Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.197
Filtrar
1.
JCO Clin Cancer Inform ; 8: e2300254, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38996196

RESUMEN

PURPOSE: Early prediction of response to immunotherapy may help guide patient management by identifying resistance to treatment and allowing adaptation of therapies. This analysis evaluated a mathematical model of response to immunotherapy that provides patient-specific prediction of outcome using the initial change in tumor size/burden from baseline to the first follow-up visit on standard imaging scans. METHODS: We applied the model to 600 patients with advanced solid tumors who received durvalumab in Study 1108, a phase I/II trial, and compared outcome prediction performance versus size-based criteria with RECIST version 1.1 best overall response (BOR), baseline circulating tumor (ct)DNA level, and other clinical/pathologic predictors of immunotherapy response. RESULTS: In multiple solid tumors, the mathematical parameter representing net tumor growth rate at the first on-treatment computed tomography (CT) scan assessed around 6 weeks after starting durvalumab (α1) had a concordance index to predict overall survival (OS) of 0.66-0.77 on multivariate analyses. This measurement of early tumor dynamics significantly improved multivariate OS models that included standard RECIST v1.1 criteria, baseline ctDNA levels, and other clinical/pathologic factors in predicting OS. Furthermore, α1 was assessed consistently at the first on-treatment CT scan, whereas all traditional RECIST BOR groups were confirmed only after this time. CONCLUSION: These results support further exploring α1 as an integral biomarker of response to immunotherapy. This biomarker may be predictive of further benefit and can be assessed before RECIST response groups can be assigned, potentially providing an opportunity to personalize oncologic management.


Asunto(s)
Anticuerpos Monoclonales , Neoplasias , Humanos , Anticuerpos Monoclonales/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Femenino , Masculino , Tomografía Computarizada por Rayos X/métodos , Resultado del Tratamiento , Antineoplásicos Inmunológicos/uso terapéutico , Persona de Mediana Edad , Modelos Teóricos , Pronóstico , Inmunoterapia/métodos , Anciano , Carga Tumoral
2.
J Immunother Cancer ; 12(7)2024 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-39032943

RESUMEN

Therapies targeting the programmed cell death protein-1/programmed death-ligand 1 (PD-L1) (abbreviated as PD-(L)1) axis are a significant advancement in the treatment of many tumor types. However, many patients receiving these agents fail to respond or have an initial response followed by cancer progression. For these patients, while subsequent immunotherapies that either target a different axis of immune biology or non-immune combination therapies are reasonable treatment options, the lack of predictive biomarkers to follow-on agents is impeding progress in the field. This review summarizes the current knowledge of mechanisms driving resistance to PD-(L)1 therapies, the state of biomarker development along this axis, and inherent challenges in future biomarker development for these immunotherapies. Innovation in the development and application of novel biomarkers and patient selection strategies for PD-(L)1 agents is required to accelerate the delivery of effective treatments to the patients most likely to respond.


Asunto(s)
Biomarcadores de Tumor , Humanos , Biomarcadores de Tumor/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/metabolismo , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia/métodos , Consenso
3.
Nat Commun ; 15(1): 5837, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38992034

RESUMEN

To inform clinical trial design and real-world precision pediatric oncology practice, we classified diagnoses, assessed the landscape of mutations, and identified genomic variants matching trials in a large unselected institutional cohort of solid tumors patients sequenced at Dana-Farber / Boston Children's Cancer and Blood Disorders Center. Tumors were sequenced with OncoPanel, a targeted next-generation DNA sequencing panel. Diagnoses were classified according to the International Classification of Diseases for Oncology (ICD-O-3.2). Over 6.5 years, 888 pediatric cancer patients with 95 distinct diagnoses had successful tumor sequencing. Overall, 33% (n = 289/888) of patients had at least 1 variant matching a precision oncology trial protocol, and 14% (41/289) were treated with molecularly targeted therapy. This study highlights opportunities to use genomic data from hospital-based sequencing performed either for research or clinical care to inform ongoing and future precision oncology clinical trials. Furthermore, the study results emphasize the importance of data sharing to define the genomic landscape and targeted treatment opportunities for the large group of rare pediatric cancers we encounter in clinical practice.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Difusión de la Información , Neoplasias , Medicina de Precisión , Humanos , Neoplasias/genética , Neoplasias/tratamiento farmacológico , Niño , Medicina de Precisión/métodos , Masculino , Preescolar , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Adolescente , Lactante , Mutación , Ensayos Clínicos como Asunto , Terapia Molecular Dirigida/métodos , Genómica/métodos , Recién Nacido
5.
Sci Rep ; 14(1): 12129, 2024 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802399

RESUMEN

Many targeted cancer therapies rely on biomarkers assessed by scoring of immunohistochemically (IHC)-stained tissue, which is subjective, semiquantitative, and does not account for expression heterogeneity. We describe an image analysis-based method for quantitative continuous scoring (QCS) of digital whole-slide images acquired from baseline human epidermal growth factor receptor 2 (HER2) IHC-stained breast cancer tissue. Candidate signatures for patient stratification using QCS of HER2 expression on subcellular compartments were identified, addressing the spatial distribution of tumor cells and tumor-infiltrating lymphocytes. Using data from trastuzumab deruxtecan-treated patients with HER2-positive and HER2-negative breast cancer from a phase 1 study (NCT02564900; DS8201-A-J101; N = 151), QCS-based patient stratification showed longer progression-free survival (14.8 vs 8.6 months) with higher prevalence of patient selection (76.4 vs 56.9%) and a better cross-validated log-rank p value (0.026 vs 0.26) than manual scoring based on the American Society of Clinical Oncology / College of American Pathologists guidelines. QCS-based features enriched the HER2-negative subgroup by correctly predicting 20 of 26 responders.


Asunto(s)
Neoplasias de la Mama , Selección de Paciente , Receptor ErbB-2 , Trastuzumab , Humanos , Femenino , Receptor ErbB-2/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Trastuzumab/uso terapéutico , Persona de Mediana Edad , Biomarcadores de Tumor/metabolismo , Adulto , Inmunoconjugados/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Anciano , Inmunohistoquímica , Camptotecina/análogos & derivados
6.
Mol Cancer ; 23(1): 115, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38811992

RESUMEN

BACKGROUND: We explored potential predictive biomarkers of immunotherapy response in patients with extensive-stage small-cell lung cancer (ES-SCLC) treated with durvalumab (D) + tremelimumab (T) + etoposide-platinum (EP), D + EP, or EP in the randomized phase 3 CASPIAN trial. METHODS: 805 treatment-naïve patients with ES-SCLC were randomized (1:1:1) to receive D + T + EP, D + EP, or EP. The primary endpoint was overall survival (OS). Patients were required to provide an archived tumor tissue block (or ≥ 15 newly cut unstained slides) at screening, if these samples existed. After assessment for programmed cell death ligand-1 expression and tissue tumor mutational burden, residual tissue was used for additional molecular profiling including by RNA sequencing and immunohistochemistry. RESULTS: In 182 patients with transcriptional molecular subtyping, OS with D ± T + EP was numerically highest in the SCLC-inflamed subtype (n = 10, median 24.0 months). Patients derived benefit from immunotherapy across subtypes; thus, additional biomarkers were investigated. OS benefit with D ± T + EP versus EP was greater with high versus low CD8A expression/CD8 cell density by immunohistochemistry, but with no additional benefit with D + T + EP versus D + EP. OS benefit with D + T + EP versus D + EP was associated with high expression of CD4 (median 25.9 vs. 11.4 months) and antigen-presenting and processing machinery (25.9 vs. 14.6 months) and MHC I and II (23.6 vs. 17.3 months) gene signatures, and with higher MHC I expression by immunohistochemistry. CONCLUSIONS: These findings demonstrate the tumor microenvironment is important in mediating better outcomes with D ± T + EP in ES-SCLC, with canonical immune markers associated with hypothesized immunotherapy mechanisms of action defining patient subsets that respond to D ± T. TRIAL REGISTRATION: ClinicalTrials.gov, NCT03043872.


Asunto(s)
Biomarcadores de Tumor , Inmunoterapia , Neoplasias Pulmonares , Carcinoma Pulmonar de Células Pequeñas , Humanos , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/patología , Carcinoma Pulmonar de Células Pequeñas/inmunología , Carcinoma Pulmonar de Células Pequeñas/terapia , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/mortalidad , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Neoplasias Pulmonares/metabolismo , Femenino , Masculino , Inmunoterapia/métodos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Persona de Mediana Edad , Anciano , Anticuerpos Monoclonales/uso terapéutico , Resultado del Tratamiento , Estadificación de Neoplasias , Anticuerpos Monoclonales Humanizados/uso terapéutico , Pronóstico , Adulto
8.
Surg Infect (Larchmt) ; 25(4): 315-321, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38696615

RESUMEN

Background: Surgical site complications (SSCs) are common, yet preventable hospital-acquired conditions. Single-use negative pressure wound therapy (sNPWT) has been shown to be effective in reducing rates of these complications. In the era of value-based care, strategic allocation of sNPWT is needed to optimize both clinical and financial outcomes. Materials and Methods: We conducted a retrospective analysis using data from the Premier Healthcare Database (2017-2021) for 10 representative open procedures in orthopedic, abdominal, cardiovascular, cesarean delivery, and breast surgery. After separating data into training and validation sets, various machine learning algorithms were used to develop pre-operative SSC risk prediction models. Model performance was assessed using standard metrics and predictors of SSCs were identified through feature importance evaluation. Highest-performing models were used to simulate the cost-effectiveness of sNPWT at both the patient and population level. Results: The prediction models demonstrated good performance, with an average area under the curve of 76%. Prominent predictors across subspecialities included age, obesity, and the level of procedure urgency. Prediction models enabled a simulation analysis to assess the population-level cost-effectiveness of sNPWT, incorporating patient and surgery-specific factors, along with the established efficacy of sNPWT for each surgical procedure. The simulation models uncovered significant variability in sNPWT's cost-effectiveness across different procedural categories. Conclusions: This study demonstrates that machine learning models can effectively predict a patient's risk of SSC and guide strategic utilization of sNPWT. This data-driven approach allows for optimization of clinical and financial outcomes by strategically allocating sNPWT based on personalized risk assessments.


Asunto(s)
Inteligencia Artificial , Terapia de Presión Negativa para Heridas , Infección de la Herida Quirúrgica , Humanos , Estudios Retrospectivos , Infección de la Herida Quirúrgica/prevención & control , Infección de la Herida Quirúrgica/economía , Infección de la Herida Quirúrgica/epidemiología , Terapia de Presión Negativa para Heridas/métodos , Terapia de Presión Negativa para Heridas/economía , Femenino , Persona de Mediana Edad , Masculino , Análisis Costo-Beneficio , Anciano , Aprendizaje Automático , Adulto , Medición de Riesgo/métodos
9.
J Am Acad Dermatol ; 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38697219

RESUMEN

Cystic fibrosis (CF) is caused by a mutation in the Cystic fibrosis transmembrane conductance regulator (CFTR) gene, and features recurrent sinus and pulmonary infections, steatorrhea, and malnutrition. CF is associated with diverse cutaneous manifestations, including transient reactive papulotranslucent acrokeratoderma of the palms, nutrient deficiency dermatoses, and vasculitis. Rarely these are presenting symptoms of CF, prior to pulmonary or gastrointestinal sequelae. Cutaneous drug eruptions are also highly common in patients with CF (PwCF) given frequent antibiotic exposure. Finally, CFTR modulating therapy, which has revolutionized CF management, is associated with cutaneous side effects ranging from acute urticaria to toxic epidermal necrolysis. Recognition of dermatologic clinical manifestations of CF is important to appropriately care for PwCF. Dermatologists may play a significant role in the diagnosis and management of CF and associated skin complications.

10.
Cancer ; 130(14): 2416-2439, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38687639

RESUMEN

Langerhans cell histiocytosis (LCH) is a myeloid neoplastic disorder characterized by lesions with CD1a-positive/Langerin (CD207)-positive histiocytes and inflammatory infiltrate that can cause local tissue damage and systemic inflammation. Clinical presentations range from single lesions with minimal impact to life-threatening disseminated disease. Therapy for systemic LCH has been established through serial trials empirically testing different chemotherapy agents and durations of therapy. However, fewer than 50% of patients who have disseminated disease are cured with the current standard-of-care vinblastine/prednisone/(mercaptopurine), and treatment failure is associated with long-term morbidity, including the risk of LCH-associated neurodegeneration. Historically, the nature of LCH-whether a reactive condition versus a neoplastic/malignant condition-was uncertain. Over the past 15 years, seminal discoveries have broadly defined LCH pathogenesis; specifically, activating mitogen-activated protein kinase pathway mutations (most frequently, BRAFV600E) in myeloid precursors drive lesion formation. LCH therefore is a clonal neoplastic disorder, although secondary inflammatory features contribute to the disease. These paradigm-changing insights offer a promise of rational cures for patients based on individual mutations, clonal reservoirs, and extent of disease. However, the pace of clinical trial development behind lags the kinetics of translational discovery. In this review, the authors discuss the current understanding of LCH biology, clinical characteristics, therapeutic strategies, and opportunities to improve outcomes for every patient through coordinated agent prioritization and clinical trial efforts.


Asunto(s)
Histiocitosis de Células de Langerhans , Humanos , Histiocitosis de Células de Langerhans/tratamiento farmacológico
11.
Lipids Health Dis ; 23(1): 94, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38566151

RESUMEN

BACKGROUND: Insulin-stimulated glucose uptake into skeletal muscle occurs via translocation of GLUT4 from intracellular storage vesicles to the plasma membrane. Elevated free fatty acid (FFA) availability via a lipid infusion reduces glucose disposal, but this occurs in the absence of impaired proximal insulin signalling. Whether GLUT4 localisation to the plasma membrane is subsequently affected by elevated FFA availability is not known. METHODS: Trained (n = 11) and sedentary (n = 10) individuals, matched for age, sex and body mass index, received either a 6 h lipid or glycerol infusion in the setting of a concurrent hyperinsulinaemic-euglycaemic clamp. Sequential muscle biopsies (0, 2 and 6 h) were analysed for GLUT4 membrane localisation and microvesicle size and distribution using immunofluorescence microscopy. RESULTS: At baseline, trained individuals had more small GLUT4 spots at the plasma membrane, whereas sedentary individuals had larger GLUT4 spots. GLUT4 localisation with the plasma membrane increased at 2 h (P = 0.04) of the hyperinsulinemic-euglycemic clamp, and remained elevated until 6 h, with no differences between groups or infusion type. The number of GLUT4 spots was unchanged at 2 h of infusion. However, from 2 to 6 h there was a decrease in the number of small GLUT4 spots at the plasma membrane (P = 0.047), with no differences between groups or infusion type. CONCLUSION: GLUT4 localisation with the plasma membrane increases during a hyperinsulinemic-euglycemic clamp, but this is not altered by elevated FFA availability. GLUT4 appears to disperse from small GLUT4 clusters located at the plasma membrane to support glucose uptake during a hyperinsulinaemic-euglycaemic clamp.


Asunto(s)
Ácidos Grasos no Esterificados , Glucosa , Humanos , Membrana Celular/metabolismo , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Insulina , Músculo Esquelético/metabolismo
12.
J Hosp Infect ; 147: 68-76, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38432585

RESUMEN

BACKGROUND: In response to identified gaps in infection prevention and control (IPC) training within Scotland, a Short Life Working Group initiated an innovative outbreak simulation training programme. AIM: To enhance the knowledge and confidence of medical microbiology and infectious diseases trainees and IPC professionals in managing healthcare-associated infection (HAI) outbreaks, employing the National Infection Prevention and Control Manual guidelines. METHODS: Participants completed prerequisite online training in epidemiology and surveillance before engaging in a meticulously crafted vancomycin-resistant enterococci outbreak simulation, which mirrored a real-life incident and adhered to the standards set by the Association for Simulated Practice in Healthcare. The programme incorporated Kolb's experiential learning cycle, fostering an authentic and engaging learning environment. A total of 41 individuals participated in the synchronous online training phase, with eight individuals involved in the pilot outbreak simulation. Evaluation of the training's efficacy followed Kirkpatrick's model, combining quantitative (five-point Likert scales) and qualitative (open-ended questions and participant reflections) data collection methods. FINDINGS: Results demonstrated significant improvements in participants' knowledge, skills, and confidence in outbreak management. Feedback highlighted the realism and educational value of the simulation, with 100% agreement on its efficacy in enhancing outbreak management capabilities. CONCLUSION: The success of this pilot study underscores the potential of simulation training in IPC and paves the way for broader implementation. It emphasizes the effectiveness of structured, experiential learning in equipping healthcare professionals with practical skills and confidence for managing complex HAI outbreaks, contributing to a more competent and prepared workforce.


Asunto(s)
Infección Hospitalaria , Brotes de Enfermedades , Control de Infecciones , Entrenamiento Simulado , Humanos , Proyectos Piloto , Escocia , Brotes de Enfermedades/prevención & control , Control de Infecciones/métodos , Entrenamiento Simulado/métodos , Infección Hospitalaria/prevención & control , Masculino , Femenino , Personal de Salud/educación , Adulto , Educación Médica/métodos
14.
Nat Commun ; 15(1): 1700, 2024 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-38402224

RESUMEN

The Ataxia telangiectasia and Rad3-related (ATR) inhibitor ceralasertib in combination with the PD-L1 antibody durvalumab demonstrated encouraging clinical benefit in melanoma and lung cancer patients who progressed on immunotherapy. Here we show that modelling of intermittent ceralasertib treatment in mouse tumor models reveals CD8+ T-cell dependent antitumor activity, which is separate from the effects on tumor cells. Ceralasertib suppresses proliferating CD8+ T-cells on treatment which is rapidly reversed off-treatment. Ceralasertib causes up-regulation of type I interferon (IFNI) pathway in cancer patients and in tumor-bearing mice. IFNI is experimentally found to be a major mediator of antitumor activity of ceralasertib in combination with PD-L1 antibody. Improvement of T-cell function after ceralasertib treatment is linked to changes in myeloid cells in the tumor microenvironment. IFNI also promotes anti-proliferative effects of ceralasertib on tumor cells. Here, we report that broad immunomodulatory changes following intermittent ATR inhibition underpins the clinical therapeutic benefit and indicates its wider impact on antitumor immunity.


Asunto(s)
Linfocitos T CD8-positivos , Indoles , Morfolinas , Neoplasias , Pirimidinas , Sulfonamidas , Humanos , Animales , Ratones , Antígeno B7-H1 , Microambiente Tumoral , Línea Celular Tumoral , Inmunoterapia , Modelos Animales de Enfermedad , Proteínas de la Ataxia Telangiectasia Mutada
15.
Nat Med ; 30(3): 716-729, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38351187

RESUMEN

For patients with non-small-cell lung cancer (NSCLC) tumors without currently targetable molecular alterations, standard-of-care treatment is immunotherapy with anti-PD-(L)1 checkpoint inhibitors, alone or with platinum-doublet therapy. However, not all patients derive durable benefit and resistance to immune checkpoint blockade is common. Understanding mechanisms of resistance-which can include defects in DNA damage response and repair pathways, alterations or functional mutations in STK11/LKB1, alterations in antigen-presentation pathways, and immunosuppressive cellular subsets within the tumor microenvironment-and developing effective therapies to overcome them, remains an unmet need. Here the phase 2 umbrella HUDSON study evaluated rational combination regimens for advanced NSCLC following failure of anti-PD-(L)1-containing immunotherapy and platinum-doublet therapy. A total of 268 patients received durvalumab (anti-PD-L1 monoclonal antibody)-ceralasertib (ATR kinase inhibitor), durvalumab-olaparib (PARP inhibitor), durvalumab-danvatirsen (STAT3 antisense oligonucleotide) or durvalumab-oleclumab (anti-CD73 monoclonal antibody). Greatest clinical benefit was observed with durvalumab-ceralasertib; objective response rate (primary outcome) was 13.9% (11/79) versus 2.6% (5/189) with other regimens, pooled, median progression-free survival (secondary outcome) was 5.8 (80% confidence interval 4.6-7.4) versus 2.7 (1.8-2.8) months, and median overall survival (secondary outcome) was 17.4 (14.1-20.3) versus 9.4 (7.5-10.6) months. Benefit with durvalumab-ceralasertib was consistent across known immunotherapy-refractory subgroups. In ATM-altered patients hypothesized to harbor vulnerability to ATR inhibition, objective response rate was 26.1% (6/23) and median progression-free survival/median overall survival were 8.4/22.8 months. Durvalumab-ceralasertib safety/tolerability profile was manageable. Biomarker analyses suggested that anti-PD-L1/ATR inhibition induced immune changes that reinvigorated antitumor immunity. Durvalumab-ceralasertib is under further investigation in immunotherapy-refractory NSCLC.ClinicalTrials.gov identifier: NCT03334617.


Asunto(s)
Antineoplásicos , Carcinoma de Pulmón de Células no Pequeñas , Indoles , Neoplasias Pulmonares , Morfolinas , Pirimidinas , Sulfonamidas , Humanos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Platino (Metal)/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Anticuerpos Monoclonales , Antineoplásicos/uso terapéutico , Biomarcadores , Antígeno B7-H1 , Microambiente Tumoral
16.
Prev Med Rep ; 38: 102588, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38283965

RESUMEN

Introduction: In the United States, safe, accessible drinking water is not equitable due to source water contamination, unreliable water treatment, or hazardous plumbing infrastructure. Drinking water free of lead, nitrates, and arsenic is vital for infant and young children's health. Methods: Researchers conducted a study combining single-case study review methods and economic evaluation for 6 US policies or programs. Researchers used case-study findings, activity-based costing, publicly available US population data, and existing literature to create 5-year cost projections (2020-2024) for strategies to address lead, nitrates, or arsenic in drinking water from private wells or community water systems for families with low incomes and young children aged 0-5y. Researchers estimated the number of households reached and the costs by activity and payer of implementing each policy or program using case-specific geographic location and eligibility criteria. Results: The total number of households reached varied from 295 to 135,000 depending on water source, population of focus, and geographic location. Focused strategies reached higher proportions of families with low incomes and young children. Community water system and state-wide strategies had the broadest reach. The total annual program cost per household that received information about their water quality ranged from $75 to $2,780. Of this cost, the portion paid by the household varied from $0.12 to $1,590, not including mitigation. Conclusions: These findings can inform local decisions about policies and programs in communities seeking to increase awareness and access to safer drinking water, particularly in homes of families with low incomes and young children.

17.
Cancer Cell ; 42(2): 209-224.e9, 2024 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-38215748

RESUMEN

Although immunotherapy with PD-(L)1 blockade is routine for lung cancer, little is known about acquired resistance. Among 1,201 patients with non-small cell lung cancer (NSCLC) treated with PD-(L)1 blockade, acquired resistance is common, occurring in >60% of initial responders. Acquired resistance shows differential expression of inflammation and interferon (IFN) signaling. Relapsed tumors can be separated by upregulated or stable expression of IFNγ response genes. Upregulation of IFNγ response genes is associated with putative routes of resistance characterized by signatures of persistent IFN signaling, immune dysfunction, and mutations in antigen presentation genes which can be recapitulated in multiple murine models of acquired resistance to PD-(L)1 blockade after in vitro IFNγ treatment. Acquired resistance to PD-(L)1 blockade in NSCLC is associated with an ongoing, but altered IFN response. The persistently inflamed, rather than excluded or deserted, tumor microenvironment of acquired resistance may inform therapeutic strategies to effectively reprogram and reverse acquired resistance.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Animales , Ratones , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Transducción de Señal , Inmunoterapia , Presentación de Antígeno , Antígeno B7-H1/metabolismo , Microambiente Tumoral
19.
Clin Pharmacol Ther ; 115(2): 349-360, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38010260

RESUMEN

This exploratory, post hoc analysis aimed to model circulating tumor DNA (ctDNA) dynamics and predict disease progression in patients with treatment-naïve locally advanced/metastatic epidermal growth factor receptor mutation (EGFRm)-positive non-small cell lung cancer, from the FLAURA trial (NCT02296125). Patients were randomized 1:1 and received osimertinib 80 mg once daily (q.d.) or comparator EGFR-TKIs (gefitinib 250 mg q.d. or erlotinib 150 mg q.d.). Plasma was collected at baseline and multiple timepoints until treatment discontinuation. Patients with Response Evaluation Criteria in Solid Tumors (RECIST) imaging data and detectable EGFR mutations (Ex19del/L858R) at baseline and ≥ 3 additional timepoints were evaluable. Joint modeling was conducted to characterize the relationship between longitudinal changes in ctDNA and probability of progression-free survival (PFS). A Bayesian joint model of ctDNA and PFS was developed solving differential equations with the ctDNA dynamics and the PFS time-to-event probability. Of 556 patients, 353 had detectable ctDNA at baseline. Evaluable patients (with available imaging and ≥ 3 additional timepoints, n = 320; ctDNA set) were divided into training (n = 259) and validation (n = 61) sets. In the validation set, the model predicted a median PFS of 17.7 months (95% confidence interval (CI): 11.9-28.3) for osimertinib (n = 23) and 9.1 months (95% CI: 6.3-14.8) for comparator (n = 38), consistent with observed RECIST PFS (16.4 months and 9.7, respectively). The model demonstrates that EGFRm ctDNA dynamics can predict the risk of disease progression in this patient population and could be used to predict RECIST-defined disease progression.


Asunto(s)
Acrilamidas , Compuestos de Anilina , Antineoplásicos , Carcinoma de Pulmón de Células no Pequeñas , ADN Tumoral Circulante , Indoles , Neoplasias Pulmonares , Pirimidinas , Humanos , Antineoplásicos/uso terapéutico , Teorema de Bayes , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , ADN Tumoral Circulante/genética , ADN Tumoral Circulante/uso terapéutico , Progresión de la Enfermedad , Receptores ErbB/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Mutación , Inhibidores de Proteínas Quinasas
20.
J Mol Diagn ; 26(1): 73-84, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37981090

RESUMEN

Comprehensive genotyping is necessary to identify therapy options for patients with advanced cancer; however, many cancers are not tested, partly because of tissue limitations. Next-generation sequencing (NGS) liquid biopsies overcome some limitations, but clinical validity is not established and adoption is limited. Herein, clinical bridging studies used pretreatment plasma samples and data from FLAURA (NCT02296125; n = 441) and AURA3 (NCT02151981; n = 450) pivotal studies to demonstrate clinical validity of Guardant360 CDx (NGS LBx) to identify patients with advanced EGFR mutant non-small-cell lung cancer who may benefit from osimertinib. The primary end point was progression-free survival (PFS). Patients with EGFR mutation as identified by NGS LBx had significant PFS benefit with first-line osimertinib over standard of care (15.2 versus 9.6 months; hazard ratio, 0.41; P < 0.0001) and with later-line osimertinib over chemotherapy (8.3 versus 4.2 months; hazard ratio, 0.34; P < 0.0001). PFS benefits were similar to the original trial cohorts selected by tissue-based EGFR testing. Analytical validation included accuracy, precision, limit of detection, and specificity. Analytical validity was established for EGFR mutation detection and pan-tumor profiling. Panel-wide limit of detection was 0.1% to 0.5%, with 98% to 100% per-sample specificity. Patients with EGFR mutant non-small-cell lung cancer by NGS LBx had improved PFS with osimertinib, confirming clinical validity. Analytical validity was established for guideline-recommended therapeutic targets across solid tumors. The resulting US Food and Drug Administration approval of NGS LBx demonstrated safety and effectiveness for its intended use and is expected to improve adherence to guideline-recommended targeted therapy use.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , Mutación , Biopsia Líquida , Secuenciación de Nucleótidos de Alto Rendimiento , Receptores ErbB/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...