Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Asian Pac J Cancer Prev ; 25(5): 1559-1566, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38809627

ABSTRACT

INTRODUCTION: Circulating tumor cells (CTCs) and Programmed death-ligand 1 (PD-L1) play pivotal roles in cancer biology and therapy response. This exploratory study aimed to elucidate the influence of neoadjuvant radiotherapy on PD-L1 expression in tumor tissues and CTCs of patients with inoperable locally advanced breast cancer. METHODS: We conducted a prospective cohort study at Universitas Andalas Hospital Padang from January to December 2022 with 27 patients. Biopsies and blood draws were executed before and after the tenth fractions of neoadjuvant radiotherapy. Following radiotherapy, CTCs were isolated using magnetic beads enrichment, followed by an RT-PCR analysis for PD-L1 expression. Correlations between PD-L1 expression and tumor response, evaluated via local response and RECIST criteria before and after radiotherapy breast CT scan, were examined using Fisher's exact and chi-square tests. RESULTS: Our data revealed no significant alterations in PD-L1 expression in either tumor tissues or CTCs during radiotherapy (p=0.848 for tissue, p=0.548 for CTCs). Notably, PD-L1 expression in tumor tissue before treatment was significantly associated with RECIST (p=0.021), while other correlations with local response and RECIST were not statistically significant. CONCLUSION: The study implies radiotherapy may not significantly influence PD-L1 expression in tumor tissue and CTCs. However, pre-treatment PD-L1 expression in tumor tissue correlates with RECIST criteria. These findings highlight the need for additional, comprehensive studies to elucidate further the interplay between PD-L1, CTCs, and radiotherapy response.


Subject(s)
B7-H1 Antigen , Biomarkers, Tumor , Breast Neoplasms , Neoplastic Cells, Circulating , Humans , B7-H1 Antigen/metabolism , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Neoplastic Cells, Circulating/radiation effects , Female , Prospective Studies , Breast Neoplasms/radiotherapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Middle Aged , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/blood , Follow-Up Studies , Prognosis , Aged , Adult , Neoadjuvant Therapy
2.
Int J Mol Sci ; 21(24)2020 Dec 16.
Article in English | MEDLINE | ID: mdl-33339353

ABSTRACT

Circulating tumor cells (CTCs) are a rare tumor cell subpopulation induced and selected by the tumor microenvironment's extreme conditions. Under hypoxia and starvation, these aggressive and invasive cells are able to invade the lymphatic and circulatory systems. Escaping from the primary tumor, CTCs enter into the bloodstream to form metastatic deposits or re-establish themselves in cancer's primary site. Although radiotherapy is widely used to cure solid malignancies, it can promote metastasis. Radiation can disrupt the primary tumor vasculature, increasing the dissemination of CTCs. Radiation also induces epithelial-mesenchymal transition (EMT) and eliminates suppressive signaling, causing the proliferation of existent, but previously dormant, disseminated tumor cells (DTCs). In this review, we collect the results and evidence underlying the molecular mechanisms of CTCs and DTCs and the effects of radiation and hypoxia in developing these cells.


Subject(s)
Neoplasms/metabolism , Neoplastic Cells, Circulating/metabolism , Tumor Hypoxia , Animals , Humans , Neoplasm Invasiveness , Neoplasms/pathology , Neoplastic Cells, Circulating/radiation effects , Radiation Tolerance
3.
Radiat Oncol ; 15(1): 137, 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-32487218

ABSTRACT

BACKGROUND: Circulating tumor cells (CTCs) are an established prognostic marker in castration-resistant prostate cancer but have received little attention in localized high-risk disease. We studied the detection rate of CTCs in patients with high-risk prostate cancer before and after androgen deprivation therapy and radiotherapy to assess its value as a prognostic and monitoring marker. PATIENTS AND METHODS: We performed a prospective analysis of CTCs in the peripheral blood of 65 treatment-naïve patients with high-risk prostate cancer. EpCAM-positive CTCs were enumerated using the CELLSEARCH system at 4 timepoints. A cut off of 0 vs ≥ 1 CTC/7.5 ml blood was defined as a threshold for negative versus positive CTCs status. RESULTS: CTCs were detected in 5/65 patients (7.5%) at diagnosis, 8/62 (12.9%) following neoadjuvant androgen deprivation and 11/59 (18.6%) at the end of radiotherapy, with a median CTC count/7.5 ml of 1 (range, 1-136). Only 1 patient presented a positive CTC result 9 months after radiotherapy. Positive CTC status (at any timepoint) was not significantly associated with any clinical or pathologic factors. However, when we analyzed variations in CTC patterns following treatment, we observed a significant association between conversion of CTCs and stages T3 (P = 0.044) and N1 (P = 0.002). Detection of CTCs was not significantly associated with overall survival (P > 0.40). CONCLUSIONS: Our study showed a low detection rate for CTCs in patients with locally advanced high-risk prostate cancer. The finding of a de novo positive CTC count after androgen deprivation therapy is probably due to a passive mechanism associated with the destruction of the tumor. Further studies with larger samples and based on more accurate detection of CTCs are needed to determine the potential prognostic and therapeutic value of this approach in non-metastatic prostate cancer. TRIAL REGISTRATION: ClinicalTrials.gov ID: NCT01800058.


Subject(s)
Biomarkers, Tumor/blood , Neoplastic Cells, Circulating/pathology , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/therapy , Aged , Androgen Antagonists/therapeutic use , Chemoradiotherapy/methods , Humans , Male , Middle Aged , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/radiation effects , Prognosis
5.
BMC Cancer ; 19(1): 816, 2019 Aug 19.
Article in English | MEDLINE | ID: mdl-31426760

ABSTRACT

BACKGROUND: Stereotactic ablative radiotherapy (SABR) has emerged as a new treatment option for patients with oligometastatic disease. SABR delivers precise, high-dose, hypofractionated radiotherapy, and achieves excellent rates of local control for primary tumors or metastases. A recent randomized phase II trial evaluated SABR in a group of patients with a small burden of oligometastatic disease (mostly with 1-3 metastatic lesions), and found that SABR was associated with benefits in progression-free survival and overall survival. The goal of this phase III trial is to assess the impact of SABR in patients with 4-10 metastatic cancer lesions. METHODS: One hundred and fifty-nine patients will be randomized in a 1:2 ratio between the control arm (consisting of standard of care palliative-intent treatments), and the SABR arm (consisting of standard of care treatment + SABR to all sites of known disease). Randomization will be stratified by two factors: histology (Group 1: prostate, breast, or renal; Group 2: all others), and type of pre-specified systemic therapy (Group 1: immunotherapy/targeted; Group 2: cytotoxic; Group 3: observation). SABR is to be completed within 2 weeks, allowing for rapid initiation of systemic therapy. Recommended SABR doses are 20 Gy in 1 fraction, 30 Gy in 3 fractions, or 35 Gy in 5 fractions, chosen to minimize risks of toxicity. The primary endpoint is overall survival, and secondary endpoints include progression-free survival, time to development of new metastatic lesions, quality of life, and toxicity. Translational endpoints include assessment of circulating tumor cells, cell-free DNA, and tumor tissue as prognostic and predictive markers, including assessment of immunological predictors of response and long-term survival. DISCUSSION: This study will provide an assessment of the impact of SABR on clinical outcomes and quality of life, to determine if long-term survival can be achieved for selected patients with 4-10 oligometastatic lesions. TRIAL REGISTRATION: Clinicaltrials.gov identifier: NCT03721341 . Date of registration: October 26, 2018.


Subject(s)
Neoplasms/diagnostic imaging , Neoplasms/radiotherapy , Neoplastic Cells, Circulating/radiation effects , Radiosurgery , Biomarkers, Tumor/blood , Dose Fractionation, Radiation , Female , Follow-Up Studies , Humans , Magnetic Resonance Imaging , Male , Neoplasms/blood , Patient Selection , Prognosis , Progression-Free Survival , Quality of Life , Surveys and Questionnaires , Tomography, X-Ray Computed , Tumor Burden
6.
Asia Pac J Clin Oncol ; 15(6): 316-322, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31309718

ABSTRACT

BACKGROUND: Both surgery and external-beam radiotherapy are effective treatments for hepatocellular carcinoma (HCC) patients with inferior vena cava/right atrium (IVC/RA) tumor thrombi. At present, it is not clear which modality is more suitable. We therefore compared outcomes between surgery and radiotherapy for these patients. METHODS: We retrospectively reviewed 108 HCC patients with IVC/RA tumor thrombi who were referred for surgery (n = 51) and external-beam radiotherapy (n = 57) at three institutions. Different surgical methods were selected according to the classification of the tumor thrombus. Radiotherapy was designed to focus on primary intrahepatic tumors and tumor thrombi. Predictors of time to progression (TTP) were identified by using univariate and multivariate analyses. RESULTS: The median TTP was significantly longer in the radiotherapy group than in the surgery group (5.0 months vs 4.2 months; P = 0.010). The multivariate analysis revealed that independent factors predicting shorter TTP were treatment with surgery (HR = 0.577; 95% CI, 0.385-0.865; P = 0.008) and intrahepatic tumor size larger than 10 cm (HR = 0.561; 95% CI, 0.342-0.919; P = 0.022). The median survival times for the radiotherapy and surgery groups were 12.8 and 14.5 months, respectively; the two groups did not have a significant difference in survival (P = 0.466). CONCLUSIONS: For HCC patients with IVC/RA tumor thrombi, treatment with external-beam radiotherapy and intrahepatic tumor size smaller than 10 cm may predict longer TTP. Compared with complicated surgery, radiotherapy, as a noninvasive treatment modality, may be more likely to be accepted.


Subject(s)
Carcinoma, Hepatocellular/secondary , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Neoplastic Cells, Circulating/pathology , Adult , Aged , Carcinoma, Hepatocellular/pathology , Female , Heart Atria/pathology , Humans , Male , Middle Aged , Multivariate Analysis , Neoplastic Cells, Circulating/radiation effects , Radiotherapy/methods , Retrospective Studies , Treatment Outcome , Vascular Surgical Procedures , Vena Cava, Inferior/pathology
7.
Cells ; 8(7)2019 06 26.
Article in English | MEDLINE | ID: mdl-31247977

ABSTRACT

Neoadjuvant chemoradiation (NCRT) followed by total mesorectal excision is the standard treatment for locally advanced rectal cancer (LARC). To justify a non-surgical approach, identification of pathologic complete response (pCR) is required. Analysis of circulating tumor cells (CTCs) can be used to evaluate pCR. We hypothesize that monitoring of thymidylate synthase (TYMS) and excision repair protein, RAD23 homolog B (RAD23B), can be used to predict resistance to chemotherapy/radiotherapy. Therefore, the aims of this study were to analyze CTCs from patients with LARC who underwent NCRT plus surgery for expression of TYMS/RAD23B and to evaluate their predictive value. Blood samples from 30 patients were collected prior to NCRT (S1) and prior to surgery (S2). CTCs were isolated and quantified by ISET®, proteins were analyzed by immunocytochemistry, and TYMS mRNA was detected by chromogenic in situ hybridization. CTC counts decreased between S1 and S2 in patients exhibiting pCR (p = 0.02) or partial response (p = 0.01). Regarding protein expression, TYMS was absent in 100% of CTCs from patients with pCR (p = 0.001) yet was expressed in 83% of non-responders at S2 (p < 0.001). Meanwhile, RAD23B was expressed in CTCs from 75% of non-responders at S1 (p = 0.01) and in 100% of non-responders at S2 (p = 0.001). Surprisingly, 100% of non-responders expressed TYMS mRNA at both timepoints (p = 0.001). In addition, TYMS/RAD23B was not detected in the CTCs of patients exhibiting pCR (p = 0.001). We found 83.3% of sensitivity for TYMS mRNA at S1 (p = 0.001) and 100% for TYMS (p = 0.064) and RAD23B (p = 0.01) protein expression at S2. Thus, TYMS mRNA and/or TYMS/RAD23B expression in CTCs, as well as CTC kinetics, have the potential to predict non-response to NCRT and avoid unnecessary radical surgery for LARC patients with pCR.


Subject(s)
Biomarkers, Tumor/metabolism , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Neoplastic Cells, Circulating/metabolism , Rectal Neoplasms/therapy , Thymidylate Synthase/metabolism , Adult , Aged , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/therapeutic use , Cell Count , Chemoradiotherapy , DNA Repair Enzymes/blood , DNA-Binding Proteins/blood , Dose Fractionation, Radiation , Drug Resistance, Neoplasm , Female , Humans , Male , Middle Aged , Neoadjuvant Therapy/methods , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/radiation effects , Preoperative Period , Proctectomy , Prognosis , Prospective Studies , Radiation Tolerance , Radiotherapy, Conformal , Rectal Neoplasms/blood , Rectal Neoplasms/pathology , Rectum/drug effects , Rectum/pathology , Rectum/radiation effects , Thymidylate Synthase/blood , Treatment Outcome
8.
Mol Biol Rep ; 46(4): 4201-4212, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31147861

ABSTRACT

Current methods, such as serum thyroglobulin measurement and medical imaging, have limitations in the routine monitoring of the disease status and treatment response of patients with differentiated thyroid cancers (DTCs), and additional methods remain to be explored. The aim of this study was to investigate the clinical value of the sodium/iodide symporter (NIS) expression and epithelial-mesenchymal transition (EMT) phenotypes of circulating tumor cells (CTCs) in monitoring the disease status and treatment response of DTC. Blood samples were obtained from DTC patients before (1 to 3 months after total thyroidectomy) and 4 to 6 months after radioactive iodine-131 (RAI) therapy for the CTC assessments. The number, NIS expression, and EMT phenotypes of CTCs were enumerated and characterized with CanPatrol™ CTC enrichment and mRNA in situ hybridization. Postoperative NIS high expression was independently correlated with a better response to first RAI therapy and good treatment efficacy. Postoperative NIS-/epithelial-/mesenchymal+ CTCs presence was independently correlated with a worse response to first RAI therapy. The numbers of total NIS+ CTCs and NIS+/epithelial+/mesenchymal+ CTCs after first RAI therapy were negatively correlated with a better response to RAI therapy only in univariate analyses. Univariate and multivariate analyses showed that a decreased or unchanged number of total NIS+ CTCs after RAI therapy may denote good efficacy and effective RAI therapy. These preliminary data suggest that assessment of the NIS expression and EMT phenotypes of CTCs may serve as potential adjuncts for predicting and monitoring the curative effect of RAI therapy in DTC patients and avoid ineffective treatment. Further validation is warranted.


Subject(s)
Iodine Radioisotopes/therapeutic use , Neoplastic Cells, Circulating/radiation effects , Symporters/genetics , Thyroid Neoplasms/genetics , Thyroid Neoplasms/radiotherapy , Adolescent , Adult , Aged , Biomarkers, Tumor/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Male , Middle Aged , Prognosis , Radiotherapy/methods , Symporters/metabolism , Thyroid Neoplasms/pathology
9.
PLoS One ; 14(5): e0216934, 2019.
Article in English | MEDLINE | ID: mdl-31136607

ABSTRACT

BACKGROUND: Radium-223 is a targeted alpha-particle therapy that improves survival in men with metastatic castration resistant prostate cancer (mCRPC), particularly in men with elevated serum levels of bone alkaline phosphatase (B-ALP). We hypothesized that osteomimicry, a form of epithelial plasticity leading to an osteoblastic phenotype, may contribute to intralesional deposition of radium-223 and subsequent irradiation of the tumor microenvironment. METHODS: We conducted a pharmacodynamic study (NCT02204943) of radium-223 in men with bone mCRPC. Prior to and three and six months after radium-223 treatment initiation, we collected CTCs and metastatic biopsies for phenotypic characterization and CTC genomic analysis. The primary objective was to describe the impact of radium-223 on the prevalence of CTC B-ALP over time. We measured radium-223 decay products in tumor and surrounding normal bone during treatment. We validated genomic findings in a separate independent study of men with bone metastatic mCRPC (n = 45) and publicly accessible data of metastatic CRPC tissues. RESULTS: We enrolled 20 men with symptomatic bone predominant mCRPC and treated with radium-223. We observed greater radium-223 radioactivity levels in metastatic bone tumor containing biopsies compared with adjacent normal bone. We found evidence of persistent Cellsearch CTCs and B-ALP (+) CTCs in the majority of men over time during radium-223 therapy despite serum B-ALP normalization. We identified genomic gains in osteoblast mimicry genes including gains of ALPL, osteopontin, SPARC, OB-cadherin and loss of RUNX2, and validated genomic alterations or increased expression at the DNA and RNA level in an independent cohort of 45 men with bone-metastatic CRPC and in 150 metastatic biopsies from men with mCRPC. CONCLUSIONS: Osteomimicry may contribute in part to the uptake of radium-223 within bone metastases and may thereby enhance the therapeutic benefit of this bone targeting radiotherapy.


Subject(s)
Biomarkers, Tumor/genetics , Bone Neoplasms/radiotherapy , Molecular Targeted Therapy/methods , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Radium/therapeutic use , Aged , Alkaline Phosphatase/blood , Alkaline Phosphatase/genetics , Biomarkers, Tumor/metabolism , Biopsy , Bone Neoplasms/genetics , Bone Neoplasms/mortality , Bone Neoplasms/secondary , Bone and Bones/metabolism , Bone and Bones/pathology , Bone and Bones/radiation effects , Cadherins/genetics , Cadherins/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Disease Progression , Gene Expression , Humans , Male , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Neoplastic Cells, Circulating/radiation effects , Osteonectin/genetics , Osteonectin/metabolism , Osteopontin/genetics , Osteopontin/metabolism , Prostate/metabolism , Prostate/pathology , Prostate/radiation effects , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/mortality , Prostatic Neoplasms, Castration-Resistant/pathology , Radium/blood , Radium/pharmacokinetics , Survival Analysis , Tumor Microenvironment/genetics , Tumor Microenvironment/radiation effects
10.
Ann Oncol ; 30(7): 1114-1120, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31020334

ABSTRACT

BACKGROUND: The clinical significance of circulating tumour cells (CTCs) in limited-stage small-cell lung cancer (LS-SCLC) is not well defined. We report a planned exploratory analysis of the prevalence and prognostic value of CTCs in LS-SCLC patients enrolled within the phase III randomised CONVERT (concurrent once-daily versus twice-daily chemoradiotherapy) trial. PATIENTS AND METHODS: Baseline blood samples were enumerated for CTCs using CellSearch in 75 patients with LS-SCLC who were enrolled in the CONVERT trial and randomised between twice- and once-daily concurrent chemoradiation. Standard statistical methods were used for correlations of CTCs with clinical factors. Log-rank test and Cox regression analyses were applied to establish the associations of 2, 15 and 50 CTC thresholds with progression-free survival (PFS) and overall survival (OS). An optimal CTC count threshold for LS-SCLC was established. RESULTS: CTCs were detected in 60% (45/75) of patients (range 0-3750). CTC count thresholds of 2, 15 and 50 CTCs all significantly correlate with PFS and OS. An optimal CTC count threshold in LS-SCLC was established at 15 CTCs, defining 'favourable' and 'unfavourable' prognostic risk groups. The median OS in <15 versus ≥15 CTCs was 26.7 versus 5.9 m (P = 0.001). The presence of ≥15 CTCs at baseline independently predicted ≤1 year survival in 70% and ≤2 years survival in 100% of patients. CONCLUSION: We report the prognostic value of baseline CTC count in an exclusive LS-SCLC population at thresholds of 2, 15 and 50 CTCs. Specific to LS-SCLC, ≥15 CTCs was associated with worse PFS and OS independent of all other factors and predicted ≤2 years survival. These results may improve disease stratification in future clinical trial designs and aid clinical decision making. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT00433563.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lung Neoplasms/blood , Lung Neoplasms/therapy , Neoplastic Cells, Circulating/pathology , Small Cell Lung Carcinoma/blood , Small Cell Lung Carcinoma/therapy , Adult , Aged , Aged, 80 and over , Chemoradiotherapy , Disease Progression , Dose Fractionation, Radiation , Female , Follow-Up Studies , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/radiation effects , Small Cell Lung Carcinoma/pathology , Survival Rate
11.
Sci Rep ; 9(1): 566, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679441

ABSTRACT

Preclinical studies demonstrated that radiation up-regulates PD-L1 expression in tumor cells, providing a rationale for combining PD-1/PD-L1 inhibitors with radiation. However this has not been validated in patients with non-small cell lung cancer due to the difficulty to obtain serial biopsies. Measuring PD-L1 expression in circulating tumor cells (CTCs), may allow real-time monitoring of immune activation in tumor. In this study, whole blood from non-metastatic NSCLC patients was collected before, during, and after radiation or chemoradiation using a microfluidic chip. PD-L1 expression in CTCs was assessed by immunofluorescence and qPCR and monitored through the course of treatment. Overall, PD-L1(+) CTCs were detected in 25 out of 38 samples (69.4%) with an average of 4.5 cells/ml. After initiation of radiation therapy, the proportion of PD-L1(+) CTCs increased significantly (median 0.7% vs. 24.7%, P < 0.01), indicating up-regulation of PD-L1 in tumor cells in response to radiation. In addition, patients positive for PD-L1 (≥5% of CTCs positive for PD-L1) at baseline had shorter PFS. Gene expression analysis revealed that higher levels of PD-L1 were associated with poor prognosis. Therefore, CTCs can be used to monitor dynamic changes of PD-L1 during radiation therapy which is potentially prognostic of response to treatment.


Subject(s)
B7-H1 Antigen/analysis , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/radiotherapy , Neoplastic Cells, Circulating/chemistry , Neoplastic Cells, Circulating/radiation effects , Carcinoma, Non-Small-Cell Lung/pathology , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Microfluidics , Prognosis , Real-Time Polymerase Chain Reaction
12.
Cytometry A ; 95(6): 664-671, 2019 06.
Article in English | MEDLINE | ID: mdl-30508273

ABSTRACT

Most cancer patients die from metastatic disease as a result of a circulating tumor cell (CTC) spreading from a primary tumor through the blood circulation to distant organs. Many studies have demonstrated the tremendous potential of using CTC counts as prognostic markers of metastatic development and therapeutic efficacy. However, it is only the viable CTCs capable of surviving in the blood circulation that can create distant metastasis. To date, little progress has been made in understanding what proportion of CTCs is viable and what proportion is in an apoptotic state. Here, we introduce a novel approach toward in situ characterization of CTC apoptosis status using a multicolor in vivo flow cytometry platform with fluorescent detection for the real-time identification and enumeration of such cells directly in blood flow. The proof of concept was demonstrated with two-color fluorescence flow cytometry (FFC) using breast cancer cells MDA-MB-231 expressing green fluorescein protein (GFP), staurosporine as an activator of apoptosis, Annexin-V apoptotic kit with orange dye color, and a mouse model. The future application of this new platform for real-time monitoring of antitumor drug efficiency is discussed. © 2018 International Society for Advancement of Cytometry.


Subject(s)
Apoptosis , Blood Vessels/diagnostic imaging , Breast Neoplasms/diagnostic imaging , Flow Cytometry/methods , Neoplastic Cells, Circulating/chemistry , Animals , Apoptosis/drug effects , Biomarkers, Tumor/blood , Blood Vessels/drug effects , Cell Count , Cell Line, Tumor , Cell Survival/drug effects , Female , Fluorescence , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , Humans , Mice , Mice, Nude , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Neoplastic Cells, Circulating/radiation effects , Staurosporine/toxicity
13.
Cytometry A ; 95(6): 657-663, 2019 06.
Article in English | MEDLINE | ID: mdl-30556307

ABSTRACT

Circulating tumor cells (CTCs) play a key role in cancer metastasis but are very difficult to detect. in vivo monitoring CTCs has been recognized as an important technique for cancer research and clinical diagnosis. Recently, a noninvasive method, in vivo flow cytometry (IVFC) has been developed to enable continuous, real-time, and long-duration detection of CTCs in animal models by detecting CTC fluorescence in blood vessels excited by lasers. In this study, we present a simple optical scheme for direct noninvasive CTC detection using confocal microscopes. We demonstrate that line scanning of confocal microscopy can provide effective and quantitative CTC detection in live mice during cancer development. Rare CTC signals can be acquired at the early stage of the tumor development after implantation of subcutaneous tumor and monitored continuously to the end. Signals from CTC clusters can also be acquired and distinguished from single CTCs. Our results suggest confocal microscopy is a simple and reliable method for biologists and doctors to use for cancer research. © 2018 International Society for Advancement of Cytometry.


Subject(s)
Blood Vessels/diagnostic imaging , Microscopy, Confocal/methods , Neoplastic Cells, Circulating/chemistry , Animals , Biomarkers, Tumor/blood , Green Fluorescent Proteins/chemistry , Humans , Injections, Subcutaneous , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Neoplastic Cells, Circulating/radiation effects , PC-3 Cells
14.
Cancer Treat Rev ; 70: 209-222, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30292979

ABSTRACT

Neuroendocrine tumours (NETs) are a heterogeneous group of neoplasms regarding their molecular biology, clinical behaviour, prognosis and response to therapy. Several attempts to establish robust predictive biomarkers have failed. Neither tissue markers nor blood borne ones have proven to be successful yet. Circulating tumour cells (CTCs) as "liquid biopsies" could provide prognostic information at the time a therapeutic decision needs to be made and could be an attractive tool for tumour monitoring throughout the treatment period. However, "liquid biopsies" are far from becoming the standard biomarker in NETs. Promising results have been presented over the last few years using a novel biomarker candidate, a multianalyte algorithm analysis PCR-based test (NETest). New technologies will open the field to different ways of approaching the biomarker conundrum in NETs. However, the complications derived from being a heterogeneous group of malignancies will remain with us forever. In summary, there is an unmet need to incorporate new biomarker candidates into clinical research trials to obtain a robust prospective validation under the most demanding scenario.


Subject(s)
Algorithms , Biomarkers, Tumor/analysis , Neoplastic Cells, Circulating/pathology , Neuroendocrine Tumors/pathology , Receptors, Peptide/chemistry , Serotonin/metabolism , Somatostatin/analogs & derivatives , Humans , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/radiation effects , Neuroendocrine Tumors/therapy , Radiopharmaceuticals/therapeutic use , Signal Transduction/drug effects , Somatostatin/therapeutic use
15.
Int J Oncol ; 53(5): 2289-2299, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30226585

ABSTRACT

Circulating epithelial tumor cells (CETCs) in peripheral blood are a prerequisite for the development of metastases. B7-H3 is an important immune checkpoint member of the B7 family and inhibits T-cell mediated antitumor immunity. Its expression is associated with a negative prognosis and a poor clinical outcome. Based on the clinical success of inhibitory immune checkpoint blockade, monoclonal antibodies (mAbs) against B7-H3 appear to be a promising therapeutic strategy. The proliferation biomarker, Ki-67, is used as a prognostic factor for breast cancer and reflects the proliferative potential of the tumor. In order to better understand the role of B7-H3 and Ki-67 in cancer development, in this study, we used a real-time biopsy for determining both biomarkers on CETCs in breast cancer patients. Blood from 50 patients suffering from breast cancer was analyzed for CETCs and the expression of B7-H3 and Ki-67 using the maintrac® method. B7-H3 expression on CETCs was found in 82% of the patients. The frequency of B7-H3- and Ki-67­positive CETCs was significantly higher in patients who had received radiation therapy compared to patients who had not received irradiation. B7-H3­positive CETCs seemed to be more aggressive as the percentage of B7-H3­positive CETCs correlated with the percentage of cells positive for the proliferation marker, Ki-67 (r=0.72 P<0.001). A significant association between the Ki-67 and B7-H3 expression level on the CETCs and nodal status was observed. On the whole, the findings of this study indicate that breast cancer patients have detectable CETCs with a high frequency of B7-H3 expression regardless of the stage of the disease. B7-H3 seems to be an important factor in immune evasion and may thus be a promising target for anticancer therapies. Radiation may lead to an upregulation of B7-H3 expression on CETCs, which could be a possible mechanism of acquired radio-resistance.


Subject(s)
B7 Antigens/blood , Breast Neoplasms/blood , Epithelial Cells/pathology , Ki-67 Antigen/blood , Neoplastic Cells, Circulating/pathology , Adult , Aged , B7 Antigens/immunology , B7 Antigens/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/immunology , Breast Neoplasms/radiotherapy , Epithelial Cells/radiation effects , Female , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Liquid Biopsy , Lymphatic Metastasis , Middle Aged , Neoplasm Staging , Neoplastic Cells, Circulating/immunology , Neoplastic Cells, Circulating/radiation effects , Prognosis , Radiation Tolerance/genetics , Tumor Escape/immunology , Tumor Escape/radiation effects , Up-Regulation
16.
Clin Genitourin Cancer ; 16(6): e1133-e1139, 2018 12.
Article in English | MEDLINE | ID: mdl-30104161

ABSTRACT

INTRODUCTION: Although increasing numbers of therapies with proven survival benefits have become available for metastatic castration-resistant prostate cancer (mCRPC), including radium-223, there is still a need for reliable biomarkers that provide information about clinically meaningful outcomes and treatment responses. MATERIALS AND METHODS: This study was a translational study that was conducted prospectively by the Spanish Oncology Genito-Urinary Group and included 45 patients with histologically confirmed mCRPC who were treated with radium-223. The primary response outcome was defined by a decline in circulating tumor cells (CTCs) of > 50% from baseline or a CTC count of ≤ 5 cells/7.5 mL at cycle 3 of radium-223. We also assessed response according to prostate-specific antigen and alkaline phosphatase levels. CTCs were evaluated as prognostic factor for treatment completion with radium-223 treatment. Kaplan-Meier estimates of survival were calculated for the global population and were correlated with biomarker response outcomes. RESULTS: Significantly, more patients with baseline CTC counts ≤ 5/7.5 mL, which are indicative of better prognoses, completed the 6 injections of therapy than those with CTC counts > 5 (16/22; 73% vs. 6/20; 30%, respectively; P = .012). The median overall survival was 16 months. Survival was significantly decreased in patients with baseline CTC counts > 5 cells/7.5 mL (7 months; P = .026) and baseline alkaline phosphatase levels ≥ 220 U/L (8 months; P = .028). CONCLUSIONS: CTCs hold significant promise as a prognostic factor for survival and completing treatment prior to the initiation of bone-targeted radium-223 therapy. These findings may help to guide the use of radium-223 in patients with mCRPC.


Subject(s)
Bone Neoplasms/radiotherapy , Neoplastic Cells, Circulating/radiation effects , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Radium/administration & dosage , Aged , Aged, 80 and over , Bone Neoplasms/blood , Bone Neoplasms/mortality , Bone Neoplasms/secondary , Cell Count , Disease Progression , Follow-Up Studies , Humans , Injections, Intravenous , Male , Prognosis , Progression-Free Survival , Prospective Studies , Prostate-Specific Antigen/blood , Prostatic Neoplasms, Castration-Resistant/blood , Prostatic Neoplasms, Castration-Resistant/mortality , Prostatic Neoplasms, Castration-Resistant/pathology , Radiotherapy Dosage
17.
Cancer Res ; 78(15): 4241-4252, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29880480

ABSTRACT

Although radiotherapy (RT) decreases the incidence of locoregional recurrence in breast cancer, patients with triple-negative breast cancer (TNBC) have increased risk of local recurrence following breast-conserving therapy. The relationship between RT and local recurrence is unknown. Here, we tested the hypothesis that recurrence in some instances is due to the attraction of circulating tumor cells to irradiated tissues. To evaluate the effect of absolute lymphocyte count on local recurrence after RT in patients with TNBC, we analyzed radiation effects on tumor and immune cell recruitment to tissues in an orthotopic breast cancer model. Recurrent patients exhibited a prolonged low absolute lymphocyte count when compared with nonrecurrent patients following RT. Recruitment of tumor cells to irradiated normal tissues was enhanced in the absence of CD8+ T cells. Macrophages (CD11b+F480+) preceded tumor cell infiltration and were recruited to tissues following RT. Tumor cell recruitment was mitigated by inhibiting macrophage infiltration using maraviroc, an FDA-approved CCR5 receptor antagonist. Our work poses the intriguing possibility that excessive macrophage infiltration in the absence of lymphocytes promotes local recurrence after RT. This combination thus defines a high-risk group of patients with TNBC.Significance: This study establishes the importance of macrophages in driving tumor cell recruitment to sites of local radiation therapy and suggests that this mechanism contributes to local recurrence in women with TNBC that are also immunosuppressed.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/15/4241/F1.large.jpg Cancer Res; 78(15); 4241-52. ©2018 AACR.


Subject(s)
Macrophages/pathology , Neoplasm Recurrence, Local/pathology , Neoplastic Cells, Circulating/pathology , Animals , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/radiation effects , Cell Line, Tumor , Cell Movement/physiology , Cell Movement/radiation effects , Female , Humans , Macrophages/metabolism , Macrophages/radiation effects , Mastectomy/methods , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Recurrence, Local/radiotherapy , Neoplastic Cells, Circulating/radiation effects , Receptors, CCR5/metabolism , Retrospective Studies , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/radiotherapy
18.
JAMA Oncol ; 4(8): e180163, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29800954

ABSTRACT

Importance: Circulating tumor cells (CTCs) represent the liquid component of solid tumors and are a surrogate marker for residual cancer burden. Although CTC status is prognostic of recurrence and death in breast cancer, its role in guiding clinical management remains unknown. Objective: To determine whether CTC status is predictive of radiotherapeutic benefit in early-stage breast cancer. Design, Setting, and Participants: The cohort studies in the present analysis included patients with stages pT1 to pT2 and pN0 to pN1 breast cancer and known CTC status from the National Cancer Database (NCDB) and the multicenter phase 3 SUCCESS clinical trial. Multivariable parametric accelerated failure time models were used to evaluate the association of CTC status and radiotherapy (RT) with survival outcomes. Data were collected from January 1, 2004, through December 31, 2014, from the NCDB cohort. The SUCCESS trial collected data from September 1, 2005, through September 30, 2013. The analyses were completed from November 1, 2016, through December 17, 2017. Exposure: Adjuvant RT. Main Outcomes and Measures: Overall survival (OS), local recurrence-free survival (LRFS), and disease-free survival (DFS). Results: A total of 1697 patients from the NCDB (16 men [0.9%] and 1681 women [99.1%]; median age, 63 years; interquartile range, 53-71 years) and 1516 patients from the SUCCESS clinical trial (median age, 52 years; interquartile range, 45-60 years) were identified. Circulating tumor cells were detected in 399 patients (23.5%) in the NCDB cohort and 294 (19.4%) in the SUCCESS cohort. The association of RT with survival was dependent on CTC status within the NCDB cohort (4-year OS, 94.9% for CTC-positive RT vs 88.0% for CTC-positive non-RT vs 93.9% for CTC-negative RT vs 93.4% for CTC-negative non-RT groups; P < .001) and 5-year DFS within the SUCCESS cohort (88.0% for CTC-positive RT vs 75.2% for CTC-positive non-RT vs 92.3% for CTC-negative RT vs 88.3% for CTC-negative non-RT; P = .04). In the NCDB cohort, RT was associated with longer OS in patients with CTCs (time ratio [TR], 2.04; 95% CI, 1.55-2.67; P < .001), but not in patients without CTCs (TR, 0.80; 95% CI, 0.52-1.25; P = .33). In the SUCCESS cohort, CTC-positive patients treated with RT exhibited longer LRFS (TR, 2.73; 95% CI, 1.62-4.80; P < .001), DFS (TR, 3.03; 95% CI, 2.22-4.13; P < .001), and OS (TR, 1.83; 95% CI, 1.23-2.72; P = .003). Among patients from both cohorts who underwent breast-conserving surgery, RT was associated with longer OS in patients with CTCs (TR, 4.37; 95% CI, 2.71-7.05; P < .001) but not in patients without CTCs (TR, 0.87; 95% CI, 0.47-1.62; P = .77). Radiotherapy was not associated with OS after mastectomy in CTC-positive or CTC-negative patients. Conclusions and Relevance: Treatment with RT was associated with longer LRFS, DFS, and OS in patients with early-stage breast cancer and detectable CTCs. These results are hypothesis generating; a prospective trial evaluating CTC-based management for RT after breast-conserving surgery in women with early-stage breast cancer is warranted.


Subject(s)
Breast Neoplasms/mortality , Carcinoma, Ductal, Breast/mortality , Carcinoma, Lobular/mortality , Neoplasm Recurrence, Local/mortality , Neoplastic Cells, Circulating/pathology , Radiotherapy, Adjuvant/mortality , Aged , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/radiotherapy , Carcinoma, Lobular/pathology , Carcinoma, Lobular/radiotherapy , Female , Follow-Up Studies , Humans , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/radiotherapy , Neoplasm Staging , Neoplastic Cells, Circulating/radiation effects , Prospective Studies , Survival Rate
19.
Vasc Endovascular Surg ; 52(6): 463-468, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29695214

ABSTRACT

Cancer-associated vein obstruction (CAVO) is a common complication in oncological patients, but the effective therapeutic options are scant. We report a patient with reiterative recurrent CAVO who was successfully treated with intraluminal brachytherapy using a self-expandable stent loaded with linear radioactive iodine-125 seeds (RIS) strand. During follow-up, her clinical symptoms were well improved. Three-month imaging follow-up revealed satisfactory patency of the iliofemoral vein, and the stents combined with RIS strands performed well. No serious complications associated with the implantation of stent and RIS strands were documented in any of the sessions. Intraluminal brachytherapy using a self-expandable stent loaded with linear RIS may be a safe and effective option for CAVO as long as it includes not only blood flow restoration but also brachytherapy administration for cancer.


Subject(s)
Brachytherapy/instrumentation , Endovascular Procedures/instrumentation , Femoral Vein/radiation effects , Iliac Vein/radiation effects , Iodine Radioisotopes/administration & dosage , Neoplastic Cells, Circulating/radiation effects , Rectal Neoplasms/radiotherapy , Stents , Vascular Diseases/radiotherapy , Aged , Computed Tomography Angiography , Constriction, Pathologic , Female , Femoral Vein/diagnostic imaging , Femoral Vein/pathology , Humans , Iliac Vein/diagnostic imaging , Iliac Vein/pathology , Neoplastic Cells, Circulating/pathology , Phlebography/methods , Radiography, Interventional , Rectal Neoplasms/complications , Rectal Neoplasms/pathology , Recurrence , Treatment Outcome , Vascular Diseases/diagnostic imaging , Vascular Diseases/etiology , Vascular Diseases/pathology , Vascular Patency
20.
Int J Radiat Oncol Biol Phys ; 100(4): 1044-1056, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29485046

ABSTRACT

PURPOSE: To investigated the influence of radiation therapy (RT), surgery (OP), radio-chemotherapy (RChT), or chemotherapy (ChT) on small cell lung cancer metastases in 2 xenograft models. METHODS AND MATERIALS: A total of 1 × 106 human small cell lung cancer cells (OH1, H69) were subcutaneously injected into severe combined immunodeficiency mice to form a local primary tumor node at the lower trunk. Radiation therapy, OP, RChT, or ChT were started after development of palpable tumors. Chemotherapy was given as a single intraperitoneal injection of cisplatin. Radiation therapy was 5 × 10 Gy on the local tumor node. Two additional groups were implemented to assess primary tumors and distant metastases in untreated mice at the beginning (control group A) and at the end of the experiment (control group B). Proapoptotic, antiproliferative, antiangiogenic, and hypoxic effects were assessed by Feulgen, Ki67, S1P1 receptor, and hypoxia-inducible factor 1α staining, respectively. Quantitative Alu-polymerase chain reaction was used to determine circulating tumor cells in the blood, and disseminated tumor cells in the lungs, bone marrow, liver, and brain. RESULTS: In both xenograft models, RT and RChT abrogated local tumor growth, indicated by increased apoptosis, decreased cell proliferation, and reduced microvessel density (equally affecting vessels of all diameters). Regarding metastases, RT and RChT not only counteracted the time-dependent increase of dissemination but also decreased the metastatic load pre-existing at therapy induction in the blood, lungs, and liver. Only in the case of relapse-free surgery could similar effects be achieved by OP. CONCLUSIONS: Our models provide evidence that RT and RChT ablate the primary tumor and inhibit metastasis development over time. Upon local recurrence, RT showed beneficial effects compared with OP with regard to suppression of circulating tumor cells and disseminated tumor cells.


Subject(s)
Bone Marrow Neoplasms/prevention & control , Brain Neoplasms/prevention & control , Chemoradiotherapy , Liver Neoplasms/prevention & control , Lung Neoplasms/therapy , Small Cell Lung Carcinoma/secondary , Small Cell Lung Carcinoma/therapy , Animals , Antineoplastic Agents/therapeutic use , Apoptosis , Bone Marrow Neoplasms/secondary , Brain Neoplasms/secondary , Cell Line, Tumor , Cell Proliferation , Cisplatin/therapeutic use , Heterografts , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Ki-67 Antigen/analysis , Liver Neoplasms/secondary , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Mice , Mice, SCID , Microvessels/pathology , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/radiation effects , Radiotherapy Dosage , Receptors, Lysosphingolipid/analysis , Small Cell Lung Carcinoma/pathology , Small Cell Lung Carcinoma/radiotherapy , Tumor Burden/drug effects , Tumor Burden/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...