Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Blood ; 132(24): 2546-2554, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30352784

RESUMEN

Selinexor is an oral inhibitor of the nuclear export protein exportin 1. Preclinical studies demonstrated synergistic antimyeloma activity between selinexor and proteasome inhibitors (PI) through suppression of NF-κB signaling and nuclear retention of tumor suppressor proteins. We tested selinexor in combination with low-dose bortezomib and dexamethasone (SVd) for the treatment of relapsed or refractory multiple myeloma (MM). The primary objectives of this study were to determine the safety profile, overall response rate (ORR), and a recommended phase 2 dose (RP2D) of SVd. We enrolled 42 patients to receive selinexor (60, 80, or 100 mg orally) plus bortezomib (1.3 mg/m2 subcutaneously) and dexamethasone (20 mg orally) once or twice weekly in 21- or 35-day cycles. Patients had a median of 3 (range 1-11) prior lines of therapy, and 50% were refractory to a PI. Treatment-related grade 3 or 4 adverse events reported in ≥10% of patients were thrombocytopenia (45%), neutropenia (24%), fatigue (14%), and anemia (12%). Incidence (4 patients, 10%) and grade (≤2) of peripheral neuropathy were low. The ORR for the entire population was 63%: 84% ORR for PI nonrefractory and 43% for PI-refractory patients. The median progression-free survival for all patients was 9.0 months; 17.8 months for PI nonrefractory, and 6.1 months for PI refractory. SVd treatment produced high response rates in patients with relapsed or refractory MM, including borezomib-refractory MM, with no unexpected side effects. The RP2D is selinexor (100 mg once weekly), bortezomib (1.3 mg/m2 once weekly for 4 weeks), and dexamethasone (40 mg once weekly) per 35-day cycle. This trial was registered at www.clinicaltrials.gov as #NCT02343042.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/mortalidad , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Bortezomib/administración & dosificación , Bortezomib/efectos adversos , Dexametasona/administración & dosificación , Dexametasona/efectos adversos , Supervivencia sin Enfermedad , Femenino , Humanos , Hidrazinas/administración & dosificación , Hidrazinas/efectos adversos , Masculino , Persona de Mediana Edad , Tasa de Supervivencia , Triazoles/administración & dosificación , Triazoles/efectos adversos
2.
Blood ; 131(8): 855-863, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29203585

RESUMEN

Novel therapies are needed for patients with relapsed or refractory multiple myeloma (MM). We conducted a multicenter, phase 1 study in advanced hematological malignancies to assess the safety, efficacy, and recommended phase 2 dose (RP2D) of oral selinexor, a selective inhibitor of the nuclear export protein XPO1. In the dose-escalation phase, 25 patients with heavily pretreated MM (22) or Waldenstrom macroglobulinemia (3) were administered selinexor (3-60 mg/m2) in 8 or 10 doses per 28-day cycle. In the dose-expansion phase, 59 patients with MM received selinexor at 45 or 60 mg/m2 with 20 mg dexamethasone, twice weekly in 28-day cycles, or selinexor (40 or 60 mg flat dose) without corticosteroids in 21-day cycles. The most common nonhematologic adverse events (AEs) were nausea (75%), fatigue (70%), anorexia (64%), vomiting (43%), weight loss (32%), and diarrhea (32%), which were primarily grade 1 or 2. The most common grade 3 or 4 AEs were hematologic, particularly thrombocytopenia (45%). Single-agent selinexor showed modest efficacy with an objective response rate (ORR) of 4% and clinical benefit rate of 21%. In contrast, the addition of dexamethasone increased the ORR with all responses of ≥partial response occurring in the 45 mg/m2 selinexor plus 20 mg dexamethasone twice weekly cohort (ORR = 50%). Furthermore, 46% of all patients showed a reduction in MM markers from baseline. Based on these findings, we conclude that selinexor in combination with dexamethasone is active in heavily pretreated MM and propose a RP2D of 45 mg/m2 (80 mg) plus 20 mg dexamethasone given twice weekly. This trial was registered at clinicaltrials.gov as #NCT01607892.


Asunto(s)
Dexametasona/uso terapéutico , Hidrazinas/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Triazoles/uso terapéutico , Macroglobulinemia de Waldenström/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos Hormonales/uso terapéutico , Quimioterapia Combinada , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Mieloma Múltiple/patología , Recurrencia Local de Neoplasia/patología , Pronóstico , Seguridad , Macroglobulinemia de Waldenström/patología
3.
Gynecol Oncol ; 156(2): 308-314, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31822399

RESUMEN

BACKGROUND: Selinexor is an oral inhibitor of the nuclear export protein Exportin 1 (XPO1) with demonstrated antitumor activity in solid and hematological malignancies. We evaluated the efficacy and safety of selinexor in heavily pretreated, recurrent gynecological malignancies. METHODS: In this phase 2 trial, patients received selinexor (35 or 50 mg/m2 twice-weekly [BIW] or 50 mg/m2 once-weekly [QW]) in 4-week cycles. Primary endpoint was disease control rate (DCR) including complete response (CR), partial response (PR) or stable disease (SD) ≥12 weeks. Secondary endpoints were progression-free survival (PFS), overall survival (OS) and safety. RESULTS: 114 patients with ovarian (N = 66), endometrial (N = 23) or cervical (N = 25) cancer were enrolled. Median number of prior regimens for ovarian, endometrial and cervical cancer was 6 (1-11), 2 (1-5), and 3 (1-6) respectively. DCR was 30% (ovarian 30%; endometrial 35%; cervical 24%), which included confirmed PRs in 8%, 9%, and 4% of patients with ovarian, endometrial, and cervical cancer respectively. Median PFS and OS for patients with ovarian, endometrial and cervical cancer were 2.6, 2.8 and 1.4 months, and 7.3, 7.0, and 5.0 months, respectively. Common Grade 3/4 adverse events (AEs) were thrombocytopenia (17%), fatigue (14%), anemia (10%), nausea (9%) and hyponatremia (9%). Patients with ovarian cancer receiving 50 mg/m2 QW had fewer high-grade AEs with similar efficacy as BIW treatment. CONCLUSIONS: Selinexor demonstrated single-agent activity and disease control in patients with heavily pretreated ovarian and endometrial cancers. Side effects were a function of dose level and treatment frequency, similar to previous reports, reversible and mitigated with supportive care.


Asunto(s)
Neoplasias de los Genitales Femeninos/tratamiento farmacológico , Hidrazinas/administración & dosificación , Carioferinas/antagonistas & inhibidores , Recurrencia Local de Neoplasia/tratamiento farmacológico , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Triazoles/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Femenino , Neoplasias de los Genitales Femeninos/metabolismo , Neoplasias de los Genitales Femeninos/patología , Humanos , Hidrazinas/efectos adversos , Carioferinas/metabolismo , Persona de Mediana Edad , Clasificación del Tumor , Recurrencia Local de Neoplasia/patología , Supervivencia sin Progresión , Receptores Citoplasmáticos y Nucleares/metabolismo , Triazoles/efectos adversos , Proteína Exportina 1
4.
Blood ; 130(9): 1132-1143, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28630120

RESUMEN

Selinexor is the first oral selective inhibitor of nuclear export compound tested for cancer treatment. Selinexor has demonstrated a safety therapy profile with broad antitumor activity against solid and hematological malignancies in phases 2 and 3 clinical trials (#NCT03071276, #NCT02343042, #NCT02227251, #NCT03110562, and #NCT02606461). Although selinexor shows promising efficacy, its primary adverse effect is high-grade thrombocytopenia. Therefore, we aimed to identify the mechanism of selinexor-induced thrombocytopenia to relieve it and improve its clinical management. We determined that selinexor causes thrombocytopenia by blocking thrombopoietin (TPO) signaling and therefore differentiation of stem cells into megakaryocytes. We then used both in vitro and in vivo models and patient samples to show that selinexor-induced thrombocytopenia is indeed reversible when TPO agonists are administered in the absence of selinexor (drug holiday). In sum, these data reveal (1) the mechanism of selinexor-induced thrombocytopenia, (2) an effective way to reverse the dose-limiting thrombocytopenia, and (3) a novel role for XPO1 in megakaryopoiesis. The improved selinexor dosing regimen described herein is crucial to help reduce thrombocytopenia in selinexor patients, allowing them to continue their course of chemotherapy and have the best chance of survival. This trial was registered at www.clinicaltrials.gov as #NCT01607905.


Asunto(s)
Hidrazinas/efectos adversos , Megacariocitos/metabolismo , Megacariocitos/patología , Transducción de Señal/efectos de los fármacos , Trombocitopenia/inducido químicamente , Trombocitopenia/metabolismo , Trombopoyesis/efectos de los fármacos , Trombopoyetina/metabolismo , Triazoles/efectos adversos , Animales , Apoptosis/efectos de los fármacos , Plaquetas/efectos de los fármacos , Plaquetas/patología , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Feto/patología , Hígado/embriología , Megacariocitos/efectos de los fármacos , Megacariocitos/ultraestructura , Ratones Noqueados , Activación Plaquetaria/efectos de los fármacos , Células Madre/citología , Trombocitopenia/sangre
6.
Eur J Cancer ; 190: 112950, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37441939

RESUMEN

DNA damage response inhibitors have a potentially important therapeutic role in paediatric cancers; however, their optimal use, including patient selection and combination strategy, remains unknown. Moreover, there is an imbalance between the number of drugs with diverse mechanisms of action and the limited number of paediatric patients available to be enrolled in early-phase trials, so prioritisation and a strategy are essential. While PARP inhibitors targeting homologous recombination-deficient tumours have been used primarily in the treatment of adult cancers with BRCA1/2 mutations, BRCA1/2 mutations occur infrequently in childhood tumours, and therefore, a specific response hypothesis is required. Combinations with targeted radiotherapy, ATR inhibitors, or antibody drug conjugates with DNA topoisomerase I inhibitor-related warheads warrant evaluation. Additional monotherapy trials of PARP inhibitors with the same mechanism of action are not recommended. PARP1-specific inhibitors and PARP inhibitors with very good central nervous system penetration also deserve evaluation. ATR, ATM, DNA-PK, CHK1, WEE1, DNA polymerase theta and PKMYT1 inhibitors are early in paediatric development. There should be an overall coordinated strategy for their development. Therefore, an academia/industry consensus of the relevant biomarkers will be established and a focused meeting on ATR inhibitors (as proof of principle) held. CHK1 inhibitors have demonstrated activity in desmoplastic small round cell tumours and have a potential role in the treatment of other paediatric malignancies, such as neuroblastoma and Ewing sarcoma. Access to CHK1 inhibitors for paediatric clinical trials is a high priority. The three key elements in evaluating these inhibitors in children are (1) innovative trial design (design driven by a clear hypothesis with the intent to further investigate responders and non-responders with detailed retrospective molecular analyses to generate a revised or new hypothesis); (2) biomarker selection and (3) rational combination therapy, which is limited by overlapping toxicity. To maximally benefit children with cancer, investigators should work collaboratively to learn the lessons from the past and apply them to future studies. Plans should be based on the relevant biology, with a focus on simultaneous and parallel research in preclinical and clinical settings, and an overall integrated and collaborative strategy.


Asunto(s)
Antineoplásicos , Neuroblastoma , Estados Unidos , Adulto , Humanos , Niño , Adolescente , Antineoplásicos/uso terapéutico , Proteína BRCA1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , United States Food and Drug Administration , Estudios Retrospectivos , Proteína BRCA2 , Neuroblastoma/tratamiento farmacológico , Biomarcadores , Daño del ADN , Proteínas de la Membrana , Proteínas Tirosina Quinasas , Proteínas Serina-Treonina Quinasas
7.
Oncogene ; 40(6): 1176-1190, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33414491

RESUMEN

Ewing sarcoma (ES) is the second most common bone tumor in children and young adults. Unfortunately, there have been minimal recent advancements in improving patient outcomes, especially in metastatic and recurrent diseases. In this study, we investigated the biological role of p21-activated kinases (PAKs) in ES, and the ability to therapeutically target them in high-risk disease. Via informatics analysis, we established the inverse association of PAK1 and PAK4 expression with clinical stage and outcome in ES patients. Through expression knockdown and small-molecule inhibition of PAKs, utilizing FRAX-597, KPT-9274, and PF-3758309 in multiple ES cell lines and patient-derived xenograft models, we further explored the role of PAKs in ES tumor growth and metastatic capabilities. In vitro studies in several ES cell lines indicated that diminishing PAK1 and PAK4 expression reduces tumor cell viability, migratory, and invasive properties. In vivo studies using PAK4 inhibitors, KPT-9274 and PF-3758309 demonstrated significant inhibition of primary and metastatic tumor formation, while transcriptomic analysis of PAK4-inhibitor-treated tumors identified concomitant suppression of Notch, ß-catenin, and hypoxia-mediated signatures. In addition, the analysis showed enrichment of anti-tumor immune regulatory mechanisms, including interferon (IFN)-É£ and IFN-α responses. Altogether, our molecular and pre-clinical studies are the first to establish a critical role for PAKs in ES development and progression, and consequently as viable therapeutic targets for the treatment of high-risk ES in the near future.


Asunto(s)
Sarcoma de Ewing/tratamiento farmacológico , Quinasas p21 Activadas/genética , Acrilamidas/farmacología , Aminopiridinas/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Interferón-alfa/genética , Interferón gamma/genética , Pirazoles/farmacología , Pirroles/farmacología , Sarcoma de Ewing/genética , Sarcoma de Ewing/patología , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasas p21 Activadas/antagonistas & inhibidores
8.
Leukemia ; 34(9): 2430-2440, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32094461

RESUMEN

Selinexor is an oral, small molecule inhibitor of the nuclear export protein exportin 1 with demonstrated activity in hematologic and solid malignancies. Side effects associated with selinexor include nausea, vomiting, fatigue, diarrhea, decreased appetite, weight loss, thrombocytopenia, neutropenia, and hyponatremia. We reviewed 437 patients with multiple myeloma treated with selinexor and assessed the kinetics of adverse events and impact of supportive care measures. Selinexor reduced both platelets and neutrophils over the first cycle of treatment and reached a nadir between 28 and 42 days. Platelet transfusions and thrombopoietin receptor agonists were effective at treating thrombocytopenia, and granulocyte colony stimulating factors were effective at resolving neutropenia. The onset of gastrointestinal side effects (nausea, vomiting, and diarrhea) was most common during the first 1-2 weeks of treatment. Nausea could be mitigated with 5-HT3 antagonists and either neurokinin 1 receptor antagonists, olanzapine, or cannbainoids. Loperamide and bismuth subsalicylate ameliorated diarrhea. The primary constitutional side effects of fatigue and decreased appetite could be managed with methylphenidate, megestrol, cannabinoids or olanzapine, respectively. Hyponatremia was highly responsive to sodium replacement. Selinexor has well-established adverse effects that mainly occur within the first 8 weeks of treatment, are reversible, and respond to supportive care.


Asunto(s)
Antineoplásicos/efectos adversos , Ensayos Clínicos como Asunto , Hidrazinas/efectos adversos , Mieloma Múltiple/tratamiento farmacológico , Triazoles/efectos adversos , Anciano , Antineoplásicos/uso terapéutico , Apetito/efectos de los fármacos , Diarrea/inducido químicamente , Fatiga/inducido químicamente , Fatiga/tratamiento farmacológico , Femenino , Humanos , Hidrazinas/uso terapéutico , Hiponatremia/inducido químicamente , Hiponatremia/terapia , Masculino , Persona de Mediana Edad , Náusea/inducido químicamente , Náusea/tratamiento farmacológico , Trombocitopenia/inducido químicamente , Triazoles/uso terapéutico
9.
Neuroscience ; 142(1): 49-58, 2006 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-16844311

RESUMEN

Human and animal studies have implicated brain-derived neurotrophic factor (BDNF) in the etiology of psychiatric disorders. It is expressed in limbic regions of the brain associated with the regulation of emotionality during fetal development and in the adult animal. To further our understanding of the role of BDNF in the modulation of mood and to distinguish its prenatal and postnatal functions, we investigated and contrasted behavioral changes elicited by its depletion from fetal or postnatal brains of mice. Two corresponding lines of BDNF conditional knockout mice were subjected to a battery of behavioral tests assessing locomotor, depressive, aggressive and anxiety-related behaviors. We found that both lines of mutants were dramatically hyperactive during the light and dark cycles and hyperaggressive. They also exhibited a depression-like phenotype in the tail suspension test but not in the forced swim test. Interestingly, depletion of BDNF from the fetal brain had more pronounced effects on aggressive and depressive-like behaviors and led to deficits in 5-HT(2A) receptor content in the medial frontal cortex, highlighting the importance of this neurotrophin during development. We conclude that expression of BDNF both pre- and postnatally is essential for normal modulation of behavior by neural circuits in the adult animal.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/fisiología , Encéfalo/metabolismo , Trastornos Mentales/genética , Trastornos Mentales/fisiopatología , Agresión/fisiología , Animales , Animales Recién Nacidos , Ansiedad/genética , Conducta Animal , Northern Blotting/métodos , Western Blotting/métodos , Factor Neurotrófico Derivado del Encéfalo/deficiencia , Depresión/genética , Femenino , Expresión Génica/genética , Suspensión Trasera/fisiología , Inmunohistoquímica/métodos , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Noqueados , Actividad Motora/genética , Desempeño Psicomotor/fisiología , Tiempo de Reacción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA