Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Leuk Lymphoma ; 63(3): 653-663, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34689693

RESUMO

Arsenic trioxide (ATO) treats Acute Promyelocytic Leukemia (APL). ATO is converted from inorganic arsenic (iAs) to methylated (MAs) and dimethylated (DMAs) metabolites, which are excreted in the urine. Methylation of iAs is important in detoxification, as iAs exposure is deleterious to health. We examined ATO metabolism in 25 APL patients, measuring iAs, MAs, and DMAs. Plasma total iAs increased after ATO administration, followed by a rapid decline, reaching trough levels by 4-6 h. We identified two patterns of iAs metabolism between 6 and 24 h after infusion: in Group 1, iAs increased and were slowly converted to MAs and DMAs, whereas in Group 2, iAs was rapidly metabolized. These patterns were associated with smoking and different treatments: ATO with all-trans retinoic acid (ATRA) alone vs. ATO preceded by ATRA and chemotherapy. Our data suggest that smoking and prior chemotherapy exposure may be associated with ATO metabolism stimulation, thus lowering the effective blood ATO dose.


Assuntos
Arsenicais , Leucemia Promielocítica Aguda , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Trióxido de Arsênio/uso terapêutico , Arsenicais/uso terapêutico , Humanos , Leucemia Promielocítica Aguda/metabolismo , Óxidos/uso terapêutico , Tretinoína/uso terapêutico
2.
Oncologist ; 24(6): e284-e291, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30975921

RESUMO

BACKGROUND: Less than 3% of older patients with cancer are enrolled in clinical trials. To reverse this underrepresentation, we compared older patients enrolled with older-patient-specific trials, defined as those designed for older patients with cancer, with those enrolled in age-unspecified trials. MATERIALS AND METHODS: We focused on individual patient data from those ≥65 years (younger patients excluded) and included all Alliance phase III adjuvant breast cancer trials from 1985-2012. RESULTS: Among 2,277 patients, 1,014 had been enrolled to older-patient-specific and 1,263 to age-unspecified trials. The median age (range) in the older-patient-specific trials was 72 (65-89) years compared with 68 (65-84) years in the cohort of older patients in age-unspecified trials; p < .0001. A greater percentage of patients 75 years or older had enrolled in older-patient-specific trials compared with the cohort of age-unspecified trials: 26% versus 6% (p < .0001). Median overall survival (OS) was 12.8 years (95% confidence interval [CI], 11.9-13.7) and 13.5 years (95% CI, 12.9-14.1) for older-patient-specific and age-unspecified trials, respectively. OS was comparable (hazard ratio [HR], 1.08; 95% CI, 0.92-1.28; p = .34; referent: age-unspecified trials), after adjusting for age, estrogen receptor status, tumor size, and lymph node status. Similar findings were reached for recurrence-free survival. A lower rate of grade 3-5 adverse events (hematologic and nonhematologic) was reported in older-patient-specific trials (43% vs. 58%; p < .0001). Sensitivity analysis with chemotherapy only trials and subset analysis, adjusted for performance score, yielded similar OS results. CONCLUSION: Older-patient-specific trials appear to address this underrepresentation of older patients with ostensibly comparable outcomes. Clinical trial identification numbers. NCT00003088 (CALGB 9741); NCT00024102 (CALGB 49907); NCT00068601 (CALGB 40401); NCT00005970 (NCCTG N9831) IMPLICATIONS FOR PRACTICE: This work underscores the importance of clinical trials that focus on the recruitment of older patients with cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Neoplasias da Mama/terapia , Ensaios Clínicos Fase III como Assunto/estatística & dados numéricos , Recidiva Local de Neoplasia/epidemiologia , Seleção de Pacientes , Ensaios Clínicos Controlados Aleatórios como Assunto/estatística & dados numéricos , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Quimioterapia Adjuvante/métodos , Ensaios Clínicos Fase III como Assunto/normas , Intervalo Livre de Doença , Feminino , Humanos , Mastectomia , Recidiva Local de Neoplasia/patologia , Modelos de Riscos Proporcionais , Ensaios Clínicos Controlados Aleatórios como Assunto/normas
3.
Mol Med ; 24(1): 50, 2018 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-30249178

RESUMO

Antibody-based therapy has revitalized the world of cancer therapeutics since rituximab was first approved for the treatment of Non-Hodgkin's Lymphoma. Monoclonal antibodies against cancer antigens have been successful strategies for only a handful of cancer types due to many reasons including lack of antibody specificity and complex nature of tumor milieu which interfere with antibody efficacy. Polyspecific antibodies are promising class of anti-cancer agents which can be directed at multiple tumor antigens to eradicate tumor cells more precisely and effectively. They may overcome some of these limitations and have already changed treatment landscape for some malignancies such as B cell acute lymphoblastic leukemia. Pre-clinical studies and early phase clinical trials have demonstrated that this approach may be an effective strategy even for solid tumors. This review focuses on the development of bispecific and trispecific antibody therapy for the treatment of solid tumor malignancies and highlights the potential they hold for future therapies to come.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Neoplasias/tratamento farmacológico , Animais , Humanos
5.
Br J Cancer ; 114(9): 956-64, 2016 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-27092785

RESUMO

BACKGROUND: To examine the effect on cognitive function of adjuvant ovarian function suppression (OFS) for breast cancer. METHODS: The Suppression of Ovarian Function (SOFT) trial randomised premenopausal women with hormone receptor-positive breast cancer to 5 years adjuvant endocrine therapy with tamoxifen+OFS, exemestane+OFS or tamoxifen alone. The Co-SOFT substudy assessed objective cognitive function and patient reported outcomes at randomisation (T0), and 1 year later (T1); the primary endpoint was change in global cognitive function, measured by the composite objective cognitive function score. Data were compared for the pooled tamoxifen+OFS and exemestane+OFS groups vs the tamoxifen alone group using the Wilcoxon rank-sum test. RESULTS: Of 86 participants, 74 underwent both T0 and T1 cognitive testing; 54 randomised to OFS+ either tamoxifen (28) or exemestane (26) and 20 randomised to tamoxifen alone. There was no significant difference in the changes in the composite cognitive function scores between the OFS+ tamoxifen or exemestane groups and the tamoxifen group (mean±s.d., -0.21±0.92 vs -0.04±0.49, respectively, P=0.71, effect size=-0.20), regardless of prior chemotherapy status, and adjusting for baseline characteristics. CONCLUSIONS: The Co-SOFT study, although limited by small samples size, provides no evidence that adding OFS to adjuvant oral endocrine therapy substantially affects global cognitive function.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/complicações , Ovário/metabolismo , Tamoxifeno/uso terapêutico , Adjuvantes Imunológicos , Adulto , Neoplasias da Mama/tratamento farmacológico , Cognição , Feminino , Humanos , Pessoa de Meia-Idade , Pré-Menopausa , Qualidade de Vida
6.
Future Oncol ; 12(9): 1151-63, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27019997

RESUMO

Inhibitory ligands on tumor cells and their corresponding receptors on T cells are collectively called immune checkpoint molecules and have emerged as druggable targets that harness endogenous immunity to fight cancer. Immune checkpoint inhibitors targeting CTLA-4, PD-1 and PD-L1 have been developed for the treatment of patients with non-small-cell lung cancer and other malignancies, with impressive clinical activity, durable responses and a favorable toxicity profile. This article reviews the development, current status and future directions for some of these agents. The efficacy and safety data for drugs such as ipilimumab, nivolumab, pembrolizumab, atezolizumab and durvalumab are reviewed, along with combination strategies and response evaluation criteria. The toxicity profiles and predictive biomarkers of response are also discussed.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/imunologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Antineoplásicos/imunologia , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Antígeno B7-H1/imunologia , Antígeno CTLA-4/imunologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/imunologia , Humanos , Receptor de Morte Celular Programada 1/imunologia
7.
Onco Targets Ther ; 6: 1613-24, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24265556

RESUMO

The histone deacetylase (HDAC) inhibitors have emerged as novel therapies for cancer. Panobinostat (LBH 589, Novartis Pharmaceuticals) is a pan-deacetylase inhibitor that is being evaluated in both intravenous and oral formulations across multiple tumor types. Comparable to the other HDACs, panobinostat leads to hyperacetylation of histones and other intracellular proteins, allowing for the expression of otherwise repressed genes, leading to inhibition of cellular proliferation and induction of apoptosis in malignant cells. Panobinostat, analogous to other HDAC inhibitors, also induces apoptosis by directly activating cellular death receptor pathways. Preclinical data suggests that panobinostat has inhibitory activity at nanomolar concentrations and appears to be the most potent clinically available HDAC inhibitor. Here we review the current status of panobinostat and discuss its role in the treatment of solid tumors.

8.
Crit Rev Oncol Hematol ; 88(2): 284-92, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23755891

RESUMO

There is no available targeted therapy for triple-negative or its more aggressive subtype, basal-like breast cancer. Multiple therapeutic strategies based on translational knowledge have not improved the treatment options for triple negative patients. As understanding of molecular pathways that drive tumor development is rapidly increasing, it is imperative to adapt our treatment strategies to perturbations in molecular pathways driving the malignant process. Basal-like breast cancers over-express EGFR (without mutations or EGFR gene amplifications) and have p53 mutations. While EGFR drives the malignant behavior in triple negative breast cancer (TNBC), anti-EGFR therapies have fallen short of the expected results in clinical trials. Here we bring evidence that the less than optimal results of the anti-EGFR therapies may be explained in part by the increased potency of the EGFR signaling due to increased endosomal recycling. The functional connection between EGFR and endosomal trafficking in TNBC is mutant p53 found in the most aggressive forms of TNBC. Mutant p53 acquires oncogenic functions and binds p63 protein, a member of p53 family with tumor suppressor activities. In the absence of functional p63 there is an upregulation of endosomal recycling EGFR and integrin to the membrane with increased proinvasive abilities of cancer cells. Blocking endosomal trafficking combined with anti-EGFR treatments may result in better clinical outcomes in TNBC.


Assuntos
Endossomos/metabolismo , Receptores ErbB/metabolismo , Mutação , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/metabolismo , Proteína Supressora de Tumor p53/genética , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Receptores ErbB/antagonistas & inibidores , Feminino , Humanos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Transporte Proteico , Transdução de Sinais , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/patologia , Proteína Supressora de Tumor p53/metabolismo
10.
Ann Hematol ; 92(6): 719-30, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23494203

RESUMO

For more than 2,000 years, arsenic and its derivatives have shown medical utility. Owing to the toxicities and potential carcinogenicity of arsenicals, their popularity has fluctuated. The exact mechanism of action of therapeutic arsenic is not well characterized but likely to involve apoptosis, generation of reactive oxygen species, inhibition of intracellular transduction pathways, and cell functions. Arsenic trioxide has received approval for use in patients with relapsed acute promyelocytic leukemia for remission induction. Arsenic has additionally shown activity in a range of solid tumors, myelodysplastic syndrome, multiple myeloma, and in autoimmune diseases. The following is a review of the history of arsenic, its cellular metabolism, pharmacology, genetic basis of disposition, associated toxicities, and clinical efficacy.


Assuntos
Antineoplásicos/uso terapêutico , Arsenicais/uso terapêutico , Neoplasias/tratamento farmacológico , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Intoxicação por Arsênico/diagnóstico , Intoxicação por Arsênico/etiologia , Trióxido de Arsênio , Arsenicais/administração & dosagem , Arsenicais/efeitos adversos , Arsenicais/farmacologia , Biotransformação/genética , Biotransformação/fisiologia , Ensaios Clínicos como Assunto , Diabetes Mellitus Tipo 2/induzido quimicamente , Gastroenteropatias/induzido quimicamente , Regulação da Expressão Gênica/efeitos dos fármacos , Cardiopatias/induzido quimicamente , Humanos , Leucemia Promielocítica Aguda/tratamento farmacológico , Leucocitose/induzido quimicamente , Redes e Vias Metabólicas/efeitos dos fármacos , Metiltransferases/genética , Metiltransferases/metabolismo , Neoplasias/induzido quimicamente , Óxidos/efeitos adversos , Óxidos/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Resultado do Tratamento
11.
Mol Med ; 18: 1473-9, 2013 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-23196784

RESUMO

Trastuzumab is a monoclonal antibody targeted against the HER2 tyrosine kinase receptor. Although trastuzumab is a very active agent in HER2-overexpressing breast cancer, the majority of patients with metastatic HER2-overexpressing breast cancer who initially respond to trastuzumab develop resistance within 1 year of initiation of treatment and, in the adjuvant setting, progress despite trastuzumab-based therapy. The antibody-drug conjugate trastuzumab-DM1 (T-DM1) was designed to combine the biological activity of trastuzumab with the targeted delivery of a highly potent antimicrotubule agent, DM1 (N-methyl-N-[3-mercapto-1-oxopropyl]-l-alanine ester of maytansinol), a maytansine derivative, to HER2-overexpressing breast cancer cells. T-DM1 is the first antibody-drug conjugate with a nonreducible thioether linker in clinical trials. Phase I and II clinical trials of T-DM1 as a single agent and in combination with paclitaxel, docetaxel and pertuzumab have shown clinical activity and a favorable safety profile in patients with HER2-positive metastatic breast cancer. Two randomized phase III trials of T-DM1 are awaiting final results; the EMILIA trial is evaluating T-DM1 compared with lapatinib plus capecitabine, and early positive results have been reported. The MARIANNE trial is evaluating T-DM1 plus placebo versus T-DM1 plus pertuzumab versus trastuzumab plus a taxane. Here, we summarize evidence from clinical studies and discuss the potential clinical implications of T-DM1.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Imunoconjugados/uso terapêutico , Maitansina/análogos & derivados , Receptor ErbB-2/metabolismo , Ado-Trastuzumab Emtansina , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/química , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Ensaios Clínicos como Assunto , Feminino , Humanos , Imunoconjugados/administração & dosagem , Imunoconjugados/efeitos adversos , Imunoconjugados/química , Maitansina/administração & dosagem , Maitansina/efeitos adversos , Maitansina/química , Maitansina/uso terapêutico , Trastuzumab , Resultado do Tratamento
12.
Support Care Cancer ; 20(11): 2865-72, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22421738

RESUMO

BACKGROUND: The benefit of adding a vena cava filter to anticoagulation in treating cancer patients with venous thromboembolism remains controversial. We initiated this study as the first prospectively randomized trial to evaluate the addition of a vena cava filter placement to anticoagulation with the factor Xa inhibitor fondaparinux sodium in patients with cancer. METHODS: Sixty-four patients with deep vein thrombosis (86 %) and/or pulmonary embolism (55 %) were randomly assigned to receive anticoagulation with fondaparinux sodium with or without a vena cava filter. Endpoints included rates of complications by treatment arm, recurrent thromboembolism, complete resolution of thromboembolism, and survival rates. RESULTS: No patient had a recurrent deep vein thrombosis; two (3 %) patients had new pulmonary emboli, one in each randomized cohort. Major bleeding occurred in three patients (5 %). Two patients on the vena cava filter arm (7 %) had complications from the filter. Median survivals were 493 days in the anticoagulation only arm and 266 days for anticoagulation + vena cava filter (p < 0.57). Complete resolution of venous thromboembolism occurred in 51 % of patients within 8 weeks of initiating anticoagulation. CONCLUSIONS: No advantage was found for placement of a vena cava filter in addition to anticoagulation with fondaparinux sodium in terms of safety, recurrent thrombosis, recurrent pulmonary embolism, or survival in this prospective randomized trial evaluating anticoagulation plus a vena cava filter in cancer patients. Favorable complete resolution rates of thrombosis were observed on both study arms.


Assuntos
Anticoagulantes/uso terapêutico , Polissacarídeos/uso terapêutico , Filtros de Veia Cava , Tromboembolia Venosa/terapia , Idoso , Idoso de 80 Anos ou mais , Anticoagulantes/efeitos adversos , Terapia Combinada , Fondaparinux , Hemorragia/induzido quimicamente , Hemorragia/epidemiologia , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/complicações , Neoplasias/patologia , Polissacarídeos/efeitos adversos , Estudos Prospectivos , Recidiva , Taxa de Sobrevida , Resultado do Tratamento , Filtros de Veia Cava/efeitos adversos , Tromboembolia Venosa/patologia
13.
Anticancer Drugs ; 23(3): 272-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23427335

RESUMO

An in-vitro 72-h assay using median effect analysis and curve shift analysis was used to evaluate the utility of potentially clinically useful combinations of agents for synergism or antagonism. Six human breast cancer cell lines, both receptor rich and receptor poor, were studied.Panobinostat (LBH-589), a pan histone deacetylase inhibitor with a multitude of biological effects, exhibits time-dependent synergistic effects in breast cancer cell lines with docetaxel, doxorubicin, or gemcitabine in clinically relevant concentrations. Survivin expression was markedly downregulated in the presence of panobinostat with gemcitabine. Bortezomib, a proteasome inhibitor,markedly enhanced the cytotoxic effects of panobinostat combined with gemcitabine. Panobinostat did not demonstrate universal enhancement of cytotoxic drugs,and therefore, synergy was dependent on the second agent selected. No synergy was noted with anti-Her2 agents in Her2 overexpressing cell lines. Metformin combined with panobinostat demonstrated no synergy in this test system. These effects were confirmed by an apoptosis assay and caspase-3 production. A positive drug interaction was identified. The triplet of panobinostat with either doxorubicin/carboplatin or gemcitabine/carboplatin was especially potent in all cell lines. As all these agents are clinically available, further studies of the potent combinations are warranted.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Proteínas Inibidoras de Apoptose/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carboplatina/administração & dosagem , Caspase 3/metabolismo , Linhagem Celular Tumoral , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Docetaxel , Regulação para Baixo/efeitos dos fármacos , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Ácidos Graxos Monoinsaturados/administração & dosagem , Feminino , Fluvastatina , Humanos , Ácidos Hidroxâmicos/administração & dosagem , Ácidos Hidroxâmicos/farmacologia , Indóis/administração & dosagem , Indóis/farmacologia , Panobinostat , Receptor ErbB-2/metabolismo , Survivina , Taxoides/administração & dosagem , Taxoides/farmacologia , Gencitabina
14.
Mol Med ; 17(7-8): 854-62, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21424107

RESUMO

The hereditary forms of breast cancer identified by BRCA1 and BRCA2 genes have a defect in homologous DNA repair and demonstrate a dependence on alternate DNA repair processes by base excision repair, which requires poly(ADP-ribose) polymerase 1 (PARP-1). siRNA and deletion mutations demonstrate that interference with PARP-1 function results in enhanced cell death when the malignancy has a defect in homologous recombination. These findings resulted in a plethora of agents in clinical trials that interfere with DNA repair, and these agents offer the potential of being more selective in their effects than classic chemotherapeutic drugs. An electronic search of the National Library of Medicine for published articles written in English used the terms "PARP inhibitors" and "breast cancer" to find prospective, retrospective and review articles. Additional searches were done for articles dealing with mechanism of action. A total of 152 articles dealing with breast cancer and PARP inhibition were identified. PARP inhibition not only affects nonhomologous repair, but also has several other nongenomic functions. Mutational resistance to these agents was seen in preclinical studies. To date, PARP-1 inhibitors were shown to enhance cytotoxic effects of some chemotherapy agents. This new class of agents may offer more therapeutic specificity by exploiting a DNA repair defect seen in some human tumors with initial clinical trials demonstrating antitumor activity. Although PARP inhibitors may offer a therapeutic option for selected malignancies, the long-term effects of these agents have not yet been defined.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Inibidores Enzimáticos/uso terapêutico , Inibidores de Poli(ADP-Ribose) Polimerases , Poli(ADP-Ribose) Polimerases/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Inibidores Enzimáticos/farmacologia , Feminino , Genes BRCA1 , Genes BRCA2 , Genes Letais/genética , Humanos , Mutação , Poli(ADP-Ribose) Polimerases/genética
15.
Invest New Drugs ; 29(6): 1224-9, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20533074

RESUMO

Although platinum based therapy has improved short term survival of patients with metastatic ovarian cancer, the majority of patients continue to relapse and eventually die of their disease. Currently, a plethora of agents are in development, but how to combine them to enhance efficacy remains largely empiric. We have used short in vitro culture of defined cell lines with application of promising agents and analysis for cell death using a MTT assay to identify potentially useful combinations. Using median effect analysis, curve shift analysis and apoptosis assays, we can identify when agents are synergistic or antagonistic when applied together. Up to three agents can be studied in combination. Using three cell lines: SK-OV3, CaOV-3, and ES-2 (a clear cell tumor), we have identified that panobinostat (LBH-589), a broad histone deacetylase inhibitor in clinical trials, demonstrates global synergy with gemcitabine, with paclitaxel, and additive to synergistic effects with 5'DFUR. The triplet of panobinostat, doxorubicin, and carboplatin is especially synergistic in these cell lines. These effects are cytotoxic and not cytostatic. As all these agents are used clinically, we have identified combinations which warrant further investigation.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sinergismo Farmacológico , Feminino , Inibidores de Histona Desacetilases/administração & dosagem , Humanos , Ácidos Hidroxâmicos/administração & dosagem , Indóis , Neoplasias Ovarianas/patologia , Panobinostat
16.
Crit Rev Oncol Hematol ; 76(3): 173-85, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20144549

RESUMO

Small regulatory ribonucleic acids (RNAs) are recently recognized as being connected with a growing list of common diseases such as: cancer, heart disease, diabetes and inflammation and to date more than 5,000 publications are recorded on PubMed alone. Specific pathways generate each class of RNAs and their activities converge in the process of silence interference. In gastrointestinal malignancies microRNAs are deregulated, sometimes found in higher or lower levels depending on the type of malignancy and stage of the disease, functioning either as tumor suppressors or as oncogenes they interact forming regulatory loops with known transcription factors and signaling pathways. MiRNAs extracted from archived tissue biopsies can be used effectively as diagnostic, prognostic tools and molecular markers because they are stable over time and resistant to RNAse degradation. The distinct physiology of small RNAs may translate in more targeted cancer therapies in the near future.


Assuntos
Neoplasias Gastrointestinais/genética , Pequeno RNA não Traduzido/genética , Animais , Neoplasias Gastrointestinais/diagnóstico , Neoplasias Gastrointestinais/terapia , Regulação Neoplásica da Expressão Gênica , Terapia Genética , Humanos , MicroRNAs/genética , Interferência de RNA , RNA Interferente Pequeno/genética
17.
Leuk Res ; 34(4): 487-91, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19748672

RESUMO

Amonafide-l-malate (amonafide) is a unique DNA intercalator that maintains activity in the presence of MDR mechanisms, a frequent cause of treatment-failure in secondary AML. 43 patients with relapsed/refractory or secondary AML or CML blast crisis were enrolled into two phase I dose-escalation studies investigating amonafide as monotherapy or in combination with cytarabine. 3/17 patients in the monotherapy trial and 10/26 patients in the combination trial achieved a complete remission. Between both trials responses occurred in 9/20 patients with secondary AML. Both trials demonstrated an acceptable safety profile and significant antileukemic activity in patients with poor-risk AML, especially those with secondary AML.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Citarabina/administração & dosagem , Leucemia Mieloide Aguda/tratamento farmacológico , Naftalimidas/administração & dosagem , Adenina , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Citarabina/efeitos adversos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Naftalimidas/efeitos adversos , Organofosfonatos , Indução de Remissão , Fatores de Risco , Resultado do Tratamento , Adulto Jovem
18.
J Clin Oncol ; 27(21): 3430-6, 2009 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-19470942

RESUMO

PURPOSE We have demonstrated that patients with HER2-amplified tumors derive more benefit from higher doses of doxorubicin-containing chemotherapy (cyclophosphamide, doxorubicin, and fluorouracil [CAF]). Because topoisomerase IIalpha (Topo-IIalpha) is a target for doxorubicin and is coamplified in 20% to 50% of HER2-amplified tumors, we postulated that Topo-IIalpha copy number might account for the benefit from CAF dose escalation in HER2-positive tumors. To address this hypothesis, we examined Topo-IIalpha and HER2 copy number, CAF dose, and clinical outcomes in Cancer and Leukemia Group B (CALGB) 8541. PATIENTS AND METHODS Topo-IIalpha and HER2 copy number were measured by fluorescent in situ hybridization (FISH) using a triple-probe system, which includes Topo-IIalpha, HER2, and chromosome 17 (CEP17). Topo-IIalpha and/or HER2 were classified as amplified (> or = two copies/CEP17, deleted (< or = 0.67 copies/CEP17) and normal copy number (> .67 to < 2.0 copies/CEP17). Results Topo-IIalpha/HER2/CEP17 measurement was successful in 624 of 687 cases. HER2 was amplified in 117 cases (19%). Topo-IIalpha was amplified in 41 cases (7%) and deleted in 69 cases (11%). Topo-IIalpha amplification was highly correlated with HER2 amplification (39 of 41; P < .0001), HER2 by immunohistochemistry, and by dual-probe FISH. Topo-IIalpha was deleted in both the HER2-amplified (30 of 69; 43%), normal (22 of 69; 32%) and HER2-deleted tumors (17 of 69; 25%). Although Topo-IIalpha-amplified tumors were nearly always HER2 amplified, these tumors did not receive benefit from increasing the dose of CAF (P = .15). CONCLUSION The correlative companion study CALGB 8541-150013 does not support the hypothesis that Topo-IIalpha amplification is the mechanism behind benefit from increased dose of anthracyclines in HER2-positive breast cancer.


Assuntos
Antraciclinas/uso terapêutico , Antígenos de Neoplasias/genética , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , DNA Topoisomerases Tipo II/genética , Proteínas de Ligação a DNA/genética , Amplificação de Genes/efeitos dos fármacos , Receptor ErbB-2/genética , Antibióticos Antineoplásicos , Antígenos de Neoplasias/fisiologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Biomarcadores Tumorais/análise , Ciclofosfamida/farmacologia , DNA Topoisomerases Tipo II/fisiologia , Proteínas de Ligação a DNA/fisiologia , Doxorrubicina/farmacologia , Interações Medicamentosas , Fluoruracila/farmacologia , Humanos , Imuno-Histoquímica , Receptor ErbB-2/metabolismo
19.
Curr Treat Options Oncol ; 10(3-4): 144-58, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19360475

RESUMO

The use of cytotoxic therapy in the fit elderly breast cancer patient has been tempered with concerns of age, physical function, and co-morbid illness. In the appropriate patient with biologically aggressive disease, such as receptor poor breast cancer, it is reasonable to consider combination chemotherapy as part of an adjuvant program. If this approach is to be employed, the physician must also consider the patient's co-morbid conditions and status of function in society as potential indicators of toxicity or lack of benefit. In this case, a formal geriatric assessment is of value. A Cancer and Leukemia Group B (CALGB) trial of monotherapy vs combination cytotoxic therapy as adjuvant treatment for localized breast cancer patients over 65 years of age has determined that the combination approach is superior to single agent therapy. In an unplanned analysis of receptor rich and receptor poor tumors, the patients with receptor poor tumors seemed to achieve the greatest benefit from combination cytotoxic therapy. Adjuvant chemotherapy can also be considered for patients with high-risk receptor rich breast cancers. However, the use of chemotherapy in the elderly patient with breast cancer is largely based upon data emerging from trials in younger patients. Studies specifically for patients over 65 years of age are urgently needed in this population to provide evidence-based proof of the current approach.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Quimioterapia Adjuvante/métodos , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica , Ensaios Clínicos como Assunto , Comorbidade , Detecção Precoce de Câncer , Feminino , Avaliação Geriátrica , Humanos , Oncologia/métodos , Pessoa de Meia-Idade
20.
Oncology ; 76(4): 231-8, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19246947

RESUMO

BACKGROUND: Hypersensitivity is a well-known complication of the platinum agents cisplatin and carboplatin. Although hypersensitivity to oxaliplatin has been noted, the incidence varies significantly in reports. Risk factors for developing reactions specifically to oxaliplatin have not been evaluated. We report the 5-year incidence of hypersensitivity to oxaliplatin in our clinical program, the patient and disease characteristics associated with its occurrence, and review the literature. METHODS: Clinical information on all patients treated with oxaliplatin between September 2002 and August 2007 was retrospectively reviewed. Data from patients who experienced hypersensitivity were compared to patients treated with this agent who did not. Risk factors investigated included age, sex, diagnosis, disease stage, presence of preexisting allergies, chemotherapy received, and use of oxaliplatin in front-line versus salvage therapy. RESULTS: 247 patients received oxaliplatin, with 29 experiencing hypersensitivity, for an incidence of 11.7% (95% CI 7.7-15.8). Grade 3/4 events occurred in 1.6%. Hypersensitivity was associated with younger mean age (54.9 +/- 12.5 vs. 60.4 +/- 12.4 years with reactions vs. those without, p = 0.02), female gender (17.2% of females vs. 6.4% of males, p = 0.01) and with use of oxaliplatin as salvage therapy (23.9% second-line or higher vs. 9.1% front-line, p = 0.01). CONCLUSIONS: Our data demonstrate an incidence of hypersensitivity to oxaliplatin of 11.7%, with grade 3/4 events in 1.6%. As use of this agent becomes more widespread, increased vigilance for this potentially serious complication should be high, especially amongst younger patients, females, and with the use of oxaliplatin as salvage therapy; three newly recognized potential risk factors.


Assuntos
Antineoplásicos/efeitos adversos , Hipersensibilidade a Drogas/epidemiologia , Compostos Organoplatínicos/efeitos adversos , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Hipersensibilidade a Drogas/etiologia , Feminino , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Oxaliplatina , Estudos Retrospectivos , Fatores de Risco , Caracteres Sexuais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA