Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Toxicol Pathol ; 42(3): 524-39, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23674391

RESUMEN

We recently reported results that erythropoiesis-stimulating agent (ESA)-related thrombotic toxicities in preclinical species were not solely dependent on a high hematocrit (HCT) but also associated with increased ESA dose level, dose frequency, and dosing duration. In this article, we conclude that sequelae of an increased magnitude of ESA-stimulated erythropoiesis potentially contributed to thrombosis in the highest ESA dose groups. The results were obtained from two investigative studies we conducted in Sprague-Dawley rats administered a low (no thrombotic toxicities) or high (with thrombotic toxicities) dose level of a hyperglycosylated analog of recombinant human erythropoietin (AMG 114), 3 times weekly for up to 9 days or for 1 month. Despite similarly increased HCT at both dose levels, animals in the high-dose group had an increased magnitude of erythropoiesis measured by spleen weights, splenic erythropoiesis, and circulating reticulocytes. Resulting prothrombotic risk factors identified predominantly or uniquely in the high-dose group were higher numbers of immature reticulocytes and nucleated red blood cells in circulation, severe functional iron deficiency, and increased intravascular destruction of iron-deficient reticulocyte/red blood cells. No thrombotic events were detected in rats dosed up to 9 days suggesting a sustained high HCT is a requisite cofactor for development of ESA-related thrombotic toxicities.


Asunto(s)
Eritropoyesis/efectos de los fármacos , Eritropoyetina/farmacología , Eritropoyetina/toxicidad , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/toxicidad , Análisis de Varianza , Animales , Plaquetas , Eritrocitos , Eritropoyetina/administración & dosificación , Hematócrito , Humanos , Hierro/sangre , Hierro/metabolismo , Masculino , Policitemia , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Reticulocitos
2.
Toxicol Pathol ; 42(3): 540-54, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23674392

RESUMEN

We previously reported an increased incidence of thrombotic toxicities in Sprague-Dawley rats administered the highest dose level of a hyperglycosylated analog of recombinant human erythropoietin (AMG 114) for 1 month as not solely dependent on high hematocrit (HCT). Thereafter, we identified increased erythropoiesis as a prothrombotic risk factor increased in the AMG 114 high-dose group with thrombotic toxicities, compared to a low-dose group with no toxicities but similar HCT. Here, we identified pleiotropic cytokines as prothrombotic factors associated with AMG 114 dose level. Before a high HCT was achieved, rats in the AMG 114 high, but not the low-dose group, had imbalanced hemostasis (increased von Willebrand factor and prothrombin time, decreased antithrombin III) coexistent with cytokines implicated in thrombosis: monocyte chemotactic protein 1 (MCP-1), MCP-3, tissue inhibitor of metalloproteinases 1, macrophage inhibitory protein-2, oncostatin M, T-cell-specific protein, stem cell factor, vascular endothelial growth factor, and interleukin-11. While no unique pathway to erythropoiesis stimulating agent-related thrombosis was identified, cytokines associated with increased erythropoiesis contributed to a prothrombotic intravascular environment in the AMG 114 high-dose group, but not in lower dose groups with a similar high HCT.


Asunto(s)
Citocinas/sangre , Citocinas/metabolismo , Eritropoyesis/efectos de los fármacos , Eritropoyetina/farmacología , Proteínas Recombinantes/farmacología , Animales , Eritropoyetina/química , Hematócrito , Humanos , Masculino , Policitemia , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/química , Reticulocitos , Trombosis
3.
Clin Cancer Res ; 25(13): 3921-3933, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30918018

RESUMEN

PURPOSE: Despite advances in the treatment of multiple myeloma, new therapies are needed to induce more profound clinical responses. T-cell-redirected lysis triggered by bispecific antibodies recruiting T cells to cancer cells is a clinically validated mechanism of action against hematologic malignancies and CD38 is a tumor-associated antigen with near-universal expression in multiple myeloma. Thus, an anti-CD38/CD3 bispecific T-cell-recruiting antibody has the potential to be an effective new therapeutic for multiple myeloma. EXPERIMENTAL DESIGN: Anti-CD38/CD3 XmAb T-cell-recruiting antibodies with different affinities for CD38 and CD3 were assessed in vitro and in vivo for their redirected T-cell lysis activity against cancer cell lines, their lower levels of cytokine release, and their potency in the presence of high levels of soluble CD38. Select candidates were further tested in cynomolgus monkeys for B-cell depletion and cytokine release properties. RESULTS: AMG 424 was selected on the basis of its ability to kill cancer cells expressing high and low levels of CD38 in vitro and trigger T-cell proliferation, but with attenuated cytokine release. In vivo, AMG 424 induces tumor growth inhibition in bone marrow-invasive mouse cancer models and the depletion of peripheral B cells in cynomolgus monkeys, without triggering excessive cytokine release. The activity of AMG 424 against normal immune cells expressing CD38 is also presented. CONCLUSIONS: These findings support the clinical development of AMG 424, an affinity-optimized T-cell-recruiting antibody with the potential to elicit significant clinical activity in patients with multiple myeloma.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos , Antineoplásicos Inmunológicos/uso terapéutico , Citocinas/biosíntesis , Mieloma Múltiple/inmunología , Mieloma Múltiple/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , ADP-Ribosil Ciclasa 1/antagonistas & inhibidores , Animales , Anticuerpos Biespecíficos/administración & dosificación , Anticuerpos Biespecíficos/efectos adversos , Afinidad de Anticuerpos/inmunología , Antineoplásicos Inmunológicos/administración & dosificación , Antineoplásicos Inmunológicos/efectos adversos , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/metabolismo , Complejo CD3/antagonistas & inhibidores , Línea Celular Tumoral , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Activación de Linfocitos/inmunología , Macaca fascicularis , Ratones , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Linfocitos T/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Pharm Sci ; 107(6): 1512-1520, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29408510

RESUMEN

Comparability studies used to assess a proposed manufacturing change for a biological product include sensitive analytical studies to confirm there are no significant differences in structural or functional attributes that may contribute to clinically meaningful changes in efficacy or safety. When a proposed change is relatively complex or when clinically relevant differences between the product before and after the change cannot be ruled out based on analytical studies, nonclinical and clinical bridging studies are generally required to confirm overall comparability. In this study, we report findings from a comparability assessment of epoetin alfa before and after a proposed manufacturing process change. Although differences in glycosylation attributes were observed, these were initially believed to be irrelevant to the product's pharmacology. This assumption was initially supported via nonclinical and clinical pharmacology studies, but a clinically meaningful difference in potency was ultimately observed in a phase 3 clinical study conducted in a sensitive patient population using a sensitive study design. These results indicate that the nonclinical assessments of structure-function relationships were insufficiently sensitive to identify clinically relevant differences resulting from differences in the glycosylation profile. This case study highlights important findings that may be relevant in the development of biosimilar epoetin alfa products.


Asunto(s)
Anemia/tratamiento farmacológico , Epoetina alfa/uso terapéutico , Hematínicos/uso terapéutico , Insuficiencia Renal Crónica/tratamiento farmacológico , Anemia/complicaciones , Animales , Biosimilares Farmacéuticos/química , Biosimilares Farmacéuticos/farmacocinética , Biosimilares Farmacéuticos/farmacología , Biosimilares Farmacéuticos/uso terapéutico , Aprobación de Drogas , Epoetina alfa/química , Epoetina alfa/farmacocinética , Epoetina alfa/farmacología , Glicosilación , Hematínicos/química , Hematínicos/farmacocinética , Hematínicos/farmacología , Humanos , Ratones , Ratones SCID , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Insuficiencia Renal Crónica/complicaciones , Proyectos de Investigación , Relación Estructura-Actividad
5.
PLoS One ; 12(8): e0183390, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28837681

RESUMEN

For targets that are homogenously expressed, such as CD19 on cells of the B lymphocyte lineage, immunotherapies can be highly effective. Targeting CD19 with blinatumomab, a CD19/CD3 bispecific antibody construct (BiTE®), or with chimeric antigen receptor T cells (CAR-T) has shown great promise for treating certain CD19-positive hematological malignancies. In contrast, solid tumors with heterogeneous expression of the tumor-associated antigen (TAA) may present a challenge for targeted therapies. To prevent escape of TAA-negative cancer cells, immunotherapies with a local bystander effect would be beneficial. As a model to investigate BiTE®-mediated bystander killing in the solid tumor setting, we used epidermal growth factor receptor (EGFR) as a target. We measured lysis of EGFR-negative populations in vitro and in vivo when co-cultured with EGFR-positive cells, human T cells and an EGFR/CD3 BiTE® antibody construct. Bystander EGFR-negative cells were efficiently lysed by BiTE®-activated T cells only when proximal to EGFR-positive cells. Our mechanistic analysis suggests that cytokines released by BiTE®-activated T-cells induced upregulation of ICAM-1 and FAS on EGFR-negative bystander cells, contributing to T cell-induced bystander cell lysis.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Efecto Espectador , Neoplasias/patología , Linfocitos T/inmunología , Animales , Técnicas de Cocultivo , Citocinas/metabolismo , Citotoxicidad Inmunológica , Receptores ErbB/metabolismo , Femenino , Xenoinjertos , Humanos , Activación de Linfocitos , Ratones , Ratones Desnudos
6.
Exp Hematol ; 43(6): 479-87, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25754671

RESUMEN

Chemotherapy-induced thrombocytopenia can lead to chemotherapy treatment delays or dose reductions. The ability of romiplostim, a thrombopoietin (TPO) mimetic, to promote platelet recovery in a mouse model of multicycle chemotherapy/radiation therapy (CRT)-induced thrombocytopenia was examined. In humans, an inverse relationship between platelet counts and endogenous TPO (eTPO) concentration exists. In a CRT mouse model, eTPO was not elevated during the first 5 days after CRT treatment (the "eTPO gap"), then increased to a peak 10 days after each CRT treatment in an inverse relationship to platelet counts seen in humans. To bridge the eTPO gap, mice were treated with 10-1,000 µg/kg of romiplostim on day 0, 1, or 2 after CRT. In some mice, the romiplostim dose was approximately divided over 3 days. Platelet recovery occurred faster with romiplostim in most conditions tested. Romiplostim doses of ≥100 µg/kg given on day 0 significantly lessened the platelet nadir. Fractionating the dose over 3 days did not appear to confer a large advantage. These data may provide a rationale for clinical studies of romiplostim in chemotherapy-induced thrombocytopenia.


Asunto(s)
Antineoplásicos/efectos adversos , Plaquetas/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Trombocitopenia/prevención & control , Trombopoyetina/farmacología , Animales , Modelos Animales de Enfermedad , Femenino , Ratones , Receptores Fc , Trombocitopenia/sangre , Trombocitopenia/inducido químicamente
7.
Cancer Res ; 70(23): 9846-54, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20935223

RESUMEN

In mammalian cells, the aurora kinases (aurora-A, -B, and -C) play essential roles in regulating cell division. The expression of aurora-A and -B is elevated in a variety of human cancers and is associated with high proliferation rates and poor prognosis, making them attractive targets for anticancer therapy. AMG 900 is an orally bioavailable, potent, and highly selective pan-aurora kinase inhibitor that is active in taxane-resistant tumor cell lines. In tumor cells, AMG 900 inhibited autophosphorylation of aurora-A and -B as well as phosphorylation of histone H3 on Ser(10), a proximal substrate of aurora-B. The predominant cellular response of tumor cells to AMG 900 treatment was aborted cell division without a prolonged mitotic arrest, which ultimately resulted in cell death. AMG 900 inhibited the proliferation of 26 tumor cell lines, including cell lines resistant to the antimitotic drug paclitaxel and to other aurora kinase inhibitors (AZD1152, MK-0457, and PHA-739358), at low nanomolar concentrations. Furthermore, AMG 900 was active in an AZD1152-resistant HCT116 variant cell line that harbors an aurora-B mutation (W221L). Oral administration of AMG 900 blocked the phosphorylation of histone H3 in a dose-dependent manner and significantly inhibited the growth of HCT116 tumor xenografts. Importantly, AMG 900 was broadly active in multiple xenograft models, including 3 multidrug-resistant xenograft models, representing 5 tumor types. AMG 900 has entered clinical evaluation in adult patients with advanced cancers and has the potential to treat tumors refractory to anticancer drugs such as the taxanes.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Ftalazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Adulto , Animales , Aurora Quinasa A , Aurora Quinasa B , Aurora Quinasas , Benzamidas/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos Clínicos como Asunto , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Células HCT116 , Células HeLa , Histonas/metabolismo , Humanos , Ratones , Ratones Desnudos , Mutación , Neoplasias/enzimología , Neoplasias/patología , Organofosfatos/farmacología , Paclitaxel/farmacología , Fosforilación/efectos de los fármacos , Piperazinas/farmacología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Pirazoles/farmacología , Quinazolinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Acta Haematol ; 113(3): 163-74, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15870486

RESUMEN

The unit of erythropoietic activity has long been the standard by which erythropoietic agents are judged, but the development of long-acting agents such as darbepoetin alfa has highlighted the shortcomings of this approach. To this point, we compared the in vivo activity of Epoetin alfa and darbepoetin alfa per microgram of protein core. Using the established mass-to-unit conversion for Epoetin alfa (1 microg congruent with 200 U), we then calculated darbepoetin alfa activity in units. Activity varied with treatment regimen (1 microg darbepoetin alfa congruent with 800 U for 3 times weekly dosing to 8,000 U for a single injection). This analysis reveals the inadequacy of evaluating darbepoetin alfa activity in terms of standard erythropoietic units. We therefore propose that for molecules with heightened biological activity, a more legitimate basis for comparison is the protein mass.


Asunto(s)
Eritropoyesis/efectos de los fármacos , Eritropoyetina/análogos & derivados , Eritropoyetina/administración & dosificación , Animales , Darbepoetina alfa , Esquema de Medicación , Epoetina alfa , Eritropoyetina/normas , Femenino , Ratones , Proteínas Recombinantes , Estándares de Referencia
9.
Br J Haematol ; 122(4): 623-36, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12899718

RESUMEN

Despite its frequency and impact on clinical outcomes, anaemia in cancer patients remains poorly understood and suboptimally treated. The definition of optimum treatment schedules with erythropoietic agents requires a suitable model of chemotherapy-induced progressive anaemia. This study investigated novel strategies such as once-per-chemotherapy-cycle dosing, synchronization between erythroid supportive care and chemotherapy, and definition of the optimum timing of erythroid support. A murine model of carboplatin chemotherapy/radiotherapy (CRT)-induced anaemia was used, which caused progressive anaemia across multiple cycles. Weekly administration of recombinant human erythropoietin (rHuEPO) was effective, but the longer-acting darbepoetin alpha resulted in superior responses. In all animals, anaemia became progressive and more refractory across cycles because of accumulated bone marrow damage. Exploiting a specific enzyme-linked immunosorbent assay, which could distinguish between darbepoetin alpha and endogenous erythropoietin, the effect of CRT upon the pharmacokinetics of darbepoetin alpha showed that clearance of darbepoetin alpha, and presumably erythropoietin, was at least partially dependent on a chemotherapy-sensitive pathway. Scheduling data suggested that administration of erythropoietic agents prior to chemotherapy was more effective than administration after chemotherapy. There was no evidence that erythropoietic agents exacerbated anaemia, even when administered immediately prior to CRT in an attempt to "prime" erythroid cells for the effects of CRT.


Asunto(s)
Anemia/prevención & control , Antineoplásicos/efectos adversos , Eritropoyetina/análogos & derivados , Eritropoyetina/uso terapéutico , Hematopoyesis/efectos de los fármacos , Traumatismos por Radiación/prevención & control , Anemia/etiología , Anemia/fisiopatología , Animales , Carboplatino/efectos adversos , Terapia Combinada , Darbepoetina alfa , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Eritropoyetina/sangre , Femenino , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/efectos de la radiación , Hemoglobinas/metabolismo , Ratones , Ratones Endogámicos , Traumatismos por Radiación/etiología , Traumatismos por Radiación/fisiopatología , Radioterapia/efectos adversos , Proteínas Recombinantes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA