Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
JCO Clin Cancer Inform ; 7: e2200170, 2023 05.
Article in English | MEDLINE | ID: mdl-37207310

ABSTRACT

PURPOSE: Cancer patient navigators (CPNs) can decrease the time from diagnosis to treatment, but workloads vary widely, which may lead to burnout and less optimal navigation. Current practice for patient distribution among CPNs at our institution approximates random distribution. A literature search did not uncover previous reports of an automated algorithm to distribute patients to CPNs. We sought to develop an automated algorithm to fairly distribute new patients among CPNs specializing in the same cancer type(s) and assess its performance through simulation on a retrospective data set. METHODS: Using a 3-year data set, a proxy for CPN work was identified and multiple models were developed to predict the upcoming week's workload for each patient. An XGBoost-based predictor was retained on the basis of its superior performance. A distribution model was developed to fairly distribute new patients among CPNs within a specialty on the basis of predicted work needed. The predicted work included the week's predicted workload from a CPN's existing patients plus that of newly distributed patients to the CPN. Resulting workload unfairness was compared between predictor-informed and random distribution. RESULTS: Predictor-informed distribution significantly outperformed random distribution for equalizing weekly workloads across CPNs within a specialty. CONCLUSION: This derivation work demonstrates the feasibility of an automated model to distribute new patients more fairly than random assignment (with unfairness assessed using a workload proxy). Improved workload management may help reduce CPN burnout and improve navigation assistance for patients with cancer.


Subject(s)
Neoplasms , Patient Navigation , Humans , Workload , Retrospective Studies , Neoplasms/diagnosis , Neoplasms/therapy
2.
Popul Health Manag ; 25(2): 244-253, 2022 04.
Article in English | MEDLINE | ID: mdl-35442784

ABSTRACT

Mammography screening rates are typically lower in those with less economic advantage (EA). This study, conducted at an integrated health care system covering a mixed rurality population, assessed the ability of interventions (text messages linking to a Web microsite, digital health care workers, and a community health fair) to affect mammography screening rates and disparity in those rates among different EA populations. Payor type served as a proxy for greater (commercially insured) versus lower (Medicaid insured) EA. 4,342 subjects were included across the preintervention ("Pre") and postintervention ("Post") periods. Interventions were prospectively applied to all Medicaid subjects and randomly selected commercial subjects. Applying interventions only to lower EA subjects reversed the screening rate disparity (2.6% Pre vs. -3.7% Post, odds ratio [OR] 2.4 P < 0.01). When intervention arms ("Least," "More," "Most") were equally applied, screening rates in both EA groups significantly increased in the More arm (Medicaid OR = 2.04 P = 0.04, Commercial OR = 3.08 P < 0.01) and Most arm (Medicaid OR 2.57 P < 0.01, Commercial OR 2.33 P < 0.01), but not in the Least (text-only) arm (Medicaid OR 1.83 P = 0.11, Commercial OR 1.72 P = 0.09), although this text-only arm was inadequately powered to detect a difference. In summary, targeting interventions to those with lower EA reversed screening rate disparities, text messaging combined with other interventions improved screening rates in both groups, and future research is needed to determine whether interventions can simultaneously improve screening rates for all without worsening the disparity.


Subject(s)
Breast Neoplasms , Early Detection of Cancer , Breast Neoplasms/diagnosis , Breast Neoplasms/prevention & control , Female , Humans , Mammography , Mass Screening , Medicaid , United States
4.
Radiother Oncol ; 118(1): 43-51, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26615717

ABSTRACT

BACKGROUND AND PURPOSE: To investigate if cranial X-irradiation reduces amyloid-ß (Aß) plaques and influences cognitive function in a transgenic mouse model of AD. METHODS AND MATERIALS: B6.Cg-Tg (APPswePSEN1dE9)85Dbo/J AD-prone mice were given cranial X-irradiation. The number of Aß plaques, along with expression of AD specific genes (84 genes: Mouse Alzheimer's Disease RT(2) Profiler), radiation-associated cytokines (Milliplex MAP Mouse Cytokine Chemokine Immunoassay) and immunohistochemistry (IL10, IL-1ß, Iba1 CD45) was assessed. Behavioral testing was performed to relate changes in Aß burden to cognitive function using a Morris water-maze task. RESULTS: Single X-ray doses reduced the number (p=0.002) and size (p=0.01) of Aß plaques. Low-dose fractionation produced greater 50.6% (1 Gy × 10), 72% (2 Gy × 5) and 78% (2 Gy × 10) reductions. Irradiation was associated with gene (Pkp4, 1.5-fold, p=0.004) and proteomic (MIP-2, 8-fold, p=0.0024) changes at 24-48 h. Microglia increased at 4 weeks post-irradiation (p=0.001). The reduction in Aß burden (2 Gy × 5) was associated with cognitive improvement (p=0.012). CONCLUSION: This is the first report that a clinically relevant course of external beam irradiation (2 Gy × 5) produces a significant reduction in AD-associated amyloid-ß plaques with a subsequent improvement in cognitive function. However, longer-term studies are needed to define the precise underlying mechanism and longevity of this response.


Subject(s)
Alzheimer Disease/radiotherapy , Behavior, Animal/radiation effects , Brain/radiation effects , Cognition/radiation effects , Cranial Irradiation/methods , Plaque, Amyloid/radiotherapy , Animals , Disease Models, Animal , Male , Mice , Mice, Transgenic
5.
J Neurosurg ; 121 Suppl: 84-90, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25434941

ABSTRACT

OBJECT: Stereotactic radiosurgery (SRS) alone is increasingly used in patients with newly diagnosed brain metastases. Stereotactic radiosurgery used together with whole-brain radiotherapy (WBRT) reduces intracranial failure rates, but this combination also causes greater neurocognitive toxicity and does not improve survival. Critics of SRS alone contend that deferring WBRT results in an increased need for salvage therapy and in higher costs. The authors compared the cost-effectiveness of treatment with SRS alone, SRS and WBRT (SRS+WBRT), and surgery followed by SRS (S+SRS) at the authors' institution. METHODS: The authors retrospectively reviewed the medical records of 289 patients in whom brain metastases were newly diagnosed and who were treated between May 2001 and December 2007. Overall survival curves were plotted using the Kaplan-Meier method. Multivariate proportional hazards analysis (MVA) was used to identify factors associated with overall survival. Survival data were complete for 96.2% of patients, and comprehensive data on the resource use for imaging, hospitalizations, and salvage therapies were available from the medical records. Treatment costs included the cost of initial and all salvage therapies for brain metastases, hospitalizations, management of complications, and imaging. They were computed on the basis of the 2007 Medicare fee schedule from a payer perspective. Average treatment cost and average cost per month of median survival were compared. Sensitivity analysis was performed to examine the impact of variations in key cost variables. RESULTS: No significant differences in overall survival were observed among patients treated with SRS alone, SRS+WBRT, or S+SRS with respective median survival of 9.8, 7.4, and 10.6 months. The MVA detected a significant association of overall survival with female sex, Karnofsky Performance Scale (KPS) score, primary tumor control, absence of extracranial metastases, and number of brain metastases. Salvage therapy was required in 43% of SRS-alone and 26% of SRS+WBRT patients (p < 0.009). Despite an increased need for salvage therapy, the average cost per month of median survival was $2412 per month for SRS alone, $3220 per month for SRS+WBRT, and $4360 per month for S+SRS (p < 0.03). Compared with SRS+WBRT, SRS alone had an average incremental cost savings of $110 per patient. Sensitivity analysis confirmed that the average treatment cost of SRS alone remained less than or was comparable to SRS+WBRT over a wide range of costs and treatment efficacies. CONCLUSIONS: Despite an increased need for salvage therapy, patients with newly diagnosed brain metastases treated with SRS alone have similar overall survival and receive more cost-effective care than those treated with SRS+WBRT. Compared with SRS+WBRT, initial management with SRS alone does not result in a higher average cost.


Subject(s)
Brain Neoplasms , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms/pathology , Radiosurgery/economics , Radiotherapy/economics , Adult , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Brain Neoplasms/secondary , Brain Neoplasms/surgery , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/secondary , Carcinoma, Non-Small-Cell Lung/surgery , Cost-Benefit Analysis , Cranial Irradiation/economics , Cranial Irradiation/mortality , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Models, Econometric , Patient Selection , Proportional Hazards Models , Radiosurgery/mortality , Radiotherapy/mortality , Retrospective Studies , Salvage Therapy/economics , Salvage Therapy/mortality
6.
Clin Breast Cancer ; 12(4): 259-63, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22658839

ABSTRACT

BACKGROUND: Limited data exist on the use of accelerated partial breast irradiation (APBI) in patients with ductal carcinoma in situ (DCIS). The purpose of this analysis was to evaluate clinical outcomes after APBI in patients with DCIS. PATIENTS AND METHODS: Between December 2002 and December 2010, 99 patients with DCIS underwent APBI as part of their breast-conserving therapy (BCT). Partial breast irradiation techniques included interstitial brachytherapy, balloon-based brachytherapy, and 3-dimensional conformal radiotherapy (3D-CRT). Clinical outcomes including local recurrence, regional recurrence, disease-free survival (DFS), cause-specific survival, and overall survival (OS) were analyzed. RESULTS: Mean follow up was 3.0 years, with a mean patient age of 61.8 years. At 5 years, the rates of local recurrence and regional recurrence were 1.4% and 0%, respectively. Overall survival was 94%, whereas cause-specific survival was 100%. No difference was noted in local control for each treatment technique. When comparing rates using the Eastern Cooperative Oncology Group (ECOG) E-5194 trial groupings, the rate of local recurrence in our cohort was 2.0% for patients with grade I/II disease < 2.5 cm and 0% for grade III < 1.0 cm, representing a 50% and 100% decrease, respectively, in local recurrence compared with excision alone. CONCLUSIONS: Patients with DCIS treated with APBI had excellent clinical outcomes regardless of the APBI technique used. Until the publication of prospective phase III trials, these data confirm previous reports highlighting the efficacy of APBI in the treatment of noninvasive carcinoma of the breast.


Subject(s)
Brachytherapy/methods , Breast Neoplasms/radiotherapy , Carcinoma, Intraductal, Noninfiltrating/radiotherapy , Radiotherapy, Conformal/methods , Aged , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Carcinoma, Intraductal, Noninfiltrating/pathology , Carcinoma, Intraductal, Noninfiltrating/surgery , Combined Modality Therapy , Disease-Free Survival , Female , Follow-Up Studies , Humans , Mastectomy, Segmental/methods , Middle Aged , Radiotherapy Dosage , Treatment Outcome , United States , Women's Health
7.
Mol Cancer Ther ; 9(3): 761-71, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20197402

ABSTRACT

Ionizing radiation is an important component of multimodal therapy for alveolar rhabdomyosarcoma (ARMS). We sought to evaluate the ability of IFN-beta to enhance the activity of ionizing radiation. Rh-30 and Rh-41 ARMS cells were treated with IFN-beta and ionizing radiation to assess synergistic effects in vitro and as orthotopic xenografts in CB17 severe combined immunodeficient mice. In addition to effects on tumor cell proliferation and xenograft growth, changes in the tumor microenvironment including interstitial fluid pressure, perfusion, oxygenation, and cellular histology were assessed. A nonlinear regression model and isobologram analysis indicated that IFN-beta and ionizing radiation affected antitumor synergy in vitro in the Rh-30 cell line; the activity was additive in the Rh-41 cell line. In vivo continuous delivery of IFN-beta affected normalization of the dysfunctional tumor vasculature of both Rh-30 and Rh-41 ARMS xenografts, decreasing tumor interstitial fluid pressure, increasing tumor perfusion (as assessed by contrast-enhanced ultrasonography), and increasing oxygenation. Tumors treated with both IFN-beta and radiation were smaller than control tumors and those treated with radiation or IFN-beta alone. Additionally, treatment with high-dose IFN-beta followed by radiation significantly reduced tumor size compared with radiation treatment followed by IFN-beta. The combination of IFN-beta and ionizing radiation showed synergy against ARMS by sensitizing tumor cells to the cytotoxic effects of ionizing radiation and by altering tumor vasculature, thereby improving oxygenation. Therefore, IFN-beta and ionizing radiation may be an effective combination for treatment of ARMS.


Subject(s)
Cell Proliferation/drug effects , Interferon-beta/pharmacology , Radiation Tolerance/drug effects , Rhabdomyosarcoma, Alveolar/radiotherapy , Tumor Burden/drug effects , Animals , Cell Line, Tumor , Humans , Male , Mice , Mice, SCID , Neovascularization, Pathologic/radiotherapy , Oxygen Consumption/drug effects , Radiation, Ionizing , Radiation-Sensitizing Agents/pharmacology , Recombinant Proteins/pharmacology , Rhabdomyosarcoma, Alveolar/blood supply , Rhabdomyosarcoma, Alveolar/pathology , Xenograft Model Antitumor Assays
8.
Int J Radiat Oncol Biol Phys ; 76(5): 1537-45, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20338480

ABSTRACT

PURPOSE: Ionizing radiation, an important component of glioma therapy, is critically dependent on tumor oxygenation. However, gliomas are notable for areas of necrosis and hypoxia, which foster radioresistance. We hypothesized that pharmacologic manipulation of the typically dysfunctional tumor vasculature would improve intratumoral oxygenation and, thus, the antiglioma efficacy of ionizing radiation. METHODS AND MATERIALS: Orthotopic U87 xenografts were treated with either continuous interferon-beta (IFN-beta) or bevacizumab, alone, or combined with cranial irradiation (RT). Tumor growth was assessed by quantitative bioluminescence imaging; the tumor vasculature using immunohistochemical staining, and tumor oxygenation using hypoxyprobe staining. RESULTS: Both IFN-beta and bevaziumab profoundly affected the tumor vasculature, albeit with different cellular phenotypes. IFN-beta caused a doubling in the percentage of area of perivascular cell staining, and bevacizumab caused a rapid decrease in the percentage of area of endothelial cell staining. However, both agents increased intratumoral oxygenation, although with bevacizumab, the effect was transient, being lost by 5 days. Administration of IFN-beta or bevacizumab before RT was significantly more effective than any of the three modalities as monotherapy or when RT was administered concomitantly with IFN-beta or bevacizumab or 5 days after bevacizumab. CONCLUSION: Bevacizumab and continuous delivery of IFN-beta each induced significant changes in glioma vascular physiology, improving intratumoral oxygenation and enhancing the antitumor activity of ionizing radiation. Additional investigation into the use and timing of these and other agents that modify the vascular phenotype, combined with RT, is warranted to optimize cytotoxic activity.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Glioma/blood supply , Neovascularization, Pathologic/drug therapy , Oxygen Consumption/drug effects , Radiation Tolerance/drug effects , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Bevacizumab , Cell Hypoxia/drug effects , Glioma/metabolism , Glioma/radiotherapy , Interferon-beta/pharmacology , Male , Mice , Mice, SCID , Transplantation, Heterologous
9.
Mol Cancer Ther ; 7(12): 3852-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19056675

ABSTRACT

Although temozolomide has shown clinical activity against neuroblastoma, this activity is likely limited by the DNA repair enzyme O6-methylguanine DNA methyltransferase (MGMT). We hypothesized that IFN-beta could sensitize neuroblastoma cells to the cytotoxic effects of temozolomide through its ability to down-regulate MGMT expression. In vitro proliferation of three neuroblastoma cell lines treated with IFN-beta and temozolomide alone or in combination was examined. Antitumor activity was assessed in both localized and disseminated neuroblastoma xenografts using single-agent and combination therapy, with continuous delivery of IFN-beta being established by a liver-targeted adeno-associated virus-mediated approach. Two neuroblastoma cell lines (NB-1691 and SK-N-AS) were found to have high baseline levels of MGMT expression, whereas a third cell line (CHLA-255) had low levels. Temozolomide had little effect on in vitro proliferation of the neuroblastoma cell lines with high MGMT expression, but pretreatment with IFN-beta significantly decreased MGMT expression and cell counts (NB-1691: 36 +/- 3% of control, P = 0.0008; SK-N-AS: 54 +/- 7% control, P = 0.003). In vivo, NB-1691 tumors in CB17-SCID mice treated with the combination of IFN-beta and temozolomide had lower MGMT expression and a significantly reduced tumor burden, both localized [percent initial tumor volume: 2,516 +/- 680% (control) versus 1,272 +/- 330% (temozolomide), P = 0.01; 1,348 +/- 220%, P = 0.03 (IFN-beta); 352 +/- 110%, P = 0.0001 (combo)] and disseminated [bioluminescent signal: control (1.32e10 +/- 6.5e9) versus IFN-beta (2.78e8 +/- 3.09e8), P = 0.025, versus temozolomide (2.06e9 +/- 1.55e9), P = 0.1, versus combination (2.13e7 +/- 7.67e6), P = 0.009]. IFN-beta appears to sensitize neuroblastoma cells to the cytotoxic effects of temozolomide through attenuation of MGMT expression. Thus, IFN-beta and temozolomide may be a useful combination for treating children with this difficult disease.


Subject(s)
Antineoplastic Agents/pharmacology , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Interferon-beta/metabolism , Interleukins/physiology , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Animals , Cell Line, Tumor , Dacarbazine/pharmacology , Humans , Male , Mice , Mice, SCID , Neoplasm Transplantation , Temozolomide
10.
J Pediatr Surg ; 43(1): 177-82; discussion 182-3, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18206478

ABSTRACT

PURPOSE: Trichostatin A (TSA) is a potent histone deacetylase inhibitor and has demonstrated significant antitumor activity against a variety of cancer cell lines. Type I interferons have also shown significant antitumor as well as antiangiogenic activity. In this study, we examined the effectiveness of combination therapy of TSA and interferon beta (IFN-beta) on human neuroblastoma cells in vitro and in vivo using a murine model of retroperitoneal neuroblastoma. MATERIALS AND METHODS: For in vitro experiments, plated human neuroblastoma cells (NB-1643 and NB-1691) were treated with vehicle or with IFN-beta, TSA, or both for 24 hours. Cytotoxicity was assessed by counting cells and expressing the results as a percentage of controls. Expression of the tumor suppressor p21(Waf1) was assessed by Western blot. For in vivo experiments, retroperitoneal neuroblastomas were established in severe combined immune deficiency (SCID) mice. Interferon beta was given using a gene therapy approach, administering 1.5 x 10(10) particles of an adeno-associated virus vector encoding human IFN-beta (AAV hIFN-beta) via tail vein as a single dose per mouse. Trichostatin A was given at a dose of 5 mg/kg every 48 hours subcutaneously. Treatment groups included controls, AAV hIFN-beta alone, TSA alone, and AAV hIFN-beta together with TSA. Tumor volume was assessed 2 weeks after the treatment began. RESULTS: After 24 hours, treatment with IFN-beta, TSA, and a combination of both resulted in a 45.3%, 68.1%, and 75% reduction in cell count relative to controls in the NB-1691 cell line. In the NB-1643 line, cell counts were reduced by 23%, 58%, and 62.3% respectively. In addition, NB-1691 cells treated with TSA showed increased expression of p21(Waf1) on Western blot. For in vivo experiments, control-, AAV hIFN-beta-, TSA-, and combination-treated tumors had the following final volumes: 1577.7 +/- 264.2 mm(3) (n = 3); 128.5 +/- 74.4 mm(3) (n = 4; P = .0001); 1248.7 +/- 673.9 mm(3) (n = 4; P = .48); and 127.5 +/- 36.8 mm(3) (n = 4; P = .0007), respectively. CONCLUSION: Neuroblastoma, because of its unique biology, continues to be a challenging tumor to treat, and many times these tumors are refractory to standard chemotherapeutic regimens. These data show that both TSA and IFN-beta inhibit neuroblastoma growth and that the combination may potentially provide a unique way to treat this difficult disease.


Subject(s)
Genetic Therapy/methods , Hydroxamic Acids/pharmacology , Interferon-beta/pharmacology , Neuroblastoma/therapy , Xenograft Model Antitumor Assays/methods , Adenoviridae , Animals , Blotting, Western , Cell Line, Tumor/cytology , Cell Line, Tumor/drug effects , Combined Modality Therapy , Disease Models, Animal , Humans , In Vitro Techniques , Male , Mice , Mice, SCID , Neuroblastoma/pathology , Probability , Random Allocation , Sensitivity and Specificity
11.
Scand J Infect Dis ; 35(9): 647-52, 2003.
Article in English | MEDLINE | ID: mdl-14620149

ABSTRACT

The variability of the course of infection by Streptococcus pneumoniae is well known but poorly understood. Most animal models of pneumonia, sepsis or meningitis have been forced to use site-specific bacterial inoculation to mimic localized human infection. This study examined the differences in the progression of disease-causing strains D39 (serotype 2), A66.1 (serotype 3) and TIGR4 (serotype 4) using isolates transformed with the Gram-positive lux transposon cassette, Tn4001 luxABCDE Km(r). Expression of the lux operon results in bioluminescence, permitting the detection of the bacteria within a living animal while using a CCD camera. Mice infected intranasally with A66.1 developed only pneumonia, those challenged with D39 experienced high-grade sepsis, while TIGR4 infection resulted in low-grade pneumonia and bacteremia ultimately progressing to meningitis. Quantitative analysis of bacterial titers confirmed these patterns, which were consistent across different lineages of mice. Mice anesthetized with ketamine and xylazine developed more severe forms of the disease compared with isoflurane. These studies unambiguously characterize 3 distinct models of the natural course of pneumococcal infection. Mapping these models provides a framework for detailed molecular modeling of pneumococcal virulence determinants at specific stages of disease.


Subject(s)
Pneumococcal Infections/physiopathology , Streptococcus pneumoniae/classification , Animals , Disease Models, Animal , Female , Lung/pathology , Mice , Mice, Inbred BALB C , Pneumococcal Infections/blood , Pneumococcal Infections/cerebrospinal fluid , Serotyping , Streptococcus pneumoniae/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...