Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Annu Rev Med ; 75: 13-29, 2024 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-37729027

RESUMO

Multiple myeloma is a cancer of bone marrow plasma cells that represents approximately 10% of hematologic malignancies. Though it is typically incurable, a remarkable suite of new therapies developed over the last 25 years has enabled durable disease control in most patients. This article briefly introduces the clinical features of multiple myeloma and aspects of multiple myeloma biology that modern therapies exploit. Key current and emerging treatment modalities are then reviewed, including cereblon-modulating agents, proteasome inhibitors, monoclonal antibodies, other molecularly targeted therapies (selinexor, venetoclax), chimeric antigen receptor T cells, T cell-engaging bispecific antibodies, and antibody-drug conjugates. For each modality, mechanism of action and clinical considerations are discussed. These therapies are combined and sequenced in modern treatment pathways, discussed at the conclusion of the article, which have led to substantial improvements in outcomes for multiple myeloma patients in recent years.


Assuntos
Mieloma Múltiplo , Humanos , Mieloma Múltiplo/tratamento farmacológico , Imunoterapia , Inibidores de Proteassoma/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Terapia Biológica
2.
Integr Cancer Ther ; 22: 15347354231159322, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36879488

RESUMO

Proteasome inhibitors are the cornerstone of multiple myeloma treatment, but challenges still remain despite the increased survival rates. We conducted a review on the role of curcumin, a natural product, as an adjunct to bortezomib and carfilzomib in preclinical multiple myeloma models. Four studies reviewed showed enhanced anticancer effects when curcumin was combined with bortezomib compared to either treatment alone. Two additional studies showed similar results with carfilzomib. Synergistic mechanisms include inhibition of NF-kB, IL-6-induced signaling pathways, JNK pathway modulation, and increased cell cycle arrest.


Assuntos
Produtos Biológicos , Curcumina , Mieloma Múltiplo , Humanos , Inibidores de Proteassoma/farmacologia , Inibidores de Proteassoma/uso terapêutico , Bortezomib/farmacologia , Bortezomib/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Curcumina/farmacologia , Curcumina/uso terapêutico
3.
Contrast Media Mol Imaging ; 2022: 6056829, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36134116

RESUMO

In order to investigate the effects of different doses of Dahuang Zhechong pills on the ubiquitin proteasome pathway/nuclear factor-κB (UPP-NF-κB) in rats with atherosclerosis (AS), 58-week-old male Wistar rats were selected and randomly divided into the normal group, model group, control group, low-dose group, and high-dose group. The model group and the drug group are given intraperitoneal injections of vitamins, and the model group and the drug group are given a high-fat diet. Rats in the low-dose group and high-dose group are given low-dose and high-dose Dahuang Zhechong pill lavage solution, respectively. Besides, the control group is given simvastatin solution by gavage, and intervention is performed once a day for 12 weeks. Ubiquitin (Ub) protein expression, ubiquitin activase (UBE1), nuclear factor-κB, nuclear inhibitory factor-κB (IκB) gene expression, total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), and serum tumor necrosis factor-α (TNF-α) are compared. The experimental result shows that Dahuang Zhechong pills can reduce inflammation and prevent and treat AS by blocking the activation of the UPP/NF-κB signaling pathway and can be used as a proteasome inhibitor in the clinical treatment of AS.


Assuntos
Aterosclerose , NF-kappa B , Animais , Aterosclerose/tratamento farmacológico , Aterosclerose/metabolismo , LDL-Colesterol/uso terapêutico , Medicamentos de Ervas Chinesas , Masculino , NF-kappa B/metabolismo , NF-kappa B/uso terapêutico , Complexo de Endopeptidases do Proteassoma/uso terapêutico , Inibidores de Proteassoma/uso terapêutico , Ratos , Ratos Wistar , Sinvastatina/uso terapêutico , Ativador de Plasminogênio Tecidual/uso terapêutico , Triglicerídeos , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/uso terapêutico , Ubiquitinas/uso terapêutico , Vitaminas/uso terapêutico
4.
Curr Cancer Drug Targets ; 22(9): 741-748, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35578889

RESUMO

Approval of the first boronic acid group-containing drug, bortezomib, in 2003 for the treatment of multiple myeloma sparked an increased interest of medicinal chemists in boronic acidbased therapeutics. As a result, another boronic acid moiety-harboring medication, ixazomib, was approved in 2015 as a second-generation proteasome inhibitor for multiple myeloma; and dutogliptin is under clinical investigation in combination therapy against myocardial infarction. Moreover, a large number of novel agents with boronic acid elements in their structure are currently in intensive preclinical studies, allowing us to suppose that at least some of them will enter clinical trials in the near future. On the other hand, only some years after bortezomib approval, direct interactions between its boronic acid group and catechol moiety of green tea catechins as well as some other common dietary flavonoids like quercetin and myricetin were discovered, leading to the formation of stable cyclic boronate esters and abolishing the anticancer activities. Although highly relevant, to date, no reports on possible co-effects of catechol group-containing flavonoids with new-generation boronic acidbased drugs can be found. However, this issue cannot be ignored, especially considering the abundance of catechol moiety-harboring flavonoids in both plant-derived food items as well as over-thecounter dietary supplements and herbal products. Therefore, in parallel with the intensified development of boronic acid-based drugs, their possible interactions with catechol groups of plant-derived flavonoids must also be clarified to provide dietary recommendations to patients for maximizing therapeutic benefits. If concurrently consumed flavonoids can indeed antagonize drug efficacy, it may pose a real risk to clinical outcomes.


Assuntos
Antineoplásicos , Mieloma Múltiplo , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Ácidos Borônicos/química , Ácidos Borônicos/farmacologia , Ácidos Borônicos/uso terapêutico , Bortezomib/farmacologia , Bortezomib/uso terapêutico , Catecóis/uso terapêutico , Flavonoides/farmacologia , Flavonoides/uso terapêutico , Humanos , Mieloma Múltiplo/tratamento farmacológico , Complexo de Endopeptidases do Proteassoma , Inibidores de Proteassoma/farmacologia , Inibidores de Proteassoma/uso terapêutico , Pirazinas/farmacologia , Pirazinas/uso terapêutico
5.
Cell Death Dis ; 13(3): 203, 2022 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-35246527

RESUMO

Despite several approved therapeutic modalities, multiple myeloma (MM) remains an incurable blood malignancy and only a small fraction of patients achieves prolonged disease control. The common anti-MM treatment targets proteasome with specific inhibitors (PI). The resulting interference with protein degradation is particularly toxic to MM cells as they typically accumulate large amounts of toxic proteins. However, MM cells often acquire resistance to PIs through aberrant expression or mutations of proteasome subunits such as PSMB5, resulting in disease recurrence and further treatment failure. Here we propose CuET-a proteasome-like inhibitor agent that is spontaneously formed in-vivo and in-vitro from the approved alcohol-abuse drug disulfiram (DSF), as a readily available treatment effective against diverse resistant forms of MM. We show that CuET efficiently kills also resistant MM cells adapted to proliferate under exposure to common anti-myeloma drugs such as bortezomib and carfilzomib used as the first-line therapy, as well as to other experimental drugs targeting protein degradation upstream of the proteasome. Furthermore, CuET can overcome also the adaptation mechanism based on reduced proteasome load, another clinically relevant form of treatment resistance. Data obtained from experimental treatment-resistant cellular models of human MM are further corroborated using rather unique advanced cytotoxicity experiments on myeloma and normal blood cells obtained from fresh patient biopsies including newly diagnosed as well as relapsed and treatment-resistant MM. Overall our findings suggest that disulfiram repurposing particularly if combined with copper supplementation may offer a promising and readily available treatment option for patients suffering from relapsed and/or therapy-resistant multiple myeloma.


Assuntos
Antineoplásicos , Mieloma Múltiplo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Bortezomib/farmacologia , Bortezomib/uso terapêutico , Linhagem Celular Tumoral , Dissulfiram/farmacologia , Reposicionamento de Medicamentos , Resistencia a Medicamentos Antineoplásicos , Humanos , Mieloma Múltiplo/patologia , Recidiva Local de Neoplasia/tratamento farmacológico , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Inibidores de Proteassoma/uso terapêutico
6.
Acta Pharmacol Sin ; 43(8): 2128-2138, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34893683

RESUMO

The ubiquitin-proteasome system (UPS) is essential for maintaining cell homeostasis by orchestrating the protein degradation, but is impaired in various diseases, including cancers. Several proteasome inhibitors, such as bortezomib, are currently used in cancer treatment, but associated toxicity limits their widespread application. Recently metal complex-based drugs have attracted great attention in tumor therapy; however, their application is hindered by low water-solubility and poor absorbency. Herein, we synthesized a new type of gold (I) complex named Na-AuPT, and further characterized its anticancer activity. Na-AuPT is highly water-soluble (6 mg/mL), and it was able to potently inhibit growth of a panel of 11 cancer cell lines (A549, SMMC7721, H460, HepG2, BEL7402, LNCap, PC3, MGC-803, SGC-7901, U266, and K562). In A549 and SMMC7721 cells, Na-AuPT (in a range of 2.5-20 µM) inhibited the UPS function in a dose-dependent fashion by targeting and inhibiting both 20 S proteasomal proteolytic peptidases and 19 S proteasomal deubiquitinases. Furthermore, Na-AuPT induced caspase-dependent apoptosis in A549 and SMMC7721 cells, which was prevented by the metal chelator EDTA. Administration of Na-AuPT (40 mg · kg-1 · d-1, ip) in nude mice bearing A549 or SMMC7721 xenografts significantly inhibited the tumor growth in vivo, accompanied by increased levels of total ubiquitinated proteins, cleaved caspase 3 and Bax protein in tumor tissue. Moreover, Na-AuPT induced cell death of primary mononuclear cells from 5 patients with acute myeloid leukemia ex vivo with an average IC50 value of 2.46 µM. We conclude that Na-AuPT is a novel metal-based proteasome inhibitor that may hold great potential for cancer therapy.


Assuntos
Antineoplásicos , Neoplasias , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Nus , Neoplasias/tratamento farmacológico , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Inibidores de Proteassoma/uso terapêutico , Ubiquitina/metabolismo , Água
7.
Hum Exp Toxicol ; 40(11): 1985-1997, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34002651

RESUMO

MG132 is a potent, reversible, and cell-permeable 20S proteasome inhibitor and it is derived from a Chinese medicinal plant. The purpose of this study is to investigate the anticancer effects of MG132 against human osteosarcoma U2OS cells. We first performed MTT and colony formation assays to investigate the anti-proliferative effects of MG132. The results demonstrated that MG132 suppressed the proliferation of U2OS cells. Furthermore, we found that treatment with MG132 increased apoptosis and induced DNA damage in U2OS cells. Additionally, zymography, wound healing, and invasion assays showed that MG132 suppressed the enzymatic activity of matrix metalloproteinases, cell migration, and invasion, respectively of U2OS cells. Furthermore, western blotting assay was performed to investigate the apoptotic signaling pathways in MG132-treated U2OS cells. Our results showed that MG132 downregulated the expression of antiapoptotic proteins, including CDK2, CDK4, Bcl-xL, and Bcl-2, whereas it upregulated the expression of proapoptotic proteins, including p21, p27, p53, p-p53 (ser15, ser20, and ser46), cleaved forms of caspase-3, caspase-7, caspase-9, and PARP, and FOXO3 in U2OS cells. These results demonstrated that MG132 activated apoptotic signaling pathways in U2OS cells. Interestingly, MG132 downregulated the phosphorylation of Akt and Erk. Taken together, our results suggest that MG132 has anticancer effects in U2OS cells. Therefore, MG132 may be a potential therapeutic agent for the treatment of osteosarcoma.


Assuntos
Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Neoplasias Ósseas/tratamento farmacológico , Linhagem Celular Tumoral/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Osteossarcoma/tratamento farmacológico , Inibidores de Proteassoma/uso terapêutico , Neoplasias Ósseas/fisiopatologia , Humanos , Medicina Tradicional Chinesa , Osteossarcoma/fisiopatologia
8.
Biomed Res Int ; 2021: 6687589, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33855081

RESUMO

The aim of this work is to evaluate the antitumor effect mediated by the proteasome inhibitors of Inula viscosa extracts on skin carcinogenesis. Female Swiss albino mice were divided into five groups depending on the combination of skin cancer-inducing 7,12-dimethylbenz(a)anthracene (DMBA) and extract of Inula viscosa treatments. Histology of the affected skin and measurement of proteasome activity were performed to demonstrate the effect of Inula viscosa on mice. The identification of the molecules responsible for this inhibitory activity was carried out through the docking studies. The results showed that Inula viscosa extracts inhibit the development of papilloma in mice. Therefore, the best chemopreventive action of Inula viscosa was observed on mice in which extract treatment was performed before and after the induction of skin carcinogenesis. It was revealed that the ingestion of extracts Inula viscosa delays the formation of skin papillomas in animals and simultaneously decreases the size and number of papillomas, which is also reflected on the skin histology of the mice treated. Structure-activity relationship information obtained from component of Inula viscosa particularly tomentosin, inuviscolide, and isocosticacid demonstrated that distinct bonding modes in ß 1, ß 2, and ß 5 subunits determine its selectivity and potent inhibition for ß 5 subunit.


Assuntos
Antineoplásicos/uso terapêutico , Inula/química , Extratos Vegetais/uso terapêutico , Complexo de Endopeptidases do Proteassoma/metabolismo , Neoplasias Cutâneas/tratamento farmacológico , 9,10-Dimetil-1,2-benzantraceno , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Células CACO-2 , Quimotripsina/metabolismo , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Camundongos , Simulação de Acoplamento Molecular , Papiloma/tratamento farmacológico , Papiloma/patologia , Extratos Vegetais/análise , Extratos Vegetais/farmacocinética , Extratos Vegetais/farmacologia , Inibidores de Proteassoma/farmacologia , Inibidores de Proteassoma/uso terapêutico , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/patologia , Testes de Toxicidade
9.
J Med Chem ; 64(9): 5905-5930, 2021 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-33904304

RESUMO

There is an urgent need for new treatments for visceral leishmaniasis (VL), a parasitic infection which impacts heavily large areas of East Africa, Asia, and South America. We previously reported on the discovery of GSK3494245/DDD01305143 (1) as a preclinical candidate for VL and, herein, we report on the medicinal chemistry program that led to its identification. A hit from a phenotypic screen was optimized to give a compound with in vivo efficacy, which was hampered by poor solubility and genotoxicity. The work on the original scaffold failed to lead to developable compounds, so an extensive scaffold-hopping exercise involving medicinal chemistry design, in silico profiling, and subsequent synthesis was utilized, leading to the preclinical candidate. The compound was shown to act via proteasome inhibition, and we report on the modeling of different scaffolds into a cryo-EM structure and the impact this has on our understanding of the series' structure-activity relationships.


Assuntos
Desenho de Fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/química , Proteínas de Protozoários/metabolismo , Animais , Antiprotozoários/química , Antiprotozoários/metabolismo , Antiprotozoários/farmacologia , Antiprotozoários/uso terapêutico , Sítios de Ligação , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Meia-Vida , Humanos , Leishmania donovani/efeitos dos fármacos , Leishmania donovani/metabolismo , Leishmaniose Visceral/tratamento farmacológico , Leishmaniose Visceral/parasitologia , Camundongos , Simulação de Dinâmica Molecular , Complexo de Endopeptidases do Proteassoma/química , Inibidores de Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Inibidores de Proteassoma/uso terapêutico , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Proteínas de Protozoários/química , Piridinas/química , Piridinas/metabolismo , Piridinas/farmacologia , Piridinas/uso terapêutico , Solubilidade , Relação Estrutura-Atividade
10.
Arthritis Rheumatol ; 71(2): 244-257, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30144298

RESUMO

OBJECTIVE: To investigate the roles of the synovial lymphatic system in the severity and progression of joint tissue damage and functional responses of synovial lymphatic endothelial cells (LECs) to macrophage subsets, and to evaluate the therapeutic potential of the proteasome inhibitor bortezomib (BTZ) in a mouse model of experimental posttraumatic osteoarthritis (OA). METHODS: C57BL/6J wild-type mice received a meniscal ligamentous injury to induce posttraumatic knee OA. Lymphangiogenesis was blocked by a vascular endothelial growth factor receptor 3 (VEGFR-3) neutralizing antibody. Synovial lymphatic drainage was examined by near-infrared imaging. Joint damage was assessed by histology. RNA-sequencing and pathway analyses were applied to synovial LECs. Macrophage subsets in the mouse synovium were identified by flow cytometry and immunofluorescence staining. M1 and M2 macrophages were induced from mouse bone marrow cells, and their effects on LECs were examined in cocultures in the presence or absence of BTZ. The effects of BTZ on joint damage, LEC inflammation, and synovial lymphatic drainage were examined. RESULTS: Injection of a VEGFR-3 neutralizing antibody into the joints of mice with posttraumatic knee OA reduced synovial lymphatic drainage and accelerated joint tissue damage. Synovial LECs from the mouse OA joints had dysregulated inflammatory pathways and expressed high levels of inflammatory genes. The number of M1 macrophages was increased in the knee joints of mice with posttraumatic OA, thereby promoting the expression of inflammatory genes by LECs; this effect was blocked by BTZ. Treatment with BTZ decreased cartilage loss, reduced the expression of inflammatory genes by LECs, and improved lymphatic drainage in the knee joints of mice with posttraumatic OA. CONCLUSION: Experimental posttraumatic knee OA is associated with decreased synovial lymphatic drainage, increased numbers of M1 macrophages, and enhanced inflammatory gene expression by LECs, all of which was improved by treatment with BTZ. Intraarticular administration of BTZ may represent a new therapy for the restoration of synovial lymphatic function in subjects with posttraumatic knee OA.


Assuntos
Bortezomib/farmacologia , Células Endoteliais/efeitos dos fármacos , Vasos Linfáticos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Osteoartrite do Joelho/imunologia , Inibidores de Proteassoma/farmacologia , Membrana Sinovial/efeitos dos fármacos , Animais , Anticorpos Neutralizantes/farmacologia , Bortezomib/uso terapêutico , Técnicas de Cocultura , Progressão da Doença , Inflamação , Traumatismos do Joelho/complicações , Linfangiogênese/efeitos dos fármacos , Vasos Linfáticos/imunologia , Macrófagos/imunologia , Camundongos , Osteoartrite do Joelho/tratamento farmacológico , Osteoartrite do Joelho/etiologia , Inibidores de Proteassoma/uso terapêutico , Espectroscopia de Luz Próxima ao Infravermelho , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/imunologia
11.
Semin Nephrol ; 38(5): 531-540, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30177025

RESUMO

Fifty years into the original description of IgA nephropathy, there is still no specific therapy for this condition and general measures including blood pressure control with blockers of the renin-angiotensin-aldosterone system and salt restriction remain the cornerstone to slow disease progression. Although the paucity in treatment advances could be related to the disease's complex pathogenesis, which requires multiple hits, heterogeneity as reflected by diverse ethnic differences, and genetic predisposition and histopathologic variations, many nonspecific and immunomodulatory agents have been tested with variable degrees of success and tribulations. Here, we review the evolution of these different therapeutic approaches over time that culminated in the 2012 Kidney Disease: Improving Global Outcomes Clinical Practice Guideline for Glomerulonephritis that presently is being updated, and provide an appraisal of recent data on various forms of immunosuppressive agents. Finally, we discuss the theoretical basis of ongoing and upcoming clinical trials that are more pathway- or cell-type-specific as knowledge in disease mechanisms advances.


Assuntos
Antagonistas de Receptores de Angiotensina/uso terapêutico , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Óleos de Peixe/uso terapêutico , Glomerulonefrite por IGA/terapia , Imunossupressores/uso terapêutico , Inibidores de Proteassoma/uso terapêutico , Corticosteroides/uso terapêutico , Azatioprina/uso terapêutico , Ciclofosfamida/uso terapêutico , Glomerulonefrite por IGA/história , História do Século XX , História do Século XXI , Humanos , Ácido Micofenólico/uso terapêutico , Tonsilectomia/história
12.
Clin Microbiol Infect ; 24 Suppl 2: S95-S107, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29427804

RESUMO

BACKGROUND: The present review is part of the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Infections in Compromised Hosts (ESGICH) consensus document on the safety of targeted and biological therapies. AIMS: To review, from an infectious diseases perspective, the safety profile of immune checkpoint inhibitors, LFA-3-targeted agents, cell adhesion inhibitors, sphingosine-1-phosphate receptor modulators and proteasome inhibitors, and to suggest preventive recommendations. SOURCES: Computer-based Medline searches with MeSH terms pertaining to each agent or therapeutic family. CONTENT: T-lymphocyte-associated antigen 4 (CTLA-4) and programmed death (PD)-1/PD-1 ligand 1 (PD-L1)-targeted agents do not appear to intrinsically increase the risk of infection but can induce immune-related adverse effects requiring additional immunosuppression. Although CD4+ T-cell lymphopenia is associated with alefacept, no opportunistic infections have been observed. Progressive multifocal leukoencephalopathy (PML) may occur during therapy with natalizumab (anti-α4-integrin monoclonal antibody (mAb)) and efalizumab (anti-CD11a mAb), but no cases have been reported to date with vedolizumab (anti-α4ß7 mAb). In patients at high risk for PML (positive anti-JC polyomavirus serology with serum antibody index >1.5 and duration of therapy ≥48 months), the benefit-risk ratio of continuing natalizumab should be carefully considered. Fingolimod induces profound peripheral blood lymphopenia and increases the risk of varicella zoster virus (VZV) infection. Prophylaxis with (val)acyclovir and VZV vaccination should be considered. Proteasome inhibitors also increase the risk of VZV infection, and antiviral prophylaxis with (val)acyclovir is recommended. Anti-Pneumocystis prophylaxis may be considered in myeloma multiple patients with additional risk factors (i.e. high-dose corticosteroids). IMPLICATIONS: Clinicians should be aware of the risk of immune-related adverse effects and PML in patients receiving immune checkpoint and cell adhesion inhibitors respectively.


Assuntos
Terapia Biológica/efeitos adversos , Adesão Celular/efeitos dos fármacos , Doenças Transmissíveis/terapia , Genes cdc/efeitos dos fármacos , Terapia de Alvo Molecular/efeitos adversos , Inibidores de Proteassoma/efeitos adversos , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/uso terapêutico , Terapia Biológica/métodos , Antígeno CTLA-4/antagonistas & inibidores , Ensaios Clínicos como Assunto , Consenso , Humanos , Hospedeiro Imunocomprometido , Leucoencefalopatia Multifocal Progressiva/terapia , Terapia de Alvo Molecular/métodos , Natalizumab/efeitos adversos , Natalizumab/uso terapêutico , Inibidores de Proteassoma/uso terapêutico , Receptores de Lisoesfingolipídeo/efeitos dos fármacos
13.
Biol Blood Marrow Transplant ; 23(2): 255-261, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27888016

RESUMO

Lapses in the prevention of graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (HSCT) warrant novel approaches. Such approaches include, among others, the use of post-transplantation cyclophosphamide (PTC) and proteasome inhibitors. Although PTC alone consistently produces low rates of chronic GVHD, the incidence of acute GVHD remains significant. Inversely, prolonged post-transplantation administration of proteasome inhibitors carries a risk of paradoxical aggravation of GVHD. We examined whether the combination of cyclophosphamide and ixazomib addresses the limitations of each of these agents when used alone to prevent GVHD in mice subjected to allogeneic HSCT across MHC barriers. We chose ixazomib, an orally bioavailable proteasome inhibitor, because of its favorable physiochemical characteristics. The combination of cyclophosphamide and ixazomib improved overall survival of mice in comparison to an untreated control group and to groups receiving either cyclophosphamide alone or ixazomib alone. Furthermore, cyclophosphamide prevented the surge of IL-1ß, GVHD aggravation, and sudden death associated with prolonged administration of ixazomib after HSCT. Finally, we demonstrated that although ixazomib was administered before cyclophosphamide, it did not impair the preferential depletion of proliferating as opposed to resting donor T cells. Our data suggest that the combination of cyclophosphamide and ixazomib for the prevention of GVHD after allogeneic HSCT is promising and merits further investigation in clinical trials.


Assuntos
Compostos de Boro/uso terapêutico , Ciclofosfamida/uso terapêutico , Glicina/análogos & derivados , Doença Enxerto-Hospedeiro/prevenção & controle , Inibidores de Proteassoma/uso terapêutico , Animais , Transplante de Medula Óssea/efeitos adversos , Compostos de Boro/administração & dosagem , Compostos de Boro/farmacologia , Ciclofosfamida/administração & dosagem , Ciclofosfamida/farmacologia , Citocinas/sangue , Esquema de Medicação , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Glicina/administração & dosagem , Glicina/farmacologia , Glicina/uso terapêutico , Doença Enxerto-Hospedeiro/tratamento farmacológico , Doença Enxerto-Hospedeiro/patologia , Intestino Delgado/patologia , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibidores de Proteassoma/administração & dosagem , Inibidores de Proteassoma/farmacologia , Quimera por Radiação , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/transplante
14.
Crit Rev Oncog ; 22(1-2): 37-47, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29604935

RESUMO

Various targeted therapies for cancer have resulted in a significant prolongation of survival and a better quality of life. However, unfortunately, a small subset of cancer patients responds to such therapies initially and then develops resistance after the initial therapies. Based on resistant mechanisms, it should be possible to develop new and specific targeted therapies effective against unresponsive patients. Our investigations and those of others have identified a gene product, Yin Yang 1 (YY1), a transcription factor that is overexpressed in many cancers and that was shown to be involved in the regulation of cell survival, cell proliferation, cell invasion, metastasis, and resistance. Several studies showed that the inhibition of YY1 resulted in significant inhibition of the tumor phenotype and reversal of resistance. Examples of such YY1 inhibitors include siRNA YY1, nitric oxide donors, proteasome inhibitors, and inhibitors of activated survival pathways such as inhibitors of nuclear factor-kappa beta. However, there is still a need to develop specific and targeted inhibitors of YY1. In this review, a general discussion is provided on the role of YY1 overexpression in cancer and the application of various inhibitors of YY1 activities and their potential as therapeutics.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias/tratamento farmacológico , Fator de Transcrição YY1/antagonistas & inibidores , Apoptose/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Humanos , NF-kappa B/genética , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Metástase Neoplásica , Neoplasias/genética , Neoplasias/patologia , Doadores de Óxido Nítrico/uso terapêutico , Inibidores de Proteassoma/uso terapêutico , RNA Interferente Pequeno/uso terapêutico , Fator de Transcrição YY1/genética
15.
Appl Physiol Nutr Metab ; 41(12): 1240-1247, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27841025

RESUMO

l-Carnitine was recently found to downregulate the ubiquitin proteasome pathway (UPP) and increase insulin-like growth factor 1 concentrations in animal models. However, the effect of l-carnitine administration on disuse muscle atrophy induced by hindlimb suspension has not yet been studied. Thus, we hypothesized that l-carnitine may have a protective effect on muscle atrophy induced by hindlimb suspension via the Akt1/mTOR and/or UPP. Male Wistar rats were assigned to 3 groups: hindlimb suspension group, hindlimb suspension with l-carnitine administration (1250 mg·kg-1·day-1) group, and pair-fed group adjusted hindlimb suspension. l-Carnitine administration for 2 weeks of hindlimb suspension alleviated the decrease in weight and fiber size in the soleus muscle. In addition, l-carnitine suppressed atrogin-1 mRNA expression, which has been reported to play a pivotal role in muscle atrophy. The present study shows that l-carnitine has a protective effect against soleus muscle atrophy caused by hindlimb suspension and decreased E3 ligase messenger RNA expression, suggesting the possibility that l-carnitine protects against muscle atrophy, at least in part, through the inhibition of the UPP. These observations suggest that l-carnitine could serve as an effective supplement in the decrease of muscle atrophy caused by weightlessness in the fields of clinical and rehabilitative research.


Assuntos
Carnitina/uso terapêutico , Suplementos Nutricionais , Repressão Enzimática , Proteínas Musculares/antagonistas & inibidores , Músculo Esquelético/metabolismo , Transtornos Musculares Atróficos/prevenção & controle , Proteínas Ligases SKP Culina F-Box/antagonistas & inibidores , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Animais , Biomarcadores/metabolismo , Elevação dos Membros Posteriores/efeitos adversos , Imuno-Histoquímica , Masculino , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Atrofia Muscular/etiologia , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia , Atrofia Muscular/prevenção & controle , Transtornos Musculares Atróficos/etiologia , Transtornos Musculares Atróficos/metabolismo , Transtornos Musculares Atróficos/patologia , Complexo de Endopeptidases do Proteassoma , Inibidores de Proteassoma/uso terapêutico , Distribuição Aleatória , Ratos , Ratos Wistar , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Ligases SKP Culina F-Box/metabolismo , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Ausência de Peso/efeitos adversos
16.
Expert Rev Hematol ; 9(9): 873-89, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27447436

RESUMO

INTRODUCTION: Proteasome inhibitors have garnered interest as novel chemotherapeutic agents based on their ability to inhibit the growth of cancer cells by altering the balance of intracellular proteins. Initial clinical trials of this drug class focused on bortezomib, a reversible inhibitor of the 20S proteasome, with promising results for the treatment of adult hematologic malignancies, including multiple myeloma and non-Hodgkin lymphoma. AREAS COVERED: This article will review the use of bortezomib and other proteasome inhibitors in both adult and pediatric populations, with a focus on their use in pediatrics. Expert commentary: Bortezomib moved into the pediatric oncology arena with encouraging results in multiple early phase trials for relapsed acute lymphoblastic leukemia and acute myeloid leukemia. Bortezomib is also being studied in the treatment of non-malignant disorders, including antibody-mediated allograft rejection, graft-versus-host disease, and autoimmune cytopenias. The numerous applications of bortezomib have inspired the development of second-generation proteasome inhibitors.


Assuntos
Antineoplásicos/uso terapêutico , Doenças Hematológicas/tratamento farmacológico , Neoplasias Hematológicas/tratamento farmacológico , Inibidores de Proteassoma/uso terapêutico , Fatores Etários , Animais , Antineoplásicos/farmacologia , Bortezomib/farmacologia , Bortezomib/uso terapêutico , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Doenças Hematológicas/diagnóstico , Doenças Hematológicas/metabolismo , Neoplasias Hematológicas/diagnóstico , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/mortalidade , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Transdução de Sinais , Resultado do Tratamento
17.
Eur J Haematol ; 96(6): 564-77, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26893241

RESUMO

Carfilzomib, a second-generation proteasome inhibitor, has been increasingly used in relapsed/refractory multiple myeloma (MM) since its approval by the American food and drug administration (FDA) in the summer of 2012. The drug is active as a single agent and in combination with other antimyeloma agents. As a result of its efficacy and safety in the relapsed/refractory setting, carfilzomib is being evaluated in patients with newly diagnosed MM as well as in high-risk smoldering MM. This review will give a comprehensive summary of the drug, including its mechanism of action, the evaluated doses and schedules as well as a summary of the main clinical trials in the relapsed/refractory and newly diagnosed settings. A focus will be placed upon certain subgroups of interest as well as a description of the toxicity associated with carfilzomib use and a clinical perspective on toxicity management.


Assuntos
Antineoplásicos/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Oligopeptídeos/uso terapêutico , Inibidores de Proteassoma/uso terapêutico , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Ensaios Clínicos Fase III como Assunto , Comorbidade , Avaliação Pré-Clínica de Medicamentos , Resistencia a Medicamentos Antineoplásicos , Humanos , Mieloma Múltiplo/complicações , Mieloma Múltiplo/patologia , Oligopeptídeos/administração & dosagem , Oligopeptídeos/efeitos adversos , Oligopeptídeos/farmacocinética , Inibidores de Proteassoma/administração & dosagem , Inibidores de Proteassoma/efeitos adversos , Inibidores de Proteassoma/farmacocinética , Recidiva , Resultado do Tratamento
18.
Drugs ; 76(3): 405-11, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26846321

RESUMO

Ixazomib (Ninlaro(®)) is an orally bioavailable, reversible proteasome inhibitor developed by Millennium Pharmaceuticals, Inc. (now Takeda Oncology). Ixazomib acts by binding to and inhibiting the ß5 subunit of the 20S proteasome. In November 2015, the US FDA approved ixazomib for use in combination with lenalidomide and dexamethasone for the treatment of patients with multiple myeloma who have received at least one prior therapy. Ixazomib is under regulatory review for this indication in the EU. Phase III development of ixazomib is underway worldwide for newly-diagnosed multiple myeloma (in patients who are not eligible for stem cell transplant, or as maintenance therapy) and for relapsed or refractory systemic light chain (AL) amyloidosis. Ixazomib is also under phase I-II development for the treatment of several other haematological and non-haematological malignancies, graft-versus-host disease and lupus nephritis. This article summarizes the milestones in the development of ixazomib leading to this first approval for multiple myeloma.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Compostos de Boro/uso terapêutico , Aprovação de Drogas , Glicina/análogos & derivados , Mieloma Múltiplo/tratamento farmacológico , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/uso terapêutico , Administração Oral , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Compostos de Boro/administração & dosagem , Compostos de Boro/efeitos adversos , Compostos de Boro/farmacocinética , Ensaios Clínicos como Assunto , Intervalo Livre de Doença , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Glicina/administração & dosagem , Glicina/efeitos adversos , Glicina/farmacocinética , Glicina/uso terapêutico , Humanos , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/mortalidade , Inibidores de Proteassoma/administração & dosagem , Inibidores de Proteassoma/efeitos adversos , Inibidores de Proteassoma/farmacocinética , Ligação Proteica
19.
Mol Neurobiol ; 52(2): 882-98, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26126513

RESUMO

Increased levels of ubiquitin and heat shock protein (HSP) 72 kD are often seen in spinal cord injury (SCI). However, their roles in cell injury or survival are not well known. Thus, we have investigated the possible relationship between ubiquitin and HSP expressions in relation to cell injury in healthy animals, or following nanoparticle (NP) intoxication in SCI animals. A focal SCI was inflicted on the T10-11 segments over the right dorsal horn; animals were allowed to survive from 5 to 8 h after trauma. Separate groups of rats were exposed to SiO2, Ag, or Cu NPs for 7 days and subjected to SCI on the eighth day. A marked increase in ubiquitin or HSP immunoreactive cells occurred in the T9 to T12 segments 5 h after the injury, which further extended to the T4 and L5 after 8 h of survival. At this time, a marked increase in blood-spinal cord barrier (BSCB) permeability to Evans blue and radioiodine, accompanied by an intense edema formation, was observed. Changes were further exacerbated in NP-treated traumatized rats. The most marked expressions of ubiquitin and HSP in SCI were seen in rats treated with SiO2, followed by Ag, and Cu NPs. Treatment with H-290/51 (50 mg/kg p.o., 30 to 60 min after injury) or carfilzomib (1 mg/kg, i.v., 30 to 60 min after trauma) significantly reduced the ubiquitin or HSP expressions, as well as the BSCB breakdown, the edema formation, and the cell injury in the cord both 5 and 8 h after the injury, in normal animals. However, a double dose of H-290/51 (100 mg/kg) or carfilzomib (2 mg/kg) is needed to reduce cord pathology or ubiquitin and HSP expressions in traumatized animals treated with NPs. H-290/51 showed superior beneficial effects in reducing cord pathology in SCI than carfilzomib. These observations are the first to demonstrate that (i) NP-treated traumatized animals induce a widespread BSCB leakage, edema formation, and cord pathology as well as an overexpression of ubiquitin and HSP, (ii) high doses of antioxidant compounds or proteasome inhibitors are required for neuroprotection in the NP-exposed traumatized group, and (iii) ubiquitin and HSP expressions play a key role in neuronal injury in SCI, not reported earlier.


Assuntos
Antioxidantes/uso terapêutico , Cobre/toxicidade , Proteínas de Choque Térmico HSP72/biossíntese , Indóis/uso terapêutico , Nanopartículas/toxicidade , Proteínas do Tecido Nervoso/biossíntese , Fármacos Neuroprotetores/uso terapêutico , Oligopeptídeos/uso terapêutico , Inibidores de Proteassoma/uso terapêutico , Prata/toxicidade , Traumatismos da Medula Espinal/tratamento farmacológico , Ubiquitina/biossíntese , Animais , Antioxidantes/farmacologia , Cobre/administração & dosagem , Avaliação Pré-Clínica de Medicamentos , Edema/etiologia , Edema/prevenção & controle , Proteínas de Choque Térmico HSP72/genética , Indóis/farmacologia , Masculino , Proteínas do Tecido Nervoso/genética , Fármacos Neuroprotetores/farmacologia , Oligopeptídeos/farmacologia , Inibidores de Proteassoma/farmacologia , Ratos , Ratos Wistar , Dióxido de Silício/administração & dosagem , Dióxido de Silício/toxicidade , Prata/administração & dosagem , Medula Espinal/metabolismo , Medula Espinal/patologia , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/etiologia , Traumatismos da Medula Espinal/metabolismo , Vértebras Torácicas , Ubiquitina/genética , Regulação para Cima
20.
Future Oncol ; 11(8): 1153-68, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25832873

RESUMO

Ixazomib is an investigational, reversible 20S proteasome inhibitor. It is the first oral proteasome inhibitor under clinical investigation in multiple myeloma (MM). Under physiological conditions, the stable citrate ester drug substance, ixazomib citrate (MLN9708), rapidly hydrolyzes to the biologically active boronic acid, ixazomib (MLN2238). Preclinical studies have demonstrated antitumor activity in MM cell lines and xenograft models. In Phase I/II clinical studies ixazomib has had generally manageable toxicities, with limited peripheral neuropathy observed to date. Preliminary data from these studies indicate ixazomib is active as a single agent in relapsed/refractory MM and as part of combination regimens in newly diagnosed patients. Phase III studies in combination with lenalidomide-dexamethasone are ongoing.


Assuntos
Compostos de Boro/uso terapêutico , Drogas em Investigação/uso terapêutico , Glicina/análogos & derivados , Mieloma Múltiplo/tratamento farmacológico , Inibidores de Proteassoma/uso terapêutico , Animais , Compostos de Boro/efeitos adversos , Avaliação Pré-Clínica de Medicamentos , Drogas em Investigação/efeitos adversos , Glicina/efeitos adversos , Glicina/uso terapêutico , Humanos , Inibidores de Proteassoma/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA