Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
JCO Oncol Pract ; 17(12): 723-730, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34077235

RESUMO

BRAF mutations in colorectal cancer have been studied over the past several decades. BRAF V600E mutation, a class I mutation, is the most common oncogenic BRAF alteration in colorectal cancer. Until recently, the BRAF V600E mutation was not among actionable genes for colorectal cancer. However, recent discoveries have revealed therapeutic opportunities. The BRAF with or without MEK inhibition combined with epidermal growth factor receptor-directed therapy was recently found to be an effective therapy choice for patients with advanced-stage BRAF V600-mutant colorectal cancer. However, it is essential to distinguish patients with BRAF V600E-mutant mismatch repair-deficient colorectal cancer from those with mismatch repair-proficient colorectal cancer, as immune checkpoint inhibitor therapy is more appealing in this subset of patients with colorectal cancer. This review article discusses the molecular characteristics of class I, II, and III BRAF mutants and their impact on the clinical behavior of colorectal cancer. We also review the recent progress in the targetability of BRAF mutations in colorectal cancer, which has led to changes in clinical practice and elaborates on innovative therapeutic approaches to enhance the efficacy of BRAF-targeting therapies, to achieve more durable responses.


Assuntos
Neoplasias Colorretais , Proteínas Proto-Oncogênicas B-raf , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Humanos , Mutação , Proteínas Proto-Oncogênicas B-raf/genética
2.
Expert Rev Gastroenterol Hepatol ; 13(10): 919-931, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31475851

RESUMO

Introduction: Metastatic colorectal cancer (CRC) remains a dilemma for cancer researchers with an increasing incidence in the younger patient population. Until the last decade, limited therapeutic options were available for metastatic CRC patients leading to relatively poor clinical outcomes.Areas covered: With advances in genome sequencing technology and reductions in the cost of next-generation sequencing, molecular profiling has become more accessible for cancer researchers and clinical investigators, which has furthered our understanding of the molecular behavior of CRC. This progress has recently translated into significant advances in molecular-based therapeutics and led to the development of new target-specific agents in metastatic CRC patients. In this review article, we extensively elaborate on genomic alterations seen in CRC patients including, but not limited to, EGFR, MMR, BRAF, HER2, NTRKs, FGFR, BRCA1/2, PALB2, POLE, and POLD1 genes, all of which are potentially actionable by either an FDA-approved agent or in a clinical trial setting.Expert opinion: We strongly recommend molecular profiling in metastatic CRC patients during the early course of their disease, as this may provide therapeutic and prognostic information that can guide clinicians to practice precision medicine. Patients with potentially actionable genes should be considered for targeting agents based on molecular alterations.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Medicina de Precisão , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Antineoplásicos Imunológicos/efeitos adversos , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Tomada de Decisão Clínica , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Humanos , Técnicas de Diagnóstico Molecular , Terapia de Alvo Molecular , Seleção de Pacientes , Valor Preditivo dos Testes , Prognóstico , Inibidores de Proteínas Quinases/efeitos adversos , Transdução de Sinais
3.
J Inorg Biochem ; 186: 176-186, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29957454

RESUMO

A water-soluble octanuclear cluster, [Fe8], was studied with regard to its properties as a potential contrast enhancing agent in magnetic resonance imaging (MRI) in magnetic fields of 1.3, 7.2 and 11.9 T and was shown to have transverse relaxivities r2 = 4.01, 10.09 and 15.83 mM s-1, respectively. A related hydrophobic [Fe8] cluster conjugated with 5 kDa hyaluronic acid (HA) was characterized by 57Fe-Mössbauer and MALDI-TOF mass spectroscopy, and was evaluated in aqueous solutions in vitro with regard to its contrast enhancing properties [r2 = 3.65 mM s-1 (1.3 T), 26.20 mM s-1 (7.2 T) and 52.18 mM s-1 (11.9 T)], its in vitro cellular cytotoxicity towards A-549 cells and COS-7 cells and its in vivo enhancement of T2-weighted images (4.7 T) of a human breast cancer xenografted on a nude mouse. The physiologically compatible [Fe8]-HA conjugate was i.v. injected to the tumor-bearing mouse, resulting in observable, heterogeneous signal change within the tumor, evident 15 min after injection and persisting for approximately 30 min. Both molecular [Fe8] and its HA-conjugate show a strong magnetic field dependence on r2, rendering them promising platforms for the further development of T2 MRI contrast agents in high and ultrahigh magnetic fields.


Assuntos
Neoplasias da Mama/diagnóstico por imagem , Meios de Contraste , Ferro , Imageamento por Ressonância Magnética , Compostos Organometálicos , Células A549 , Animais , Neoplasias da Mama/metabolismo , Células COS , Chlorocebus aethiops , Meios de Contraste/síntese química , Meios de Contraste/química , Meios de Contraste/farmacologia , Feminino , Xenoenxertos , Humanos , Ferro/química , Ferro/farmacologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Compostos Organometálicos/síntese química , Compostos Organometálicos/química , Compostos Organometálicos/farmacologia
4.
Oncotarget ; 8(65): 109596-109608, 2017 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-29312632

RESUMO

Small cell lung cancer (SCLC) is the most aggressive lung-cancer subtype and so far, no favorable therapeutic strategy has been established for chemo-resistant SCLC. Cisplatin is one of the most important components among all standard poly-chemotherapeutic regimens for SCLC; therefore, this study focused on revealing Cisplatin-resistance mechanism(s) in this disease. Cisplatin-resistant SCLC cells were generated in the NCI-H69 xenograft model in nude mice by continuous intravenous administration of Cisplatin; Cisplatin resistance of the tumor cells was confirmed by in vitro and in vivo tests, and the gene expression profile of the resistant cells was determined using microarray analysis. A significantly higher expression of tribbles pseudokinase 2 (TRIB2) mRNA in the Cisplatin-resistant cells was found compared to parental H69 cells. Further, the Cisplatin-resistance level was decreased when TRIB2 expression was knocked down. The mRNA and protein levels of CCAAT/enhancer binding protein alpha (CEBPA), known to be a transcription factor regulating cell differentiation and a target for degradation by TRIB2, as well as selected cancer stem cell makers in the Cisplatin-resistant cells, were measured. We found that CEBPA protein levels could be upregulated by knocking down the overexpressed TRIB2, which also reversed the Cisplatin-resistance of these cells; further, the Cisplatin-resistant SCLC cells demonstrated certain cancer stem cell-like properties. Similar patterns were also observed in limited human tumor specimens of chemo-resistant SCLC patients: namely, overexpressed TRIB2 and undetected CEBPA proteins. Our study revealed a possible molecular mechanism for Cisplatin-resistant SCLC involving induced TRIB2 overexpression and downregulation of CEBPA protein. We propose that this mechanism is a potential therapeutic target to circumvent chemo-resistance in SCLC.

5.
Expert Opin Ther Targets ; 19(12): 1587-91, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26374284

RESUMO

In the treatment of cancers, the dual goals of drug targeting are to deliver therapeutic agents more selectively to tumor tissue and to minimize exposure of normal tissues and organs to those agents, the latter causing toxicities that limit treatment and thereby attenuate clinical efficacy. CD44, a transmembrane proteoglycan, has been considered as a targetable candidate to generate a cancer-specific drug delivery axis. Although numerous preclinical studies showed promising results exploiting CD44 as such a target, results of two clinical trials, including a Phase III registration trial, have been very disappointing. Herein, we discuss the potential underlying causes that might have led to such failures, as well as the future of CD44-targeted cancer treatment.


Assuntos
Antineoplásicos/farmacologia , Receptores de Hialuronatos/metabolismo , Neoplasias/tratamento farmacológico , Animais , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Humanos , Terapia de Alvo Molecular , Neoplasias/patologia
6.
Future Med Chem ; 6(17): 1909-26, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25495984

RESUMO

Breast cancer is among the most commonly diagnosed cancer types in women worldwide and is the second leading cause of cancer-related disease in the USA. SH2 domains recruit signaling proteins to phosphotyrosine residues on aberrantly activated growth factor and cytokine receptors and contribute to cancer cell cycling, metastasis, angiogenesis and so on. Herein we review phosphopeptide mimetic and small-molecule approaches targeting the SH2 domains of Grb2, Grb7 and STAT3 that inhibit their targets and reduce proliferation in in vitro breast cancer models. Only STAT3 inhibitors have been evaluated in in vivo models and have led to tumor reduction. Taken together, these studies suggest that targeting SH2 domains is an important approach to the treatment of breast cancer.


Assuntos
Proteína Adaptadora GRB2/antagonistas & inibidores , Proteína Adaptadora GRB7/antagonistas & inibidores , Fator de Transcrição STAT3/antagonistas & inibidores , Domínios de Homologia de src , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Feminino , Proteína Adaptadora GRB2/metabolismo , Proteína Adaptadora GRB7/metabolismo , Humanos , Fosfopeptídeos/química , Fosfopeptídeos/metabolismo , Fosfopeptídeos/uso terapêutico , Ligação Proteica , Fator de Transcrição STAT3/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/metabolismo , Bibliotecas de Moléculas Pequenas/uso terapêutico
7.
Clin Cancer Res ; 18(15): 4114-21, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22693353

RESUMO

PURPOSE: Most primary human ovarian tumors and peritoneal implants, as well as tumor vascular endothelial cells, express the CD44 family of cell surface proteoglycans, the natural ligand for which is hyaluronic acid. Metronomic dosing, the frequent administration of chemotherapeutics at substantially lower than maximum tolerated doses (MTD), has been shown to result in reduced normal tissue toxicity and to minimize "off-treatment" exposure resulting in an improved therapeutic ratio. EXPERIMENTAL DESIGN: We tested the hypothesis that hyaluronic acid (HA) conjugates of paclitaxel (TXL; HA-TXL) would exert strong antitumor effects with metronomic (MET) dosing and induce antiangiogenic effects superior to those achieved with MTD administration or with free TXL. Female nude mice bearing SKOV3ip1 or HeyA8 ovarian cancer cells were treated intraperitoneally (i.p.) with MET HA-TXL regimens (or MTD administration) to determine therapeutic and biologic effects. RESULTS: All MET HA-TXL-treated mice and the MTD group revealed significantly reduced tumor weights and nodules compared with controls (all P values < 0.05) in the chemotherapy-sensitive models. However, the MTD HA-TXL-treated mice showed significant weight loss compared with control mice, whereas body weights were not affected in the metronomic groups in HeyA8-MDR model, reflecting reduced toxicity. In the taxane-resistant HeyA8-MDR model, significant reduction in tumor weight and nodule counts was noted in the metronomic groups whereas the response of the MTD group did not achieve significance. While both MTD and metronomic regimens reduced proliferation (Ki-67) and increased apoptosis (TUNEL, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling), only metronomic treatment resulted in significant reductions in angiogenesis (CD31, microvessel density). Moreover, metronomic treatment resulted in substantial increases in thrombospondin-1 (Tsp-1), an inhibitor of angiogenesis. CONCLUSIONS: This study showed that MET HA-TXL regimens have substantial antitumor activity in ovarian carcinoma, likely via a predominant antiangiogenic mechanism.


Assuntos
Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Paclitaxel/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citocinas/sangue , Relação Dose-Resposta a Droga , Esquema de Medicação , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Ácido Hialurônico/administração & dosagem , Ácido Hialurônico/química , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Nus , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Paclitaxel/administração & dosagem , Paclitaxel/química , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Trombospondina 1/metabolismo , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Expert Opin Ther Targets ; 16(7): 635-50, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22621669

RESUMO

INTRODUCTION: Advances in cancer therapeutics, namely more effective and less toxic treatments, will occur with targeting strategies that enhance the tumor biodistribution and thwart normal tissue exposure of the drug. This review focuses on cancer drug targeting approaches that exploit the expression of the cell-surface proteoglycan family, CD44, on the tumor cell surface followed by some form of ligand binding and induced CD44 internalization and intracellular drug release: in effect using this as a 'Trojan Horse' to more selectively access tumor cells. AREAS COVERED: This review defines the origins of evidence for a linkage between CD44 expression and malignancy, and invokes contemporary views of the importance of putative CD44(+) cancer stem cells in disease resistance. Although the primary emphasis is on the most advanced and developed paths, those that have either made it to the clinic or are well-poised to get there, a wide scope of additional approaches at various preclinical stages is also briefly reviewed. EXPERT OPINION: The future should see development of drug targeting approaches that exploit CD44 expression on CSCs/TICs, including applications to cytotoxic agents currently in the clinic.


Assuntos
Receptores de Hialuronatos/metabolismo , Neoplasias/metabolismo , Animais , Antineoplásicos/administração & dosagem , Portadores de Fármacos/administração & dosagem , Humanos , Receptores de Hialuronatos/genética , Ácido Hialurônico/metabolismo , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Ligação Proteica
9.
Nanomedicine ; 8 Suppl 1: S37-50, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22640907

RESUMO

Nanotechnology holds the promise of novel and more effective treatments for vexing human health issues. Among these are the use of nanoparticle platforms for site-specific delivery of therapeutics to tumors, both by passive and active mechanisms; the latter includes magnetic vectoring of magnetically responsive nanoparticles (MNP) that are functionalized to carry a drug payload that is released at the tumor. The conceptual basis, which actually dates back a number of decades, resides in physical (magnetic) enhancement, with magnetic field gradients aligned non-parallel to the direction of flow in the tumor vasculature, of existing passive mechanisms for extravasation and accumulation of MNP in the tumor interstitial fluid, followed by MNP internalization. In this review, we will assess the most recent developments and current status of this approach, considering MNP that are composed of one or more of the three elements that are ferromagnetic at physiological temperature: nickel, cobalt and iron. The effects on cellular functions in vitro, the ability to successfully vector the platform in vivo, the anti-tumor effects of such localized nano-vectors, and any associated toxicities for these MNP will be presented. The merits and shortcomings of nanomaterials made of each of the three elements will be highlighted, and a roadmap for moving this long-established approach forward to clinical evaluation will be put forth.


Assuntos
Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Imãs/química , Nanopartículas/química , Neoplasias/tratamento farmacológico , Animais , Cobalto/química , Cobalto/toxicidade , Humanos , Ferro/química , Ferro/toxicidade , Magnetismo/métodos , Imãs/toxicidade , Nanomedicina/métodos , Nanopartículas/toxicidade , Níquel/química , Níquel/toxicidade
10.
Maturitas ; 73(1): 33-44, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22402027

RESUMO

Nanotechnology holds the promise of novel and more effective treatments for vexing human health issues. Among these are the use of nanoparticle platforms for site-specific delivery of therapeutics to tumors, both by passive and active mechanisms; the latter includes magnetic vectoring of magnetically responsive nanoparticles (MNP) that are functionalized to carry a drug payload that is released at the tumor. The conceptual basis, which actually dates back a number of decades, resides in physical (magnetic) enhancement, with magnetic field gradients aligned non-parallel to the direction of flow in the tumor vasculature, of existing passive mechanisms for extravasation and accumulation of MNP in the tumor interstitial fluid, followed by MNP internalization. In this review, we will assess the most recent developments and current status of this approach, considering MNP that are composed of one or more of the three elements that are ferromagnetic at physiological temperature: nickel, cobalt and iron. The effects on cellular functions in vitro, the ability to successfully vector the platform in vivo, the anti-tumor effects of such localized nano-vectors, and any associated toxicities for these MNP will be presented. The merits and shortcomings of nanomaterials made of each of the three elements will be highlighted, and a roadmap for moving this long-established approach forward to clinical evaluation will be put forth.


Assuntos
Sistemas de Liberação de Medicamentos , Campos Magnéticos , Nanopartículas Metálicas/uso terapêutico , Neoplasias/tratamento farmacológico , Animais , Cobalto , Extravasamento de Materiais Terapêuticos e Diagnósticos , Humanos , Ferro , Nanopartículas Metálicas/toxicidade , Nanomedicina/tendências , Níquel
11.
Nanomedicine (Lond) ; 7(2): 289-99, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22339137

RESUMO

Superparamagnetic iron oxide nanoparticles (SPIONs) are being developed as vehicles for the selective targeting of therapeutics and bioactive compounds. Presented herein is a brief review of the history of approaches to magnetic-based drug delivery platforms, leading to current concepts of magnetically vectored therapeutics via functionalized SPION-prodrugs. With this background, recent experimental results are discussed that demonstrate the use of shaped external magnetic field gradients, generated by designed configurations of permanent magnets, to drive the concentration/accumulation of modified SPION-prodrug constructs at a tumor site, followed by tumor extravasation and activation of the prodrug within the tumor microenvironment. In order to successfully translate this approach to clinical application, one of the key requirements is the ability to magnetically drive ('vector') the SPION to human-scale tumor settings. In this review, various configurations of permanent magnets are described and models are presented that demonstrate that magnetic field gradients can potentially be focused and extended to lengths of several inches in vivo. This modification thereby increases the range of the delivery platform, and offers the potential for the treatment of visceral as well as superficial tumors and for translation from preclinical animal tumor models to clinical settings. The methodology of magnetically vectored prodrug therapeutics, as a means for selective localized targeting of tumor tissue, and minimizing harm to normal tissue, has the additional advantage of raising the therapeutic index compared with that of free drugs, thus, offering great potential as a cancer treatment modality.


Assuntos
Antineoplásicos/administração & dosagem , Previsões , Nanopartículas de Magnetita/uso terapêutico , Terapia de Alvo Molecular/tendências , Nanocápsulas/uso terapêutico , Neoplasias/tratamento farmacológico , Humanos
12.
J Exp Ther Oncol ; 10(2): 155-62, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23350355

RESUMO

Signal transducer and activator of transcription 3 (Stat3) is constitutively activated in a number of human cancers and cancer cell lines. Via its Src homology 2 (SH2) domain, Stat3 is recruited to phosphotyrosine residues on intracellular domains of cytokine and growth factor receptors, whereupon it is phosphorylated on Tyr705, dimerizes, translocates to the nucleus and is reported to participate in the expression of genes related to angiogenesis, metastasis, growth and survival. To block this process, we are developing cell-permeable, phosphatase-stable phosphopeptide mimics, targeted to the SH2 domain of Stat3, that inhibit the phosphorylation of Tyr705 of Stat3 in cultured tumor cells (Mandal et al., J. Med. Chem. 54, 3549-5463, 2011). At concentrations that inhibit tyrosine phosphorylation, these materials were not cytotoxic, similar to recent reports on JAK inhibitors. At higher concentrations, cytotoxicity was accompanied by off-target effects. We report that treatment of MDA-MB-468 human breast cancer xenografts in mice with peptidomimetic PM-73G significantly inhibited tumor growth, which was accompanied by reduction in VEGF production and microvessel density. No evidence of apoptosis or changes in the expression of the canonical genes cyclin D1 or survivin were observed. Thus selective inhibition of Stat3 Tyr705 phosphorylation may be a novel anti-angiogenesis strategy for the treatment of cancer.


Assuntos
Biomimética , Neoplasias da Mama/prevenção & controle , Neovascularização Patológica/prevenção & controle , Fosfopeptídeos/farmacologia , Pró-Fármacos/farmacologia , Fator de Transcrição STAT3/metabolismo , Domínios de Homologia de src/efeitos dos fármacos , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Feminino , Humanos , Camundongos , Camundongos Nus , Fosforilação/efeitos dos fármacos , Fosfotirosina/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
13.
JAKSTAT ; 1(4): 263-347, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24058783

RESUMO

Herein we review our progress on the development of phosphopeptide-based prodrugs targeting the SH2 domain of STAT3 to prevent recruitment to cytokine and growth factor receptors, activation, nuclear translocation and transcription of genes involved in cancer. We developed high affinity phosphopeptides (K I = 46-200 nM). Corresponding prodrugs inhibited constitutive and IL-6 induced Tyr705 phosphorylation at 0.5-1 µM in a variety of human cancer cell lines. They were not cytotoxic at 5 µM in vitro but they inhibited tumor growth in a human xenograft breast cancer model in mice, accompanied by reduced VEGF expression and angiogenesis.

14.
PLoS One ; 6(9): e24397, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21935404

RESUMO

Cancer stem cells (CSCs) possess high tumor-initiating capacity and have been reported to be resistant to therapeutics. Vice versa, therapy-resistant cancer cells seem to manifest CSC phenotypes and properties. It has been generally assumed that drug-resistant cancer cells may all be CSCs although the generality of this assumption is unknown. Here, we chronically treated Du145 prostate cancer cells with etoposide, paclitaxel and some experimental drugs (i.e., staurosporine and 2 paclitaxel analogs), which led to populations of drug-tolerant cells (DTCs). Surprisingly, these DTCs, when implanted either subcutaneously or orthotopically into NOD/SCID mice, exhibited much reduced tumorigenicity or were even non-tumorigenic. Drug-tolerant DLD1 colon cancer cells selected by a similar chronic selection protocol also displayed reduced tumorigenicity whereas drug-tolerant UC14 bladder cancer cells demonstrated either increased or decreased tumor-regenerating capacity. Drug-tolerant Du145 cells demonstrated low proliferative and clonogenic potential and were virtually devoid of CD44(+) cells. Prospective knockdown of CD44 in Du145 cells inhibited cell proliferation and tumor regeneration, whereas restoration of CD44 expression in drug-tolerant Du145 cells increased cell proliferation and partially increased tumorigenicity. Interestingly, drug-tolerant Du145 cells showed both increases and decreases in many "stemness" genes. Finally, evidence was provided that chronic drug exposure generated DTCs via epigenetic mechanisms involving molecules such as CD44 and KDM5A. Our results thus reveal that 1) not all DTCs are necessarily CSCs; 2) conventional chemotherapeutic drugs such as taxol and etoposide may directly target CD44(+) tumor-initiating cells; and 3) DTCs generated via chronic drug selection involve epigenetic mechanisms.


Assuntos
Antineoplásicos/uso terapêutico , Epigênese Genética/genética , Receptores de Hialuronatos/metabolismo , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Etoposídeo/farmacologia , Etoposídeo/uso terapêutico , Imunofluorescência , Humanos , Receptores de Hialuronatos/genética , Immunoblotting , Masculino , Camundongos , Camundongos SCID , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , Proteína 2 de Ligação ao Retinoblastoma/genética , Proteína 2 de Ligação ao Retinoblastoma/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estaurosporina/farmacologia , Estaurosporina/uso terapêutico
15.
Oral Oncol ; 47(11): 1039-47, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21903450

RESUMO

Chemotherapeutic regimens incorporating taxanes significantly improve outcomes for patients with squamous cell carcinomas of the head and neck (SCCHN). However, treatment with taxanes is limited by toxicities, including bone marrow suppression and peripheral neuropathies. We proposed that conjugating taxanes to targeting carrier molecules would increase antitumor efficacy and decrease toxicity. The cell surface proteoglycan, CD44, is expressed on most SCCHNs, and we hypothesized that it is an attractive candidate for targeted therapy via its natural ligand, hyaluronic acid (HA). We determined whether HA-paclitaxel conjugates were able to decrease tumor growth and improve survival in orthotopic nude mouse human SCCHN xenograft models. HA-paclitaxel concentration-dependent growth inhibition of human SCCHN cell lines OSC-19 and HN5 in vitro, very similarly to free paclitaxel treatment. Tumor cell uptake of FITC-labeled HA-paclitaxel was significantly blocked with free HA, indicating the dependence of uptake on CD44. HA-paclitaxel administered intravenously once per week for three weeks at 120 mg/kg paclitaxel equivalents, far above the paclitaxel maximum tolerated dose, exerted superior tumor growth control to that of paclitaxel in both orthotopic OSC-19-luciferase and HN5 xenograft models in vivo. Mouse survival following HA-paclitaxel administration was prolonged compared with that of controls in mice implanted with either of these xenografts. Mice treated with HA-paclitaxel displayed increased TUNEL(+) cells in tumor tissue, as well as markedly reduced microvessel density compared to those treated with free paclitaxel. No acute histopathological changes were observed in mice treated with HA-paclitaxel. Thus, we conclude that HA-paclitaxel effectively inhibits tumor growth in human SCCHN xenografts via an HA-mediated mechanism and this conjugate should be considered for further preclinical development for this disease.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Ácido Hialurônico/farmacologia , Paclitaxel/farmacologia , Neoplasias da Língua/tratamento farmacológico , Animais , Carcinoma de Células Escamosas/metabolismo , Modelos Animais de Doenças , Humanos , Receptores de Hialuronatos/efeitos dos fármacos , Receptores de Hialuronatos/metabolismo , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Nus , Neoplasias da Língua/metabolismo , Transplante Heterólogo , Resultado do Tratamento , Células Tumorais Cultivadas/efeitos dos fármacos
16.
Biochemistry ; 49(42): 9106-12, 2010 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-20843027

RESUMO

CD44, the primary receptor for hyaluronic acid, plays an important role in tumor growth and metastasis. CD44-hyaluronic acid interactions can be exploited for targeted delivery of anticancer agents specifically to cancer cells. Although various splicing variants of CD44 are expressed on the plasma membrane of cancer cells, the hyaluronic acid binding domain (HABD) is highly conserved among the CD44 splicing variants. Using a novel two-step process, we have identified monothiophosphate-modified aptamers (thioaptamers) that specifically bind to the CD44's HABD with high affinities. Binding affinities of the selected thioaptamers for the HABD were in the range of 180-295 nM, an affinity significantly higher than that of hyaluronic acid (K(d) above the micromolar range). The selected thioaptamers bound to CD44 positive human ovarian cancer cell lines (SKOV3, IGROV, and A2780) but failed to bind the CD44 negative NIH3T3 cell line. Our results indicated that thio substitution at specific positions of the DNA phosphate backbone results in specific and high-affinity binding of thioaptamers to CD44. The selected thioaptamers will be of great interest for further development as a targeting or imaging agent for the delivery of therapeutic payloads for cancer tissues.


Assuntos
Aptâmeros de Nucleotídeos/química , Aptâmeros de Nucleotídeos/metabolismo , Receptores de Hialuronatos/química , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/metabolismo , Animais , Aptâmeros de Nucleotídeos/genética , Sequência de Bases , Sítios de Ligação , Biomarcadores Tumorais/química , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Receptores de Hialuronatos/genética , Técnicas In Vitro , Cinética , Camundongos , Células NIH 3T3 , Conformação de Ácido Nucleico , Neoplasias Ovarianas/metabolismo , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Técnica de Seleção de Aptâmeros
17.
P R Health Sci J ; 29(3): 223-31, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20799509

RESUMO

Magnetic resonance imaging (MRI) is occupying an increasing niche in the clinical diagnostic workup of several cancers, including breast cancers. Despite the high level of implementation of mammography, it has become apparent that MRI can play at least a complementary role in the imaging and diagnosis of primary breast cancers, including ductal carcinoma in situ, the earliest stage of breast cancer that is associated with an increased risk of invasive breast cancer. This can also be said of inflammatory breast cancer, of low incidence but with high impact on overall breast cancer mortality rates, and for which mammography is not ideal due to the typically diffused nature of this disease. Much of the value of breast MRI is dependent on its high sensitivity, resulting from the use of contrast agent enhancement in the detection of breast cancer. Interest has also increased in the application of diffusion-weighted MRI for early assessment of treatment response in this disease. Regarding ovarian and other gynecological cancers, MRI has already demonstrated value in the evaluation of patients with ovarian masses, uterine leiomyoma, endometrioma, and cervical cancer. Features on MRI suggestive of malignant ovarian tumors are varied, and span irregular or solid components to a cystic mass, prominent septations, evidence of peritoneal, hematogenous, or lymphatic spread, or local invasion. The majority of ovarian malignancies are diagnosed in advanced, incurable stages, where exploratory laparotomy provides the opportunity for maximal debulking. Although a role for MRI has yet to be established in this initial setting or in staging, some studies have shown that high sensitivity may be achieved with contrast agent-enhanced MRI for detection of recurrent disease, including demonstration of macroscopic intraabdominal dissemination and the hallmark omental "cake". Efforts in recent years have been focused on design of MRI contrast agents (MRI-CAs), which either target biomarkers, or take advantage of the different physiology of cancerous cells. MRI-CAs based on gadolinium complexes, ferrumoxides, or other metallic nanoparticles have been investigated. This review will focus on the recent progress in the application of MRI to the imaging of breast and ovarian cancers, and present a possible role for molecularly-targeted contrast agents in enriching the context for MRI-based diagnosis.


Assuntos
Neoplasias da Mama/diagnóstico , Imageamento por Ressonância Magnética/tendências , Neoplasias Ovarianas/diagnóstico , Feminino , Previsões , Humanos
18.
Bioorg Med Chem Lett ; 19(3): 1012-7, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19097786

RESUMO

Poly-L-glutamic acid (PGA) has previously been demonstrated to be an effective backbone for creating a hydrophilic prodrug of the established anti-tumor agent, paclitaxel, the active agent in Taxol; this approach has obviated the need for the toxic Cremophor excipient, used to enhance the solubility of paclitaxel in the clinical formulation. In order to form hydrophilic prodrugs of the hydrophobic pro-apoptotic sphingolipid, N,N-dimethylsphingosine (DMSP), PGA was condensed with DMSP, previously modified with coumarin to allow spectroscopic detection during conjugate synthesis, to yield PGA-DMSP. Conjugates with different loadings of DMSP were prepared and evaluated for in vitro cytotoxicity against two human breast adenocarcinoma cell lines. Time- and loading-dependent expression of cytotoxicity was observed, such that endpoints essentially equivalent to those observed with free-DMSP were achieved, but in a more protracted manner, consistent with prodrug behavior. PGA-DMSP was initially evaluated for toxicity in female nude mice, and administration of high net levels of DMSP, exceeding those achievable with free-DMSP, was well-tolerated. We propose that PGA-DMSP conjugates merit evaluation for anti-tumor efficacy in pre-clinical tumor models.


Assuntos
Química Farmacêutica/métodos , Ácido Poliglutâmico/química , Esfingosina/análogos & derivados , Animais , Apoptose , Linhagem Celular Tumoral , Desenho de Fármacos , Feminino , Humanos , Concentração Inibidora 50 , Camundongos , Camundongos Nus , Modelos Químicos , Pró-Fármacos/química , Espectrofotometria/métodos , Esfingosina/química
19.
Neoplasia ; 9(6): 479-86, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17603630

RESUMO

Numerous human tumor types, including ovarian cancer, display a significant expression of the CD44 family of cell surface proteoglycans. To develop tumor-targeted drugs, we have initially evaluated whether the CD44 ligand hyaluronic acid (HA) could serve as a backbone for paclitaxel (TXL) prodrugs. HA-TXL was prepared by modification of previous techniques. The in vitro cytotoxicity of HA-TXL against the CD44(+) human ovarian carcinoma cell lines SKOV-3ip and NMP-1 could be significantly blocked by preincubation with a molar excess of free HA. Female nude mice bearing intraperitoneal implants of NMP-1 cells were treated intraperitoneally with a single sub-maximum tolerated dose dose of HA-TXL or with multiple-dose regimens of paclitaxel (Taxol; Mead Johnson, Princeton, NJ) to determine the effects of these regimens on host survival and intraperitoneal tumor burden, with the latter being assessed by magnetic resonance imaging. NMP-1 xenografts were highly resistant to Taxol regimens, as host survival was only nominally improved compared to controls (T//C approximately 120), whereas single-dose HA-TXL treatment significantly improved survival in this model (T//C approximately 140; P = .004). In both NMP-1 and SKOV-3ip models, MR images of abdomens of HA-TXL-treated mice obtained shortly before controls required humane sacrifice revealed markedly reduced tumor burdens compared to control mice. This study is among the first to demonstrate that HA-based prodrugs administered locoregionally have antitumor activity in vivo.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/química , Neoplasias Ovarianas/tratamento farmacológico , Paclitaxel/uso terapêutico , Animais , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Injeções Intraperitoneais , Imageamento por Ressonância Magnética , Camundongos , Camundongos Nus , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Pró-Fármacos/administração & dosagem , Carga Tumoral , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Bioconjug Chem ; 18(3): 731-5, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17432826

RESUMO

Coumarin derivatives of N,N-dimethylsphingosine (DMSP) were prepared and chemically characterized. They were apparently biologically equivalent to DMSP in terms of tumor cell cytotoxicity and were used to establish the rapid mitochondrial localization of this sphingolipid in tumor cells, followed closely by its marked reduction of mitochondrial membrane potential.


Assuntos
Cumarínicos/metabolismo , Cumarínicos/farmacologia , Mitocôndrias/metabolismo , Esfingosina/análogos & derivados , Linhagem Celular Tumoral , Cumarínicos/química , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Microscopia de Fluorescência , Mitocôndrias/química , Esfingosina/química , Esfingosina/metabolismo , Esfingosina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA