Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Medicine (Baltimore) ; 100(41): e27521, 2021 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-34731142

RESUMO

ABSTRACT: To investigate the effect of a combined immune score including the lymphocyte-to-monocyte ratio (LMR) and uninvolved immunoglobulin (u-Ig) levels on the prognosis of newly diagnosed multiple myeloma (NDMM) patients treated with bortezomib.Clinical data of 201 NDMM patients were retrospectively analyzed. Patients with LMR ≥ 3.6 and LMR < 3.6 were scored 0 and 1, respectively. Patients with preserved u-Ig levels, suppression of 1 u-Ig, and suppression of at least 2 u-Igs were scored 0, 1, and 2, respectively. The immune score, established from these individual scores, was used to separate patients into good (0-1 points), intermediate (2 points), and poor (3 points) risk groups. The baseline data, objective remission rate (ORR), whether receive maintenance treatment regularly and overall survival of patients before treatment were analyzed.The ORR of the good-risk group was significantly higher than that of the intermediate-risk group (75.6% vs 57.7%, P = .044) and the poor-risk group (75.6% vs 48.2%, P = .007). The multivariate analysis results showed that age ≥ 65 years, International Staging System stage III, platelet count ≤ 100 × 109/L, lactate dehydrogenase (LDH) > 250 U/L, serum calcium > 2.75 mmol/L, no receipt of regular maintenance treatment, LMR < 3.6, suppressed u-Igs = 1, suppressed u-Igs ≥ 2, intermediate-risk group and poor-risk group were independent predictors of poor overall survival.In the bortezomib era, the LMR, u-Ig levels, and the immune score play an important role in the prognosis of NDMM patients. Among them, the immune score showed the strongest prognostic value, and it could be a beneficial supplement for the early identification of high-risk patients.


Assuntos
Antineoplásicos/uso terapêutico , Bortezomib/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/mortalidade , Fatores Etários , Idoso , Antineoplásicos/administração & dosagem , Bortezomib/administração & dosagem , Cálcio/sangue , Estudos de Casos e Controles , Feminino , Humanos , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/imunologia , Imunoglobulinas/efeitos dos fármacos , Imunoglobulinas/imunologia , L-Lactato Desidrogenase/análise , Linfócitos/citologia , Masculino , Pessoa de Meia-Idade , Monócitos/citologia , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/imunologia , Estadiamento de Neoplasias/métodos , Contagem de Plaquetas/estatística & dados numéricos , Contagem de Plaquetas/tendências , Valor Preditivo dos Testes , Prognóstico , Estudos Retrospectivos , Fatores de Risco
2.
Biol Pharm Bull ; 44(2): 271-274, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33518680

RESUMO

The anticancer agents including oxaliplatin, paclitaxel, and bortezomib cause severe peripheral neuropathy. The Kampo medicine Sokeikakketsuto (SOKT) has been widely used to treat several types of pain. In this study, the analgesic effects of SOKT on oxaliplatin-, paclitaxel-, and bortezomib-induced peripheral neuropathy were investigated in rat models. Rats were treated with oxaliplatin (4 mg/kg, intraperitoneally (i.p.), twice a week for four weeks), paclitaxel (4 mg/kg, i.p., twice a week for two weeks), or bortezomib (0.2 mg/kg, i.p., twice a week for two weeks). SOKT (0.3 or 1.0 g/kg) or duloxetine hydrochloride (30 mg/kg, as a positive control) was administered orally after neuropathy developed. Mechanical allodynia and cold hyperalgesia were assessed using the von Frey test and the acetone test, respectively. These tests were performed immediately before and 30, 60, 90, and 120 min after the administration of the drugs. Repeated treatment of oxaliplatin induced mechanical allodynia and cold hyperalgesia. A single administration of SOKT (1 g/kg, per os (p.o.)), as well as duloxetine, temporarily reversed both the mechanical allodynia and the cold hyperalgesia. Repeated administration of paclitaxel and bortezomib also induced the mechanical allodynia. SOKT and duloxetine reversed the mechanical allodynia caused by bortezomib, but not by paclitaxel. SOKT might have the potential to become a new drug to relieve the symptom of oxaliplatin- or bortezomib-induced peripheral neuropathy.


Assuntos
Analgésicos/farmacologia , Antineoplásicos/efeitos adversos , Temperatura Baixa/efeitos adversos , Medicamentos de Ervas Chinesas/farmacologia , Hiperalgesia/tratamento farmacológico , Analgésicos/uso terapêutico , Animais , Antineoplásicos/administração & dosagem , Bortezomib/administração & dosagem , Bortezomib/efeitos adversos , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/uso terapêutico , Cloridrato de Duloxetina/farmacologia , Cloridrato de Duloxetina/uso terapêutico , Humanos , Hiperalgesia/induzido quimicamente , Hiperalgesia/diagnóstico , Masculino , Medicina Kampo/métodos , Oxaliplatina/administração & dosagem , Oxaliplatina/efeitos adversos , Paclitaxel/administração & dosagem , Paclitaxel/efeitos adversos , Medição da Dor , Ratos , Ratos Sprague-Dawley
3.
FASEB J ; 35(2): e21231, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33428275

RESUMO

Tumors of the nervous system including glioblastoma multiforme (GBM) are the most frequent and aggressive form of brain tumors; however, little is known about the impact of the circadian timing system on the formation, growth, and treatment of these tumors. We investigated day/night differences in tumor growth after injection of A530 glioma cells isolated from malignant peripheral nerve sheath tumor (MPNSTs) of NPcis (Trp53+/- ; Nf1+/- ) mice. Synchronized A530 cell cultures expressing typical glial markers were injected at the beginning of the day or night into the sciatic nerve zone of C57BL/6 mice subject to a 12:12 hours light/dark (LD) cycle or after being released to constant darkness (DD). Tumors generated in animals injected early at night in the LD cycle or in DD showed higher growth rates than in animals injected diurnally. No differences were found when animals were injected at the same time with cultures synchronized 12 hours apart. Similar experiments performed with B16 melanoma cells showed higher tumor growth rates in animals injected at the beginning of the night compared to those injected in the daytime. A higher tumor growth rate than that in controls was observed when mice were injected with knocked-down clock gene Bmal1 cells. Finally, when we compared day/night administration of different doses of the proteasome inhibitor Bortezomib (0.5-1.5 mg/kg) in tumor-bearing animals, we found that low-dose chemotherapy displayed higher efficacy when administered at night. Results suggest the existence of a precise temporal control of tumor growth and of drug efficacy in which the host state and susceptibility are critical.


Assuntos
Neoplasias Encefálicas/patologia , Ritmo Circadiano , Glioblastoma/patologia , Fotoperíodo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Fatores de Transcrição ARNTL/genética , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Bortezomib/administração & dosagem , Bortezomib/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Esquema de Medicação , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Camundongos , Camundongos Endogâmicos C57BL , Neurofibromina 1/genética , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Ensaios Antitumorais Modelo de Xenoenxerto/normas
4.
Hum Cell ; 34(1): 271-278, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32997328

RESUMO

Atypical teratoid/rhabdoid tumor (AT/RT) is a rare intracranial tumor occurring predominantly in young children. The prognosis is poor, and no effective treatment is currently available. To develop novel effective therapies, there is a need for experimental models for AT/RT. In this research, we established a cell line from a patient's AT/RT tissue (designated ATRT_OCGH) and performed drug screening using 164 FDA-approved anti-cancer agents, to identify candidates for therapeutic options. We found that bortezomib, a proteasome inhibitor, was among the agents for which the cell line showed high sensitivity, along with tyrosine kinase inhibitors, topoisomerase inhibitors, and histone deacetylase inhibitors, which are known to exert anti-AT/RT effects. Concomitant use of panobinostat potentiated the inhibitory effect of bortezomib on AT/RT cell proliferation. Our findings may provide a rationale for considering combination therapy of panobinostat and bortezomib for treatment of AT/RT.


Assuntos
Antineoplásicos/farmacologia , Bortezomib/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Inibidores de Proteassoma/farmacologia , Tumor Rabdoide/tratamento farmacológico , Tumor Rabdoide/patologia , Teratoma/tratamento farmacológico , Teratoma/patologia , Antineoplásicos/administração & dosagem , Bortezomib/administração & dosagem , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Panobinostat/administração & dosagem , Panobinostat/farmacologia , Prognóstico , Inibidores de Proteassoma/administração & dosagem
5.
PLoS One ; 15(2): e0229469, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32084254

RESUMO

Treatment of transplant-ineligible (TNE) newly diagnosed multiple myeloma (NDMM) requires a balance between disease control and maintaining quality of life (QoL). Patients value treatment-free remission periods in this incurable condition, as they are associated with better QoL. We set out to study clinical outcomes of consecutive TNE NDMM patients in routine care treated in Thames Valley Cancer Network between 2009 and 2017. The primary outcome was the evaluation of the treatment-free interval (TFI) after 1st and subsequent lines of therapy in the total cohort and in individual subgroups, according to age (≤75 vs. >75 years), and co-morbidities using Charlson Co-morbidity Index (CCI): mild: 0-2 vs. moderate: 3-4 vs. severe: ≥5). Secondary outcomes include response rates, overall survival (OS) and progression-free survival (PFS) between subgroups: according to age and according to co-morbidities. In a total cohort of 292 patients, median TFI (IQR) was longest after first-line therapy 6.9 months (1.4-16.9), reducing after second line therapy to 1.8 months (.7-6.9), and after third line therapy to 0.6 months (0.2-1.5). Median TFI followed the same trend across the different subgroups, by age (≤75, >75 years) and by CCI (0-2, 3-4, ≥5). Overall response rate (ORR) to first line therapy for total cohort was 67%, with responses categorised as complete response (CR): 21%, very good partial response: 16%, partial response: 30%, stable disease: 18%, and progressive disease: 8%. ORR in individual subgroups by age were (≤75: 70% vs. >75: 63%), and by CCI (0-2: 65% vs. 3-4: 71% vs. ≥5: 77%). Median OS and PFS for the total cohort were (30.2 months, 95% CI: 23.8-36.9), and (9 months, 95% CI: 7.9-9.8), respectively. Patients aged >75 years showed a significant reduction in OS and PFS compared to those ≤75 years of age: OS (49.0 vs. 22.4 months, p<0.0001, HR: 2.08, 95% CI: 1.5-2.8), PFS (9.7 vs. 8.0 months, p<0.01, HR: 1.47, 95% CI: 1.1-1.9). Median OS was significantly reduced with worsening co-morbidities: (CCI 0-2: 52.4 months vs. CCI 3-4: 33.0 months vs. CCI ≥5: 24.0 months, p = 0.01, HR: 1.43, 95% CI: 1.1-1.9). Median PFS was significantly reduced in the severely co-morbid subgroup (CCI 0-2: 9.4 months vs. CCI 3-4: 9.6 months vs. CCI ≥5: 7.1 months, p = 0.025, HR: 1.3, 95% CI: 1.0-1.6). This study demonstrated that first line therapy in the TNE NDMM setting resulted in the longest TFI which was modest at a median of 6.9 months, and decreased significantly following subsequent lines of therapy and across the different subgroups by age and by co-morbidities. Therapy objective should be to maximise the benefit of first line treatment. We envisage that the recent shift towards a continuous therapeutic approach will benefit TNE patients in view of improved survival data demonstrated by a number phase 3 trials. When continuous therapy is not appropriate due to patient choice or toxicities, an efficacious (not limited to thalidomide and bortezomib) but tolerable first line FDT strategy, which can maximise TFI and maintain a good QoL, remains a reasonable alternative approach.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Qualidade de Vida , Idoso , Idoso de 80 Anos ou mais , Bortezomib/administração & dosagem , Esquema de Medicação , Feminino , Seguimentos , Humanos , Lenalidomida/administração & dosagem , Masculino , Mieloma Múltiplo/epidemiologia , Mieloma Múltiplo/patologia , Indução de Remissão , Estudos Retrospectivos , Taxa de Sobrevida , Talidomida/administração & dosagem , Fatores de Tempo , Resultado do Tratamento , Reino Unido/epidemiologia
8.
Colloids Surf B Biointerfaces ; 160: 73-83, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28917152

RESUMO

Pancreatic cancer has extremely poor prognosis with an 85% mortality rate that results from aggressive and asymptomatic growth, high metastatic potential, and rapid development of resistance to already ineffective chemotherapy. In this study, plasmonic hollow gold nanoshells (GNS) coated with PEGylated thermosensitive lipids were prepared as an efficient platform to ratiometrically co-deliver two drugs, bortezomib and gemcitabine (GNS-L/GB), for combinational chemotherapy and photothermal therapy of pancreatic cancer. Bortezomib was loaded within the lipid bilayers, while gemcitabine was loaded into the hydrophilic interior of the porous GNS via an ammonium sulfate-driven pH gradient method. Physicochemical characterizations and biological studies of GNS-L/GB were performed, with the latter using cytotoxicity assays, cellular uptake and apoptosis assays, live/dead assays, and western blot analysis of pancreatic cancer cell lines (MIA PaCa-2 and PANC-1). The nanoshells showed remotely controllable drug release when exposed to near-infrared laser for site-specific delivery. GNS-L/GB showed synergistic cytotoxicity and improved internalization by cancer cells. High-powered near-infrared continuous wave laser (λ=808nm) effectively killed cancer cells via the photothermal effect of GNS-L/GB, irrespective of cell type in a power density-, time-, and GNS dose-dependent manner. These results suggest that this method can provide a novel approach to achieve synergistic combinational chemotherapy and photothermal therapy, even with resistant pancreatic cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ouro/química , Lipídeos/química , Nanoconchas/química , Neoplasias Pancreáticas/tratamento farmacológico , Fototerapia/métodos , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Bortezomib/administração & dosagem , Bortezomib/química , Linhagem Celular Tumoral , Terapia Combinada , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Desoxicitidina/química , Sistemas de Liberação de Medicamentos/métodos , Liberação Controlada de Fármacos/efeitos da radiação , Humanos , Interações Hidrofóbicas e Hidrofílicas , Raios Infravermelhos , Nanoconchas/ultraestrutura , Polietilenoglicóis/química , Temperatura , Gencitabina
9.
Br J Haematol ; 178(1): 61-71, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28382618

RESUMO

This phase II study explored the effects of bortezomib consolidation versus observation on myeloma-related bone disease in patients who had a partial response or better after frontline high-dose therapy and autologous stem cell transplantation. Patients were randomized to receive four 35-day cycles of bortezomib 1·6 mg/m2 intravenously on days 1, 8, 15 and 22, or an equivalent observation period, and followed up for disease status/survival. The modified intent-to-treat population included 104 patients (51 bortezomib, 53 observation). There were no meaningful differences in the primary endpoint of change from baseline to end of treatment in bone mineral density (BMD). End-of-treatment rates (bortezomib versus observation) of complete response/stringent complete response were 22% vs. 11% (P = 0·19), very good partial response or better of 80% vs. 68% (P = 0·17), and progressive disease of 8% vs. 23% (P = 0·06); median progression-free survival was 44·9 months vs. 21·8 months (P = 0·22). Adverse events observed ≥15% more frequently with bortezomib versus observation were diarrhoea (37% vs. 0), peripheral sensory neuropathy (20% vs. 4%), nausea (18% vs. 0) and vomiting (16% vs. 0). Compared with observation, bortezomib appeared to have little impact on bone metabolism/health, but was associated with trends for improved myeloma response and survival.


Assuntos
Antineoplásicos/uso terapêutico , Bortezomib/uso terapêutico , Quimioterapia de Consolidação/métodos , Mieloma Múltiplo/tratamento farmacológico , Osteólise/tratamento farmacológico , Adulto , Idoso , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Biomarcadores/sangue , Bortezomib/administração & dosagem , Bortezomib/efeitos adversos , Esquema de Medicação , Feminino , Seguimentos , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/complicações , Mieloma Múltiplo/fisiopatologia , Osteólise/etiologia , Osteólise/fisiopatologia , Transplante de Células-Tronco , Resultado do Tratamento
10.
Blood Cancer J ; 6(11): e492, 2016 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-27813535

RESUMO

The survival of Waldenstrom macroglobulinemia (WM) tumor cells hinges on aberrant B-cell receptor (BCR) and MYD88 signaling. WM cells upregulate the proteasome function to sustain the BCR-driven growth while maintaining homeostasis. Clinically, two treatment strategies are used to disrupt these complementary yet mutually exclusive WM survival pathways via ibrutinib (targets BTK/MYD88 node) and bortezomib (targets 20 S proteasome). Despite the success of both agents, WM patients eventually become refractory to treatment, highlighting the adaptive plasticity of WM cells and underscoring the need for development of new therapeutics. Here we provide a comprehensive preclinical report on the anti-WM activity of VLX1570, a novel small-molecule inhibitor of the deubiquitinating enzymes (DUBs), ubiquitin-specific protease 14 (USP14) and ubiquitin carboxyl-terminal hydrolase isozyme L5 (UCHL5). Both DUBs reside in the 19 S proteasome cap and their inhibition by VLX1570 results in rapid and tumor-specific apoptosis in bortezomib- or ibrutinib-resistant WM cells. Notably, treatment of WM cells with VLX1570 downregulated BCR-associated elements BTK, MYD88, NFATC, NF-κB and CXCR4, the latter whose dysregulated function is linked to ibrutinib resistance. VLX1570 administered to WM-xenografted mice resulted in decreased tumor burden and prolonged survival (P=0.0008) compared with vehicle-treated mice. Overall, our report demonstrates significant value in targeting USP14/UCHL5 with VLX1570 in drug-resistant WM and carries a high potential for clinical translation.


Assuntos
Azepinas/administração & dosagem , Compostos de Benzilideno/administração & dosagem , Ubiquitina Tiolesterase/antagonistas & inibidores , Macroglobulinemia de Waldenstrom/tratamento farmacológico , Adenina/análogos & derivados , Apoptose/efeitos dos fármacos , Bortezomib/administração & dosagem , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Enzimas Desubiquitinantes/antagonistas & inibidores , Enzimas Desubiquitinantes/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Piperidinas , Pirazóis/administração & dosagem , Pirimidinas/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Ubiquitina Tiolesterase/genética , Macroglobulinemia de Waldenstrom/genética , Macroglobulinemia de Waldenstrom/patologia
11.
Zhonghua Xue Ye Xue Za Zhi ; 37(9): 784-789, 2016 Sep 14.
Artigo em Chinês | MEDLINE | ID: mdl-27719722

RESUMO

Objective: To determine the anti-tumor effects of 13-cis-retinoic acid (13cRA) combined with interferonα-2b (IFNα-2b) in mantle cell lymphoma (MCL) animal model. Methods: The animal model of MCL was established by introducing Jeko-1 cell line into severe combined immunodeficiency disease mice. The successfully tumor-developed mice were assigned to different groups treated with negative control group (solvents), 13cRA (high dose: 200mg/kg; middle dose: 100mg/kg; low dose: 50 mg/kg) alone, IFNα-2b alone or combination of different dose of 13cRA with IFNα-2b, and positive control group (bortezomib, rituximab, cyclophosphamide), respectively. Variations of tumor volume were observed regularly. The relative tumor proliferation rate and tumor inhibition rate were calculated. Immunohistochemistry stain was used to detect the Ki-67 expression and TUNEL was applied to measure the apoptosis of tumor cells. Furthermore, the levels of Cyclin D1, caspase 9 and Rb protein were measured by Western-blot method. Results: ① The relative tumor proliferation rates (T/C%)were 30%, 37%, 32% and 33% in middle dose, high dose groups of 13cRA as well as their combination with IFN α-2b, respectively. ② Comparing with the negative control, both 13cRA at different doses and its combination with IFNα-2b remarkably inhibited the tumor growth (P<0.05), while no statistic significance existed in different dose group of 13cRA. IFN-α 2b alone didn't demonstrate the tumor-inhibition effects (P>0.05). Middle dose of 13cRA and its combination with IFN-α-2b demonstrated relatively high tumor-inhibition effects (59.2% and 62.6% respectively), which were similar to the effects in positive control (69.4%). ③ There was no statistic difference of Ki-67 in each experimental group. ④ Comparing with negative control group, all doses of 13cRA and their combinations with IFNα-2b remarkably increased the apoptosis (P<0.05), similar to the positive control group (P>0.05). However, IFNα-2b alone didn' t promote the apoptosis of tumor tissue (P=0.098). ⑤ Comparing with negative control group, IFNα-2b combined with each dose of 13cRA significantly decreased the levels of cycling D1 and procaspase-9, while increased the level of cleaved caspase-9 (P<0.05), which were similar to the positive control group (P>0.05). Nevertheless, 13cRA alone didn't demonstrate such effects. Conclusion: In the MCL animal model, IFNα-2b alone showed no effects, but combined with IFNα-2b, 13cRA displayed anti-tumor effects at different doses. The anti-tumor mechanism of 13cRA combined with IFNα-2b was probably down-regulation of the cyclin D1 expression, inhibition of cell proliferation and induction of apoptosis by activating caspase-9.


Assuntos
Interferon-alfa/uso terapêutico , Isotretinoína/uso terapêutico , Linfoma de Célula do Manto/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Bortezomib/administração & dosagem , Caspase 9/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Ciclina D1/efeitos dos fármacos , Ciclina D1/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Humanos , Camundongos
12.
Lima; s.n; jul. 2016. tab.
Não convencional em Espanhol | LILACS, BRISA | ID: biblio-847613

RESUMO

INTRODUCCIÓN: Antecedentes: El presente dictamen expone la evaluación de tecnología de la eficacia y seguridad de Bortezomib para su uso en pacientes con mieloma múltiple recidivante y/o refractario a uno o más tratamientos previos. Como antecedente relevante, existe un dictamen emitido por IETSI-ESSALUD sobre el uso de Bortezomib como primera línea de tratamiento en pacientes con mieloma múltiple. Generalidades: El mieloma múltiple es una neoplasia hematológica caracterizada por la proliferación de células plasmáticas que producen inmunoglobulinas monoclonales. Estas células, proliferan en la medula ósea y suelen producir destrucción ósea masiva manifestada por lesiones osteolíticas, osteopenia e incluso fracturas patológicas. Clínicamente suele presentarse con dolor óseo y lesiones líticas, proteínas séricas totales incrementadas, proteínas monoclonales incrementadas en suero u orina, anemia de origen incierto, hipercalcemia e incluso insuficiencia renal aguda o síndrome nefrótico por amiloidosis primaria concurrente. Tecnología Sanitaria de Interés: sobre Bortezomib: Bortezomib es un inhibidor de proteosomas, el primero de su clase, que actúa en la homeostasis proteica celular bloqueando específicamente el proteosoma 26. Este proteosoma 26 es una enzima que se encarga normalmente de catalizar todas las proteínas anormales que son generadas usualmente en el metabolismo. La inhibición de este proteosoma conlleva a apoptosis celular y es por lo tanto un blanco interesante en terapia del cáncer. Estrategia de Busqueda: Se realizó una estrategia de búsqueda sistemática de la evidencia científica con respecto al uso de bortezomib en pacientes con mieloma múltiple recidivante y/o refractario a uno o más tratamientos previos. Las siguientes fuentes han sido revisadas y consultadas: Medline/Pubmed, Trip Database, The Cochrane Library, The National Institute for Health and Care Excellence (NICE) del Reino Unido, The Scottish Intercollegiate Guidelines Network (SIGN) de Escocia, The National Guideline Clearinghouse (NCG) de los Estados Unidos, The American Society of Clinical Oncology (ASCO) de los Estados Unidos, The National Comprehensive Cancer Network (NCCN) de los Estados Unidos. RESULTADOS: Sinopsis de la Evidencia: Se realizó la búsqueda y revisión de la evidencia científica actual para la evaluación de la eficacia y seguridad de Bortezomib en pacientes con mieloma múltiple recidivante y/o refractario a uno o más tratamientos previos. Se presenta la información encontrada de acuerdo al tipo de evidencia revisada: Guías de Práctica Clínica (GPC): Se describió la GPC de la Evaluaciones de Tecnologías Sanitarias (ETS): Se encontró una evaluación de tecnología elaborada por la NICE del Reino Unido del año 2007 (Documento TA 129 de la NICE).NCCN Versión 3 del año 2016; Revisiones Sistemáticas (RS) o Meta-análisis: Se encontró la RS de Scott et al., 2016 del grupo colaborativo Cochrane; Estudios Primarios: Se consideraron tres ensayos clínicos pertenecientes a Richardson, et al., 2005, Garderet et al., 2012, Hjorth et al., 2012. CONCLUSIONES: El objetivo del presente dictamen fue evaluar la eficacia y seguridad de Bortezomib para su uso en pacientes con mieloma múltiple recidivante y/o refractario a uno o más tratamientos previos. Esta indicación actualmente no está contemplada en el petitorio de medicamentos. En la bibliografía identificada, existen recomendaciones vigentes para el uso de Bortezomib como tratamiento, o parte del tratamiento, farmacológico para pacientes con mieloma múltiple que han recibido tratamiento (esquemas) previos. Sin embargo la cantidad de evidencia es mucho menor con respecto al uso de este mismo fármaco como primera línea de tratamiento. Se identificó una revisión sistemática Cochrane, la misma que incluyó 3 estudios clínicos aleatorizados cuya población fue compatible con la pregunta PICO del presente dictamen, en donde el sub-análisis de sobrevida global y sobrevida libre de progresión favoreció a bortezomib sobre otros esquemas sin bortezomib. De los 3 estudios identificados solamente un estudio reportó calidad de vida y esta no fue diferente entre los dos grupos de estudio. Por lo expuesto, el Instituto de Evaluación de Tecnologías en Salud e Investigación ­ IETSI, aprueba el uso de Bortezomib en pacientes con mieloma múltiple recidivante y/o refractario a uno o más tratamientos previos. La vigencia del presente dictamen preliminar es de dos años.


Assuntos
Humanos , Mieloma Múltiplo/tratamento farmacológico , Recidiva Local de Neoplasia/tratamento farmacológico , Bortezomib/administração & dosagem , Quimioterapia Combinada , Peru , Avaliação da Tecnologia Biomédica , Resultado do Tratamento
13.
Am J Health Syst Pharm ; 73(7): 441-50, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-27001985

RESUMO

PURPOSE: The mechanism of action, pharmacodynamics, pharmacokinetics, clinical efficacy, interaction potential, adverse effects, and place in therapy of panobinostat are reviewed. SUMMARY: Panobinostat (Farydak, Novartis) is a novel pan-deacetylase inhibitor approved for use in combination with bortezomib and dexamethasone in patients with relapsed or refractory multiple myeloma (RRMM) who have received at least two regimens containing an immunomodulatory drug and bortezomib. National Comprehensive Cancer Network (NCCN) guidelines recommend the use of panobinostat plus bortezomib and dexamethasone as a preferred regimen for previously treated multiple myeloma (MM). A Phase III trial comparing panobinostat or placebo use in combination with bortezomib and dexamethasone demonstrated improved median progression-free survival in the panobinostat group (11.99 months [95% CI, 10.33-12.94 months] versus 8.08 months [95% CI, 7.56-9.23 months]; hazard ratio, 0.63 [95% CI, 0.52-0.76]; p < 0.0001), as well as a significantly higher rate of complete or near complete response (27.6% [95% CI, 23.2-32.4%] versus 15.7% [95% CI, 12.2-19.8%]; p = 0.00006). Common grade 3 or 4 laboratory abnormalities and adverse events associated with panobinostat include thrombocytopenia, lymphopenia, diarrhea, asthenia, fatigue, and peripheral neuropathy. CONCLUSION: Panobinostat is a promising alternative to well-studied, NCCN-recommended regimens for the treatment of RRMM. It has demonstrated efficacy when used in combination with bortezomib and dexamethasone for the treatment of patients with MM who have received at least two prior regimens including bortezomib and an immunomodulatory agent. Despite the observed benefits, concern regarding toxicity may limit panobinostat use in practice.


Assuntos
Inibidores de Histona Desacetilases/administração & dosagem , Ácidos Hidroxâmicos/administração & dosagem , Indóis/administração & dosagem , Mieloma Múltiplo/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Bortezomib/administração & dosagem , Ensaios Clínicos Fase II como Assunto/métodos , Ensaios Clínicos Fase III como Assunto/métodos , Dexametasona/administração & dosagem , Humanos , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/epidemiologia , Panobinostat , Recidiva , Resultado do Tratamento
14.
Expert Opin Investig Drugs ; 25(6): 743-9, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26998658

RESUMO

INTRODUCTION: Sorafenib is an orally available compound that acts predominantly by targeting the Ras/Raf/MEK/ERK pathway and by inhibiting the vascular endothelial growth factor (VEGF). Since the Ras/Raf/MEK/ERK pathway is implicated in the proliferation of multiple myeloma (MM) cells and VEGF in bone marrow neovascularization, sorafenib is a drug offering the potential for targeting two important pathogenetic mechanisms involved in MM. Thus, sorafenib is being proposed for use in MM. AREAS COVERED: In this review, the authors discuss the rationale for the use of sorafenib in MM. They then summarize the clinical development of sorafenib in MM, from initial Phase I to Phase II studies. A systematic literature review of the trials was performed using PubMed. EXPERT OPINION: Preliminary data from phase I/II trials showed that sorafenib had a good safety profile but minimal anti-myeloma activity as a single agent in relapsed/refractory patients. Results of phase II trials, evaluating sorafenib combined with new drugs, such as bortezomib and lenalidomide are eagerly awaited.


Assuntos
Antineoplásicos/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Niacinamida/análogos & derivados , Compostos de Fenilureia/uso terapêutico , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Bortezomib/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Humanos , Lenalidomida , Mieloma Múltiplo/patologia , Niacinamida/efeitos adversos , Niacinamida/farmacologia , Niacinamida/uso terapêutico , Compostos de Fenilureia/efeitos adversos , Compostos de Fenilureia/farmacologia , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Sorafenibe , Talidomida/administração & dosagem , Talidomida/análogos & derivados
15.
J Med Econ ; 19(3): 243-58, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26517601

RESUMO

OBJECTIVE: To conduct a cost-effectiveness assessment of lenalidomide plus dexamethasone (Rd) vs bortezomib plus melphalan and prednisone (VMP) as initial treatment for transplant-ineligible patients with newly-diagnosed multiple myeloma (MM), from a U.S. payer perspective. METHODS: A partitioned survival model was developed to estimate expected life-years (LYs), quality-adjusted LYs (QALYs), direct costs and incremental costs per QALY and LY gained associated with use of Rd vs VMP over a patient's lifetime. Information on the efficacy and safety of Rd and VMP was based on data from multinational phase III clinical trials and a network meta-analysis. Pre-progression direct costs included the costs of Rd and VMP, treatment of adverse events (including prophylaxis) and routine care and monitoring associated with MM. Post-progression direct costs included costs of subsequent treatment(s) and routine care and monitoring for progressive disease, all obtained from published literature and estimated from a U.S. payer perspective. Utilities were obtained from the aforementioned trials. Costs and outcomes were discounted at 3% annually. RESULTS: Relative to VMP, use of Rd was expected to result in an additional 2.22 LYs and 1.47 QALYs (discounted). Patients initiated with Rd were expected to incur an additional $78,977 in mean lifetime direct costs (discounted) vs those initiated with VMP. The incremental costs per QALY and per LY gained with Rd vs VMP were $53,826 and $35,552, respectively. In sensitivity analyses, results were found to be most sensitive to differences in survival associated with Rd vs VMP, the cost of lenalidomide and the discount rate applied to effectiveness outcomes. CONCLUSIONS: Rd was expected to result in greater LYs and QALYs compared with VMP, with similar overall costs per LY for each regimen. Results of this analysis indicated that Rd may be a cost-effective alternative to VMP as initial treatment for transplant-ineligible patients with MM, with an incremental cost-effectiveness ratio well within the levels for recent advancements in oncology.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/economia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Bortezomib/administração & dosagem , Dexametasona/administração & dosagem , Melfalan/administração & dosagem , Mieloma Múltiplo/tratamento farmacológico , Prednisona/administração & dosagem , Talidomida/análogos & derivados , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Análise Custo-Benefício , Feminino , Humanos , Lenalidomida , Masculino , Cadeias de Markov , Anos de Vida Ajustados por Qualidade de Vida , Talidomida/administração & dosagem , Resultado do Tratamento , Estados Unidos
16.
Acta Biomater ; 31: 122-133, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26687978

RESUMO

The study describes the design and synthesis of an implantable smart magnetic nanofiber device for endoscopic hyperthermia treatment and tumor-triggered controlled drug release. This device is achieved using a two-component smart nanofiber matrix from monodisperse iron oxide nanoparticles (IONPs) as well as bortezomib (BTZ), a chemotherapeutic drug. The IONP-incorporated nanofiber matrix was developed by electrospinning a biocompatible and bioresorbable polymer, poly (d,l-lactide-co-glycolide) (PLGA), and tumor-triggered anticancer drug delivery is realized by exploiting mussel-inspired surface functionalization using 2-(3,4-dihydroxyphenyl)ethylamine (dopamine) to conjugate the borate-containing BTZ anticancer drug through a catechol metal binding in a pH-sensitive manner. Thus, an implantable smart magnetic nanofiber device can be exploited to both apply hyperthermia with an alternating magnetic field (AMF) and to achieve cancer cell-specific drug release to enable synergistic cancer therapy. These results confirm that the BTZ-loaded mussel-inspired magnetic nanofiber matrix (BTZ-MMNF) is highly beneficial not only due to the higher therapeutic efficacy and low toxicity towards normal cells but also, as a result of the availability of magnetic nanoparticles for repeated hyperthermia application and tumor-triggered controlled drug release. STATEMENT OF SIGNIFICANCE: The current work report on the design and development of a smart nanoplatform responsive to a magnetic field to administer both hyperthermia and pH-dependent anticancer drug release for the synergistic anticancer treatment. The iron oxide nanoparticles (IONPs) incorporated nanofiber matrix was developed by electrospinning a biocompatible polymer, poly (d,l-lactide-co-glycolide) (PLGA), and tumor-triggered anticancer drug delivery is realized by surface functionalization using 2-(3,4-dihydroxyphenyl)ethylamine (dopamine) to conjugate the boratecontaining anticancer drug bortezomib through a catechol metal binding in a pH-sensitive manner. This implantable magnetic nanofiber device can be exploited to apply hyperthermia with an alternating magnetic field and to achieve cancer cell-specific drug release to enable synergistic cancer therapy, which results in an improvement in both quality of life and patient compliance.


Assuntos
Sistemas de Liberação de Medicamentos , Endoscopia/métodos , Hipertermia Induzida/métodos , Nanofibras/química , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Materiais Biocompatíveis/química , Bivalves , Bortezomib/administração & dosagem , Bortezomib/química , Catecóis/química , Linhagem Celular Tumoral , Dopamina/química , Liberação Controlada de Fármacos , Endoscópios , Compostos Férricos/química , Concentração de Íons de Hidrogênio , Ácido Láctico/química , Magnetismo , Nanopartículas de Magnetita/química , Camundongos , Células NIH 3T3 , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Termogravimetria
17.
Clin Exp Med ; 16(1): 99-101, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25600700

RESUMO

We retrospectively analyzed the medical history of 19 elderly myeloma patients treated with the "novel subcutaneous formulation of bortezomib." In our experience, two patients (10 %) discontinued treatment for paralytic ileus. The exact pathogenetic mechanisms of toxic megacolon and paralytic ileus due to "novel subcutaneous formulation of bortezomib" are unclear. Probably, it may be related to possible damage of the autonomic nerve fibers that control organ functions. Adequate prevention and management of the gastrointestinal (GI) toxicities with the use of fluid intake and prokinetic and laxative drugs (at least two types of agents in a suboptimal dose) especially in patients with risk factors for GI side effects (anti-myeloma novel agents, opioids or antiemetics, iron supplements, spinal and cord compression, immobility, history of constipation) can decrease the possibility of interruption of administration of drug and increase adherence to treatment. Clearly this complication must be borne in mind whenever a patient develops acute abdominal pain and distension.


Assuntos
Antineoplásicos/administração & dosagem , Bortezomib/administração & dosagem , Pseudo-Obstrução Intestinal/induzido quimicamente , Mieloma Múltiplo/tratamento farmacológico , Idoso , Antineoplásicos/efeitos adversos , Bortezomib/efeitos adversos , Gerenciamento Clínico , Feminino , Humanos , Injeções Subcutâneas , Pseudo-Obstrução Intestinal/patologia , Pseudo-Obstrução Intestinal/terapia , Mieloma Múltiplo/patologia
18.
Oncologist ; 20(4): 370-1, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25777345

RESUMO

BACKGROUND: Sorafenib is an orally active multikinase inhibitor, and bortezomib is a proteasome inhibitor that affects multiple signaling pathways. Sorafenib has clinical activity in renal cell carcinoma (RCC), whereas bortezomib has demonstrated activity against RCC cell lines in vitro, with in vitro studies showing synergism between the two agents in the induction of apoptosis in neoplastic cell lines. In this phase II study, we explored the efficacy and toxicity of this regimen. METHODS: Adult patients with cytologically confirmed clear cell RCC with no prior chemotherapy, Zubrod performance status of 0-1, serum creatinine <1.5 mg/dL, and normal liver function tests were treated with sorafenib 200 mg orally b.i.d. with bortezomib 1 mg/m(2) intravenously on days 1, 4, 8, and 11 every 21 days. Treatment was continued until disease progression or unacceptable toxicity. The primary objective was median progression-free survival (PFS) of at least 70 weeks. RESULTS: Seventeen patients were enrolled between April 2011 and January 2013. Median age was 62 years (range: 44-75 years). Four of 17 patients had known brain metastasis on enrollment. Median number of cycles was 4 (range: 1 to ≥45). No patient had complete response, 1 had partial response, 12 had stable disease, and 4 had progressive disease (response rate of 6%; 95% confidence interval: 0%-29%) with treatment. Median PFS was 13.7 weeks, and median overall survival was 110 weeks. The study was halted for futility. CONCLUSION: The combination of sorafenib and bortezomib was well tolerated; however, response rate and PFS were comparable to sorafenib monotherapy. This regimen is not recommended for further development.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Renais/tratamento farmacológico , Neoplasias Renais/tratamento farmacológico , Adulto , Idoso , Bortezomib/administração & dosagem , Carcinoma de Células Renais/mortalidade , Carcinoma de Células Renais/patologia , Carcinoma de Células Renais/cirurgia , Intervalo Livre de Doença , Humanos , Estimativa de Kaplan-Meier , Neoplasias Renais/mortalidade , Neoplasias Renais/patologia , Neoplasias Renais/cirurgia , Pessoa de Meia-Idade , Niacinamida/administração & dosagem , Niacinamida/análogos & derivados , Compostos de Fenilureia/administração & dosagem , Sorafenibe , Resultado do Tratamento
19.
Surg Today ; 45(12): 1481-8, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25673034

RESUMO

Biliary cancer and pancreatic cancer are considered to be difficult diseases to cure. Although complete resection provides the only means of curing these cancers, the rate of resectability is not high. Therefore, chemotherapy is often selected in patients with advanced unresectable biliary-pancreatic cancer. Many combination chemotherapy regimens have been applied in clinical trials. However, the survival time is not satisfactory. On the other hand, most chemotherapeutic agents induce anti-apoptotic transcriptional factor nuclear factor kappa b (NF-κB) activation, and agent-induced NF-κB activation is deeply involved in the onset of chemoresistance. Recently, novel approaches to potentiating chemosensitivity in cases of biliary-pancreatic cancer using NF-κB inhibitors with cytotoxic agents have been reported, most of which comprise translational research, although some clinical trials have also been conducted. Nevertheless, to date, there is no breakthrough chemotherapy regimen for these diseases. As some reports show promising data, combination chemotherapy consisting of a NF-κB inhibitor with chemotherapeutic agents seems to improve chemosensitivity and prolong the survival time of biliary-pancreatic cancer patients.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias do Sistema Biliar/tratamento farmacológico , Neoplasias do Sistema Biliar/genética , Terapia de Alvo Molecular , NF-kappa B/antagonistas & inibidores , NF-kappa B/fisiologia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Benzamidinas , Bortezomib/administração & dosagem , Bortezomib/uso terapêutico , Camptotecina/administração & dosagem , Camptotecina/análogos & derivados , Curcumina/uso terapêutico , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Ditiocarb/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Guanidinas/administração & dosagem , Humanos , Irinotecano , Paclitaxel/administração & dosagem , Pregnenodionas/administração & dosagem , Resultado do Tratamento , Gencitabina
20.
Clin Cancer Res ; 21(12): 2802-10, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25398450

RESUMO

PURPOSE: Novel therapeutic agents have significantly improved the survival of patients with multiple myeloma. Nonetheless, the prognosis of patients with multiple myeloma who become refractory to the novel agents lenalidomide and bortezomib is very poor, indicating the urgent need for new therapeutic options for these patients. The human CD38 monoclonal antibody daratumumab is being evaluated as a novel therapy for multiple myeloma. Prompted with the encouraging results of ongoing clinical phase I/II trials, we now addressed the potential value of daratumumab alone or in combination with lenalidomide or bortezomib for the treatment of lenalidomide- and bortezomib-refractory patients. EXPERIMENTAL DESIGN: In ex vivo assays, mainly evaluating antibody-dependent cell-mediated cytotoxicity, and in an in vivo xenograft mouse model, we evaluated daratumumab alone or in combination with lenalidomide or bortezomib as a potential therapy for lenalidomide- and bortezomib-refractory multiple myeloma patients. RESULTS: Daratumumab induced significant lysis of lenalidomide/bortezomib-resistant multiple myeloma cell lines and of primary multiple myeloma cells in the bone marrow mononuclear cells derived from lenalidomide- and/or bortezomib-refractory patients. In these assays, lenalidomide but not bortezomib, synergistically enhanced daratumumab-mediated multiple myeloma lysis through activation of natural killer cells. Finally, in an in vivo xenograft model, only the combination of daratumumab with lenalidomide effectively reduced the tumorigenic growth of primary multiple myeloma cells from a lenalidomide- and bortezomib-refractory patient. CONCLUSIONS: Our results provide the first preclinical evidence for the benefit of daratumumab plus lenalidomide combination for lenalidomide- and bortezomib-refractory patients.


Assuntos
ADP-Ribosil Ciclase 1/antagonistas & inibidores , ADP-Ribosil Ciclase 1/metabolismo , Bortezomib/farmacologia , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/metabolismo , Talidomida/análogos & derivados , Adulto , Idoso , Animais , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Bortezomib/administração & dosagem , Linhagem Celular Tumoral , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Feminino , Humanos , Imunoterapia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Lenalidomida , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Masculino , Camundongos , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/terapia , Talidomida/administração & dosagem , Talidomida/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA