Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 147
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Sci Rep ; 13(1): 12069, 2023 07 26.
Article in English | MEDLINE | ID: mdl-37495623

ABSTRACT

Metastasis is a leading cause of mortality in patients with lung adenocarcinoma. Histone deacetylases have emerged as promising targets for anti-tumor drugs, with histone deacetylase inhibitors (HDACi) being an active area of research. However, the precise mechanisms by which HDACi inhibits lung cancer metastasis remain incompletely understood. In this study, we employed a range of techniques, including qPCR, immunoblotting, co-immunoprecipitation, chromatin-immunoprecipitation, and cell migration assays, in conjunction with online database analysis, to investigate the role of HDACi and HDAC2/YY1 in the process of lung adenocarcinoma migration. The present study has demonstrated that both trichostatin A (TSA) and sodium butyrate (NaBu) significantly inhibit the invasion and migration of lung cancer cells via Histone deacetylase 2 (HDAC2). Overexpression of HDAC2 promotes lung cancer cell migration, whereas shHDAC2 effectively inhibits it. Further investigation revealed that HDAC2 interacts with YY1 and deacetylates Lysine 27 and Lysine9 of Histone 3, thereby inhibiting Cdh1 transcriptional activity and promoting cell migration. These findings have shed light on a novel functional mechanism of HDAC2/YY1 in lung adenocarcinoma cell migration.


Subject(s)
Adenocarcinoma of Lung , Antigens, CD , Cadherins , Histone Deacetylase 2 , Histone Deacetylase Inhibitors , Neoplasm Metastasis , YY1 Transcription Factor , Humans , Animals , Mice , Female , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/enzymology , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Cell Movement/drug effects , Butyric Acid/pharmacology , Butyric Acid/therapeutic use , Transforming Growth Factor beta/metabolism , Epithelial-Mesenchymal Transition/drug effects , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase 2/metabolism , YY1 Transcription Factor/metabolism , Cadherins/genetics , Cadherins/metabolism , Antigens, CD/metabolism , Protein Binding , Transcription, Genetic , Gene Expression Regulation, Neoplastic , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control
2.
Cancer Lett ; 567: 216261, 2023 07 28.
Article in English | MEDLINE | ID: mdl-37302563

ABSTRACT

Tumor-derived exosome (TDE)-mediated premetastatic niche (PMN) formation is a potential mechanism underlying the organotropic metastasis of primary tumors. Traditional Chinese medicine (TCM) has shown considerable success in preventing and treating tumor metastasis. However, the underlying mechanisms remain elusive. In this review, we discussed PMN formation from the perspectives of TDE biogenesis, cargo sorting, and TDE recipient cell alterations, which are critical for metastatic outgrowth. We also reviewed the metastasis-preventive effects of TCM, which act by targeting the physicochemical materials and functional mediators of TDE biogenesis, regulating the cargo sorting machinery and secretory molecules in TDEs, and targeting the TDE-recipient cells involved in PMN formation.


Subject(s)
Exosomes , Neoplasms , Humans , Exosomes/pathology , Medicine, Chinese Traditional , Tumor Microenvironment , Cell Communication , Neoplasms/drug therapy , Neoplasms/prevention & control , Neoplasms/pathology , Neoplasm Metastasis/pathology
3.
Dis Colon Rectum ; 64(12): 1463-1470, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34508014

ABSTRACT

BACKGROUND: Retrospective studies suggest that watch-and-wait is a safe alternative to total mesorectal excision in selected patients with a clinical complete response after chemoradiotherapy. OBJECTIVE: This study aimed to determine the proportion of patients with rectal cancer who may benefit from watch-and-wait. DESIGN: This study is a retrospective analysis of data from prospectively maintained databases. SETTING: This study was conducted at a comprehensive cancer center. PATIENTS: Consecutive patients with stage II or III rectal adenocarcinoma who were treated with total neoadjuvant therapy using induction chemotherapy between 2012 and 2019 under the care of the same surgeon were included. INTERVENTION: Induction-type total neoadjuvant therapy consisted of 8 cycles of leucovorin-fluorouracil-oxaliplatin or 5 cycles of capecitabine-oxaliplatin before chemoradiotherapy. Patients with a clinical complete response were offered watch-and-wait, and patients with residual tumor were offered total mesorectal excision. MAIN OUTCOMES AND MEASURES: Tumor response was assessed with a digital rectal examination, endoscopy, and MRI. Patient characteristics and recurrence-free survival were compared between the watch-and-wait group and the total mesorectal excision group. RESULTS: A total of 88 patients were included in the analysis. One (1%) died during neoadjuvant therapy. Fifty-five patients (62.5%) had an incomplete clinical response and underwent surgery, 10 (18%) of the 55 developed distant metastasis, and 3 (5%) developed local recurrence. The remaining 32 patients (36.3%) had a clinical complete response and underwent watch-and-wait. On average, patients in the watch-and-wait group were older and had smaller, more distal tumors compared with patients in the surgery group. The median radiation dose, number of chemotherapy cycles, rate of adverse events, and length of follow-up did not differ substantively between the total mesorectal excision group and the watch-and-wait group. In the watch-and-wait group, 2 (6%) patients developed tumor regrowth, and one of them had distant metastasis. Recurrence-free survival was significantly higher in the watch-and-wait group. LIMITATIONS: Generalizability, sample size, and follow-up duration were limitations of this study. CONCLUSIONS: Approximately one-third of patients with stage II or III rectal cancer can benefit from a watch-and-wait approach with the aim of preserving the rectum if treated with induction-type total neoadjuvant therapy and followed by an experienced multidisciplinary team. See Video Abstract at http://links.lww.com/DCR/B688. CONSERVACIN DE RGANOS EN PACIENTES CON CNCER DE RECTO TRATADOS CON TERAPIA NEOADYUVANTE TOTAL: ANTECEDENTES:Estudios retrospectivos sugieren que observar y esperar es una alternativa segura a la escisión mesorrectal total en pacientes seleccionados con una respuesta clínica completa después de la quimiorradioterapia.OBJETIVO:Determinar la proporción de pacientes con cáncer de recto que pueden beneficiarse de observar y esperar.DISEÑO:Análisis retrospectivo de datos de bases de datos mantenidas de forma prospectiva.ESCENARIO:Centro Oncológico Integral.PACIENTES:Pacientes consecutivos con adenocarcinoma de recto en estadio II o III tratados con TNT utilizando quimioterapia de inducción entre 2012 y 2019 bajo el cuidado del mismo cirujano.INTERVENCIÓN:La terapia neoadyuvante total de tipo inducción consistió en ocho ciclos de leucovorín-fluorouracilo-oxaliplatino o cinco ciclos de capecitabina-oxaliplatino antes de la quimiorradioterapia. A los pacientes con una respuesta clínica completa se les ofreció observar y esperar, y a los pacientes con tumor residual se les ofreció la escisión mesorrectal total.PRINCIPALES RESULTADOS Y MEDIDAS:La respuesta del tumor se evaluó con un tacto rectal, endoscopia y resonancia magnética. Se compararon las características de los pacientes y la supervivencia libre de recurrencia entre el grupo de observación y espera y el grupo de escisión mesorrectal total.RESULTADOS:Se incluyó en el análisis a un total de 88 pacientes. Uno (1%) murió durante la terapia neoadyuvante. Cincuenta y cinco pacientes (62.5%) tuvieron una respuesta clínica incompleta y se sometieron a cirugía; 10 (18%) de los 55 desarrollaron metástasis a distancia y 3 (5%) desarrollaron recidiva local. Los 32 pacientes restantes (36.3%) tuvieron una cCR (respuesta clínica completa) y se sometieron a observar y esperar. En promedio, los pacientes del grupo de observación y espera eran mayores y tenían tumores más pequeños y distales en comparación con el grupo de cirugía. La dosis mediana de radiación, el número de ciclos de quimioterapia, la tasa de eventos adversos y la duración del seguimiento no difirieron sustancialmente entre el grupo de escisión mesorrectal total y el grupo de observación y espera. En el grupo de observación y espera, 2 (6%) pacientes desarrollaron recrecimiento del tumor y uno de ellos tuvo metástasis a distancia. La supervivencia libre de recurrencia fue significativamente mayor en el grupo de observación y espera.LIMITACIONES:Generalizabilidad, tamaño de la muestra, duración del seguimiento.CONCLUSIONES:Aproximadamente un tercio de los pacientes con cáncer de recto en estadio II o III pueden beneficiarse de un abordaje de observación y espera con el objetivo de preservar el recto si se tratan con terapia neoadyuvante total de tipo inducción y son seguidos por un equipo multidisciplinario experimentado. Consulte Video Resumen en http://links.lww.com/DCR/B688.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Neoadjuvant Therapy/methods , Neoplasm, Residual/surgery , Adenocarcinoma/diagnosis , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Case-Control Studies , Chemoradiotherapy/methods , Disease-Free Survival , Female , Humans , Induction Chemotherapy/methods , Male , Middle Aged , Neoadjuvant Therapy/mortality , Neoplasm Metastasis/pathology , Neoplasm Recurrence, Local/epidemiology , Neoplasm Staging , Organ Preservation , Proctectomy/methods , Proctectomy/statistics & numerical data , Rectal Neoplasms/pathology , Retrospective Studies , Watchful Waiting
4.
FASEB J ; 35(9): e21834, 2021 09.
Article in English | MEDLINE | ID: mdl-34403553

ABSTRACT

Two distinct genetic mutational pathways characterized by either chromosomal instability or high-frequency microsatellite instability (MSI-H) are recognized in the pathogenesis of colorectal cancer (CRC). Recently, it has been shown that patients with primary CRC that displays MSI-H have a significant, stage-independent, multivariate survival advantage. Biological properties of CMS1 (MSI-H type) can affect therapeutic efficiencies of agents used in the treatment of CRC, and therefore become a new predictive factor of the treatment. But, the predictive impact of MSI-H status for adjuvant chemotherapy remains controversial. This study will assess whether there is any unnecessary or inappropriate use of treatment agents recommended for adjuvant therapy of stage 2 and 3 of disease and for palliative or curative treatment of liver metastatic disease in microsatellite instability high group, a molecular subtype of colon cancer. Within this scope, the efficiencies of fluorouracil- and oxaliplatin-based chemotherapeutic agents will be shown on stage 3 microsatellite instability high colon tumor cell lines first, and then a microfluidic model will be created, imitating the metastasis of colon cancer to the liver. In the microfluidic chip model, we will create in liver tissue, where the metastasis of microsatellite instability high colon cancer will be simulated; the effectiveness of chemotherapeutic agents, immunotherapy agents, and targeted agents on tumor cells as well as drug response will be assessed according to cell viability through released biomarkers from the cells. The proposed hypothesis study includes the modeling and treatment of patient-derived post-metastatic liver cancer in microfluidics which has priority at the global and our region and consequently develop personal medication.


Subject(s)
Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Lab-On-A-Chip Devices , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Microsatellite Instability , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/genetics , Antineoplastic Agents/therapeutic use , Chemotherapy, Adjuvant , Colonic Neoplasms/pathology , Drug Screening Assays, Antitumor , Fluorouracil/therapeutic use , Humans , Liver/drug effects , Liver/immunology , Liver/pathology , Liver Neoplasms/secondary , Models, Biological , Neoplasm Metastasis/pathology , Organ Specificity , Oxaliplatin/therapeutic use
5.
Clin Adv Hematol Oncol ; 19 Suppl 3(1): 1-20, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33843898

ABSTRACT

In the setting of metastatic colorectal cancer, many gains in patient outcomes have been achieved throughout the last 2 decades. A primary driver of these gains is access to more lines of therapy. In the palliative metastatic setting, all patients ultimately progress and require continued treatment sequencing. The goal is to expose patients to all lines of available therapies. It is now possible to better select patients for each therapy. Treatment selection algorithms encompass disease factors and patient characteristics, such as overall condition and age. Appropriate molecular profiling assessments should be available early in the treatment course, to drive decision-making and allow use of alternative therapies when possible. The transition to third-line therapy can be prompted by changes in imaging scans or laboratory tests, as well as changes in the patient's symptom burden. It can be problematic to delay initiation of third-line therapy when it is clinically indicated. Many oncologists will consider rechallenging patients with the same chemotherapy that did not work earlier. Although this strategy is reasonable, it should not necessarily take precedence over use of agents with proven efficacy in later lines of therapy in randomized clinical trials, such as regorafenib and trifluridine/tipiracil. Clinicians now commonly adjust the dose of regorafenib. A delay in the initiation of these third-line agents can allow the patient's performance status to decrease, thus diminishing the opportunity for a successful outcome.


Subject(s)
Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Neoplasm Metastasis/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Clinical Decision-Making , Disease Management , Drug Combinations , Humans , Neoplasm Metastasis/pathology , Palliative Care , Phenylurea Compounds/therapeutic use , Pyridines/therapeutic use , Pyrrolidines/therapeutic use , Thymine/therapeutic use , Trifluridine/therapeutic use
6.
Lima; Instituto Nacional de Salud; ene. 2021.
Non-conventional in Spanish | BRISA | ID: biblio-1354015

ABSTRACT

INTRODUCCIÓN: Este documento técnico se realiza a solicitud del Hospital Santa Rosa, a través de la Gerencia de Riesgos y Evaluación de las Prestaciones del Seguro Integral de Salud; la pregunta PICO fue la siguiente: P: adultos con cáncer colorrectal metastásico de localización izquierda y genes RAS no mutados; I: cetuximab + quimioterapia (C+Qt); C: quimioterapia (Qt) o panitumumab + quimioterapia (P+Qt); O: Sobrevida global, sobrevida libre de progresión, tasa de respuesta y eventos adversos. a. Cuadro clínico: En Perú, el cáncer colorrectal constituye el quinto tipo de cáncer más frecuente y el sexto con mayor mortalidad. En el diagnóstico inicial, un 25% de pacientes presenta enfermedad metastásica distante. En los últimos veinte años, la sobrevida global de pacientes con cáncer colorrectal metastásico se ha extendido, gracias a la aprobación de nuevas terapias administradas en combinación con agentes citotóxicos y la identificación de mutaciones genéticas que pueden predecir la respuesta al tratamiento. b. Tecnología sanitaria: Cetuximab es un antagonista del receptor de factor del crecimiento epidérmico, indicado para el tratamiento del cáncer colorrectal metastásico en tumores con sobreexpresión del EGFR y de tipo KRAS no mutado. Actúa mediante inhibición del crecimiento celular, inducción de apoptosis y disminución de la producción de metaloproteinasas de matriz y del factor de crecimiento endotelial vascular. Se recomienda una dosis inicial de 400 mg/m2, seguida de dosis semanales de 250 mg/m2, administradas en monoterapia o en combinación con quimioterapia. Cuenta con aprobación de la FDA desde el año 2004. En Perú, cuenta con dos registros sanitarios con vigencia prorrogada provisional. OBJETIVO: Describir la evidencia científica disponible sobre la eficacia y seguridad de cetuximab para el tratamiento en primera línea de pacientes con cáncer colorrectal metastásico de localización izquierda y genes RAS no mutados. METODOLOGÍA: Búsqueda sistemática en Medline, EMBASE, The Cochrane Library y LILACS, complementada con la búsqueda en páginas de agencias gubernamentales y buscadores genéricos. La calidad se valoró usando AMSTAR 2 para revisiones sistemáticas (RS), la herramienta de la colaboración Cochrane para ensayos clínicos y AGREE II para las Guías de Práctica Clínica (GPC). RESULTADOS: Sobrevida global: C+Qt mejoró la sobrevida global, en comparación con solo Qt, tanto en combinación con FOLFIRI (hazard ratio [HR]: 0,65; IC 95%: 0,50 a 0,86; 28,7 meses vs. 21,7 meses), como con FOLFOX (HR: 0,69; IC 95%: 0,53 a 0,90; 22 meses vs. 18,7 meses). No se observó diferencias entre C+Qt y P+Qt. Sobrevida libre de progresión: C+Qt mejoró la sobrevida global, en comparación con solo Qt, tanto en combinación con FOLFIRI (HR: 0,50; IC 95%: 0,34 a 0,72; 12 meses vs. 8,9 meses) como con FOLFOX (HR: 0,68; IC 95%: 0,50 a 0,91; 9,2 meses vs. 7,6 meses). No se observó diferencias entre C+Qt y P+Qt. Tasa de respuesta objetiva: C+Qt produjo una tasa de respuesta objetiva significativamente más alta, en comparación con solo Qt, tanto en combinación con FOLFIRI (odds ratio [OR]: 3,99; IC 95%: 2,40 a 6,62), como con FOLFOX (OR: 2,60; IC 95%: 1,64 a 4,14). No se observó diferencias entre C+Qt y P+Qt. Eventos adversos grado: C+Qt incrementó hasta el doble la probabilidad de sufrir algún evento adverso, en comparación con solo Qt, con un riesgo incrementado de toxicidad dermatológica (RR: 20,76; IC 95%: 3,87 a 111,33), diarrea (RR: 1,48; IC 95%: 1,33 a 1,64), hipertensión (RR: 1,69; IC 95%: 1,17 a 2,46), anorexia (RR: 1,57; IC 95%: 1,18 a 2,10) y mucositis/estomatitis (RR: 2,69; IC 95%: 1,90 a 3,80). Recomendaciones en GPC: Sólo NCCN y ASCO incluyen recomendaciones específicas para cáncer colorrectal metastásico con tumores RAS no mutadas de localización izquierda. Ambas incluyen como opciones de tratamiento a C+Qt y P+Qt. Evaluaciones de Tecnología Sanitaria: Dos ETS nacionales no recomiendan dar cobertura a C+Qt en cáncer colorrectal metastásico RAS de tipo salvaje, citando aspectos relacionados con su eficacia y costo-efectividad. NICE recomienda, tanto C+Qt como P+Qt sin especificar la lateralidad del tumor. Evaluación de la calidad metodológica: Dos RS fueron consideradas como nivel de confianza bajo y dos como nivel de confianza críticamente bajo. Las GPC obtuvieron un puntaje en la valoración global de calidad que varío entre 60,1% y 84,6%. CONCLUSIONES: La evidencia sobre el uso de C+Qt para cáncer colorrectal de lado izquierdo es limitada. Los desenlaces de eficacia que comparan C+Qt provienen de análisis de subgrupos no especificados de dos ensayos clínicos, mientras que los desenlaces de seguridad provienen de evidencia que no considera la lateralidad del tumor. Las comparaciones entre C+Qt y P+Qt proceden de meta-análisis en red de comparaciones indirectas. En pacientes con cáncer colorrectal metastásico de lado izquierdo con genes RAS no mutados, la adición de cetuximab a la quimioterapia incrementó la sobrevida global alrededor de 7 meses cuando se combinó con FOLFIRI y alrededor de 3,3 meses cuando se combinó con FOLFOX. No se hallaron diferencias entre cetuximab y panitumumab. La sobrevida libre de progresión se incrementó en alrededor de 3,1 meses tras la adición de cetuximab a FOLFIRI y alrededor de 1,6 meses cuando se adicionó a FOLFOX. No se hallaron diferencias entre cetuximab y panitumumab. Los pacientes tratados con cetuximab más quimioterapia tuvieron el doble de probabilidad de sufrir algún evento adverso, en comparación con solo quimioterapia, siendo común observar un riesgo incrementado de presentar toxicidad dermatológica, diarrea, hipertensión, anorexia y mucositis/dermatitis. Cetuximab y panitumumab presentaron diferente perfil de eventos adversos. Sólo las GPC de NCCN y ASCO incluyen recomendaciones específicas para cáncer colorrectal metastásico con tumores RAS no mutados de localización izquierda. Ambas GPC incluyen como opciones de tratamiento a cetuximab y panitumumab acompañados por quimioterapia. Dos ETS nacionales recomiendan no dar cobertura a C+Qt en pacientes con cáncer colorrectal metastásico con tumores RAS de tipo salvaje, citando aspectos relacionados con su eficacia y costo-efectividad. La ETS de NICE recomienda, tanto C+Qt como P+Qt, para cáncer colorrectal metastásico con tumores RAS de tipo salvaje sin especificar la lateralidad del tumor. Dos RS fueron consideradas como nivel de confianza bajo y dos como nivel de confianza críticamente bajo. Las GPC obtuvieron un puntaje en la valoración global de calidad que varío entre 60,1% y 84,6%. Ambas RS fueron consideradas como nivel de confianza bajo. Las GPC incluidas obtuvieron un puntaje en la valoración global de calidad que varío grandemente entre 60,1% y 84,6%.


Subject(s)
Colorectal Neoplasms/drug therapy , Genes, ras , Cetuximab/therapeutic use , Neoplasm Metastasis/pathology , Efficacy , Cost-Benefit Analysis
7.
Oncol Res ; 28(7): 685-700, 2021 Sep 07.
Article in English | MEDLINE | ID: mdl-32886059

ABSTRACT

Curcumol (Cur), isolated from the Traditional Chinese Medical plant Rhizoma Curcumae, is the bioactive component of sesquiterpene reported to possess antitumor activity. However, its bioactivity and mechanisms against lung adenocarcinoma are still unclear. We investigated its effect on lung adenocarcinoma and elucidated its underlying molecular mechanisms. In vitro, Cur effectively suppressed proliferation, migration, and invasion of lung adenocarcinoma cells A549 and H460, which were associated with the altered expressions of signaling molecules, including p-AKT, p-PI3K, p-LRP5/6, AXIN, APC, GSK3 and p--catenin, matrix metalloproteinase (MMP)-2, and MMP-9. Furthermore, Cur significantly induced cell apoptosis of A549 and H460 by promoting the expression of Bax, caspase 3, and caspase 9 and suppressing the expression of Bcl-2, and arrested the cell cycle at the G0/G1 phase by lowering the levels of cyclin D1, CDK1, and CDK4. In vivo experiment revealed that Cur could inhibit lung tumor growth and lung metastasis, which were consistent with these in vitro results. In xenograft model mice, Cur strongly decreased tumor weight and tumor volume, which may be related to the downregulation of p-AKT and p-PI3K by immunofluorescence analysis. In addition, a lung metastasis model experiment suggested that Cur dramatically decreased the ratio of lung/total weight, tumor metastatic nodules, and the expressions of MMP-2 and MMP-9 in lung tissues compared with the control. Overall, these data suggested that the inhibitory activity of Cur on lung adenocarcinoma via the inactivation of PI3K/Akt and Wnt/-catenin pathways, at least in part, indicates that curcumol may be a potential antitumor agent for lung adenocarcinoma therapy.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Lung Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sesquiterpenes/pharmacology , Wnt Signaling Pathway/drug effects , A549 Cells , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Catenins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Curcuma/chemistry , Drugs, Chinese Herbal/pharmacology , Female , Glycogen Synthase Kinase 3/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Neoplasm Metastasis/pathology
8.
J Nutr Biochem ; 89: 108556, 2021 03.
Article in English | MEDLINE | ID: mdl-33249185

ABSTRACT

Prostate cancer (PCa) is a very prevalent male-specific malignancy; most PCa patients eventually die as a result of metastasis. L-theanine (C7H14N2O3), a nonprotein amino acid derivative from green tea leaves, has been demonstrated to act as an anticarcinogen through proapoptotic and antiproliferative effects. However, the antimetastatic effect of L-theanine in tumor cells and its underlying mechanism are still unclear. Here, we found that L-theanine could suppress invasion, migration, and increase cell-cell adhesion of prostate cancer cells in vitro and in vivo. We also found that L-theanine could inhibit the epithelial-mesenchymal transition process in PCa. Our study revealed that L-theanine could downregulate MMP9, N-cadherin, Vimentin, Snail, and upregulate E-cadherin. Furthermore, L-theanine suppressed the transcription of MMP9 and Snail by significantly inhibiting the ERK/NF-κB signaling pathway and the binding activity of p65 to the promoter regions of MMP9 and Snail. All of these findings suggest that L-theanine has therapeutic potential for metastatic PCa and may be considered a promising candidate for antimetastatic therapy of prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Glutamates/pharmacology , Matrix Metalloproteinase 9/metabolism , Neoplasm Metastasis/pathology , Prostatic Neoplasms/pathology , Snail Family Transcription Factors/metabolism , Animals , Antineoplastic Agents/metabolism , Cadherins/metabolism , Cell Movement/drug effects , Down-Regulation , Epithelial-Mesenchymal Transition/drug effects , Glutamates/metabolism , Humans , Male , Mice , NF-kappa B/metabolism , PC-3 Cells , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Signal Transduction/drug effects , Tea/chemistry , Vimentin/metabolism
9.
Biomolecules ; 10(12)2020 12 10.
Article in English | MEDLINE | ID: mdl-33321708

ABSTRACT

Metastasis represents a major obstacle in cancer treatment and the leading cause of cancer-related deaths. Therefore, the identification of compounds targeting the multi-step and complex process of metastasis could improve outcomes in the management of cancer patients. Carotenoids are naturally occurring pigments with a plethora of biological activities. Carotenoids exert a potent anti-cancer capacity in various cancer models in vitro and in vivo, mediated by the modulation of signaling pathways involved in the migration and invasion of cancer cells and metastatic progression, including key regulators of the epithelial-mesenchymal transition and regulatory molecules, such as matrix metalloproteinases (MMPs), tissue inhibitors of metalloproteinases (TIMPs), urokinase plasminogen activator (uPA) and its receptor (uPAR), hypoxia-inducible factor-1α (HIF-1α), and others. Moreover, carotenoids modulate the expression of genes associated with cancer progression and inflammatory processes as key mediators of the complex process involved in metastasis. Nevertheless, due to the predominantly preclinical nature of the known anti-tumor effects of carotenoids, and unclear results from certain carotenoids in specific cancer types and/or specific parts of the population, a precise analysis of the anti-cancer effects of carotenoids is essential. The identification of carotenoids as effective compounds targeting the complex process of cancer progression could improve the outcomes of advanced cancer patients.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Carotenoids/therapeutic use , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Metastasis/drug therapy , Neoplasms/drug therapy , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/classification , Carotenoids/chemistry , Carotenoids/classification , Chemotherapy, Adjuvant , Epithelial-Mesenchymal Transition/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Machine Learning , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Precision Medicine , Receptors, Urokinase Plasminogen Activator/genetics , Receptors, Urokinase Plasminogen Activator/metabolism , Signal Transduction , Tissue Inhibitor of Metalloproteinases/genetics , Tissue Inhibitor of Metalloproteinases/metabolism , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
10.
CPT Pharmacometrics Syst Pharmacol ; 9(11): 628-638, 2020 11.
Article in English | MEDLINE | ID: mdl-33015996

ABSTRACT

This study aimed to explore the currently competing and new semimechanistic clearance models for monoclonal antibodies and the impact of clearance model misspecification on exposure metrics under different study designs exemplified for cetuximab. Six clearance models were investigated under four different study designs (sampling density and single/multiple-dose levels) using a rich data set from two cetuximab clinical trials (226 patients with metastatic colorectal cancer) and using the nonlinear mixed-effects modeling approach. A two-compartment model with parallel Michaelis-Menten and time-decreasing linear clearance adequately described the data, the latter being related to post-treatment response. With respect to bias in exposure metrics, the simplified time-varying linear clearance (CL) model was the best alternative. Time-variance of the linear CL component should be considered for biotherapeutics if response impacts pharmacokinetics. Rich sampling at steady-state was crucial for unbiased estimation of Michaelis-Menten elimination in case of the reference (parallel Michaelis-Menten and time-varying linear CL) model.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacokinetics , Biological Therapy/methods , Cetuximab/pharmacokinetics , Colorectal Neoplasms/drug therapy , Administration, Intravenous , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/therapeutic use , Cetuximab/administration & dosage , Cetuximab/therapeutic use , Colorectal Neoplasms/genetics , Colorectal Neoplasms/secondary , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Female , Humans , Kinetics , Linear Models , Male , Medical Oncology/statistics & numerical data , Middle Aged , Models, Biological , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/pathology , Nonlinear Dynamics
11.
Theranostics ; 10(23): 10652-10664, 2020.
Article in English | MEDLINE | ID: mdl-32929372

ABSTRACT

Rationale: Tumors are commonly treated by resection, which usually leads to massive hemorrhage and tumor cell residues, thereby increasing the risk of local recurrence and distant metastasis. Methods: Herein, an intelligent 3D-printed poly(lactic-co-glycolic acid), gelatin, and chitosan scaffold loaded with anti-cancer drugs was prepared that showed hemostatic function and good pH sensitivity. Results: Following in situ implantation in wounds, the scaffolds absorbed hemorrhage and cell residues after surgery, and promoted wound healing. In an in vivo environment, the scaffold responded to the slightly acidic environment of the tumor to undergo sustained drug release to significantly inhibit the recurrence and growth of the tumor, and reduced drug toxicity, all without causing damage to healthy tissues and with good biocompatibility. Conclusions: The multifunctional intelligent scaffold represents an excellent treatment modality for breast cancer following resection, and provides great potential for efficient cancer therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/therapy , Drug Carriers/chemistry , Neoplasm Recurrence, Local/prevention & control , Smart Materials/chemistry , Surgical Wound/therapy , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Chemotherapy, Adjuvant/methods , Chitosan/chemistry , Doxycycline/administration & dosage , Drug Compounding/methods , Female , Fluorouracil/administration & dosage , Gelatin/chemistry , Humans , Mammary Glands, Animal/pathology , Mammary Glands, Animal/surgery , Mastectomy , Mice , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Printing, Three-Dimensional , Tissue Scaffolds/chemistry , Wound Healing/drug effects , Xenograft Model Antitumor Assays
12.
Pathol Res Pract ; 216(9): 153082, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32825950

ABSTRACT

BACKGROUND: Curcumin is a natural phytochemical polyphenol with significant anti-cancer effects and negligible side effects. In this study, the therapeutic capacity of nanomicellar-curcumin for treating lung metastasis was evaluated in an immunocompetent mouse model of metastatic melanoma. MARTIALS AND METHODS: Two doses of nanomicellar-curcumin (i.e. 10 and 20 µM) were used to induce cytotoxicity in 3 melanoma cell lines. A total of 60 mice were allocated to 20 mice in each of three groups (10 for survival and 10 for assays). Groups were no treatment control, PBS control, nanomicellar-curcumin 20 mg/kg IP 4 times a week, for three weeks). Immunohistochemistry, TUNEL assay, and Western blots were used on lung samples. RESULTS: Nanomicellar-curcumin inhibited the in vitro growth of B16 F10 melanoma cells at 20 µM over 72 h. In vivo, 20 mg/kg nanomicellar-curcumin injected IP, delayed tumor cell growth and significantly extended mouse survival rate. Tumor infiltration of regulatory T cells and angiogenesis were reduced, while IFN-γ and CXCL10 were increased. CONCLUSION: Nanomicellar-curcumin can inhibit lung metastasis and growing melanoma via activation of apoptosis, activated T cells and inhibition of angiogenesis, tumor growth and regulatory T cells.


Subject(s)
Apoptosis/drug effects , Curcumin/pharmacology , Lung Neoplasms/drug therapy , Neoplasm Metastasis/pathology , T-Lymphocytes, Regulatory/metabolism , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Humans , Lung Neoplasms/metabolism , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Neovascularization, Pathologic/drug therapy , T-Lymphocytes, Regulatory/drug effects
13.
Biochim Biophys Acta Rev Cancer ; 1874(1): 188380, 2020 08.
Article in English | MEDLINE | ID: mdl-32461135

ABSTRACT

Cellular communication through gap junctions and hemichannels formed by connexins and through channels made by pannexins allows for metabolic cooperation and control of cellular activity and signalling. These channel proteins have been described to be tumour suppressors that regulate features such as cell death, proliferation and differentiation. However, they display cancer type-dependent and stage-dependent functions and may facilitate tumour progression through junctional and non-junctional pathways. The accumulated knowledge and emerging strategies to target connexins and pannexins are providing novel clinical opportunities for the treatment of cancer. Here, we provide an updated overview of the role of connexins and pannexins in malignant melanoma. We discuss how targeting of these channel proteins may be used to potentiate antitumour effects in therapeutic settings, including through improved immune-mediated tumour elimination.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Connexins/metabolism , Melanoma/secondary , Skin Neoplasms/pathology , Skin/pathology , Animals , Antineoplastic Agents, Immunological/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/immunology , Carcinogenesis/pathology , Cell Communication/drug effects , Cell Communication/immunology , Cell Line, Tumor , Connexins/agonists , Connexins/antagonists & inhibitors , Disease Models, Animal , Disease Progression , Gap Junctions/drug effects , Gap Junctions/pathology , Host Microbial Interactions/drug effects , Host Microbial Interactions/immunology , Humans , Melanoma/drug therapy , Melanoma/immunology , Melanoma/mortality , Microbiota/immunology , Neoplasm Invasiveness/immunology , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control , Neoplasm Staging , Signal Transduction/drug effects , Signal Transduction/immunology , Skin/cytology , Skin/microbiology , Skin Neoplasms/drug therapy , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
14.
Am J Chin Med ; 48(3): 703-718, 2020.
Article in English | MEDLINE | ID: mdl-32329642

ABSTRACT

Cinobufacini is a well-known Chinese medicine extracted from Venenum Bufonis, also called Chan Su. It has been used clinically for various cancers, including colon cancer. However, the function of Cinobufacini on colon cancer invasion and metastasis, and its underlying molecular mechanism, is still not clear. In this study, we investigated the function and mechanism of Cinobufacini on colon cancer invasion and metastasis both in vitro and in vivo studies. Human colon cancer cells were cultured. CCK assay was used to detect the effect of Cinobufacini on colon cancer cells proliferation. The invasion and migration abilities were observed by transwell assays, and the expression of invasion and migration related genes MMP2, MMP9, and epithelial-to-mesenchymal transition (EMT) relate genes were observed by Western blot assays. An orthotopic xenograft model in nude mice was established using colon cancer HCT116 cells, and the function of Cinobufacini on colon cancer invasion and metastasis were observed in vivo. We found Cinobufacini significantly inhibited colon cancer cell proliferation in a dose/time-dependent manner; the invasion and migration abilities of colon cancer were decreased after treated with Cinobufacini. The metastasis and EMT related genes MMP9, MMP2, N-cadherin and Snail were obviously down-regulated, while the expression of E-cadherin was up-regulated after treatment with Cinobufacini. The Wnt/ß-catenin signaling pathway related genes were observed using WB,and results show that the expression of ß-catenin, wnt3a, c-myc, cyclin D1, and MMP7 were all down-regulated after being treated with cinobufacini, while the expression of APC was up-regulated. In vivo studies of the volume and weight of orthotopic xenograft tumors showed significantly shrinkage in the Cinobufacini group compared to the control group. The enterocoelia and liver metastasis tumors were significantly decreased, and the expression of MMP9, MMP2, and ß-catenin were also down-regulated, while E-cadherin was up-regulated in vivo after the treatment with Cinobufacini. Our data proves that Cinobufacini can inhibit colon cancer invasion and metastasis both in vitro and in vivo; the mechanism is related by suppressing the Wnt/ß-catenin signaling pathway and then inhibiting the EMT of CRC.


Subject(s)
Amphibian Venoms/pharmacology , Amphibian Venoms/therapeutic use , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Neoplasm Metastasis/pathology , Phytotherapy , Signal Transduction/drug effects , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Cell Proliferation/drug effects , Colonic Neoplasms/genetics , HCT116 Cells , Humans , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis/genetics
15.
Adv Exp Med Biol ; 1220: 61-80, 2020.
Article in English | MEDLINE | ID: mdl-32304080

ABSTRACT

Metastasis is the major cause of breast cancer death worldwide. In metastatic breast cancer, circulating tumor cells (CTCs) can be captured from patient blood samples sequentially over time and thereby serve as surrogates to assess the biology of surviving cancer cells that may still persist in solitary or multiple metastatic sites following treatment. CTCs may thus function as potential real-time decision-making guides for selecting appropriate therapies during the course of disease or for the development and testing of new treatments. The heterogeneous nature of CTCs warrants the use of single cell platforms to better inform our understanding of these cancer cells. Current techniques for single cell analyses and techniques for investigating interactions between cancer and immune cells are discussed. In addition, methodologies for growing patient-derived CTCs in vitro or propagating them in vivo to facilitate CTC drug testing are reviewed. We advocate the use of CTCs in appropriate microenvironments to appraise the effectiveness of cancer chemotherapies, immunotherapies, and for the development of new cancer treatments, fundamental to personalizing and improving the clinical management of metastatic breast cancer.


Subject(s)
Breast Neoplasms/pathology , Drug Evaluation, Preclinical , Neoplasm Metastasis/pathology , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Precision Medicine , Single-Cell Analysis , Biomarkers, Tumor , Breast Neoplasms/diagnosis , Breast Neoplasms/therapy , Clinical Decision-Making , Humans , Neoplasm Metastasis/diagnosis
16.
Histopathology ; 77(2): 284-292, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32285460

ABSTRACT

AIMS: Perineural invasion (PNI) by prostatic adenocarcinoma is debated as a prognostic parameter. This study investigates the prognostic predictive value of PNI in a series of patients with locally advanced prostate cancer treated with radiotherapy and androgen deprivation using 10 years outcome data from the TROG 03.04 RADAR trial. METHODS: Diagnostic prostate biopsies from 976 patients were reviewed and the presence of PNI noted. Patients were followed for 10 years according to the trial protocol or until death. The primary endpoint for the study was time to bone metastasis. Secondary endpoints included time to soft tissue metastasis, transition to castration resistance, prostate cancer-specific mortality and all-cause mortality. RESULTS: PNI was detected in 449 cases (46%), with 234 cases (24%) having PNI in more than one core. The presence of PNI was significantly associated with higher ISUP grade, clinical T staging category, National Comprehensive Cancer Network risk group, and percent positive biopsy cores. The cumulative probability of bone metastases according to PNI status was significant over the 10 years follow-up interval of the study (log-rank test P < 0.0001). PNI was associated with all endpoints on univariable analysis. After adjusting for baseline clinicopathological and treatment factors, bone metastasis was the only endpoint in which PNI retained its prognostic significance (hazard ratio 1.42, 95% confidence interval 1.05-1.92, P = 0.021). CONCLUSIONS: The association between PNI and the development of bone metastases supports the inclusion of this parameter as a component of the routine histology report. Further this association suggests that evaluation of PNI may assist in selecting those patients who should be monitored more closely during follow-up.


Subject(s)
Adenocarcinoma/pathology , Peripheral Nerves/pathology , Prostatic Neoplasms/pathology , Adenocarcinoma/complications , Aged , Biopsy, Needle , Bone Neoplasms/etiology , Bone Neoplasms/pathology , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Prognosis , Prostate/pathology , Prostate-Specific Antigen , Prostatic Neoplasms/complications
17.
Curr Drug Metab ; 21(1): 6-10, 2020.
Article in English | MEDLINE | ID: mdl-31987018

ABSTRACT

BACKGROUND: Genistein being a phytoestrogen imitates the characteristics of estrogen, which can be useful to treat conditions by reducing the estrogen levels at the time of menopause, osteoporosis and high risk for breast cancer. OBJECTIVE: The superior binding of genistein to ERß might help in reducing breast malignancy risk. CONCLUSION: Genistein induces cell cycle arrest, anti-metastatic properties and ultimately affects the breast cancer cell growth by multiple mechanisms. Genistein-mediated anti-proliferative or anti-growth effects are usually observed at higher concentrations. These signaling pathways involve the decrease of NF-κB, HIF-1α, VEGF, and an increase of tumor suppressor p21. This will provide further insight into understanding the biology of transcription factors NF-κB, and HIF-1α in breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Genistein/therapeutic use , Neoplasm Metastasis/drug therapy , Breast Neoplasms/metabolism , Cell Cycle Checkpoints/drug effects , Estrogen Receptor beta/metabolism , Female , Humans , Neoplasm Metastasis/pathology , Phytoestrogens/therapeutic use , Signal Transduction/drug effects
18.
Altern Ther Health Med ; 26(2): 18-22, 2020 Mar.
Article in English | MEDLINE | ID: mdl-29477137

ABSTRACT

CONTEXT: Taiwanofungus camphoratus is a parasitic mushroom found in the heartwood of Cinnamomum kanehirai and is used as a nutritional supplement. It has an anticancer action, both alone and synergistically with amphotericin B (AmB). OBJECTIVE: The study intended to assess the efficacy of a T camphoratus ethanol extract (TCEE) combined with AmB for patients with metastatic cancer whose cancer did not respond to multiline chemotherapy or who were unwilling to receive chemotherapy. DESIGN: The research team performed a retrospective analysis as a pilot study. SETTING: The study took place at a single hospital (Taipei Medical University Hospital, Taipei, Taiwan). PARTICIPANTS: Participants were 9 patients at the hospital who were terminally ill with metastatic cancer. INTERVENTIONS: The participants had received daily doses of 2-3 g of the TCEE in combination with a weekly dose of 20-25 mg of AmB in 500 cc of 5% glucose water, given intravenously in 4-6 h. OUTCOME MEASURES: Outcome measures included (1) a primary evaluation index measuring the efficacy of the treatment; (2) a measure of tumor burden that was estimated using the response evaluation criteria in solid tumors (RECIST 1.1), (3) a secondary evaluation index measuring survival duration, and (4) safety. RESULTS: The mean treatment time was 54.4 ± 18.3 wk. At the end of the study, 2 patients showed a continued complete response, 1 patient had a continued partial response, and 1 patient showed a stable disease. The other 5 participants had times to progression ranging from 24 to 48 wk, with a mean of 35.6 wk. The mean survival time was 57.8 ± 18.5 wk, and 5 patients were still alive at the end of the study. CONCLUSIONS: For patients whose metastatic cancer did not respond to multiline chemotherapy or who were unwilling to receive chemotherapy, the use of TCEE as an adjuvant therapy to AmB resulted in tumor suppression and a delay in time to disease progression. The preliminary results reported here can be used to guide a future, more extensive clinical study of the combination.


Subject(s)
Amphotericin B/therapeutic use , Antifungal Agents/therapeutic use , Antrodia/chemistry , Biological Products/pharmacology , Neoplasm Metastasis/pathology , Neoplasms/drug therapy , Amphotericin B/administration & dosage , Antifungal Agents/administration & dosage , Biological Products/administration & dosage , Ethanol , Humans , Neoplasms/pathology , Pilot Projects , Retrospective Studies , Taiwan , Treatment Outcome
19.
Cancer Res ; 80(4): 659-662, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31831463

ABSTRACT

Looking beyond tumor angiogenesis, the past decade has witnessed a fundamental change of paradigm with the discovery that the vascular endothelium does not just respond to exogenous cytokines, but exerts active "angiocrine" gatekeeper roles, controlling their microenvironment in an instructive manner. While vascular niches host disseminated cancer cells and promote their stemness, endothelial cell-derived angiocrine signals orchestrate a favorable immune milieu to facilitate metastatic growth. Here, we discuss recent advances in the field of tumor microenvironment research and propose angiocrine signals as promising targets of future mechanism-driven antimetastatic therapies, which may prove useful to synergistically combine with chemotherapy and immunotherapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinogenesis/drug effects , Endothelium, Vascular/pathology , Neoplasm Metastasis/prevention & control , Neoplasms/drug therapy , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinogenesis/pathology , Disease Models, Animal , Disease Progression , Drug Evaluation, Preclinical , Endothelial Cells/pathology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Neoplasm Metastasis/pathology , Neoplasms/blood supply , Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Vascular Endothelial Growth Factor A/metabolism
20.
Clin Adv Hematol Oncol ; 18 Suppl 15(9): 1-20, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33843838

ABSTRACT

Metastatic human epidermal growth factor receptor 2 (HER2)-positive breast cancer is currently incurable. The primary goals of treatment are to prolong survival while optimizing quality of life. Several agents are now available in this setting, including neratinib, tucatinib, ado-trastuzumab emtansine, and trastuzumab deruxtecan. Neratinib in combination with capecitabine was recently approved for the treatment of adult patients with advanced or metastatic breast cancer who have received 2 or more prior anti-HER2-based regimens in the metastatic setting. Neratinib is an oral pan-HER inhibitor that binds covalently to the kinase site, providing irreversible binding. Phase 3 data showed that the combination of neratinib plus capecitabine improved progression-free survival vs lapatinib plus capecitabine. The duration of response was longer among patients in the neratinib arm. Neratinib plus capecitabine was also active against brain metastases associated with refractory, HER2-positive breast cancer, and this combination is listed in guidelines from the National Comprehensive Cancer Network for this indication. When combined with fulvestrant, neratinib demonstrated efficacy in patients with HER2-positive breast cancer, regardless of their hormone receptor status. Ongoing trials are evaluating the ability of neratinib to treat brain metastases, as well as the efficacy and safety of the triplet combination of neratinib, fulvestrant, and trastuzumab in this setting.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Neoplasm Metastasis/drug therapy , Receptor, ErbB-2/analysis , Ado-Trastuzumab Emtansine/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/pathology , Capecitabine/therapeutic use , Clinical Trials as Topic , Disease Management , Female , Fulvestrant/therapeutic use , Humans , Neoplasm Metastasis/pathology , Oxazoles/therapeutic use , Pyridines/therapeutic use , Quality of Life , Quinazolines/therapeutic use , Quinolines/therapeutic use , Trastuzumab/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL