Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Melanoma Res ; 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39012389

ABSTRACT

Using a customized, harmonized US electronic health record database, real-world prescription patterns of first-line adjuvant immunotherapy and targeted therapy were retrospectively assessed for BRAFV600-mutated melanoma. Adults with BRAFV600 mutation-positive stage IIIA-D cutaneous melanoma who received first-line adjuvant immunotherapy (nivolumab or pembrolizumab) or targeted therapy (dabrafenib plus trametinib) between 1 January 2014 and 30 August 2020 in the NOBLE database were included. Patients were followed from first-line adjuvant therapy initiation for at least 6 months, until death, progression, follow-up loss, or data cutoff. Primary endpoints were proportion of patients receiving either therapy in first-line and second-line, treatment switching, treatment timing, and status at the end of first-line therapy. Secondary endpoints included discontinuation rates, recurrence-free survival (RFS), and overall survival (OS). Of 318 patients evaluated, 67.6% received nivolumab, 14.2% pembrolizumab, and 18.2% targeted therapy as first-line adjuvant therapy. Median treatment duration was longest for nivolumab (292 days) and shortest for targeted therapy (115 days). Reason for discontinuation was recorded for 195 of 274 patients who discontinued first-line therapy; most common reasons were treatment completion and treatment-related toxicity [87/158 (55.0%) and 29/158 (18.4%), respectively, in immunotherapy-treated patients; 9/37 (24.3%) and 21/37 (56.8%) in targeted therapy-treated patients]. Median RFS and OS for targeted therapy and nivolumab were not reached and were 34.6 and 38.1 months, respectively, for pembrolizumab. These results inform on prescription preferences and clinical outcomes for BRAFV600-mutated melanoma patients in the first-line adjuvant setting.

2.
J Clin Invest ; 134(10)2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38502231

ABSTRACT

Neurofibromatosis type 1 (NF1) is caused by mutations in the NF1 gene that encodes neurofibromin, a RAS GTPase-activating protein. Inactivating NF1 mutations cause hyperactivation of RAS-mediated signaling, resulting in the development of multiple neoplasms, including malignant peripheral nerve sheath tumors (MPNSTs). MPNSTs are an aggressive tumor and the main cause of mortality in patients with NF1. MPNSTs are difficult to resect and refractory to chemo- and radiotherapy, and no molecular therapies currently exist. Immune checkpoint blockade (ICB) is an approach to treat inoperable, undruggable cancers like MPNST, but successful outcomes require an immune cell-rich tumor microenvironment. While MPNSTs are noninflamed "cold" tumors, here, we converted MPNSTs into T cell-inflamed "hot" tumors by activating stimulator of IFN genes (STING) signaling. Mouse genetic and human xenograft MPNST models treated with a STING agonist plus ICB exhibited growth delay via increased apoptotic cell death. This strategy offers a potential treatment regimen for MPNSTs.


Subject(s)
Immunotherapy , Membrane Proteins , Neurofibromatosis 1 , Neurofibromin 1 , Tumor Microenvironment , Animals , Tumor Microenvironment/immunology , Humans , Mice , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/immunology , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology , Neurofibromatosis 1/immunology , Neurofibromatosis 1/therapy , Neurofibromatosis 1/metabolism , Neurofibromin 1/genetics , Immune Checkpoint Inhibitors/pharmacology , Nerve Sheath Neoplasms/genetics , Nerve Sheath Neoplasms/pathology , Nerve Sheath Neoplasms/therapy , Nerve Sheath Neoplasms/immunology , Nerve Sheath Neoplasms/metabolism , Cell Line, Tumor , Xenograft Model Antitumor Assays , Signal Transduction/immunology
3.
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
4.
Cancers (Basel) ; 15(13)2023 Jun 22.
Article in English | MEDLINE | ID: mdl-37444395

ABSTRACT

VIP (vasoactive intestinal peptide) is a 28-amino acid peptide hormone expressed by cancer and the healthy nervous system, digestive tract, cardiovascular, and immune cell tissues. Many cancers express VIP and its surface receptors VPAC1 and VPAC2, but the role of autocrine VIP signaling in cancer as a targetable prognostic and predictive biomarker remains poorly understood. Therefore, we conducted an in silico gene expression analysis to study the mechanisms of autocrine VIP signaling in cancer. VIP expression from TCGA PANCAN tissue samples was analyzed against the expression levels of 760 cancer-associated genes. Of the 760 genes, 10 (MAPK3, ZEB1, TEK, NOS2, PTCH1 EIF4G1, GMPS, CDK2, RUVBL1, and TIMELESS) showed statistically meaningful associations with the VIP (Pearson's R-coefficient > |0.3|; p < 0.05) across all cancer histologies. The strongest association with the VIP was for the epithelial-mesenchymal transition regulator ZEB1 in gastrointestinal malignancies. Similar positive correlations between the VIP and ZEB1 expression were also observed in healthy gastrointestinal tissues. Gene set analysis indicates the VIP is involved in the EMT and cell cycle pathways, and a high VIP and ZEB1 expression is associated with higher median estimate and stromal scores These findings uncover novel mechanisms for VIP- signaling in cancer and specifically suggest a role for VIP as a biomarker of ZEB1-mediated EMT. Further studies are warranted to characterize the specific mechanism of this interaction.

5.
Am J Infect Control ; 51(7): 807-811, 2023 07.
Article in English | MEDLINE | ID: mdl-36306861

ABSTRACT

BACKGROUND: Hospital acquired infections pose a significant risk for patients undergoing hematopoietic stem cell transplantation. Horizontal transfer of antimicrobial resistance genes contributes to prevalence of multidrug-resistant infections in this patient population. METHODS: At an academic bone marrow transplantation center, we performed whole genome DNA sequencing (WGS) on commonly used physician items, including badges, stethoscopes, soles of shoes, and smart phones from 6 physicians. Data were analyzed to determine antimicrobial resistance and virulence factor genes. RESULTS: A total of 1,126 unique bacterial species, 495 distinct bacteriophages, 91 unique DNA viruses, and 175 fungal species were observed. Every item contained bacteria with antibiotic and/or antiseptic resistance genes. Stethoscopes contained greatest frequency of antibiotic resistance and more plasmid-carriage of antibiotic resistance. DISCUSSION AND CONCLUSIONS: These data indicate that physician examination tools and personal items possess potentially pathogenic microbes. Infection prevention policies must consider availability of resources to clean physical examination tools as well as provider awareness when enacting hospital policies. Additionally, the prevalence of antimicrobial resistance genes (eg, encoding resistance to aminoglycosides, ß-lactams, and quinolones) reinforces need for antimicrobial stewardship, including for immunocompromised patients. Further research is needed to assess whether minute quantities of microbes on physician objects detectable by WGS represents clinically significant inoculums for immunocompromised patients.


Subject(s)
Anti-Bacterial Agents , Bacteria , Humans , Plasmids , Anti-Bacterial Agents/therapeutic use , Bacteria/genetics , Drug Resistance, Microbial , beta-Lactams/pharmacology , Drug Resistance, Multiple, Bacterial , Microbial Sensitivity Tests
6.
IEEE J Biomed Health Inform ; 26(8): 3779-3790, 2022 08.
Article in English | MEDLINE | ID: mdl-35594223

ABSTRACT

The determination of step length, an important gait parameter, has been a challenging task. Although unobtrusive sensors (inertial measurement units) have been developed recently, they cannot facilitate the automatic estimation of step length. In this article, we use a model-based technique to determine the step length using the Unscented Kalman Filter with angular velocity from a gyroscope inside the thigh pocket. We then propose a novel covariance estimation algorithm based on a screening technique that performs a search for the optimal Process Noise Covariance matrix. Upon implementing the Unscented Kalman Filter, the step length is found using the horizontal position of the foot relative to the hip using a patient-independent robust peak detection algorithm. This research article paves the way for algorithms that are computationally much faster than black box methods, with more scope for the development of better algorithms for covariance estimation using the one proposed in this article as a foundation.


Subject(s)
Thigh , Wearable Electronic Devices , Algorithms , Foot , Gait , Humans
7.
Annu Int Conf IEEE Eng Med Biol Soc ; 2022: 1125-1128, 2022 07.
Article in English | MEDLINE | ID: mdl-36086518

ABSTRACT

Step Length is an important metric that can be used for the analysis and assessment of the gait. Proper dynamical models are not available in current literature associated with the wrist that can adequately determine the step length using recursive estimation techniques. This study presents a method to estimate the step length using angular velocity data from the wrist sensor. The technique maps the dynamical region corresponding to periods of activity of the gait manifested in angular velocity from the inertial measurement unit located at the wrist to that of the thigh using an artificial neural network, upon which an unscented Kalman filter is used to determine the horizontal position of the foot relative to the hip, and consequently, determine step length. The results for Step Length indicate an average accuracy of 81.8% and 91.1% for the young and elderly, respectively, when compared to a reference system, which, in our study, is data from a treadmill.


Subject(s)
Wearable Electronic Devices , Wrist , Aged , Foot , Gait , Humans , Wrist Joint
8.
Front Immunol ; 12: 718621, 2021.
Article in English | MEDLINE | ID: mdl-34512641

ABSTRACT

PI3K-δ and PI3K-γ are critical regulators of T-cell differentiation, senescence, and metabolism. PI3K-δ and PI3K-γ signaling can contribute to T-cell inhibition via intrinsic mechanisms and regulation of suppressor cell populations, including regulatory T-cells and myeloid derived suppressor cells in the tumor. We examine an exciting new role for using selective inhibitors of the PI3K δ- and γ-isoforms as modulators of T-cell phenotype and function in immunotherapy. Herein we review the current literature on the implications of PI3K-δ and -γ inhibition in T-cell biology, discuss existing challenges in adoptive T-cell therapies and checkpoint blockade inhibitors, and highlight ongoing efforts and future directions to incorporate PI3K-δ and PI3K-γ as synergistic T-cell modulators in immunotherapy.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/immunology , Class Ib Phosphatidylinositol 3-Kinase/immunology , Immunotherapy/methods , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Biomarkers , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/metabolism , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Disease Management , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy, Adoptive , Lymphocyte Activation/immunology , Molecular Targeted Therapy , Signal Transduction , Translational Research, Biomedical
9.
Mol Cancer Res ; 17(12): 2395-2409, 2019 12.
Article in English | MEDLINE | ID: mdl-31548239

ABSTRACT

Molecular events activating the PI3K pathway are frequently detected in human tumors and the activation of PI3K signaling alters numerous cellular processes including tumor cell proliferation, survival, and motility. More recent studies have highlighted the impact of PI3K signaling on the cellular response to interferons and other immunologic processes relevant to antitumor immunity. Given the ability of IFNγ to regulate antigen processing and presentation and the pivotal role of MHC class I (MHCI) and II (MHCII) expression in T-cell-mediated antitumor immunity, we sought to determine the impact of PI3K signaling on MHCI and MHCII induction by IFNγ. We found that the induction of cell surface MHCI and MHCII molecules by IFNγ is enhanced by the clinical grade PI3K inhibitors dactolisib and pictilisib. We also found that PI3K inhibition increases STAT1 protein levels following IFNγ treatment and increases accessibility at genomic STAT1-binding motifs. Conversely, we found that pharmacologic activation of PI3K signaling can repress the induction of MHCI and MHCII molecules by IFNγ, and likewise, the loss of PTEN attenuates the induction of MHCI, MHCII, and STAT1 by IFNγ. Consistent with these in vitro studies, we found that within human head and neck squamous cell carcinomas, intratumoral regions with high phospho-AKT IHC staining had reduced MHCI IHC staining. IMPLICATIONS: Collectively, these findings demonstrate that MHC expression can be modulated by PI3K signaling and suggest that activation of PI3K signaling may promote immune escape via effects on antigen presentation.


Subject(s)
Interferon-gamma/pharmacology , Phosphatidylinositol 3-Kinase/genetics , STAT1 Transcription Factor/genetics , Squamous Cell Carcinoma of Head and Neck/drug therapy , Antigen Presentation/genetics , Antigen Presentation/immunology , Binding Sites/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , Genes, MHC Class I/genetics , Genes, MHC Class I/immunology , Genes, MHC Class II/genetics , Genes, MHC Class II/immunology , Genomics , Humans , Interferon-gamma/genetics , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinase/immunology , Protein Binding/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/drug effects , Signal Transduction/immunology , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/pathology
11.
Cancer Res ; 74(8): 2152-9, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24525740

ABSTRACT

Blood tests to detect circulating tumor cells (CTC) offer great potential to monitor disease status, gauge prognosis, and guide treatment decisions for patients with cancer. For patients with brain tumors, such as aggressive glioblastoma multiforme, CTC assays are needed that do not rely on expression of cancer cell surface biomarkers like epithelial cell adhesion molecules that brain tumors tend to lack. Here, we describe a strategy to detect CTC based on telomerase activity, which is elevated in nearly all tumor cells but not normal cells. This strategy uses an adenoviral detection system that is shown to successfully detect CTC in patients with brain tumors. Clinical data suggest that this assay might assist interpretation of treatment response in patients receiving radiotherapy, for example, to differentiate pseudoprogression from true tumor progression. These results support further development of this assay as a generalized method to detect CTC in patients with cancer.


Subject(s)
Brain Neoplasms/blood , Brain Neoplasms/pathology , Glioma/blood , Glioma/pathology , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Telomerase/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Brain Neoplasms/enzymology , Female , Glioma/metabolism , Heterografts , Humans , Male , Mice , Promoter Regions, Genetic , Telomerase/analysis , Telomerase/genetics , Xenograft Model Antitumor Assays
12.
Cancer Biol Ther ; 15(6): 683-7, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24618718

ABSTRACT

BACKGROUND: Muscle invasive bladder carcinoma is an often lethal disease that requires aggressive treatment. Improved assays would contribute to better risk prediction and clinical management of this disease. A telomerase-based assay to detect circulating tumor cells (CTCs) may usefully fulfill this role. METHODS: Two patients (C1 and C2) were enrolled onto an IRB-approved bladder biomarker study before initiating post-operative radiation therapy (RT) for muscle invasive bladder carcinoma. Blood samples were taken at predefined intervals: before, during, and after RT and then retrospectively correlated with imaging studies and disease course. RESULTS: C1 began RT for positive resection margins on surgical pathology, at which time CTCs were undetectable and pelvic imaging demonstrated no evidence of disease. However, following the completion of treatment, the patient's CTC count was found to have increased to 202 CTCs/mL, and MRI demonstrated new abdominal and pelvic masses consistent with progressive disease. C1 ultimately died of disease with distant and local failure. Conversely, C2 was found to have 632 CTCs/mL before the initiation of RT for positive surgical margins, although imaging demonstrated no visible masses. At the conclusion of RT, repeat imaging showed changes that were indeterminate for either tumor recurrence or post-radiation effects. However, the patient's CTC count had dropped to 184 CTCs/mL. Furthermore, a second follow-up assay performed 6 months later revealed no detectable CTCs and repeat imaging showed complete resolution of worrisome imaging changes, thus excluding tumor progression. CONCLUSIONS: To our knowledge this is the first report of a telomerase-based assay to identify CTCs in bladder cancer patients. Further studies are required to fully determine the ultimate clinical utility of this assay. However, the two patient vignettes described here illustrate how serial CTC assays may track the disease course and inform the management of bladder cancer patients undergoing adjuvant RT and potentially chemotherapy.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Transitional Cell/diagnosis , Neoplastic Cells, Circulating/metabolism , Telomerase/metabolism , Urinary Bladder Neoplasms/diagnosis , Aged , Carcinoma, Transitional Cell/enzymology , Carcinoma, Transitional Cell/therapy , Enzyme Assays , Fatal Outcome , Female , Humans , Male , Middle Aged , Radiotherapy, Adjuvant , Treatment Outcome , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/therapy
13.
Transl Oncol ; 5(4): 230-7, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22937174

ABSTRACT

Preclinical studies of cranial radiation therapy (RT) using animal brain tumor models have been hampered by technical limitations in the delivery of clinically relevant RT. We established a bioimageable mouse model of glioblastoma multiforme (GBM) and an image-guided radiation delivery system that facilitated precise tumor localization and treatment and which closely resembled clinical RT. Our novel radiation system makes use of magnetic resonance imaging (MRI) and bioluminescent imaging (BLI) to define tumor volumes, computed tomographic (CT) imaging for accurate treatment planning, a novel mouse immobilization system, and precise treatments delivered with the Small Animal Radiation Research Platform. We demonstrated that, in vivo, BLI correlated well with MRI for defining tumor volumes. Our novel restraint system enhanced setup reproducibility and precision, was atraumatic, and minimized artifacts on CT imaging used for treatment planning. We confirmed precise radiation delivery through immunofluorescent analysis of the phosphorylation of histone H2AX in irradiated brains and brain tumors. Assays with an intravenous near-infrared fluorescent probe confirmed that radiation of orthografts increased disruption of the tumor blood-brain barrier (BBB). This integrated model system, which facilitated delivery of precise, reproducible, stereotactic cranial RT in mice and confirmed RT's resultant histologic and BBB changes, may aid future brain tumor research.

14.
Neoplasia ; 9(4): 341-8, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17460778

ABSTRACT

Pancreatic and colorectal carcinomas frequently express oncogenic/mutant K-Ras that contributes to both tumorigenesis and clinically observed resistance to radiation treatment. We have previously shown that farnesyltransferase inhibitors (FTI) radiosensitize many pancreatic and colorectal cancer cell lines that express oncogenic K-ras at doses that inhibit the prenylation and activation of H-Ras but not K-Ras. In the present study, we have examined the mechanism of FTI-mediated radiosensitization in cell lines that express oncogenic K-Ras and found that wild-type H-Ras is a contributor to radiation survival in tumor cells that express oncogenic K-Ras. In these experiments, inhibiting the expression of oncogenic K-Ras, wild-type H-Ras, or epidermal growth factor receptor (EGFR) led to similar levels of radiosensitization as treatment with the FTI tipifarnib. Treatment with the EGFR inhibitor gefitinib led to similar levels of radiosensitization, and the combinations of tipifarnib or gefitinib plus inhibition of K-Ras, H-Ras, or EGFR expression did not provide additional radiosensitization compared with tipifarnib or gefitinib alone. Finally, supplementing culture medium with the EGFR ligand transforming growth factor alpha was able to reverse the radiosensitizing effect of inhibiting K-ras expression. Taken together, these findings suggest that EGFR-activated H-Ras signaling is initiated by oncogenic K-Ras to promote radiation survival in pancreatic and colorectal cancers.


Subject(s)
Colorectal Neoplasms/pathology , ErbB Receptors/physiology , ErbB Receptors/radiation effects , Genes, ras/physiology , Genes, ras/radiation effects , Pancreatic Neoplasms/pathology , Signal Transduction/genetics , Signal Transduction/radiation effects , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/radiation effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/radiotherapy , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/radiotherapy
SELECTION OF CITATIONS
SEARCH DETAIL