Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Future Oncol ; 18(10): 1185-1198, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35034502

RESUMEN

Cabozantinib inhibits multiple receptor tyrosine kinases, including the TAM kinase family, and may enhance response to immune checkpoint inhibitors. One cohort of the ongoing phase Ib COSMIC-021 study (NCT03170960) evaluating cabozantinib plus the PD-L1 inhibitor atezolizumab in men with metastatic castration-resistant prostate cancer (mCRPC) that has progressed in soft tissue on/after enzalutamide and/or abiraterone treatment for metastatic disease has shown promising efficacy. Here, we describe the rationale and design of a phase III trial of cabozantinib plus atezolizumab versus a second novel hormone therapy (NHT) in patients who have previously received an NHT for mCRPC, metastatic castration-sensitive PC or nonmetastatic CRPC and have measurable visceral disease and/or extrapelvic adenopathy - a population with a significant unmet need for treatment options. Trial Registration Clinical Trial Registration: NCT04446117 (ClinicalTrials.gov) Registered on 24 June 2020.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Anilidas/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos Hormonales/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Piridinas/uso terapéutico , Adenocarcinoma/patología , Androstenos/uso terapéutico , Benzamidas/uso terapéutico , Humanos , Masculino , Metástasis de la Neoplasia , Nitrilos/uso terapéutico , Feniltiohidantoína/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/patología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores
2.
Lancet Oncol ; 17(1): 27-38, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26671818

RESUMEN

BACKGROUND: Bortezomib with dexamethasone is a standard treatment option for relapsed or refractory multiple myeloma. Carfilzomib with dexamethasone has shown promising activity in patients in this disease setting. The aim of this study was to compare the combination of carfilzomib and dexamethasone with bortezomib and dexamethasone in patients with relapsed or refractory multiple myeloma. METHODS: In this randomised, phase 3, open-label, multicentre study, patients with relapsed or refractory multiple myeloma who had one to three previous treatments were randomly assigned (1:1) using a blocked randomisation scheme (block size of four) to receive carfilzomib with dexamethasone (carfilzomib group) or bortezomib with dexamethasone (bortezomib group). Randomisation was stratified by previous proteasome inhibitor therapy, previous lines of treatment, International Staging System stage, and planned route of bortezomib administration if randomly assigned to bortezomib with dexamethasone. Patients received treatment until progression with carfilzomib (20 mg/m(2) on days 1 and 2 of cycle 1; 56 mg/m(2) thereafter; 30 min intravenous infusion) and dexamethasone (20 mg oral or intravenous infusion) or bortezomib (1·3 mg/m(2); intravenous bolus or subcutaneous injection) and dexamethasone (20 mg oral or intravenous infusion). The primary endpoint was progression-free survival in the intention-to-treat population. All participants who received at least one dose of study drug were included in the safety analyses. The study is ongoing but not enrolling participants; results for the interim analysis of the primary endpoint are presented. The trial is registered at ClinicalTrials.gov, number NCT01568866. FINDINGS: Between June 20, 2012, and June 30, 2014, 929 patients were randomly assigned (464 to the carfilzomib group; 465 to the bortezomib group). Median follow-up was 11·9 months (IQR 9·3-16·1) in the carfilzomib group and 11·1 months (8·2-14·3) in the bortezomib group. Median progression-free survival was 18·7 months (95% CI 15·6-not estimable) in the carfilzomib group versus 9·4 months (8·4-10·4) in the bortezomib group at a preplanned interim analysis (hazard ratio [HR] 0·53 [95% CI 0·44-0·65]; p<0·0001). On-study death due to adverse events occurred in 18 (4%) of 464 patients in the carfilzomib group and in 16 (3%) of 465 patients in the bortezomib group. Serious adverse events were reported in 224 (48%) of 463 patients in the carfilzomib group and in 162 (36%) of 456 patients in the bortezomib group. The most frequent grade 3 or higher adverse events were anaemia (67 [14%] of 463 patients in the carfilzomib group vs 45 [10%] of 456 patients in the bortezomib group), hypertension (41 [9%] vs 12 [3%]), thrombocytopenia (39 [8%] vs 43 [9%]), and pneumonia (32 [7%] vs 36 [8%]). INTERPRETATION: For patients with relapsed or refractory multiple myeloma, carfilzomib with dexamethasone could be considered in cases in which bortezomib with dexamethasone is a potential treatment option. FUNDING: Onyx Pharmaceuticals, Inc., an Amgen subsidiary.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Anciano , Anemia/inducido químicamente , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Bortezomib/administración & dosificación , Bortezomib/efectos adversos , Dexametasona/administración & dosificación , Dexametasona/efectos adversos , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Hipertensión/inducido químicamente , Masculino , Oligopéptidos/administración & dosificación , Oligopéptidos/efectos adversos , Neumonía/inducido químicamente , Retratamiento , Tasa de Supervivencia , Trombocitopenia/inducido químicamente
3.
J Environ Public Health ; 2022: 6745813, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35342432

RESUMEN

Introduction: Spatial presentation is considered a useful tool for analyzing and mapping the frequencies of incidences of different pathogens. Khartoum State accounted for 78% of the overall cases of COVID-19 in Sudan. The aim of this study was to present the spatial extent of healthcare services of a private isolation center during the pandemic at the locality level. Materials and Methods: A spatial descriptive study was conducted using ArcGIS to present the locations of all COVID-19 patients who attended Imperial Hospital isolation center on November-December 2020 in Khartoum, Sudan. Results: Patients diagnosed with COVID-19 during the study period were 188; they had attended Imperial Hospital from 9 states. Patients from Khartoum State were 167 patients. Of those 167 patients, 75 were from the Khartoum locality; it is the locality in which Imperial Hospital is located, followed by Khartoum Bahri (34 patients), Omdurman (19 patients), and South Khartoum (14 patients), while 10 patients each were from the Sharg En Nile and Karary localities. Conclusion: Patients from 8 different states of Sudan had travelled to reach Khartoum State to get health services. At the state level, Khartoum State was the most benefited state from the healthcare services of Imperial Hospital. At a locality level, Khartoum locality was the most benefited one.


Asunto(s)
COVID-19 , COVID-19/epidemiología , Atención a la Salud , Servicios de Salud , Hospitales Privados , Humanos , Sudán/epidemiología
4.
Eur J Med Chem ; 123: 191-201, 2016 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-27484508

RESUMEN

Efforts to develop new antitumor agents are now directed towards multitarget therapies that are believed to have high potency and low tendency to resistance compared to conventional drugs. Herein, we highlighted the synthesis and antitumor activity of five series of phthalazine-based compounds featuring a variety of bioactive chemical fragments at position 1 of the phthalazine nucleus. The antitumor activity of the target compounds was performed against fourteen cancer cell lines where all compounds were active in the nanomolar level. In addition, the mechanism of action of the target compounds was investigated through an enzymatic inhibitory assay against VEGFR-2 and EGFR kinases, revealing potent and preferential activity toward VEGFR-2. Binding mode of the most active compounds was studied using docking experiment.


Asunto(s)
Antineoplásicos/química , Receptores ErbB/antagonistas & inhibidores , Ftalazinas/farmacología , Inhibidores de Proteínas Quinasas/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Antineoplásicos/farmacología , Línea Celular Tumoral , Humanos , Simulación del Acoplamiento Molecular , Ftalazinas/química , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Relación Estructura-Actividad
5.
Curr Opin Mol Ther ; 7(2): 144-50, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15844622

RESUMEN

Heteropolymer (HP) technology is a novel cassette technology which is being developed for the treatment of infectious and autoimmune diseases. HPs are dual antibody conjugates, composed of a monoclonal antibody (mAb) directed against the complement receptor type 1 (CR1) on primate red blood cells (RBCs) chemically cross-linked to mAbs that recognize blood-borne antigens. Upon administration of an HP, the target is bound to its counterpart mAb in the HP and immobilized on an RBC by binding of the anti-CR1 mAb to CR1 in a complement-independent manner, forming an immune complex. When the RBC traverses the liver during circulation, the immune complex is recognized by fixed tissue macrophages, the CR1 molecule is cleaved and the HP-pathogen complex is phagocytosed and destroyed. Due to rapid binding and immobilization of the target by HPs, the anti-target mAb used in the HP need not be directed to a neutralizing epitope on the target organism. Studies in animal models have shown that HPs are effective in treating infectious diseases and autoimmune diseases such as systemic lupus erythematosus. HPs are advantageous over natural immune adherence or conventional mAb therapies due to their complement-independent mechanism, low therapeutic dose and lack of the need for neutralizing mAbs against the target.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Patógenos Transmitidos por la Sangre , Enfermedades Transmisibles/tratamiento farmacológico , Receptores de Complemento/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Antígenos/inmunología , Enfermedades Transmisibles/inmunología , Humanos , Ratones , Receptores de Complemento/genética
7.
Infect Immun ; 73(2): 795-802, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15664918

RESUMEN

We have developed a therapeutic for the treatment of anthrax using an affinity-enhanced monoclonal antibody (ETI-204) to protective antigen (PA), which is the central cell-binding component of the anthrax exotoxins. ETI-204 administered preexposure by a single intravenous injection of a dose of between 2.5 and 10 mg per animal significantly protected rabbits from a lethal aerosolized anthrax spore challenge ( approximately 60 to 450 times the 50% lethal dose of Bacillus anthracis Ames). Against a similar challenge, ETI-204 administered intramuscularly at a 20-mg dose per animal completely protected rabbits from death (100% survival). In the postexposure setting, intravenous administration of ETI-204 provided protection 24 h (8 of 10) and 36 h (5 of 10) after spore challenge. Administration at 48 h postchallenge, when 3 of 10 animals had already succumbed to anthrax infection, resulted in the survival of 3 of 7 animals (43%) for the duration of the study (28 days). Importantly, surviving ETI-204-treated animals were free of bacteremia by day 10 and remained so until the end of the studies. Only 11 of 51 ETI-204-treated rabbits had positive lung cultures at the end of the studies. Also, rabbits that were protected from inhalational anthrax by administration of ETI-204 developed significant titers of PA-specific antibodies. Presently, the sole therapeutic regimen available to treat infection by inhalation of B. anthracis spores is a 60-day course of antibiotics that is effective only if administered prior to or shortly after exposure. Based upon results reported here, ETI-204 is an effective therapy for prevention and treatment of inhalational anthrax.


Asunto(s)
Carbunco/prevención & control , Anticuerpos Monoclonales/inmunología , Antígenos Bacterianos/inmunología , Bacillus anthracis/inmunología , Toxinas Bacterianas/inmunología , Animales , Carbunco/inmunología , Carbunco/veterinaria , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacocinética , Antitoxinas , Sangre/microbiología , Inyecciones Intramusculares , Conejos , Esporas Bacterianas , Factores de Tiempo
8.
Cancer Immunol Immunother ; 54(12): 1172-9, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15846490

RESUMEN

Complement-dependent cytotoxicity (CDC) is a key mechanism of Rituximab (RTX) action in killing non-Hodgkin's lymphoma (NHL) cells both in vitro and probably in vivo. A DeImmunized, mouse/human chimeric monoclonal antibody (Mab), H17, specific for cell-associated complement C3 cleavage products, C3b and iC3b, was generated to enhance RTX-mediated killing of target cells by CDC. When NHL cell lines were treated with RTX and H17 in the presence of complement for 1 h, there was 40-70% more cell death than that observed with RTX alone. The enhancing effect of H17 was also seen over longer treatment periods. H17 was tested ex vivo against primary cells from NHL and chronic lymphocytic leukemia (CLL) patients. In RTX-resistant NHL samples, H17 enhanced RTX-mediated killing; in the remaining samples RTX + complement alone promoted more than 80% killing, and no significant enhancement was observed. The H17 antibody also increased RTX-mediated killing in four out of nine CLL samples. H17 may have therapeutic applications in NHL and CLL treatment as an adjunctive therapy to RTX. It might also enhance the activity of other therapeutic antibodies that work through CDC.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Activación de Complemento , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Linfoma no Hodgkin/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales de Origen Murino , Proteínas del Sistema Complemento/inmunología , Humanos , Ratones , Rituximab , Células Tumorales Cultivadas
9.
Infect Immun ; 72(6): 3276-83, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15155630

RESUMEN

We investigated the ability of using monoclonal antibodies (MAbs) against anthrax protective antigen (PA), an anthrax exotoxin component, to modulate exotoxin cytotoxic activity on target macrophage cell lines. Anthrax PA plays a critical role in the pathogenesis of Bacillus anthracis infection. PA is the cell-binding component of the two anthrax exotoxins: lethal toxin (LeTx) and edema toxin. Several MAbs that bind the PA component of LeTx are known to neutralize LeTx-mediated killing of target macrophages. Here we describe for the first time an overlooked population of anti-PA MAbs that, in contrast, function to increase the potency of LeTx against murine macrophage cell lines. The results support a possible mechanism of enhancement: binding of MAb to PA on the macrophage cell surface stabilizes the PA by interaction of MAb with macrophage Fcgamma receptors. This results in an increase in the amount of PA bound to the cell surface, which in turn leads to an enhancement in cell killing, most likely due to increased internalization of LF. Blocking of PA-receptor binding eliminates enhancement by MAb, demonstrating the importance of this step for the observed enhancement. The additional significance of these results is that, at least in mice, immunization with PA appears to elicit a poly-clonal response that has a significant prevalence of MAbs that enhance LeTx-mediated killing in macrophages.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/inmunología , Acrecentamiento Dependiente de Anticuerpo , Toxinas Bacterianas/toxicidad , Macrófagos/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Monoclonales/sangre , Antígenos Bacterianos/inmunología , Bacillus anthracis/metabolismo , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/inmunología , Células CHO , Línea Celular , Cricetinae , Humanos , Macrófagos/fisiología , Ratones , Receptores Fc/metabolismo , Células U937
10.
Clin Immunol ; 105(2): 141-54, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12482388

RESUMEN

Autoantibodies that react with double-stranded DNA (dsDNA) are a hallmark for diagnosis of systemic lupus erythematosus (SLE) and are also considered the pathogenic subset that is most associated with lupus nephritis. As an agent to remove the pathogenic dsDNA antibodies from the circulation of SLE patients, we are developing an antigen-based heteropolymer (AHP). The AHP consists of a monoclonal antibody to the complement receptor (CR1) cross-linked to salmon testis dsDNA to effect clearance of anti-DNA antibodies by binding them to erythrocyte CR1. Utilizing a cynomolgus monkey model for SLE in which we infused plasma from SLE patients containing a high titer of high-avidity anti-dsDNA antibody, we have evaluated the safety and efficacy of AHP infusion. The results demonstrate that AHP rapidly (within 2 min of infusion) binds to monkey erythrocytes without causing any toxicological effects. We also demonstrate that human Ig (G+M) antibodies are rapidly bound to the AHP-erythrocyte complex. These events are mirrored in their kinetics by a substantial drop in the level of high-avidity dsDNA antibody in the plasma.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Lupus Eritematoso Sistémico/terapia , Animales , Anticuerpos Antinucleares/sangre , Afinidad de Anticuerpos , Antígenos , ADN/inmunología , ADN/uso terapéutico , Modelos Animales de Enfermedad , Eritrocitos/inmunología , Femenino , Humanos , Lupus Eritematoso Sistémico/inmunología , Macaca fascicularis , Masculino , Receptores de Complemento 3b/inmunología , Salmón
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA