Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Oncologist ; 21(1): 84-94, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26614709

ABSTRACT

UNLABELLED: : Circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and messenger RNA (mRNA), collectively termed circulating tumor products (CTPs), represent areas of immense interest from scientists' and clinicians' perspectives. In melanoma, CTP analysis may have clinical utility in many areas, from screening and diagnosis to clinical decision-making aids, as surveillance biomarkers or sources of real-time genetic or molecular characterization. In addition, CTP analysis can be useful in the discovery of new biomarkers, patterns of treatment resistance, and mechanisms of metastasis development. Here, we compare and contrast CTCs, ctDNA, and mRNA, review the extent of translational evidence to date, and discuss how future studies involving both scientists and clinicians can help to further develop this tool for the benefit of melanoma patients. IMPLICATIONS FOR PRACTICE: Scientific advancement has enabled the rapid development of tools to analyze circulating tumor cells, tumor DNA, and messenger RNA, collectively termed circulating tumor products (CTPs). A variety of techniques have emerged to detect and characterize melanoma CTPs; however, only a fraction has been applied to human subjects. This review summarizes the available human data that investigate clinical utility of CTP in cancer screening, melanoma diagnosis, prognosis, prediction, and genetic or molecular characterization. It provides a rationale for how CTPs may be useful for future research and discusses how clinicians can be involved in developing this exciting new technology.


Subject(s)
Biomarkers, Tumor/blood , DNA, Neoplasm/blood , Melanoma/blood , RNA, Messenger/blood , Humans , Melanoma/genetics , Melanoma/pathology , Neoplastic Cells, Circulating/pathology , Prognosis
2.
Cancer ; 121(1): 139-49, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25241991

ABSTRACT

BACKGROUND: Assays identifying circulating tumor cells (CTCs) allow noninvasive and sequential monitoring of the status of primary or metastatic tumors, potentially yielding clinically useful information. However, to the authors' knowledge, the effect of radiation therapy (RT) on CTCs in patients with non-small cell lung cancer (NSCLC) has not been previously explored. METHODS: This report describes results from a pilot study of 30 patients with NSCLC who received RT. Peripheral blood samples obtained from these patients were assayed for CTCs using an assay that identified live cells using an adenoviral probe that detected the elevated telomerase activity present in almost all cancer cells, but not in normal cells, and the validity of the assay was confirmed with secondary tumor-specific markers. Patients were assayed before initiation of RT (pre-RT), during the RT course, and/or after the completion of RT (post-RT). RESULTS: The assay successfully detected CTCs in the majority of patients, including 65% of patients before the start of RT, and in patients with both epidermal growth factor receptor wild-type and mutation-positive tumors. The median CTC counts in patients before RT was 9.1 CTCs per mL (range, undetectable to 571 CTCs per mL) and was significantly higher than the average post-RT count of 0.6 CTCs per mL (range, undetectable to 1.8 CTCs per mL; P<.001). Sequential CTC counts were available in a subset of patients and demonstrated decreases after RT, except for 1 patient who subsequently developed distant failure. CONCLUSIONS: The current pilot data suggest that CTC counts appear to reflect response to RT in patients with localized NSCLC. On the basis of these promising results, the authors have launched a more comprehensive and detailed clinical trial.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnosis , Lung Neoplasms/diagnosis , Neoplastic Cells, Circulating/metabolism , Telomerase/metabolism , Aged , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Line, Tumor , Female , Humans , Lung Neoplasms/blood , Lung Neoplasms/radiotherapy , Male , Middle Aged , Pilot Projects , Telomerase/blood , Treatment Outcome
3.
Small ; 11(7): 834-43, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25264301

ABSTRACT

Gold nanoparticles have garnered interest as both radiosensitzers and computed tomography (CT) contrast agents. However, the extremely high concentrations of gold required to generate CT contrast is far beyond that needed for meaningful radiosensitization, which limits their use as combined therapeutic-diagnostic (theranostic) agents. To establish a theranostic nanoplatform with well-aligned radiotherapeutic and diagnostic properties for better integration into standard radiation therapy practice, a gold- and superparamagnetic iron oxide nanoparticle (SPION)-loaded micelle (GSM) is developed. Intravenous injection of GSMs into tumor-bearing mice led to selective tumoral accumulation, enabling magnetic resonance (MR) imaging of tumor margins. Subsequent irradiation leads to a 90-day survival of 71% in GSM-treated mice, compared with 25% for irradiation-only mice. Furthermore, measurements of the GSM-enhanced MR contrast are highly predictive of tumor response. Therefore, GSMs may not only guide and enhance the efficacy of radiation therapy, but may allow patients to be managed more effectively.


Subject(s)
Diagnostic Imaging , Nanoparticles/chemistry , Radiotherapy , Animals , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , Dextrans/pharmacokinetics , Dextrans/pharmacology , Female , Gold/pharmacokinetics , Gold/pharmacology , Humans , Kaplan-Meier Estimate , Magnetite Nanoparticles , Mice, Nude , Micelles , Polymers/chemistry , Radiation-Sensitizing Agents/pharmacology , Tissue Distribution/drug effects , Treatment Outcome
4.
Radiother Oncol ; 190: 110030, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38008414

ABSTRACT

PURPOSE: To explore the association of the effective dose to immune cells (EDIC) with disease control, lymphopenia, and toxicity in patients with non-small cell lung cancer (NSCLC) and identify methods to reduce EDIC. METHODS: We abstracted data from all patients with locally advanced NSCLC treated with chemoradiation with or without consolidative immunotherapy over a ten-year period. Associations between EDIC and progression-free survival (PFS) and overall survival (OS) were modeled with Cox proportional hazards and Kaplan-Meier method. Logistic regression was used to model predictors of lymphopenia and higher EDIC. Analyses were performed with EDIC as a continuous and categorical variable. Lymphopenia was graded per CTCAE v5.0. RESULTS: Overall, 786 patients were included (228 of which received consolidative immunotherapy); median EDIC was 4.7 Gy. Patients with EDIC < 4.7 Gy had a longer median PFS (15.3 vs. 9.0 months; p < 0.001) and OS (34.2 vs. 22.4 months; p < 0.001). On multivariable modeling, EDIC correlated with inferior PFS (HR 1.08, 95 % CI 1.01-1.14, p = 0.014) and OS (HR 1.10, 95 % CI 1.04-1.18, p = 0.002). EDIC was predictive of grade 4 lymphopenia (OR 1.16, 95 % CI 1.02-1.33, p = 0.026). EDIC ≥ 4.7 Gy was associated with increased grade 2 + pneumonitis (6-month incidence: 26 % vs 20 %, p = 0.04) and unplanned hospitalizations (90-day incidence: 40 % vs 30 %, p = 0.002). Compared to protons, photon therapy was associated with EDIC ≥ 4.7 Gy (OR 5.26, 95 % CI 3.71-7.69, p < 0.001) in multivariable modeling. CONCLUSIONS: EDIC is associated with inferior disease outcomes, treatment-related toxicity, and the development of severe lymphopenia. Proton therapy is associated with lower EDIC. Further investigations to limit radiation dose to the immune system appear warranted.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Lymphopenia , Humans , Lymphopenia/etiology , Chemoradiotherapy/adverse effects , Chemoradiotherapy/methods , Radiation Dosage
5.
ScientificWorldJournal ; 2013: 796029, 2013.
Article in English | MEDLINE | ID: mdl-23690748

ABSTRACT

Introduction. PET imaging is a useful clinical tool for studying tumor progression and treatment effects. Conventional (18)F-FDG-PET imaging is of limited usefulness for imaging Glioblastoma Multiforme (GBM) due to high levels of glucose uptake by normal brain and the resultant signal-to-noise intensity. (18)F-Fluorothymidine (FLT) in contrast has shown promise for imaging GBM, as thymidine is taken up preferentially by proliferating cells. These studies were undertaken to investigate the effectiveness of (18)F-FLT-PET in a GBM mouse model, especially after radiation therapy (RT), and its correlation with useful biomarkers, including proliferation and DNA damage. Methods. Nude/athymic mice with human GBM orthografts were assessed by microPET imaging with (18)F-FDG and (18)F-FLT. Patterns of tumor PET imaging were then compared to immunohistochemistry and immunofluorescence for markers of proliferation (Ki-67), DNA damage and repair (γH2AX), hypoxia (HIF-1α), and angiogenesis (VEGF). Results. We confirmed that (18)F-FLT-PET uptake is limited in healthy mice but enhanced in the intracranial tumors. Our data further demonstrate that (18)F-FLT-PET imaging usefully reflects the inhibition of tumor by RT and correlates with changes in biomarker expression. Conclusions. (18)F-FLT-PET imaging is a promising tumor imaging modality for GBM, including assessing RT effects and biologically relevant biomarkers.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/radiotherapy , Brain/metabolism , Brain/radiation effects , Glioblastoma/metabolism , Glioblastoma/radiotherapy , Radiotherapy, Conformal/methods , Animals , Brain/diagnostic imaging , Brain Neoplasms/diagnostic imaging , Cell Line, Tumor , Dideoxynucleosides/pharmacokinetics , Female , Glioblastoma/diagnostic imaging , Humans , Metabolic Clearance Rate/radiation effects , Mice , Mice, Nude , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Radiotherapy Dosage , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution/radiation effects
6.
Clin Transl Radiat Oncol ; 39: 100581, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36691564

ABSTRACT

Background and purpose: Prior studies have examined associations of cardiovascular substructure dose with overall survival (OS) or cardiac events after chemoradiotherapy (CRT) for non-small cell lung cancer (NSCLC). Herein, we investigate an alternative endpoint, death without cancer progression (DWP), which is potentially more specific than OS and more sensitive than cardiac events for understanding CRT toxicity. Materials and methods: We retrospectively reviewed records of 187 patients with locally advanced or oligometastatic NSCLC treated with definitive CRT from 2008 to 2016 at a single institution. Dosimetric parameters to the heart, lung, and ten cardiovascular substructures were extracted. Charlson Comorbidity Index (CCI), excluding NSCLC diagnosis, was used to stratify patients into CCI low (0-2; n = 66), CCI intermediate (3-4; n = 78), and CCI high (≥5; n = 43) groups. Primary endpoint was DWP, modeled with competing risk regression. Secondary endpoints included OS. An external cohort consisted of 140 patients from another institution. Results: Median follow-up was 7.3 years for survivors. Death occurred in 143 patients (76.5 %), including death after progression in 118 (63.1 %) and DWP in 25 (13.4 %). On multivariable analysis, increasing CCI stratum and mean heart dose were associated with DWP. For mean heart dose ≥ 10 Gy vs < 10 Gy, DWP was higher (5-year rate, 16.9 % vs 6.7 %, p = 0.04) and OS worse (median, 22.9 vs 34.1 months, p < 0.001). Ventricle (left, right, and bilateral) and pericardial but not atrial substructure dose were associated with DWP, whereas all three were inversely associated with OS. Cutpoint analysis identified right ventricle mean dose ≥ 5.5 Gy as a predictor of DWP. In the external cohort, we confirmed an association of ventricle, but not atrial, dose with DWP. Conclusion: Cardiovascular substructure dose showed distinct associations with DWP. Future cardiotoxicity studies in NSCLC could consider DWP as an endpoint.

7.
J Cell Biol ; 177(3): 413-24, 2007 May 07.
Article in English | MEDLINE | ID: mdl-17485487

ABSTRACT

hSgo2 (previously annotated as Tripin) was recently reported to be a new inner centromere protein that is essential for centromere cohesion (Kitajima et al., 2006). In this study, we show that hSgo2 exhibits a dynamic distribution pattern, and that its localization depends on the BUB1 and Aurora B kinases. hSgo2 is concentrated at the inner centromere of unattached kinetochores, but extends toward the kinetochores that are under tension. This localization pattern is reminiscent of MCAK, which is a microtubule depolymerase that is believed to be a key component of the error correction mechanism at kinetochores. Indeed, we found that hSgo2 is essential for MCAK to localize to the centromere. Delocalization of MCAK accounts for why cells depleted of hSgo2 exhibit kinetochore attachment defects that go uncorrected, despite a transient delay in the onset of anaphase. Consequently, these cells exhibit a high frequency of lagging chromosomes when they enter anaphase. We confirmed that hSgo2 is associated with PP2A, and we propose that it contributes to the spatial regulation of MCAK activity within inner centromere and kinetochore.


Subject(s)
Anaphase/physiology , Cell Cycle Proteins/metabolism , Kinesins/metabolism , Kinetochores/metabolism , Aurora Kinase B , Aurora Kinases , HeLa Cells , Humans , Kinetochores/ultrastructure , Phosphoprotein Phosphatases/metabolism , Protein Binding/physiology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Transport/physiology
8.
Int J Radiat Oncol Biol Phys ; 112(4): 1045-1054, 2022 03 15.
Article in English | MEDLINE | ID: mdl-34775000

ABSTRACT

PURPOSE: We develop a deep learning (DL) radiomics model and integrate it with circulating tumor cell (CTC) counts as a clinically useful prognostic marker for predicting recurrence outcomes of early-stage (ES) non-small cell lung cancer (NSCLC) patients treated with stereotactic body radiation therapy (SBRT). METHODS AND MATERIALS: A cohort of 421 NSCLC patients was used to train a DL model for gleaning informative imaging features from computed tomography (CT) data. The learned imaging features were optimized on a cohort of 98 ES-NSCLC patients treated with SBRT for predicting individual patient recurrence risks by building DL models on CT data and clinical measures. These DL models were validated on the third cohort of 60 ES-NSCLC patients treated with SBRT to predict recurrent risks and stratify patients into subgroups with distinct outcomes in conjunction with CTC counts. RESULTS: The DL model obtained a concordance-index of 0.880 (95% confidence interval, 0.879-0.881). Patient subgroups with low and high DL risk scores had significantly different recurrence outcomes (P = 3.5e-04). The integration of DL risk scores and CTC measures identified 4 subgroups of patients with significantly different risks of recurrence (χ2 = 20.11, P = 1.6e-04). Patients with positive CTC measures were associated with increased risks of recurrence that were significantly different from patients with negative CTC measures (P = 0.0447). CONCLUSIONS: In this first-ever study integrating DL radiomics models and CTC counts, our results suggested that this integration improves patient stratification compared with either imagining data or CTC measures alone in predicting recurrence outcomes for patients treated with SBRT for ES-NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Deep Learning , Lung Neoplasms , Neoplastic Cells, Circulating , Radiosurgery , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/radiotherapy , Humans , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Radiosurgery/methods
9.
Semin Radiat Oncol ; 31(2): 149-154, 2021 04.
Article in English | MEDLINE | ID: mdl-33610272

ABSTRACT

Recent treatment advances have improved outcomes for patients with non-small cell lung cancer (NSCLC), often utilizing tumor molecular characterization to identify targetable mutations. This is further enhanced by advancements in "liquid biopsies", using peripheral blood for noninvasive, serial sampling of tumor biology. While tumor genomic alterations have established therapeutic implications in metastatic NSCLC, research is also ongoing to develop applications for tissue and liquid biomarkers in earlier stage disease, such as patients treated with radiation for early stage or locoregional NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/radiotherapy , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Mutation , Prognosis
10.
Int J Radiat Oncol Biol Phys ; 109(5): 1647-1656, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33333202

ABSTRACT

PURPOSE: To predict overall survival of patients receiving stereotactic body radiation therapy (SBRT) for early-stage non-small cell lung cancer (ES-NSCLC), we developed a radiomic model that integrates risk of death estimates and changes based on pre- and posttreatment computed tomography (CT) scans. We hypothesize this innovation will improve our ability to stratify patients into various oncologic outcomes with greater accuracy. METHODS AND MATERIALS: Two cohorts of patients with ES-NSCLC uniformly treated with SBRT (a median dose of 50 Gy in 4-5 fractions) were studied. Prediction models were built on a discovery cohort of 100 patients with treatment planning CT scans, and then were applied to a separate validation cohort of 60 patients with pre- and posttreatment CT scans for evaluating their performance. RESULTS: Prediction models achieved a c-index up to 0.734 in predicting survival outcomes of the validation cohort. The integration of the pretreatment risk of survival measures (risk-high vs risk-low) and changes (risk-increase vs risk-decrease) in risk of survival measures between the pretreatment and posttreatment scans further stratified the patients into 4 subgroups (risk: high, increase; risk: high, decrease; risk: low, increase; risk: low, decrease) with significant difference (χ2 = 18.549, P = .0003, log-rank test). There was also a significant difference between the risk-increase and risk-decrease groups (χ2 = 6.80, P = .0091, log-rank test). In addition, a significant difference (χ2 = 7.493, P = .0062, log-rank test) was observed between the risk-high and risk-low groups obtained based on the pretreatment risk of survival measures. CONCLUSION: The integration of risk of survival measures estimated from pre- and posttreatment CT scans can help differentiate patients with good expected survival from those who will do more poorly following SBRT. The analysis of these radiomics-based longitudinal risk measures may help identify patients with early-stage NSCLC who will benefit from adjuvant treatment after lung SBRT, such as immunotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/mortality , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/mortality , Radiosurgery/methods , Tomography, X-Ray Computed , Aged , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cohort Studies , Dose Fractionation, Radiation , Female , Forecasting/methods , Humans , Kaplan-Meier Estimate , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Male , Models, Theoretical , Postoperative Care , Preoperative Care , Prognosis , Radiosurgery/mortality , Treatment Outcome
11.
Int J Radiat Oncol Biol Phys ; 110(5): 1451-1465, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33662459

ABSTRACT

PURPOSE: The main objective of the present study was to integrate 18F-FDG-PET/CT radiomics with multiblock discriminant analysis for predicting circulating tumor cells (CTCs) in early-stage non-small cell lung cancer (ES-NSCLC) treated with stereotactic body radiation therapy (SBRT). METHODS: Fifty-six patients with stage I NSCLC treated with SBRT underwent 18F-FDG-PET/CT imaging pre-SBRT and post-SBRT (median, 5 months; range, 3-10 months). CTCs were assessed via a telomerase-based assay before and within 3 months after SBRT and dichotomized at 5 and 1.3 CTCs/mL. Pre-SBRT, post-SBRT, and delta PET/CT radiomics features (n = 1548 × 3/1562 × 3) were extracted from gross tumor volume. Seven feature blocks were constructed including clinical parameters (n = 12). Multiblock data integration was performed using block sparse partial least squares-discriminant analysis (sPLS-DA) referred to as Data Integration Analysis for Biomarker Discovery Using Latent Components (DIABLO) for identifying key signatures by maximizing common information between different feature blocks while discriminating CTC levels. Optimal input blocks were identified using a pairwise combination method. DIABLO performance for predicting pre-SBRT and post-SBRT CTCs was evaluated using combined AUC (area under the curve, averaged across different blocks) analysis with 20 × 5-fold cross-validation (CV) and compared with that of concatenation-based sPLS-DA that consisted of combining all features into 1 block. CV prediction scores between 1 class versus the other were compared using the Wilcoxon rank sum test. RESULTS: For predicting pre-SBRT CTCs, DIABLO achieved the best performance with combined pre-SBRT PET radiomics and clinical feature blocks, showing CV AUC of 0.875 (P = .009). For predicting post-SBRT CTCs, DIABLO achieved the best performance with combined post-SBRT CT and delta CT radiomics feature blocks, showing CV AUCs of 0.883 (P = .001). In contrast, all single-block sPLS-DA models could not attain CV AUCs higher than 0.7. CONCLUSIONS: Multiblock integration with discriminant analysis of 18F-FDG-PET/CT radiomics has the potential for predicting pre-SBRT and post-SBRT CTCs. Radiomics and CTC analysis may complement and together help guide the subsequent management of patients with ES-NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/radiotherapy , Lung Neoplasms/blood , Lung Neoplasms/radiotherapy , Neoplastic Cells, Circulating , Positron Emission Tomography Computed Tomography , Aged , Aged, 80 and over , Area Under Curve , Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung/pathology , Discriminant Analysis , Female , Fluorodeoxyglucose F18 , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Prospective Studies , Radiopharmaceuticals , Statistics, Nonparametric , Tumor Burden
12.
Virol J ; 7: 237, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20846395

ABSTRACT

HIV-1 integration is mediated by the HIV-1 integrase protein, which joins 3'-ends of viral DNA to host cell DNA. To complete the integration process, HIV-1 DNA has to be joined to host cell DNA also at the 5'-ends. This process is called post-integration repair (PIR). Integration and PIR involve a number of cellular co-factors. These proteins exhibit different degrees of involvement in integration and/or PIR. Some are required for efficient integration or PIR. On the other hand, some reduce the efficiency of integration. Finally, some are involved in integration site selection. We have studied the role of the histone deacetylase HDAC4 in these processes. HDAC4 was demonstrated to play a role in both cellular double-strand DNA break repair and transcriptional regulation. We observed that HDAC4 associates with viral DNA in an integrase-dependent manner. Moreover, infection with HIV-1-based vectors induces foci of the HDAC4 protein. The related histone deacetylases, HDAC2 and HDAC6, failed to associate with viral DNA after infection. These data suggest that HDAC4 accumulates at integration sites. Finally, overexpression studies with HDAC4 mutants suggest that HDAC4 may be required for efficient transduction by HIV-1-based vectors in cells that are deficient in other DNA repair proteins. We conclude that HDAC4 is likely involved in PIR.


Subject(s)
HIV Integrase/metabolism , HIV-1/physiology , Histone Deacetylases/metabolism , Host-Pathogen Interactions , Repressor Proteins/metabolism , Virus Integration , DNA, Viral/metabolism , Genetic Vectors , HIV-1/genetics , HeLa Cells , Histone Deacetylase 2/metabolism , Histone Deacetylase 6 , Humans , Protein Binding , Transduction, Genetic
13.
J Support Oncol ; 8(1): 28-34, 2010.
Article in English | MEDLINE | ID: mdl-20235421

ABSTRACT

Cancer patients are at increased risk for potentially life-threatening infections. Patient safety goals recently issued by the Joint Commission on the Accreditation of Healthcare Organizations (JCAHO) and current Centers for Disease Control and Prevention (CDC) guidelines recommend vaccinations for all cancer patients over the age of 65 (for Pneumococcus) and 50 years of age (annually, for Influenza). The authors investigated vaccination practices in patients over a season of risk at a university-based outpatient cancer treatment clinic. Of 204 patients recruited, 196 (93%) completed the survey. Overall, 30% of patients reported never receiving the Influenza vaccine (33% of patients >50 years old), and 56% reported never receiving the Pneumococcal vaccine (30% of patients >65 years old). Only 7% of patients reported being asked or informed about vaccination by their oncologists. Substantial proportions of patients undergoing cancer treatment have not received vaccinations as recommended by national guidelines. The reasons cited for lack of compliance seem correctable, and doing so would potentially prevent mortality and morbidity, thereby improving the care of cancer patients. Recommended vaccinations may now include that for the Influenza A virus (H1N1).


Subject(s)
Influenza, Human/prevention & control , Neoplasms/prevention & control , Neoplasms/radiotherapy , Patient Compliance , Pneumonia, Pneumococcal/prevention & control , Vaccination/standards , Adult , Aged , Aged, 80 and over , Attitude of Health Personnel , Female , Guidelines as Topic , Health Surveys , Humans , Influenza Vaccines/administration & dosage , Male , Middle Aged , Pneumococcal Vaccines/administration & dosage , Young Adult
14.
Clin Cancer Res ; 26(10): 2372-2380, 2020 05 15.
Article in English | MEDLINE | ID: mdl-31969332

ABSTRACT

PURPOSE: Although stereotactic body radiotherapy (SBRT) is effective in early-stage non-small cell lung cancer (NSCLC), approximately 10%-15% of patients will fail regionally and 20%-25% distantly. We evaluate a novel circulating tumor cell (CTC) assay as a prognostic marker for increased risk of recurrence following SBRT. EXPERIMENTAL DESIGN: Ninety-two subjects (median age, 71 years) with T1a (64%), T1b (23%), or T2a (13%) stage I NSCLC treated with SBRT were prospectively enrolled. CTCs were enumerated by utilizing a GFP-expressing adenoviral probe that detects elevated telomerase activity in cancer cells. Samples were obtained before, during, and serially up to 24 months after treatment. SBRT was delivered to a median dose of 50 Gy (range, 40-60 Gy), mostly commonly in four to five fractions (92%). RESULTS: Thirty-eight of 92 subjects (41%) had a positive CTC test prior to SBRT. A cutoff of ≥5 CTCs/mL before treatment defined favorable (n = 78) and unfavorable (n = 14) prognostic groups. Increased risk of nodal (P = 0.04) and distant (P = 0.03) failure was observed in the unfavorable group. Within 3 months following SBRT, CTCs continued to be detected in 10 of 35 (29%) subjects. Persistent detection of CTCs was associated with increased risk of distant failure (P = 0.04) and trended toward increased regional (P = 0.08) and local failure (P = 0.16). CONCLUSIONS: Higher pretreatment CTCs and persistence of CTCs posttreatment is significantly associated with increased risk of recurrence outside the targeted treatment site. This suggests that CTC analysis may potentially identify patients at higher risk for regional or distant recurrences and who may benefit from either systemic therapy and/or timely locoregional salvage treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/radiotherapy , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Neoplasm Recurrence, Local/pathology , Neoplastic Cells, Circulating/pathology , Radiosurgery/methods , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/surgery , Disease Progression , Female , Humans , Lung Neoplasms/blood , Lung Neoplasms/surgery , Male , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/surgery , Neoplasm Staging , Retrospective Studies , Survival Rate , Telomerase/blood , Treatment Outcome
15.
J Cell Biol ; 160(7): 1017-27, 2003 Mar 31.
Article in English | MEDLINE | ID: mdl-12668657

ABSTRACT

Anumber of proteins are recruited to nuclear foci upon exposure to double-strand DNA damage, including 53BP1 and Rad51, but the precise role of these DNA damage-induced foci remain unclear. Here we show in a variety of human cell lines that histone deacetylase (HDAC) 4 is recruited to foci with kinetics similar to, and colocalizes with, 53BP1 after exposure to agents causing double-stranded DNA breaks. HDAC4 foci gradually disappeared in repair-proficient cells but persisted in repair-deficient cell lines or cells irradiated with a lethal dose, suggesting that resolution of HDAC4 foci is linked to repair. Silencing of HDAC4 via RNA interference surprisingly also decreased levels of 53BP1 protein, abrogated the DNA damage-induced G2 delay, and radiosensitized HeLa cells. Our combined results suggest that HDAC4 is a critical component of the DNA damage response pathway that acts through 53BP1 and perhaps contributes in maintaining the G2 cell cycle checkpoint.


Subject(s)
Carrier Proteins/metabolism , DNA Damage , Histone Deacetylases/metabolism , Intracellular Signaling Peptides and Proteins , Nuclear Proteins/metabolism , Phosphoproteins , Repressor Proteins/metabolism , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Radiation , Etoposide/pharmacology , G2 Phase , Gamma Rays/adverse effects , Histone Deacetylases/drug effects , Histone Deacetylases/radiation effects , Humans , Hydroxamic Acids/pharmacology , Kinetics , Mutation , Nuclear Proteins/genetics , Nucleic Acid Synthesis Inhibitors/pharmacology , Protein Synthesis Inhibitors/pharmacology , RNA, Small Interfering/metabolism , Repressor Proteins/drug effects , Repressor Proteins/radiation effects , Tumor Cells, Cultured , Tumor Suppressor p53-Binding Protein 1
16.
Mol Biol Cell ; 17(2): 585-97, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16280357

ABSTRACT

Histone deacetylases mediate critical cellular functions but relatively little is known about mechanisms controlling their expression, including expression of HDAC4, a class II HDAC implicated in the modulation of cellular differentiation and viability. Endogenous HDAC4 mRNA, protein levels and promoter activity were all readily repressed by mithramycin, suggesting regulation by GC-rich DNA sequences. We validated consensus binding sites for Sp1/Sp3 transcription factors in the HDAC4 promoter through truncation studies and targeted mutagenesis. Specific and functional binding by Sp1/Sp3 at these sites was confirmed with chromatin immunoprecipitation (ChIP) and electromobility shift assays (EMSA). Cotransfection of either Sp1 or Sp3 with a reporter driven by the HDAC4 promoter led to high activities in SL2 insect cells (which lack endogenous Sp1/Sp3). In human cells, restored expression of Sp1 and Sp3 up-regulated HDAC4 protein levels, whereas levels were decreased by RNA-interference-mediated knockdown of either protein. Finally, variable levels of Sp1 were in concordance with that of HDAC4 in a number of human tissues and cancer cell lines. These studies together characterize for the first time the activity of the HDAC4 promoter, through which Sp1 and Sp3 modulates expression of HDAC4 and which may contribute to tissue or cell-line-specific expression of HDAC4.


Subject(s)
Gene Expression Regulation, Enzymologic , Histone Deacetylases/genetics , Repressor Proteins/genetics , Sp1 Transcription Factor/physiology , Sp3 Transcription Factor/physiology , Animals , Base Sequence , Binding Sites , Cells, Cultured , Consensus Sequence/physiology , Drosophila/cytology , Gene Expression Regulation, Enzymologic/drug effects , HeLa Cells , Histone Deacetylases/drug effects , Histone Deacetylases/metabolism , Humans , Male , Molecular Sequence Data , Plicamycin/pharmacology , Promoter Regions, Genetic , Repressor Proteins/drug effects , Repressor Proteins/metabolism , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor/genetics , Tissue Distribution
17.
Cancer Res ; 67(18): 8791-9, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875720

ABSTRACT

For patients with solid tumors, the tolerance of surrounding tissues often limits the dose of radiation that can be delivered. Thus, agents that preferentially increase the cytotoxic effects of radiation toward tumor cells would significantly alter the therapeutic ratio and improve patient survival. Using a high-throughput, unbiased screening approach, we have identified 4'-bromo-3'-nitropropiophenone (NS-123) as a radiosensitizer of human glioma cells in vitro and in vivo. NS-123 radiosensitized U251 glioma cells in a dose-dependent and time-dependent manner, with dose enhancement ratios ranging from 1.3 to 2.0. HT-29 colorectal carcinoma and A549 lung adenocarcinoma cells were also radiosensitized by NS-123 in vitro, whereas NS-123 did not increase the radiation sensitivity of normal human astrocytes or developmental abnormalities or lethality of irradiated Zebrafish embryos. In a novel xenograft model of U251 cells implanted into Zebrafish embryos, NS-123 enhanced the tumor growth-inhibitory effects of ionizing radiation (IR) with no apparent effect on embryo development. Similar results were obtained using a mouse tumor xenograft model in which NS-123 sensitized U251 tumors to IR while exhibiting no overt toxicity. In vitro pretreatment with NS-123 resulted in accumulation of unrepaired IR-induced DNA strand breaks and prolonged phosphorylation of the surrogate markers of DNA damage H2AX, ataxia telangiectasia mutated protein, DNA-dependent protein kinase, and CHK2 after IR, suggesting that NS-123 inhibits a critical step in the DNA repair pathway. These results show the potential of this cell-based, high-throughput screening method to identify novel radiosensitizers and suggest that NS-123 and similar nitrophenol compounds may be effective in antiglioma modalities.


Subject(s)
Neoplasms/drug therapy , Neoplasms/radiotherapy , Propiophenones/pharmacology , Radiation-Sensitizing Agents/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Animals , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/radiotherapy , Combinatorial Chemistry Techniques/methods , Combined Modality Therapy , DNA Breaks, Double-Stranded , DNA Repair/drug effects , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/radiation effects , Female , Glioma/drug therapy , Glioma/radiotherapy , HT29 Cells , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Mice , Mice, Nude , Xenograft Model Antitumor Assays , Zebrafish/embryology
18.
Neurooncol Adv ; 1(1): vdz030, 2019.
Article in English | MEDLINE | ID: mdl-31807732

ABSTRACT

BACKGROUND: Amplification of the epidermal growth factor receptor (EGFR) gene is commonly found in glioblastoma (GBM). About 57% GBM overexpresses EGFR and are associated with tumor progression, poor prognosis, and shorter life expectancy. Molecular profiling of solid tumors usually takes several weeks and may be biased by intrinsic tumor heterogeneity. METHODS: The unique sequence created by the fusion of exon 1 and exon 8 in EGFRvIII was used to guide the design of primers and a Minor Groove Binder (MGB) probe. Extracted total RNA was reverse transcribed and pre-amplified by PCR, followed by detection of the EGFRvIII mutation by dPCR. RESULTS: The lowest limit of quantification of our EGFRvIII assay was 0.003%. The EGFRvIII variant was identified in patient-derived glioma neurosphere cell lines, xenograft mouse model, and patient-derived tumor specimens. The overall workflow can be accomplished within 24 hours. In certain samples, EGFRvIII was detected when next-generation sequencing was unable to identify the variant. This finding highlights the ability of the dPCR assay to identify EGFRvIII mutations in heterogeneous solid tumors such as GBM in a rapid fashion by profiling samples from spatially distinct areas of tumors from the same patient. CONCLUSIONS: In this study, we developed a highly sensitive digital PCR (dPCR) platform and leveraged our assay to detect the variant III alteration of EGFR (EGFRvIII) and amplified EGFR in patient-derived glioma neurosphere cell lines, orthotopic xenograft GBM mouse models, and patient-derived tumor specimens in less than 24 hours from minute quantities of starting material.

19.
Clin Lung Cancer ; 20(5): 384-390.e2, 2019 09.
Article in English | MEDLINE | ID: mdl-31221522

ABSTRACT

BACKGROUND: Assays to identify circulating tumor cells (CTCs) might allow for noninvasive and sequential monitoring of lung cancer. We investigated whether serial CTC analysis could complement conventional imaging for detecting recurrences after treatment in patients with locally advanced non-small-cell lung cancer (LA-NSCLC). PATIENTS AND METHODS: Patients with LA-NSCLC (stage II-III) who definitively received concurrent chemoradiation were prospectively enrolled, with CTCs from peripheral blood samples identified using an adenoviral probe that detects elevated telomerase activity present in nearly all lung cancer cells. A "detectable" CTC level was defined as 1.3 green flourescent protein-positive cells per milliliter of collected blood. Samples were obtained before, during (at weeks 2, 4, and 6), and after treatment (post-radiation therapy [RT]; at months 1, 3, 6, 12, 18, and 24). RESULTS: Forty-eight patients were enrolled. At a median follow-up of 10.9 months, 22 (46%) patients had disease recurrence at a median time of 7.6 months post-RT (range, 1.3-32.0 months). Of the 20 of 22 patients for whom post-RT samples were obtained, 15 (75%) had an increase in CTC counts post-RT. In 10 of these 15 patients, CTCs were undetectable on initial post-RT draw but were then detected again before radiographic detection of recurrence, with a median lead time of 6.2 months and mean lead time of 6.1 months (range, 0.1-12.0 months) between CTC count increase and radiographic evidence of recurrence. One patient with an early recurrence (4.7 months) had persistently elevated detectable CTC levels during and after treatment. CONCLUSION: These results indicate that longitudinal CTC monitoring in patients with LA-NSCLC treated with chemoradiation is feasible, and that detectable CTC levels in many patients meaningfully precede radiologic evidence of disease recurrence.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnosis , Cell Count/methods , Lung Neoplasms/diagnosis , Neoplastic Cells, Circulating/pathology , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Chemoradiotherapy , Feasibility Studies , Female , Humans , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Male , Middle Aged , Neoplasm Recurrence, Local , Neoplasm Staging
20.
Mol Cell Biol ; 25(21): 9232-48, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16227576

ABSTRACT

The fidelity of chromosomal duplication is monitored by cell cycle checkpoints operational during mitosis. One such cell cycle delay is invoked by microtubule-targeting agents such as nocodazole or paclitaxel (Taxol) and is mediated by mitotic checkpoint proteins that include BubR1. Relatively little is known about the regulation of expression and stability of BubR1 (or other checkpoint proteins) and how these factors dictate the durability of the cell cycle delay. We report here that treatment of HeLa cells with spindle-disrupting agents resulted in caspase activation and precipitated the cleavage of BubR1. This mechanism ultimately leads to reduced levels of full-length protein, which are accompanied by abrogation of the mitotic block; the checkpoint abrogation is substantially accelerated by inhibition of de novo protein synthesis. In contrast, inhibition of caspase activity blocked BubR1 degradation and prolonged mitosis. To confirm a direct link between caspase activity and BubR1 protein expression, we identified by site-directed mutagenesis the specific caspase cleavage sites cleaved after exposure to paclitaxel. Surprisingly, BubR1 has two sites of cleavage: primarily at Asp607/Asp610 and secondarily at Asp576/Asp579. BubR1 mutated at both locations (BubR1Delta579Delta610) was resistant to paclitaxel-induced degradation. Expression of BubR1Delta579Delta610 augmented the mitotic delay induced by spindle disruption in transfected cells as well as in clones engineered to inducibly express the mutant protein upon exposure to doxycycline and ultimately led to increased aneuploidy. Underscoring the importance of these caspase cleavage sites, both tetrapeptide motifs are identified in the amino acid sequences of human, mouse, chicken, and Xenopus BubR1. These results are potentially the first to link the control of the stability of a key mitotic checkpoint protein to caspase activation, a regulatory pathway that may be involved in killing defective cells and that has been evolutionarily conserved.


Subject(s)
Caspases/metabolism , Cell Cycle/physiology , Protein Kinases/metabolism , Spindle Apparatus/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Caspase 3 , Caspase Inhibitors , Cell Cycle/drug effects , Cell Cycle Proteins , Chickens , Conserved Sequence , Doxycycline/pharmacology , Enzyme Activation , HeLa Cells , Humans , Mice , Microtubules/drug effects , Microtubules/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Nocodazole/pharmacology , Paclitaxel/pharmacology , Protein Kinases/biosynthesis , Protein Kinases/genetics , Protein Serine-Threonine Kinases , Sequence Homology, Amino Acid , Spindle Apparatus/drug effects , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL