Skip to main content
JCO Precision Oncology logoLink to JCO Precision Oncology
. 2023 Dec 14;7:e2300303. doi: 10.1200/PO.23.00303

Prospective Characterization of Circulating Tumor Cell Kinetics in Patients With Oligometastatic Disease Receiving Definitive Intent Radiation Therapy

Shivani Sud 1, Michael J Poellmann 2,3, Jacob Hall 1, Xianming Tan 1, Jiyoon Bu 2, Ja Hye Myung 4, Andrew Z Wang 1,5, Seungpyo Hong 2,3,4,6, Dana L Casey 1,
PMCID: PMC10730071  PMID: 38096474

Abstract

PURPOSE

There are currently no predictive molecular biomarkers to identify patients with oligometastatic disease (OMD) who will benefit from definitive-intent radiation therapy (RT). We prospectively characterized circulating tumor cell (CTC) kinetics in patients with OMD undergoing definitive-intent RT.

METHODS

This prospective correlative biomarker study included patients with any solid malignancy ≤5 metastatic sites in ≤3 anatomic organ systems undergoing definitive-intent RT to all disease sites. Circulating tumor cells (CTCs) were captured and enumerated using a biomimetic cell rolling and nanotechnology-based assay functionalized with antibodies against epithelial cell adhesion molecule, against human epidermal growth factor receptor 2, and against epidermal growth factor receptor before and during RT and at follow-up visits up to 2 years post-RT.

RESULTS

We enrolled 43 patients with a median follow-up of 14.3 months. The pretreatment CTC level (cells captured/mL) was not associated with the number of disease sites (median one metastatic site/patient, range 1-5) or metastasis location (bone, brain, visceral) on Wilcoxon signed-rank test, P > .05. Post-RT, 56% of patients received systemic therapy, and 72% of patients experienced subsequent local or systemic progression. For 90% of patients, a CTC level <15 within 130 days post-RT corresponded to a durable control of irradiated lesions. Patients with a favorable versus an unfavorable clearance profile experienced significantly longer progression-free survival after RT (median 13 v 4 months, log-rank test, P = .0011). On logistic regression, CTC level >15 at a given time point was associated with clinical disease progression within the subsequent 6 months (odds ratio 3.31, P = .007). In 26% of patients with disease progression, a CTC level >15 preceded radiographic or clinical progression.

CONCLUSION

CTCs may serve as a biomarker for disease control in OMD and may predict disease progression before standard assessments for patients receiving diverse cancer-directed therapies.

INTRODUCTION

Patients with oligometastatic disease (OMD), an intermediate state between localized and wide-spread metastatic disease, may experience a progression-free survival (PFS) or overall survival (OS) benefit from aggressive metastasis-directed therapy.1-5 Guidelines define OMD as ≤5 metastatic lesions on imaging, an optional controlled primary tumor, where all metastatic sites are safely treatable on the basis of expert consensus given the absence of rigorously validated criteria.2 Thus, this definition encompasses true OMD and occult polymetastatic disease which is treated per a vastly different paradigm. There are presently no clinically available assays to assist in defining OMD, selecting appropriate candidates for OMD-directed therapy, nor identifying patients with persistent disease requiring prompt initiation of systemic therapy, highlighting an urgent need for biomarkers for management of OMD.

CONTEXT

  • Key Objective

  • There are currently no predictive molecular biomarkers to identify patients with oligometastatic disease (OMD) who will benefit from definitive-intent radiation therapy (RT). We prospectively characterized circulating tumor cell (CTC) kinetics in patients with OMD undergoing definitive-intent RT to all disease sites.

  • Knowledge Generated

  • Patients with a favorable versus an unfavorable clearance profile experienced significantly longer progression-free survival after RT. On logistic regression, elevations in CTC levels at a given time point were associated with clinical disease progression within the subsequent 6 months including a subset of cases where elevation in CTC level preceded radiographic or clinical progression.

  • Relevance

  • CTCs may serve as a biomarker for disease control in OMD and have potential to further define the role and timing of RT and systemic therapy for OMD by predicting disease progression prior to standard assessments for patients receiving diverse cancer-directed therapies.

Circulating biomarkers including cell-free DNA (cfDNA) and circulating tumor cells (CTCs) show potential as liquid biopsies to diagnose cancer, monitor response, detect residual disease, and recognize recurrence before standard surveillance practices.6 Although cfDNA has substantial emerging indications, it is challenging to apply in the setting of OMD given the wide array of histologies under investigation and the ability to only detect a priori specified mutations that may not reflect clones surviving therapy. In contrast, sensitive CTC capture assays are capable of detecting CTCs from a wide range of histologies and mutational profiles in the peripheral circulation.7,8

We performed a prospective correlative biomarker study inclusive of patients with OMD receiving definitive-intent radiation therapy (RT) to all disease sites in which we characterize CTC profiles associated with treatment outcomes.

METHODS

Patients and Biospecimens

This prospective single-institution correlative biomarker study included patients with any solid malignancy and OMD defined as ≤5 metastatic sites in ≤3 anatomic organ systems receiving definitive-intent RT to all active disease sites. Patients with de novo oligometastatic, oligorecurrent, oligoprogressive, and oligopersistent disease meeting the above criteria were included.1 Subsequent lines of therapy were given per treating physician's discretion. Whole blood was collected in sodium heparin tubes. Biospecimen collections occurred before RT (baseline), during RT, and at standard-of-care follow-up visits up to 24 months post-RT. Enrollment was from October 5, 2015, to October 28, 2020. Providers were blinded to CTC enumeration results.

Ethics Approval

All appropriate ethics approvals and consents were obtained from the University of North Carolina Institutional Review Board.

CTC Enumeration

In brief, CTCs were captured and enumerated using a previously reported biomimetic cell rolling and nanotechnology-based assay. The capture surfaces were functionalized with three antibodies against epithelial cell adhesion molecule (aEPCAM), against human epidermal growth factor receptor 2 (aHER2), and against epidermal growth factor receptor (aEGFR) on poly(amidoamine) (PAMAM) dendrimers-coated surface for strong multivalent binding, along with recombinant human E-selectin that induces cell rolling.7 The surfaces after capture were imaged after immunocytochemistry for pan-cytokeratin (cytokeratin [CK]), leukocyte marker CD45, and 4’,6-diamidino-2-phenylindole (DAPI) nuclear stain. CTC levels were determined from DAPI+/CK+/CD45– cells and expressed per mL of whole blood.

CTC Capture Surface Preparation

Capture surfaces were fabricated as previously described.7 Generation 7 PAMAM dendrimer (Dendritech, Midland, MI) was purified by centrifugal filtration, lyophilized, and partially carboxylated by reaction with succinic anhydride (Sigma-Aldrich, St Louis, MO) in dimethyl sulfoxide at a molar ratio of 358:1, resulting in approximately 70% of terminal amines converted to carboxyl groups. Epoxy-functionalized glass slides (Tekdon, Myakka City, FL) were treated overnight with 5,000 MW and heterobifunctional amine-poly(ethylene glycol)-carboxymethyl polymers (JenKem, Plano, TX) at 0.5 mg mL−1 in distilled deionized (DDI) water. The polyethylene glycol was activated with 15 mM 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDC, Sigma-Aldrich) and 25 mM N-hydroxysuccinimide (NHS, Sigma-Aldrich) in DDI water for 30 minutes. PAMAM-COOH was added at 0.1 mg mL−1 in sodium phosphate buffer, pH 8 with 150 mM sodium chloride and incubated overnight. The surfaces were activated once more with EDC and NHS for 30 minutes before treatment for 60 minutes with a mixture of mouse anti-human monoclonal antibodies (R&D Systems, Minneapolis, MN) each at 1 μg mL−1 in sodium phosphate, pH 8 with 150 mM sodium chloride. Mouse anti-human monoclonal antibodies consisted of aEPCAM (R&D Systems AF960), aHER2 (R&D Systems AF1129), and aEGFR (R&D Systems AF231).9 Finally, surfaces were incubated with 1 μg mL−1 recombinant human e-selectin Fc chimera protein (R&D Systems 724-ES) in phosphate buffered saline (PBS) for 60 minutes. Slides were stored at 4°C in PBS for up to 4 weeks before use.

CTC Purification and Quantification

Blood tubes were gently agitated, and 6 mL whole blood was separated by Ficoll-Paque PLUS (GE Healthcare, Uppsala, Sweden). The buffy coat was purified twice by centrifugation to isolate mononuclear cells and resuspended in 0.1 mL Dulbecco's Minimum Essential Medium (Corning Life Sciences, Tewksbury, MA). The cell suspension was pumped through a flow chamber8 containing the capture slide. The flow chamber consisted of a pair of 55 mm long, 5 mm wide, and 0.15 mm tall channels connected by a length of 0.16ʺ inner diameter tubing. The flow rate was controlled by a NE-1000 syringe pump (New Era, Farmingdale, NY) at 0.025 mL/min for 20 minutes, followed by 0.090 mL/minutes for 15 minutes, followed by a rinse in the reverse direction with PBS at 0.090 mL/minutes for 15 minutes.

Slides were removed from the chamber, fixed with 4% paraformaldehyde (Polysciences, Warrington, PA), permeabilized with 0.2% Triton X-100 (Sigma-Aldrich), and treated with 2 wt.% bovine serum albumin (BSA, Sigma-Aldrich) to reduce nonspecific binding. Cells were then sequentially stained with the following antibodies: rabbit anti-human pan CK (1:50, Abcam 9377), goat anti-human AlexaFluor 568 or AlexaFluor 647 conjugated secondary antibody (1:500, Thermo Fisher Scientific, Waltham, MA), mouse anti-human CD45 (1:500, BD Biosciences 555480, San Jose, CA), and goat anti-mouse AlexaFluor 488 conjugated secondary antibody (1:500, Thermo Fisher Scientific). Nuclei were labeled with DAPI (Thermo Fisher Scientific), and slides were sealed with mounting media under a cover glass.

Slide surfaces were imaged with a Zeiss 701 confocal laser scanning microscope with 405-nm diode UV laser, 488-nm mutliline Ar laser, and 561-nm solid state diode laser at 20×/0.8 Plan-Apocromat objective (Carl Zeiss, Munich, Germany) or Zeiss Axio Observer wide field microscope with Colibri 7 LED light source with a 90 HE filter set and either a 20×/0.5 or 10×/0.3 Plan-Neofluar objective and Axiocam 503 mono CCD Camera (Carl Zeiss). A motorized stage on both microscopes enabled collection of the entire slide surface in tiled images. After color balancing, each color channel was exported as a jpeg and analyzed using a custom algorithm in Image J (NIH). The algorithm identified particles in the cytokeratin channel with brightness and circularity similar to a cell. Any CK-positive particles with a minimum signal in the CD45 channel under or immediately around the particle were discarded. Finally, particles were identified in the DAPI using brightness and circularity thresholds. Particles were identified as CTCs if the centroid of a DAPI particle was within half the radius of the centroid of a CK particle.

End Points and Statistical Analysis

Disease status was assessed per standardized RECIST 1.1 criteria at each time point with available CTC level and clinical evaluation/imaging obtained per standard of care. Clinical documentation, radiology reports, and primary images were reviewed by two clinicians to determine whether target lesions met criteria of complete response, progressive disease, or noncomplete response/nonprogressive disease. On exploratory analysis, disease status was correlated with CTCs as a continuous and ordinal variable (cut point upper bound of the third quartile). Regarding kinetics, a favorable (v unfavorable) CTC clearance profile was defined as a decrease (v increase) in CTCs between pretreatment and the end of treatment. PFS was estimated from the end date of RT to the date of the event. Data were analyzed with a generalized linear mixed model. A two-sided P value of <.05 was considered significant. Statistical analyses were performed with R version 4.0.2.10

RESULTS

We enrolled 43 patients with a median follow-up of 14.3 months post-RT (range, 1.4-24.7 months) corresponding to 255 CTC measurements. Samples from 16 time points, 6% of samples, were excluded from analysis because of technical issues including errors during sample processing, shipping delays, sample coagulation, and insufficient biospecimen. Patient characteristics are summarized in Table 1. The median number of distinct metastatic lesions per patient was 1 (range, 1-5). Most patients received stereotactic body RT (SBRT; 72%) versus conventional or hypofractionated courses of RT (28%).

TABLE 1.

Patient Characteristics

graphic file with name po-7-e2300303-g001.jpg

At baseline, all patients had detectable CTCs (median baseline, 28; range, 0.17-1,085). There was no significant association between the pretreatment CTC level and presence of one versus ≥2 metastatic sites (median CTC 22 v 42, Wilcoxon signed-rank test, P = .37), Figure 1. The location of the metastatic site was not associated with the CTC level when categorized as bone, brain, or visceral (median CTC 36 versus 5 v 28, Wilcoxon signed-rank test, P > .05 for all comparisons).

FIG 1.

FIG 1.

Baseline CTC levels and association with disease state. All patients had detectable CTCs before radiation therapy. There was no association between pretreatment CTC count and the total number of active oligometastatic sites. There was also no association between the pretreatment CTC count and the location of metastatic sites with regards to bone, brain, and visceral metastatic disease. CTC, circulating tumor cell.

During RT, the median CTC level declined: baseline 22 (range, 0.17-99; SD, 30), end of RT 11 (range, 0.00-242; SD, 58), and first post-treatment follow-up within 130 days post-RT median 11 (range, 0.00-157; SD, 37) for 17 patients evaluable at all time points.

Therapy after RT was determined per physician discretion independent from CTC levels. After RT, 72% of patients experienced disease progression (n = 31); 3% (n = 1) in the irradiated lesion alone and 33% in the irradiated lesion as well as systemic disease (n = 10), and 64% experienced systemic progression alone (n = 20). For 90% of patients (n = 27 of 30), CTC ≤15 within 130 days post-RT corresponded to long-term local control of irradiated lesions. Among patients with disease progression, 26% experienced an increase in CTC >15 before radiographic or clinical detection of disease progression (n = 8 of 31). Patients with a favorable versus unfavorable clearance profile experienced significantly longer PFS post-RT (median 13 v 4 months, log-rank test, P = .0011; Fig 2).

FIG 2.

FIG 2.

CTC kinetics during RT and association with PFS. During treatment, a favorable kinetic profile defined as a decrease in CTC count between pretreatment and end of treatment versus an unfavorable profile defined as the opposite was associated with a durable response to therapy as evidenced by the difference in PFS between patients with the two CTC profiles. Median PFS for patients with a favorable clearance profile was 13 months compared with 4 months for those with an unfavorable clearance profile. CTC, circulating tumor cell; PFS, progression-free survival; RT, radiation therapy.

At each follow-up time point, we evaluated whether patients experienced disease progression allowing for multiple progression events per patient during their disease course. There were 43 progression events versus 109 nonprogression events in total. On logistic regression, CTC >15 at a given time point was associated with clinical disease progression within the subsequent 6 months (odds ratio, 3.31; P = .007). During this period, 56% (n = 24) patients received systemic therapy (cytotoxic chemotherapy, hormone therapy, immunotherapy, kinase inhibitors) either immediately after RT or at the time of progression. This association remained when controlling for prior CTC levels and type of systemic therapy.

DISCUSSION

Recent randomized clinical trials of SBRT for OMD-directed therapy show favorable outcomes for a subset of patients.3-5 Prognostic and predictive biomarkers are needed to identify patients with OMD versus occult polymetastatic disease who will benefit from OMD-directed therapy.

In this correlative biomarker study, we characterize CTC profiles associated with favorable response versus disease progression in a prospective cohort of patients receiving definitive-intent RT for OMD. These data demonstrate that optimized CTC enumeration technologies can detect CTCs in the setting of OMD, and furthermore, CTC profiles during cancer-directed therapies are strongly associated with, and potentially predictive of, therapeutic response. The ability to distinguish patients with response versus nonresponse to OMD-directed therapy is critical for identifying patients for whom prompt initiation of systemic therapy is indicated.

These findings are consistent with studies of widely metastatic breast, colorectal, and castrate-resistant prostate cancers wherein CTC enumeration is prognostic of PFS or OS using the CellSearch platform.11-13 However, the utility of existing commercial platforms in predictive and low-disease burden settings is limited in part due to assay sensitivity and specificity. Here, we used a second-generation assay with biomimetic rolling and antibodies targeted to various antigens expressed on CTCs.7,14,15

Our study has important limitations. First, the sample size is modest and includes various histologies. Second, the RT dose and fractionation were not standardized. Third, after RT, additional lines of therapy and follow-up intervals were variable. Bearing these limitations, our findings show potential for CTC profiles to predict response to OMD-directed RT and subsequent recurrence.

In conclusion, CTCs may serve as a biomarker for disease control in OMD and have potential to further define the role of both RT and systemic therapy for OMD.

ACKNOWLEDGMENT

The authors acknowledge Sin-Jung Park for performing experiments for capture surface preparation and CTC enumeration/analysis.

Shivani Sud

Employment: UNC Health

Research Funding: Naveris (Inst)

Michael J. Poellmann

Research Funding: Capio Biosciences

Other Relationship: Capio Biosciences

(OPTIONAL) Uncompensated Relationships: Capio Biosciences

Andrew Z. Wang

Leadership: Capio BioSciences, Archimmune Therapeutics, Nanorobotics, Novartis

Stock and Other Ownership Interests: Capio BioSciences, Archimmune Therapeutics, Archimmune Therapeutics

Consulting or Advisory Role: Capio BioSciences, Merck KGaA, Regeneron, Mirati Therapeutics, Janssen, QED Therapeutics, Dendreon, Seagen, Eisai, Aravive, Sanofi/Aventis, MJH Associates, Aptitude Health, PlatformQ Health, AVEO, Gerson Lehrman Group, Peerview, Bayer, Kidney Cancer Association

Research Funding: Archimmune Therapeutics, Varian Medical Systems

Patents, Royalties, Other Intellectual Property: Biomatrix patent licensed to PhoenixSongs

Travel, Accommodations, Expenses: Pfizer, Sanofi, Exelixis, Genentech

Seungpyo Hong

Employment: Capio Biosciences, Dongkook Pharma

Leadership: Capio BioSciences

Stock and Other Ownership Interests: Capio BioSciences

Honoraria: Dongkook Pharma

Consulting or Advisory Role: Dongkook Pharma

Research Funding: Capio BioSciences (Inst), Dongkook Pharma (Inst), Werfen (Inst)

Patents, Royalties, Other Intellectual Property: Have multiple patents that have been licensed to Capio Biosciences

Travel, Accommodations, Expenses: Capio BioSciences

Dana L. Casey

Consulting or Advisory Role: EMD Serono

No other potential conflicts of interest were reported.

PRIOR PRESENTATION

Presented at ASTRO 2021, Chicago, IL, October 24, 2021 and ASCO Annual Meeting, Chicago, IL, June 5, 2022.

SUPPORT

Supported by the University Cancer Research Fund from the University of North Carolina. A.Z.W. was also supported by the National Institutes of Health Center for Nanotechnology Excellence Grant U54-CA151652 and R01CA178748. S.H. was also supported by NIH SPORE (P50CA278595), NIH 1R01CA262292, D2P SEED Fund from University of Wisconsin, and Milton J. Henrichs Chair fund. S.S. was supported by the Lung Cancer Initiative of North Carolina Lung Cancer Research Fellowship grant.

*

S.S. and M.J.P contributed equally to this work.

DATA SHARING STATEMENT

Research data are stored in an institutional repository and will be shared on request to the corresponding author.

AUTHOR CONTRIBUTIONS

Conception and design: Shivani Sud, Jacob Hall, Jiyoon Bu, Ja Hye Myung, Andrew Z. Wang, Seungpyo Hong, Dana L. Casey

Financial support: Andrew Z. Wang, Seungpyo Hong, Dana L. Casey

Provision of study materials or patients: Andrew Z. Wang, Seungpyo Hong, Dana L. Casey

Collection and assembly of data: Shivani Sud, Michael J. Poellmann, Jacob Hall, Jiyoon Bu, Ja Hye Myung, Andrew Z. Wang, Dana L. Casey

Data analysis and interpretation: Shivani Sud, Michael J. Poellmann, Jacob Hall, Xianming Tan, Andrew Z. Wang, Seungpyo Hong, Dana L. Casey

Manuscript writing: All authors

Final approval of manuscript: All authors

Accountable for all aspects of the work: All authors

AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST

The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/po/author-center.

Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments).

Shivani Sud

Employment: UNC Health

Research Funding: Naveris (Inst)

Michael J. Poellmann

Research Funding: Capio Biosciences

Other Relationship: Capio Biosciences

(OPTIONAL) Uncompensated Relationships: Capio Biosciences

Andrew Z. Wang

Leadership: Capio BioSciences, Archimmune Therapeutics, Nanorobotics, Novartis

Stock and Other Ownership Interests: Capio BioSciences, Archimmune Therapeutics, Archimmune Therapeutics

Consulting or Advisory Role: Capio BioSciences, Merck KGaA, Regeneron, Mirati Therapeutics, Janssen, QED Therapeutics, Dendreon, Seagen, Eisai, Aravive, Sanofi/Aventis, MJH Associates, Aptitude Health, PlatformQ Health, AVEO, Gerson Lehrman Group, Peerview, Bayer, Kidney Cancer Association

Research Funding: Archimmune Therapeutics, Varian Medical Systems

Patents, Royalties, Other Intellectual Property: Biomatrix patent licensed to PhoenixSongs

Travel, Accommodations, Expenses: Pfizer, Sanofi, Exelixis, Genentech

Seungpyo Hong

Employment: Capio Biosciences, Dongkook Pharma

Leadership: Capio BioSciences

Stock and Other Ownership Interests: Capio BioSciences

Honoraria: Dongkook Pharma

Consulting or Advisory Role: Dongkook Pharma

Research Funding: Capio BioSciences (Inst), Dongkook Pharma (Inst), Werfen (Inst)

Patents, Royalties, Other Intellectual Property: Have multiple patents that have been licensed to Capio Biosciences

Travel, Accommodations, Expenses: Capio BioSciences

Dana L. Casey

Consulting or Advisory Role: EMD Serono

No other potential conflicts of interest were reported.

REFERENCES

  • 1.Guckenberger M, Lievens Y, Bouma AB, et al. : Characterisation and classification of oligometastatic disease: A European Society for Radiotherapy and Oncology and European Organisation for Research and Treatment of Cancer consensus recommendation. Lancet Oncol 21:e18-e28, 2020 [DOI] [PubMed] [Google Scholar]
  • 2.Lievens Y, Guckenberger M, Gomez D, et al. : Defining oligometastatic disease from a radiation oncology perspective: An ESTRO-ASTRO consensus document. Radiother Oncol 148:157-166, 2020 [DOI] [PubMed] [Google Scholar]
  • 3.Palma DA, Olson R, Harrow S, et al. : Stereotactic ablative radiotherapy versus standard of care palliative treatment in patients with oligometastatic cancers (SABR-COMET): A randomised, phase 2, open-label trial. Lancet 393:2051-2058, 2019 [DOI] [PubMed] [Google Scholar]
  • 4.Parker CC, James ND, Brawley CD, et al. : Radiotherapy to the primary tumour for newly diagnosed, metastatic prostate cancer (STAMPEDE): A randomised controlled phase 3 trial. Lancet 392:2353-2366, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Gomez DR, Tang C, Zhang J, et al. : Local consolidative therapy vs. maintenance therapy or observation for patients with oligometastatic non-small-cell Lung cancer: Long-term results of a multi-institutional, phase II, randomized study. J Clin Oncol 37:1558-1565, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Moon DH, Lindsay DP, Hong S, et al. : Clinical indications for, and the future of, circulating tumor cells. Adv Drug Deliv Rev 125:143-150, 2018 [DOI] [PubMed] [Google Scholar]
  • 7.Myung JH, Eblan MJ, Caster JM, et al. : Multivalent binding and biomimetic cell rolling improves the sensitivity and specificity of circulating tumor cell capture. Clin Cancer Res 24:2539-2547, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Myung JH, Launiere CA, Eddington DT, et al. : Enhanced tumor cell isolation by a biomimetic combination of E-selectin and anti-EpCAM: Implications for the effective separation of circulating tumor cells (CTCs). Langmuir 26:8589-8596, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hong S, Lee D, Zhang H, et al. : Covalent immobilization of P-selectin enhances cell rolling. Langmuir 23:12261-12268, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.R Core Team : R: A Language and Environment for Statistical Computing. Vienna, Austria, R Foundation for Statistical Computing, 2020. https://www.R-project.org/ [Google Scholar]
  • 11.Cristofanilli M, Budd GT, Ellis MJ, et al. : Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med 351:781-791, 2004 [DOI] [PubMed] [Google Scholar]
  • 12.Cohen SJ, Punt CJA, Iannotti N, 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 26:3213-3221, 2008 [DOI] [PubMed] [Google Scholar]
  • 13.de Bono JS, Scher HI, Montgomery RB, et al. : Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin Cancer Res 14:6302-6309, 2008 [DOI] [PubMed] [Google Scholar]
  • 14.Yokobori T, Iinuma H, Shimamura T, et al. : Plastin3 is a novel marker for circulating tumor cells undergoing the epithelial–mesenchymal transition and is associated with colorectal cancer prognosis. Cancer Res 73:2059-2069, 2013 [DOI] [PubMed] [Google Scholar]
  • 15.Satelli A, Li S: Vimentin in cancer and its potential as a molecular target for cancer therapy. Cell Mol Life Sci 68:3033-3046, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Research data are stored in an institutional repository and will be shared on request to the corresponding author.


Articles from JCO Precision Oncology are provided here courtesy of American Society of Clinical Oncology

RESOURCES