Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Biol Chem ; 293(4): 1163-1177, 2018 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-29162724

RESUMEN

Distal metastasis of luminal breast cancer is frequent and incurable, yet the metastasis mechanisms are poorly understood. Estrogen, even at postmenopausal concentrations, suppresses invasiveness of luminal breast cancer cells through the estrogen receptor (ER). Invasive tumors overexpress the short progesterone receptor A (PR-A) isoform. Even at postmenopausal concentrations, progesterone activates PR-A, inducing invasiveness by counteracting estrogen's effects, particularly when cells are hypersensitized to progesterone by PR-A overexpression. To interrogate the role of this cross-talk in metastasis, we investigated selective cross-talk mechanisms of PR-A with ER. We developed a quantitative PCR-based lymph node infiltration assay to address the slowness of metastasis of tumor xenografts. We found that 15 microRNAs (miRNAs) are regulated by progesterone via PR-A, but not the longer PR-B isoform, with increased progesterone sensitivity when PR-A was overexpressed. Two of these miRNAs whose induction (miR-92a-3p) or repression (miR-26b-5p) by estrogen was suppressed by progesterone plus PR-A were critical for the PR-A-ER cross-talk causing a gene-regulatory pattern of invasiveness and metastasis and complete rescue of invasiveness in vitro Constitutive expression of miR-92a-3p or inhibition of miR-26b-5p profoundly suppressed metastasis. Finally, in primary breast tumors, PR-A expression was correlated negatively with miR-92a-3p expression and positively with miR-26b-5p expression. Therefore, hormonal cross-talk of PR-A with ER is probably a fundamental mechanism that enables metastasis of luminal breast cancer. Moreover, miRNA biomarkers of hyperactive PR-A may help predict metastatic potential of luminal breast tumors. Further, miR-92a-3p and miR-26b-5p may reveal target pathways for selective intervention to suppress hormone-regulated metastasis, both pre- and postmenopause.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Estrógenos/farmacología , MicroARNs/metabolismo , Proteínas de Neoplasias/metabolismo , ARN Neoplásico/metabolismo , Receptores de Progesterona/metabolismo , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Humanos , MicroARNs/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , ARN Neoplásico/genética , Receptores de Progesterona/genética
2.
Breast Cancer Res ; 20(1): 130, 2018 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-30359299

RESUMEN

BACKGROUND: De novo or acquired resistance in breast cancer leads to treatment failures and disease progression. In human epidermal growth factor receptor 2 (HER2)-positive (HER2+) breast cancer, Src, a non-receptor tyrosine kinase, is identified as a major mechanism of trastuzumab resistance, with its activation stabilizing aberrant HER2 signaling, thus making it an attractive target for inhibition. Here, we explored the causal relationship between Src and HER2 by examining the potential of 89Zr-trastuzumab as a surrogate imaging marker of Src activity upon inhibition with dasatinib in HER2+ breast cancer. METHODS: HER2+ primary breast cancer cell lines BT-474 and trastuzumab-resistant JIMT-1 were treated with dasatinib and assessed for expression and localization of HER2, Src, and phosphorylated Src (pSrc) (Y416) through western blots and binding assays. Mice bearing BT-474 or JIMT-1 tumors were treated for 7 or 14 days with dasatinib. At the end of each treatment, tumors were imaged with 89Zr-trastuzumab. The results of 89Zr-trastuzumab positron emission tomography (PET) was compared against tumor uptake of fluorodeoxyglucose (18F-FDG) obtained the day before in the same group of mice. Ex vivo western blots and immunohistochemical staining (IHC) were performed for validation. RESULTS: In BT-474 and JIMT-1 cells, treatment with dasatinib resulted in a decrease in internalized 89Zr-trastuzumab. Confirmation with immunoblots displayed abrogation of pSrc (Y416) signaling; binding assays in both cell lines demonstrated a decrease in cell surface and internalized HER2-bound tracer. In xenograft models, dasatinib treatment for 7 days (BT-474, 11.05 ± 2.10 % injected dose per gram of tissue %(ID)/g; JIMT-1, 3.88 ± 1.47 %ID/g)) or 14 days (BT-474, 9.20 ± 1.85 %ID/g; JIMT-1, 4.45 ± 1.23 %ID/g) resulted in a significant decrease in 89Zr-trastuzumab uptake on PET compared to untreated control (BT-474, 17.88 ± 2.18 %ID/g; JIMT-1, 8.04 ± 1.47 %ID/g). No difference in 18F-FDG uptake was observed between control and treated cohorts. A parallel decrease in membranous HER2 and pSrc (Y416) staining was observed in tumors post treatment on IHC. Immunoblots further validated the 89Zr-trastuzumab-PET readout. Positive correlation was established between 89Zr-trastuzumab tumor uptake versus tumor regression, pSrc and pHER2 expression. CONCLUSIONS: 89Zr-trastuzumab can potentially assess tumor response to dasatinib in HER2+ breast cancer and could be used as a surrogate tool to monitor early changes in Src signaling downstream of HER2.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Imagen Molecular/métodos , Tomografía de Emisión de Positrones/métodos , Inhibidores de Proteínas Quinasas/farmacología , Familia-src Quinasas/análisis , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/farmacocinética , Mama/diagnóstico por imagen , Mama/patología , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/patología , Línea Celular Tumoral , Dasatinib/farmacología , Dasatinib/uso terapéutico , Resistencia a Antineoplásicos , Estudios de Factibilidad , Femenino , Fluorodesoxiglucosa F18/administración & dosificación , Fluorodesoxiglucosa F18/farmacocinética , Humanos , Ratones , Ratones Desnudos , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptor ErbB-2/metabolismo , Distribución Tisular , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto , Familia-src Quinasas/antagonistas & inhibidores
3.
J Labelled Comp Radiopharm ; 61(9): 727-738, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29341222

RESUMEN

Therapeutic monoclonal antibodies have been used in cancer treatment for 30 years, with around 24 mAb and mAb:drug conjugates approved by the FDA to date. Despite their specificity, efficacy has remained limited, which, in part, derails nascent initiatives towards precision medicine. An image-guided approach to reinforce treatment decisions using immune positron emission tomography (immunoPET) companion diagnostic is warranted. This review provides a general overview of current translational research using Zr-89 immunoPET and opportunities for utilizing and harnessing this tool to its full potential. Patient case studies are cited to illustrate immunoPET probes as tools for profiling molecular signatures. Discussions on its utility in reinforcing clinical decisions as it relates to histopathological tumor assessment and standard diagnostic methods, and its potential as predictive biomarkers, are presented. We finally conclude with an overview of practical considerations to its utility in the clinic.


Asunto(s)
Desarrollo de Medicamentos , Tomografía de Emisión de Positrones/métodos , Radioisótopos , Circonio , Animales , Humanos , Distribución Tisular
4.
Proc Natl Acad Sci U S A ; 111(20): 7254-9, 2014 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-24785505

RESUMEN

Generally, solid tumors (>400 mm(3)) are inherently acidic, with more aggressive growth producing greater acidity. If the acidity could be targeted as a biomarker, it would provide a means to gauge the pace of tumor growth and degree of invasiveness, as well as providing a basis for predicting responses to pH-dependent chemotherapies. We have developed a (64)Cu pH (low) insertion peptide (pHLIP) for targeting, imaging, and quantifying acidic tumors by PET, and our findings reveal utility in assessing prostate tumors. The new pHLIP version limits indiscriminate healthy tissue binding, and we demonstrate its targeting of extracellular acidification in three different prostate cancer models, each with different vascularization and acid-extruding protein carbonic anhydrase IX (CAIX) expression. We then describe the tumor distribution of this radiotracer ex vivo, in association with blood perfusion and known biomarkers of acidity, such as hypoxia, lactate dehydrogenase A, and CAIX. We find that the probe reveals metabolic variations between and within tumors, and discriminates between necrotic and living tumor areas.


Asunto(s)
Tomografía de Emisión de Positrones , Neoplasias de la Próstata/diagnóstico por imagen , Radiofármacos/farmacología , Animales , Antígenos de Neoplasias/metabolismo , Anhidrasa Carbónica IX , Anhidrasas Carbónicas/metabolismo , Línea Celular Tumoral , Quelantes/farmacología , Radioisótopos de Galio/farmacología , Compuestos Heterocíclicos con 1 Anillo/farmacología , Humanos , Concentración de Iones de Hidrógeno , Hipoxia , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Lactato Deshidrogenasa 5 , Masculino , Proteínas de la Membrana/farmacología , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Fenotipo
5.
Bioconjug Chem ; 27(9): 2014-23, 2016 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-27396694

RESUMEN

pH (low) insertion peptides (pHLIP peptides) target acidic extracellular environments in vivo due to pH-dependent cellular membrane insertion. Two variants (Var3 and Var7) and wild-type (WT) pHLIP peptides have shown promise for in vivo imaging of breast cancer. Two positron emitting radionuclides ((64)Cu and (18)F) were used to label the NOTA- and NO2A-derivatized Var3, Var7, and WT peptides for in vivo biodistribution studies in 4T1 orthotopic tumor-bearing BALB/c mice. All of the constructs were radiolabeled with (64)Cu or [(18)F]-AlF in good yield. The in vivo biodistribution of the 12 constructs in 4T1 orthotopic allografted female BALB/c mice indicated that NO2A-cysVar3, radiolabeled with either (18)F (4T1 uptake; 8.9 ± 1.7%ID/g at 4 h p.i.) or (64)Cu (4T1 uptake; 8.2 ± 0.9%ID/g at 4 h p.i. and 19.2 ± 1.8% ID/g at 24 h p.i.), shows the most promise for clinical translation. Additional studies to investigate other tumor models (melanoma, prostate, and brain tumor models) indicated the universality of tumor targeting of these tracers. From this study, future clinical translation will focus on (18)F- or (64)Cu-labeled NO2A-cysVar3.


Asunto(s)
Radioisótopos de Cobre , Espacio Extracelular/química , Radioisótopos de Flúor , Proteínas de la Membrana , Tomografía de Emisión de Positrones/métodos , Animales , Línea Celular Tumoral , Femenino , Humanos , Masculino , Proteínas de la Membrana/química , Proteínas de la Membrana/farmacocinética , Ratones , Ratones Endogámicos BALB C , Trazadores Radiactivos , Relación Estructura-Actividad , Distribución Tisular
6.
Mol Pharm ; 11(11): 3965-73, 2014 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-24779727

RESUMEN

Engineered antibody fragments offer faster delivery with retained tumor specificity and rapid clearance from nontumor tissues. Here, we demonstrate that positron emission tomography (PET) based detection of prostate specific membrane antigen (PSMA) in prostatic tumor models using engineered bivalent antibodies built on single chain fragments (scFv) derived from the intact antibody, huJ591, offers similar tumor delineating properties but with the advantage of rapid targeting and imaging. (89)Zr-radiolabeled huJ591 scFv (dimeric scFv-CH3; (89)Zr-Mb) and cysteine diabodies (dimeric scFv; (89)Zr-Cys-Db) demonstrated internalization and similar Kds (∼2 nM) compared to (89)Zr-huJ591 in PSMA(+) cells. Tissue distribution assays established the specificities of both (89)Zr-Mb and (89)Zr-Cys-Db for PSMA(+) xenografts (6.2 ± 2.5% ID/g and 10.2 ± 3.4% ID/g at 12 h p.i. respectively), while minimal accumulation in PSMA(-) tumors was observed. From the PET images, (89)Zr-Mb and (89)Zr-Cys-Db exhibited faster blood clearance than the parent huJ591 while tumor-to-muscle ratios for all probes show comparable values across all time points. Ex vivo autoradiography and histology assessed the distribution of the probes within the tumor. Imaging PSMA-expressing prostate tumors with smaller antibody fragments offers rapid tumor accumulation and accelerated clearance; hence, shortened wait periods between tracer administration and high-contrast tumor imaging and lower dose-related toxicity are potentially realized.


Asunto(s)
Anticuerpos Monoclonales , Antígenos de Superficie/inmunología , Glutamato Carboxipeptidasa II/inmunología , Imagen Molecular/métodos , Neoplasias de la Próstata/diagnóstico por imagen , Radiofármacos , Anticuerpos de Cadena Única , Circonio , Animales , Anticuerpos Monoclonales/farmacocinética , Humanos , Fragmentos de Inmunoglobulinas , Radioisótopos de Yodo/farmacocinética , Masculino , Ratones , Tomografía de Emisión de Positrones/métodos , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Radiofármacos/farmacocinética , Anticuerpos de Cadena Única/farmacocinética , Distribución Tisular , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Circonio/farmacocinética
7.
Inorg Chem ; 53(4): 1880-99, 2014 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-24313747

RESUMEN

Over the past several decades, radionuclides have matured from largely esoteric and experimental technologies to indispensible components of medical diagnostics. Driving this transition, in part, have been mutually necessary advances in biomedical engineering, nuclear medicine, and cancer biology. Somewhat unsung has been the seminal role of inorganic chemistry in fostering the development of new radiotracers. In this regard, the purpose of this Forum Article is to more visibly highlight the significant contributions of inorganic chemistry to nuclear imaging by detailing the development of five metal-based imaging agents: (64)Cu-ATSM, (68)Ga-DOTATOC, (89)Zr-transferrin, (99m)Tc-sestamibi, and (99m)Tc-colloids. In a concluding section, several unmet needs both in and out of the laboratory will be discussed to stimulate conversation between inorganic chemists and the imaging community.


Asunto(s)
Química Inorgánica/tendencias , Imagen Molecular/tendencias , Animales , Quelantes/química , Química Inorgánica/normas , Sistemas de Liberación de Medicamentos , Ligandos , Compuestos Macrocíclicos/química , Ratones , Tomografía de Emisión de Positrones , Radiofármacos/química
8.
Bioconjug Chem ; 24(11): 1784-9, 2013 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-24180480

RESUMEN

Liposomes are attractive vehicles for the controlled release of drugs and cytotoxins and have a long-standing history in medical research and clinical practice. In addition to established therapeutic indications, liposomes have several favorable properties for molecular imaging, including high stability and the ability to be labeled with radioisotopes, as well as paramagnetic and fluorescent contrast agents. However, long circulation times and difficulties in creating targeted liposomes have proven challenges for imaging. In this study, we have addressed these limitations using a recently developed strategy for bioorthogonal conjugation, the reaction between tetrazines and trans-cyclooctenes. By coating radiolabeled liposomes with trans-cyclooctene and pretargeting with a tetrazine coupled to a targeted peptide, we were able to selectively enhance the retention of liposomes and bind them to tumor tissue in live animals. The rapid reaction between tetrazines and trans-cyclooctenes allowed imaging to be performed with the short-lived PET tracer (18)F, yielding signal-to-background activity ratios of 7:1. The covalent, bioorthogonally driven tumor-targeting of liposomes by in vivo click chemistry is promising and should be explored for more selective and rapid delivery of radiodiagnostics and radiotherapeutics, two classes of drugs which particularly benefit from fast clearance, low nonspecific binding, and the associated reduced toxicity to kidneys and bone marrow.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Radioisótopos de Flúor/metabolismo , Liposomas/química , Liposomas/metabolismo , Animales , Línea Celular Tumoral , Química Clic , Ciclooctanos/química , Humanos , Liposomas/farmacocinética , Ratones , Modelos Moleculares , Estructura Molecular , Neoplasias Experimentales/diagnóstico , Neoplasias Experimentales/metabolismo , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/metabolismo , Tomografía de Emisión de Positrones , Tetrazoles/química
9.
Cancer Res ; 78(19): 5706-5717, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30115693

RESUMEN

IFNγ is an attractive target for imaging active antitumor immunity due to its function in the T-cell signaling axis. Here, we test an IFNγ immuno-PET (immunoPET) probe for its capacity to identify adaptive immunotherapy response after HER2/neu vaccination in both spontaneous salivary and orthotopic neu+ mouse mammary tumors. IFNγ immunoPET detected elevated cytokine levels in situ after vaccination, which inversely correlated with tumor growth rate, an indicator of response to therapy. In a model of induced T-cell anergy where CD8 T cells infiltrate the tumor, but upregulate PD-1, IFNγ tracer uptake was equivalent to isotype control, illustrating a lack of antitumor T-cell activity. The IFNγ immunoPET tracer detected IFNγ protein sequestered on the surface of tumor cells, likely in complex with the IFNγ receptor, which may explain imaging localization of this soluble factor in vivo Collectively, we find that the activation status of cytotoxic T cells is annotated by IFNγ immunoPET, with reduced off-target binding to secondary lymphoid tissues compared with imaging total CD3+ tumor-infiltrating lymphocytes. Targeting of soluble cytokines such as IFNγ by PET imaging may provide valuable noninvasive insight into the function of immune cells in situ Significance: This study presents a novel approach to monitor therapeutic outcomes via IFNγ-targeted positron emission tomography. Cancer Res; 78(19); 5706-17. ©2018 AACR.


Asunto(s)
Inmunoterapia , Interferón gamma/metabolismo , Neoplasias/diagnóstico por imagen , Neoplasias/terapia , Tomografía de Emisión de Positrones , Animales , Unión Competitiva , Complejo CD3/metabolismo , Linfocitos T CD8-positivos/citología , Línea Celular Tumoral , Islas de CpG , Femenino , Heterocigoto , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Mamarias Animales/diagnóstico por imagen , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Animales/terapia , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Receptor de Muerte Celular Programada 1/metabolismo , Radiofármacos , Saliva/metabolismo , Linfocitos T Citotóxicos/citología
10.
Sci Rep ; 8(1): 9043, 2018 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-29899472

RESUMEN

Tumor resistance to treatment paved the way toward the development of single agent drugs that target multiple molecular signatures amplified within the malignancy. The discovered crosstalk between EGFR and HER3 as well as the role of HER3 in mediating EGFR resistance made these two receptor tyrosine kinases attractive targets. MEHD7945A or duligotuzumab is a single immunotherapy agent that dually targets both molecular signatures. In this study, a positron emission tomography (PET) companion diagnostic to MEHD7945A is reported and evaluated in pancreatic cancer. Tumor accretion and whole body pharmacokinetics of 89Zr-MEHD7945A were established. Specificity of the probe for EGFR and/or HER3 was further examined.


Asunto(s)
Inmunoglobulina G/farmacología , Neoplasias Pancreáticas/terapia , Tomografía de Emisión de Positrones/métodos , Receptor ErbB-3/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/farmacología , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Línea Celular Tumoral , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Femenino , Humanos , Inmunoglobulina G/química , Ratones SCID , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/metabolismo , Radioisótopos/química , Receptor ErbB-3/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Circonio/química
11.
ACS Omega ; 2(10): 6314-6320, 2017 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-29104950

RESUMEN

Vitamin B12, or cobalamin (Cbl), is an essential nutrient. Acquisition, transport, and cellular internalization of Cbl are dependent on specific binding proteins and associated receptors. The circulating transport protein transcobalamin (TC) promotes cellular uptake via binding to specific receptors such as CD320, a receptor upregulated in several cancer cell lines. In this study, we report the successful synthesis of 89Zirconium-labeled Cbl that was derivatized with desferrioxamine (89Zr-Cbl). We document the purity of the tracer and its binding to TC compared with that of unmodified cyano-Cbl (CN-Cbl). In vitro studies employing the CD320 receptor-positive breast cancer cell line MDA-MB-453 showed a 6- to 10-fold greater uptake of 89Zr-Cbl when compared with the uptake in the presence of 200-fold excess of CN-Cbl at 37 °C. We used nude mice with MDA-MB-453 tumors to study the feasibility of employing the tracer to visualize CD320 positive tumors. In vivo positron emission tomography images displayed a clear visualization of the tumor with 1.42 ± 0.48 %ID/g uptake (n = 3) at 4 h after injection (p.i.) with the tracer retained at 48 h p.i. Ex vivo biodistribution studies using 89Zr-Cbl exhibited the highest uptake in kidney and liver at 48 h p.i. Results document the feasibility of synthesizing a Cbl-based tracer suitable for both in vivo and ex vivo studies of Cbl trafficking and with the potential to visualize tumors expressing TC receptors, such as CD320.

12.
Sci Transl Med ; 7(283): 283ra51, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25877889

RESUMEN

Activating mutations of PIK3CA are the most frequent genomic alterations in estrogen receptor (ER)-positive breast tumors, and selective phosphatidylinositol 3-kinase α (PI3Kα) inhibitors are in clinical development. The activity of these agents, however, is not homogeneous, and only a fraction of patients bearing PIK3CA-mutant ER-positive tumors benefit from single-agent administration. Searching for mechanisms of resistance, we observed that suppression of PI3K signaling results in induction of ER-dependent transcriptional activity, as demonstrated by changes in expression of genes containing ER-binding sites and increased occupancy by the ER of promoter regions of up-regulated genes. Furthermore, expression of ESR1 mRNA and ER protein were also increased upon PI3K inhibition. These changes in gene expression were confirmed in vivo in xenografts and patient-derived models and in tumors from patients undergoing treatment with the PI3Kα inhibitor BYL719. The observed effects on transcription were enhanced by the addition of estradiol and suppressed by the anti-ER therapies fulvestrant and tamoxifen. Fulvestrant markedly sensitized ER-positive tumors to PI3Kα inhibition, resulting in major tumor regressions in vivo. We propose that increased ER transcriptional activity may be a reactive mechanism that limits the activity of PI3K inhibitors and that combined PI3K and ER inhibition is a rational approach to target these tumors.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptor alfa de Estrógeno/metabolismo , Mutación , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Antineoplásicos/química , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos , Retículo Endoplásmico/metabolismo , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Trasplante de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proyectos de Investigación , Transducción de Señal , Tiazoles/farmacología
13.
Sci Signal ; 7(318): ra29, 2014 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-24667376

RESUMEN

Both abundant epidermal growth factor receptor (EGFR or ErbB1) and high activity of the phosphatidylinositol 3-kinase (PI3K)-Akt pathway are common and therapeutically targeted in triple-negative breast cancer (TNBC). However, activation of another EGFR family member [human epidermal growth factor receptor 3 (HER3) (or ErbB3)] may limit the antitumor effects of these drugs. We found that TNBC cell lines cultured with the EGFR or HER3 ligand EGF or heregulin, respectively, and treated with either an Akt inhibitor (GDC-0068) or a PI3K inhibitor (GDC-0941) had increased abundance and phosphorylation of HER3. The phosphorylation of HER3 and EGFR in response to these treatments was reduced by the addition of a dual EGFR and HER3 inhibitor (MEHD7945A). MEHD7945A also decreased the phosphorylation (and activation) of EGFR and HER3 and the phosphorylation of downstream targets that occurred in response to the combination of EGFR ligands and PI3K-Akt pathway inhibitors. In culture, inhibition of the PI3K-Akt pathway combined with either MEHD7945A or knockdown of HER3 decreased cell proliferation compared with inhibition of the PI3K-Akt pathway alone. Combining either GDC-0068 or GDC-0941 with MEHD7945A inhibited the growth of xenografts derived from TNBC cell lines or from TNBC patient tumors, and this combination treatment was also more effective than combining either GDC-0068 or GDC-0941 with cetuximab, an EGFR-targeted antibody. After therapy with EGFR-targeted antibodies, some patients had residual tumors with increased HER3 abundance and EGFR/HER3 dimerization (an activating interaction). Thus, we propose that concomitant blockade of EGFR, HER3, and the PI3K-Akt pathway in TNBC should be investigated in the clinical setting.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor ErbB-3/antagonistas & inhibidores , Transducción de Señal/fisiología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados , Western Blotting , Línea Celular Tumoral , Cetuximab , Dimerización , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Humanos , Inmunoglobulina G/farmacología , Indazoles/farmacología , Neurregulina-1/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Piperazinas/farmacología , Pirimidinas/farmacología , Receptor ErbB-3/metabolismo , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología
14.
J Nucl Med ; 54(6): 936-43, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23578997

RESUMEN

UNLABELLED: We evaluated the ability of the PET imaging agent (89)Zr-trastuzumab to delineate HER2-positive gastric cancer and to monitor the pharmacodynamic effects of the epidermal growth factor receptor (EGFR)/human epidermal growth factor receptor 2 (HER2) tyrosine kinase inhibitor afatinib. METHODS: Using (89)Zr-trastuzumab, (18)F-FDG, or 3'-deoxy-3'-(18)F-fluorothymidine ((18)F-FLT PET), we imaged HER2-positive NCI-N87 and HER2-negative MKN74 gastric cancer xenografts in mice. Next, we examined the pharmacodynamic effects of afatinib in NCI-N87 xenografts using (89)Zr-trastuzumab and (18)F-FDG PET and comparing imaging results to changes in tumor size and in protein expression as monitored by Western blot and histologic studies. RESULTS: Although (18)F-FDG uptake in NCI-N87 tumors did not change, a decrease in (89)Zr-trastuzumab uptake was observed in the afatinib-treated versus control groups (3.0 ± 0.0 percentage injected dose per gram (%ID/g) vs. 21.0 ± 3.4 %ID/g, respectively; P < 0.05). (89)Zr-trastuzumab PET results corresponded with tumor reduction, apoptosis, and downregulation of HER2 observed on treatment with afatinib. Downregulation of total HER2, phosphorylated (p)-HER2, and p-EGFR occurred within 24 h of the first dose of afatinib, with a sustained effect over 21 d of treatment. CONCLUSION: Afatinib demonstrated antitumor activity in HER2-positive gastric cancer in vivo. (89)Zr-trastuzumab PET specifically delineated HER2-positive gastric cancer and can be used to measure the pharmacodynamic effects of afatinib.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Fluorodesoxiglucosa F18 , Tomografía de Emisión de Positrones , Quinazolinas/farmacología , Receptor ErbB-2/metabolismo , Neoplasias Gástricas/diagnóstico por imagen , Neoplasias Gástricas/tratamiento farmacológico , Afatinib , Animales , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/farmacocinética , Línea Celular Tumoral , Transformación Celular Neoplásica , Humanos , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinazolinas/uso terapéutico , Radioquímica , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Resultado del Tratamiento
15.
J Nucl Med ; 54(11): 1876-82, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24029655

RESUMEN

UNLABELLED: Despite their considerable advantages, many circulating biomarkers have well-documented limitations. One prominent shortcoming in oncology is a high frequency of false-positive indications for malignant disease in upfront diagnosis. Because one common cause of false positivism is biomarker production from benign disorders in unrelated host tissues, we hypothesized that probing the sites of biomarker secretion with an imaging tool could be a broadly useful strategy to deconvolute the meaning of foreboding but inconclusive circulating biomarker levels. METHODS: In preparation to address this hypothesis clinically, we developed (89)Zr-5B1, a fully human, antibody-based radiotracer targeting tumor-associated CA19.9 in the preclinical setting. RESULTS: (89)Zr-5B1 localized to multiple tumor models representing diseases with undetectable and supraphysiologic serum CA19.9 levels. Among these, (89)Zr-5B1 detected orthotopic models of pancreatic ductal adenocarcinoma, an elusive cancer for which the serum assay is measured in humans but with limited specificity in part because of the frequency of CA19.9 secretion from benign hepatic pathologies. CONCLUSION: In this report, a general strategy to supplement some of the shortcomings of otherwise highly useful circulating biomarkers with immunoPET is described. To expedite the clinical validation of this model, a human monoclonal antibody to CA19.9 (a highly visible but partially flawed serum biomarker for several cancers) was radiolabeled and evaluated, and the compelling preclinical evidence suggests that the radiotracer may enhance the fidelity of diagnosis and staging of pancreatic ductal adenocarcinoma, a notoriously occult cancer.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores/sangre , Biomarcadores de Tumor/sangre , Neoplasias/sangre , Neoplasias/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Radioisótopos , Circonio , Animales , Anticuerpos Monoclonales/farmacocinética , Línea Celular Tumoral , Transformación Celular Neoplásica , Femenino , Humanos , Ratones , Neoplasias/patología
16.
J Med Chem ; 52(16): 5253-61, 2009 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-19627091

RESUMEN

The intrinsic factor (IF) vitamin B(12) ileum anchored receptor, cubilin, mediates endocytotic uptake of the IF complex of vitamin B(12) to the blood serum. This receptor was targeted for the selective delivery and accumulation of a new bioprobe, a B(12) conjugate of rhenium 2, in the cubilin expressing placental choriocarcinoma BeWo cell line. Competitive uptake and cytotoxicity assays of 2 were investigated and interactions with nuclear DNA explored. In addition, the mechanism of internalization of 2 was confirmed to proceed in an IF-cubilin mediated fashion via siRNA transfection experiments. These studies show the great potential of cubilin as a new target for the delivery of B(12) based conjugates for cancer diagnostics and/or treatment.


Asunto(s)
Antineoplásicos/síntesis química , Complejos de Coordinación/síntesis química , Colorantes Fluorescentes/síntesis química , Sondas Moleculares/síntesis química , Compuestos Organometálicos/síntesis química , Receptores de Superficie Celular/metabolismo , Renio , Vitamina B 12/análogos & derivados , Vitamina B 12/síntesis química , Animales , Antineoplásicos/farmacología , Células CHO , Línea Celular Tumoral , Complejos de Coordinación/farmacología , Cricetinae , Cricetulus , Femenino , Colorantes Fluorescentes/farmacología , Humanos , Factor Intrinseco/metabolismo , Modelos Moleculares , Sondas Moleculares/farmacología , Compuestos Organometálicos/farmacología , Embarazo , ARN Interferente Pequeño/genética , Receptores de Superficie Celular/genética , Vitamina B 12/farmacología
17.
ChemMedChem ; 3(9): 1387-94, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18651631

RESUMEN

The synthesis, characterization, in vitro imaging, and cytotoxic properties of a new folate conjugate of rhenium(I) are reported. The conjugate [FA-PEG-BQAV-Re(CO)3]+ (gamma-4) was screened against an adriamycin- and cisplatin-resistant human ovarian cancer cell line (A2780/AD) that overexpresses the folate receptor (FR). Compound gamma-4 was internalized by a folate-receptor-mediated endocytotic pathway, which results in internal accumulation of gamma-4. This was contrasted with a FR-negative Chinese hamster ovary cell line in which no internalization of gamma-4 was observed. gamma-4 was found to be cytotoxic with IC(50) values of 189 and 78 microM at 6 and 24 h, respectively, toward the FR-positive cell line. This is in contrast to the IC(50) value of 502 microM at 6 h and 84 microM at 24 h for cisplatin in the same cell line, with a significantly greater toxicity at the earlier time point. The cytotoxicity of gamma-4 as explained by interactions that occur between the rhenium(I) complex moiety and DNA is described.


Asunto(s)
Proteínas Portadoras/biosíntesis , Proteínas Portadoras/efectos de los fármacos , Ácido Fólico/química , Compuestos Organometálicos/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/efectos de los fármacos , Renio/química , Animales , Células CHO , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cricetinae , Cricetulus , ADN/química , ADN/efectos de los fármacos , ADN-Topoisomerasas de Tipo I/química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Ensayo de Cambio de Movilidad Electroforética/métodos , Femenino , Receptores de Folato Anclados a GPI , Ácido Fólico/análogos & derivados , Ácido Fólico/síntesis química , Humanos , Concentración 50 Inhibidora , Sustancias Intercalantes/síntesis química , Sustancias Intercalantes/química , Sustancias Intercalantes/farmacología , Microscopía Confocal , Estructura Molecular , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/química , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Relación Estructura-Actividad , Inhibidores de Topoisomerasa I
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA