable and proliferative micrometastatic cells from early-stage breast cancer patients
.
Clin Cancer Res
2004
;
10
:
3457
64
.
30.
Stoecklein
NH
,
Hosch
SB
,
Bezler
M
,
Stern
F
,
Hartmann
CH
,
Vay
C
, et al
Direct genetic analysis of single disseminated cancer cells for prediction of outcome and therapy selection in esophageal cancer
.
Cancer Cell
2008
;
13
:
441
53
.
31.
Fuhrmann
C
,
Schmidt-Kittler
O
,
Stoecklein
NH
,
Petat-Dutter
K
,
Vay
C
,
Bockler
K
, et al
High-resolution array comparative genomic hybridization of single micrometastatic tumor cells
.
Nucleic Acids Res
2008
;
36
:
e39
.
32.
Geigl
JB
,
Speicher
MR
. 
Single-cell isolation from cell suspensions and whole genome amplification from single cells to provide templates for CGH analysis
.
Nat Protoc
2007
;
2
:
3173
84
.
33.
Heitzer
E
,
Auer
M
,
Gasch
C
,
Pichler
M
,
Ulz
P
,
Hoffmann
EM
, et al
Complex tumor genomes inferred from single circulating tumor cells by array-CGH and next-generation sequencing
.
Cancer Res
2013
;
73
:
2965
75
.
34.
Gasch
C
,
Bauernhofer
T
,
Pichler
M
,
Langer-Freitag
S
,
Reeh
M
,
Seifert
AM
, et al
Heterogeneity of epidermal growth factor receptor status and mutations of KRAS/PIK3CA in circulating tumor cells of patients with colorectal cancer
.
Clin Chem
2013
;
59
:
252
60
.
35.
Mostert
B
,
Jiang
Y
,
Sieuwerts
AM
,
Wang
H
,
Bolt-de Vries
J
,
Biermann
K
, et al
KRAS and BRAF mutation status in circulating colorectal tumor cells and their correlation with primary and metastatic tumor tissue
.
Int J Cancer
2013
;
133
:
130
41
.
36.
Misale
S
,
Yaeger
R
,
Hobor
S
,
Scala
E
,
Janakiraman
M
,
Liska
D
, et al
Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer
.
Nature
2012
;
486
:
532
6
.
37.
Müller
V
,
Stahmann
N
,
Riethdorf
S
,
Rau
T
,
Zabel
T
,
Goetz
A
, et al
Circulating tumor cells in breast cancer: correlation to bone marrow micrometastases, heterogeneous response to systemic therapy and low proliferative activity
.
Clin Cancer Res
2005
;
11
:
3678
85
.
38.
Guo
Q
,
Xia
B
,
Zhang
F
,
Richardson
MM
,
Li
M
,
Zhang
JS
, et al
Tetraspanin CO-029 inhibits colorectal cancer cell movement by deregulating cell-matrix and cell-cell adhesions
.
PLoS ONE
2012
;
7
:
e38464
.
39.
Gesierich
S
,
Paret
C
,
Hildebrand
D
,
Weitz
J
,
Zgraggen
K
,
Schmitz-Winnenthal
FH
, et al
Colocalization of the tetraspanins, CO-029 and CD151, with integrins in human pancreatic adenocarcinoma: impact on cell motility
.
Clin Cancer Res
2005
;
11
:
2840
52
.
40.
Lin
PC
,
Lin
SC
,
Lee
CT
,
Lin
YJ
,
Lee
JC
. 
Dynamic change of tetraspanin CD151 membrane protein expression in colorectal cancer patients
.
Cancer Invest
2011
;
29
:
542
7
.
41.
Chien
CW
,
Lin
SC
,
Lai
YY
,
Lin
BW
,
Lin
SC
,
Lee
JC
, et al
Regulation of CD151 by hypoxia controls cell adhesion and metastasis in colorectal cancer
.
Clin Cancer Res
2008
;
14
:
8043
51
.
42.
Chang
YS
,
di Tomaso
E
,
McDonald
DM
,
Jones
R
,
Jain
RK
,
Munn
LL
. 
Mosaic blood vessels in tumors: frequency of cancer cells in contact with flowing blood
.
Proc Natl Acad Sci USA
2000
;
97
:
14608
13
.
43.
Kim
MJ
,
Lee
JC
,
Lee
JJ
,
Kim
S
,
Lee
SG
,
Park
SW
, et al
Association of CD47 with natural killer cell-mediated cytotoxicity of head-and-neck squamous cell carcinoma lines
.
Tumour Biol
2008
;
29
:
28
34
.
44.
Legrand
N
,
Huntington
ND
,
Nagasawa
M
,
Bakker
AQ
,
Schotte
R
,
Strick-Marchand
H
, et al
Functional CD47/signal regulatory protein alpha (SIRP(alpha)) interaction is required for optimal human T- and natural killer- (NK) cell homeostasis in vivo
.
Proc Natl Acad Sci U S A
2011
;
108
:
13224
9
.
45.
Baccelli
I
,
Schneeweiss
A
,
Riethdorf
S
,
Stenzinger
A
,
Schillert
A
,
Vogel
V
, et al
Identification of a population of blood circulating tumor cells from breast cancer patients that initiates metastasis in a xenograft assay
.
Nat Biotechnol
2013
;
31
:
539
44
.
46.
Jiang
P
,
Lagenaur
CF
,
Narayanan
V
. 
Integrin-associated protein is a ligand for the P84 neural adhesion molecule
.
J Biol Chem
1999
;
274
:
559
62
.
47.
Brown
EJ
,
Frazier
WA
. 
Integrin-associated protein (CD47) and its ligands
.
Trends Cell Biol
2001
;
11
:
130
5
.
48.
Avice
MN
,
Rubio
M
,
Sergerie
M
,
Delespesse
G
,
Sarfati
M
. 
CD47 ligation selectively inhibits the development of human naive T cells into Th1 effectors
.
J Immunol
2000
;
165
:
4624
31
.
49.
Armant
M
,
Avice
MN
,
Hermann
P
,
Rubio
M
,
Kiniwa
M
,
Delespesse
G
, et al
CD47 ligation selectively downregulates human interleukin 12 production
.
J Exp Med
1999
;
190
:
1175
82
.
50.
Nagahara
M
,
Mimori
K
,
Kataoka
A
,
Ishii
H
,
Tanaka
F
,
Nakagawa
T
, et al
Correlated expression of CD47 and SIRPA in bone marrow and in peripheral blood predicts recurrence in breast cancer patients
.
Clin Cancer Res
2010
;
16
:
4625
35
.
51.
Chao
MP
,
Tang
C
,
Pachynski
RK
,
Chin
R
,
Majeti
R
,
Weissman
IL
. 
Extranodal dissemination of non-Hodgkin lymphoma requires CD47 and is inhibited by anti-CD47 antibody therapy
.
Blood
2011
;
118
:
4890
901
.
52.
Chao
MP
,
Alizadeh
AA
,
Tang
C
,
Myklebust
JH
,
Varghese
B
,
Gill
S
, et al
Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma
.
Cell
2010
;
142
:
699
713
.
53.
Zeng
Y
,
Chen
X
,
Larmonier
N
,
Larmonier
C
,
Li
G
,
Sepassi
M
, et al
Natural killer cells play a key role in the antitumor immunity generated by chaperone-rich cell lysate vaccination
.
Int J Cancer
2006
;
119
:
2624
31
.
54.
Gardai
SJ
,
McPhillips
KA
,
Frasch
SC
,
Janssen
WJ
,
Starefeldt
A
,
Murphy-Ullrich
JE
, et al
Cell-surface calreticulin initiates clearance of viable or apoptotic cells through trans-activation of LRP on the phagocyte
.
Cell
2005
;
123
:
321
34
.

Supplementary data