Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Oncol Pharm Pract ; 28(2): 421-424, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33611974

RESUMEN

INTRODUCTION: Immunomodulatory drugs used to treat multiple myeloma carry an increased risk of venous thromboembolic (VTE) disease. Previously published guidelines outline consensus opinion on how to mitigate this risk. METHODS: We collected baseline data to analyze how these strategies are utilized at our single institution and sought to improve the rates of anticoagulation for high-risk patients. This was done through a quality improvement project that added pharmacy/haematology oversight to the VTE risk assessment. RESULTS: Thirty-nine patients newly started on IMiDs were assessed for VTE risk. This information was passed on to the myeloma provider for consideration. Twenty-two patients were classified as high risk for VTE. Of the high-risk patients, 14 (64%) were placed on an anticoagulant for thromboprophylaxis. Eleven (79%) of the 14 used direct oral anticoagulants (DOACs). Eight high-risk patients did not receive an anticoagulant for thromboprophylaxis; 4 of these developed VTE. No patients on anticoagulation developed a VTE. This strategy had rare minor bleeding complications. CONCLUSION: This quality action verifies guideline-based thromboprophylaxis in multiple myeloma and supports the benefit of pharmacy oversight in improving VTE rates. The use of DOACs in myeloma should be further explored.


Asunto(s)
Mieloma Múltiple , Farmacia , Tromboembolia Venosa , Anticoagulantes/uso terapéutico , Humanos , Agentes Inmunomoduladores , Mieloma Múltiple/complicaciones , Mieloma Múltiple/tratamiento farmacológico , Factores de Riesgo , Tromboembolia Venosa/prevención & control
2.
Curr Diab Rep ; 21(12): 62, 2021 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-34902069

RESUMEN

PURPOSE OF REVIEW: There is a bidirectional relationship between cancer and diabetes, with one condition influencing the prognosis of the other. Multiple cancer therapies cause diabetes including well-established medications such as glucocorticoids and novel cancer therapies such as immune checkpoint inhibitors (CPIs) and phosphoinositide 3-kinase (PI3K) inhibitors. RECENT FINDINGS: The nature and severity of diabetes caused by each therapy differ, with some predominantly mediated by insulin resistance, such as PI3K inhibitors and glucocorticoids, while others by insulin deficiency, such as CPIs. Studies have demonstrated diabetes from CPIs to be more rapidly progressing than conventional type 1 diabetes. There remains a scarcity of published guidance for the screening, diagnosis, and management of hyperglycemia and diabetes from these therapies. The need for such guidance is critical because diabetes management in the cancer patient is complex, individualized, and requires inter-disciplinary care. In the present narrative review, we synthesize and summarize the most relevant literature pertaining to diabetes and hyperglycemia in the setting of these cancer therapies and provide an updated patient-centered framework for their evaluation and management.


Asunto(s)
Diabetes Mellitus Tipo 1 , Hiperglucemia , Neoplasias , Diabetes Mellitus Tipo 1/inducido químicamente , Glucocorticoides/efectos adversos , Glucocorticoides/uso terapéutico , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/complicaciones , Neoplasias/epidemiología , Neoplasias/terapia , Atención Dirigida al Paciente , Inhibidores de las Quinasa Fosfoinosítidos-3/efectos adversos , Inhibidores de las Quinasa Fosfoinosítidos-3/uso terapéutico
3.
Am J Hematol ; 94(11): 1244-1253, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31456261

RESUMEN

We tested the hypothesis that using CXCR4 inhibition to target the interaction between the tumor cells and the microenvironment leads to sensitization of the tumor cells to apoptosis. Eligibility criteria included multiple myeloma (MM) patients with 1-5 prior lines of therapy. The purposes of the phase I study were to evaluate the safety and maximal-tolerated dose (MTD) of the combination. The treatment-related adverse events and response rate of the combination were assessed in the phase II study. A total of 58 patients were enrolled in the study. The median age of the patients was 63 years (range, 43-85), and 78% of them received prior bortezomib. In the phase I study, the MTD was plerixafor 0.32 mg/kg, and bortezomib 1.3 mg/m2 . The overall response rate for the phase II study was 48.5%, and the clinical benefit rate 60.6%. The median disease-free survival was 12.6 months. The CyTOF analysis demonstrated significant mobilization of plasma cells, CD34+ stem cells, and immune T cells in response to plerixafor. This is an unprecedented study that examines therapeutic targeting of the bone marrow microenvironment and its interaction with the tumor clone to overcome resistance to therapy. Our results indicate that this novel combination is safe and that the objective response rate is high even in patients with relapsed/refractory MM. ClinicalTrials.gov, NCT00903968.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Mieloma Múltiple/tratamiento farmacológico , Proteínas de Neoplasias/antagonistas & inhibidores , Receptores CXCR4/antagonistas & inhibidores , Terapia Recuperativa , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Bencilaminas , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Bortezomib/administración & dosificación , Bortezomib/efectos adversos , Terapia Combinada , Ciclamas , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Enfermedades Gastrointestinales/inducido químicamente , Enfermedades Hematológicas/inducido químicamente , Trasplante de Células Madre Hematopoyéticas , Compuestos Heterocíclicos/administración & dosificación , Compuestos Heterocíclicos/efectos adversos , Humanos , Estimación de Kaplan-Meier , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Mieloma Múltiple/genética , Mieloma Múltiple/terapia , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/efectos de los fármacos , Recurrencia , Microambiente Tumoral/efectos de los fármacos
4.
Am J Hematol ; 90(2): 100-4, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25349134

RESUMEN

Disease assessment in Waldenstrom Macroglobulinemia (WM) is dependent on the percent involvement of B-cell neoplasm in the bone marrow and IgM paraprotein in the serum. A subset of patients also demonstrates extramedullary involvement, which is infrequently examined. The role of extramedullary involvement in the diagnosis and prognosis of WM is poorly understood. The purpose of this study is to report the characteristics of WM patients with extramedullary disease (EMD). Nine hundred and eight-five patients with WM were evaluated at one academic center and the presence of EMD was assessed in these patients. Forty-three (4.4%) patients were identified to have EMD. Nine (21%) patients presented with involvement at WM diagnosis, while 34 (79%) developed EMD post-therapy for WM. Most frequent EMD sites involved were pulmonary (30%), soft tissue (21%), cerebrospinal fluid (23%), renal (8%), and bone (9%). The median overall survival at 10 years was 79% (95% CI: 57-90%). This is the first study to describe the clinical characteristics, response and overall survival in patients with extramedullary WM. Further studies to define the molecular characteristics of this entity and mechanisms of its development are warranted.


Asunto(s)
Linfocitos B/patología , Médula Ósea/patología , Huesos/patología , Riñón/patología , Pulmón/patología , Ganglios Linfáticos/patología , Macroglobulinemia de Waldenström/patología , Adulto , Anciano , Antineoplásicos/uso terapéutico , Linfocitos B/inmunología , Médula Ósea/inmunología , Huesos/inmunología , Femenino , Humanos , Inmunoglobulina M/sangre , Riñón/inmunología , Pulmón/inmunología , Ganglios Linfáticos/inmunología , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Análisis de Supervivencia , Macroglobulinemia de Waldenström/tratamiento farmacológico , Macroglobulinemia de Waldenström/inmunología , Macroglobulinemia de Waldenström/mortalidad
5.
Biol Blood Marrow Transplant ; 20(2): 285-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24269706

RESUMEN

Hematopoietic stem cell transplantation (HSCT) recipients are at risk for varicella-zoster virus (VZV) reactivation. Vaccination may help restore VZV immunity; however, the available live attenuated VZV vaccine (Zostavax) is contraindicated in immunocompromised hosts. We report our experience with using a single dose of VZV vaccine in 110 adult autologous and allogeneic HSCT recipients who were about 2 years after transplantation, free of graft-versus-host disease, and not receiving immunosuppression. One hundred eight vaccine recipients (98.2%) had no clinically apparent adverse events with a median follow-up period of 9.5 months (interquartile range, 6 to 16; range, 2 to 28). Two vaccine recipients (1.8%) developed a skin rash (one zoster-like rash with associated pain, one varicella-like) within 42 days post-vaccination that resolved with antiviral therapy. We could not confirm if these rashes were due to vaccine (Oka) or wild-type VZV. No other possible cases of VZV reactivation have occurred with about 1178 months of follow-up. Live attenuated zoster vaccine appears generally safe in this population when vaccinated as noted; the overall vaccination risk needs to be weighed against the risk of wild-type VZV disease in this high-risk population.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Vacuna contra el Herpes Zóster/uso terapéutico , Vacunas Atenuadas/uso terapéutico , Adulto , Anciano , Femenino , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Vacuna contra el Herpes Zóster/administración & dosificación , Humanos , Masculino , Persona de Mediana Edad , Vacunas Atenuadas/administración & dosificación , Adulto Joven
7.
JCO Oncol Pract ; 19(2): e298-e305, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36409966

RESUMEN

PURPOSE: More oncologists desire to treat their patients with immune checkpoint inhibitors (ICIs) in the inpatient setting as their use has become more widespread for numerous oncologic indications. This is cost-prohibitive to patients and institutions because of high drug cost and lack of reimbursement in the inpatient setting. We sought to examine current practice of inpatient ICI administration to determine if and in which clinical scenarios it may provide significant clinical benefit and therefore be warranted regardless of cost. METHODS: We conducted a retrospective chart review of adult patients who received at least one dose of an ICI for treatment of an active solid tumor malignancy during hospitalization at a single academic medical center between January 2017 and June 2018. Patient, disease, and admission characteristics including mortality data were examined, and cost analysis was performed. RESULTS: Sixty-five doses of ICIs were administered to 58 patients during the study period. Nearly 40% and 80% of patients died within 30 days and 180 days of ICI administration, respectively. There was a trend toward longer overall survival in patients with good prognostic factors including positive programmed death-ligand 1 (PD-L1) expression or microsatellite instability-high (MSI-H) status. Slightly over 70% of patients were discharged within 7 days of ICI administration. The total cost of inpatient ICI administration over the 18-month study period was $615,016 US dollars. CONCLUSION: Inpatient ICI administration is associated with high costs and poor outcomes in acutely ill hospitalized patients with advanced solid tumor malignancies and therefore should largely be avoided. Careful discharge planning to expedite outpatient treatment after discharge will be paramount in ensuring patients with good prognostic features who will benefit most from ICI therapy can be promptly treated in the outpatient setting as treating very close to discharge in the inpatient setting appears to be unnecessary, regardless of tumor features.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Neoplasias , Adulto , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Estudios Retrospectivos , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Pronóstico , Hospitalización
8.
Blood Cancer J ; 13(1): 31, 2023 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-36849497

RESUMEN

Thrombotic microangiopathy (TMA) has been reported to occur in multiple myeloma (MM) patients in association with treatment with carfilzomib, an irreversible proteasome inhibitor (PI). The hallmark of TMA is vascular endothelial damage leading to microangiopathic hemolytic anemia, platelet consumption, fibrin deposition and small-vessel thrombosis with resultant tissue ischemia. The molecular mechanisms underlying carfilzomib-associated TMA are not known. Germline mutations in the complement alternative pathway have been recently shown to portend increased risk for the development of atypical hemolytic uremic syndrome (aHUS) and TMA in the setting of allogeneic stem cell transplant in pediatric patients. We hypothesized that germline mutations in the complement alternative pathway may similarly predispose MM patients to carfilzomib-associated TMA. We identified 10 MM patients with a clinical diagnosis of TMA in the context of carfilzomib treatment and assessed for the presence of germline mutations in the complement alternative pathway. Ten, matched MM patients exposed to carfilzomib but without clinical TMA were used as negative controls. We identified a frequency of deletions in the complement Factor H genes 3 and 1 (delCFHR3-CFHR1) and genes 1 and 4 (delCFHR1-CFHR4) in MM patients with carfilzomib-associated TMA that was higher as compared to the general population and matched controls. Our data suggest that complement alternative pathway dysregulation may confer susceptibility to vascular endothelial injury in MM patients and predispose to development of carfilzomib-associated TMA. Larger, retrospective studies are needed to evaluate whether screening for complement mutations may be indicated to properly counsel patients about TMA risk with carfilzomib use.


Asunto(s)
Mieloma Múltiple , Microangiopatías Trombóticas , Humanos , Niño , Vía Alternativa del Complemento , Mieloma Múltiple/complicaciones , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mutación , Microangiopatías Trombóticas/inducido químicamente , Microangiopatías Trombóticas/genética
9.
Curr Opin Hematol ; 18(4): 260-5, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21519243

RESUMEN

PURPOSE OF REVIEW: Waldenstrom macroglobulinemia is a distinct low-grade lymphoproliferative disease. There have been recent significant advances in understanding the underlying pathogenesis of this disease, including genetic and epigenetic regulators of tumor progression. RECENT FINDINGS: Current studies have shown that the tumor microenvironment plays a critical role in cell proliferation, dissemination, and drug resistance. SUMMARY: This review provides an update of the advances in the pathogenesis of factors both intrinsic (in the tumor clone) and extrinsic (in the bone marrow microenvironment) that regulate tumor progression in Waldenstrom macroglobulinemia. We next discuss novel agents that have been recently tested in clinical trials based on the advances observed in the pathogenesis of Waldenstrom macroglobulinemia.


Asunto(s)
Macroglobulinemia de Waldenström/etiología , Macroglobulinemia de Waldenström/terapia , Epigenómica , Humanos , Macroglobulinemia de Waldenström/genética , Macroglobulinemia de Waldenström/patología
10.
Lancet Haematol ; 9(5): e340-e349, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35344689

RESUMEN

BACKGROUND: Prevalence estimates for monoclonal gammopathy of undetermined significance (MGUS) are based on predominantly White study populations screened by serum protein electrophoresis supplemented with immunofixation electrophoresis. A prevalence of 3% is reported for MGUS in the general population of European ancestry aged 50 years or older. MGUS prevalence is two times higher in individuals of African descent or with a family history of conditions related to multiple myeloma. We aimed to evaluate the prevalence and clinical implications of monoclonal gammopathies in a high-risk US population screened by quantitative mass spectrometry. METHODS: We used quantitative matrix-assisted laser desorption ionisation-time of flight (MALDI-TOF) mass spectrometry and EXENT-iQ software to screen for and quantify monoclonal gammopathies in serum from 7622 individuals who consented to the PROMISE screening study between Feb 26, 2019, and Nov 4, 2021, and the Mass General Brigham Biobank (MGBB) between July 28, 2010, and July 1, 2021. M-protein concentrations at the monoclonal gammopathy of indeterminate potential (MGIP) level were confirmed by liquid chromatography mass spectrometry testing. 6305 (83%; 2211 from PROMISE, 4094 from MGBB) of 7622 participants in the cohorts were at high risk for developing a monoclonal gammopathy on the basis of Black race or a family history of haematological malignancies and fell within the eligible high-risk age range (30 years or older for PROMISE cohort and 18 years or older for MGBB cohort); those over 18 years were also eligible if they had two or more family members with a blood cancer (PROMISE cohort). Participants with a plasma cell malignancy diagnosed before screening were excluded. Longitudinal clinical data were available for MGBB participants with a median follow-up time from serum sample screening of 4·5 years (IQR 2·4-6·7). The PROMISE study is registered with ClinicalTrials.gov, NCT03689595. FINDINGS: The median age at time of screening was 56·0 years (IQR 46·8-64·1). 5013 (66%) of 7622 participants were female, 2570 (34%) male, and 39 (<1%) unknown. 2439 (32%) self-identified as Black, 4986 (65%) as White, 119 (2%) as other, and 78 (1%) unknown. Using serum protein electrophoresis with immunofixation electrophoresis, the MGUS prevalence was 6% (101 of 1714) in high-risk individuals aged 50 years or older. Using mass spectrometry, we observed a total prevalence of monoclonal gammopathies of 43% (1788 of 4207) in this group. We termed monoclonal gammopathies below the clinical immunofixation electrophoresis detection level (<0·2 g/L) MGIPs, to differentiate them from those with higher concentrations, termed mass-spectrometry MGUS, which had a 13% (592 of 4207) prevalence by mass spectrometry in high-risk individuals aged 50 years or older. MGIP was predominantly of immunoglobulin M isotype, and its prevalence increased with age (19% [488 of 2564] for individuals aged <50 years, 29% [1464 of 5058] for those aged ≥50 years, and 37% [347 of 946] for those aged ≥70 years). Mass-spectrometry MGUS prevalence increased with age (5% [127 of 2564] for individuals aged <50 years, 13% [678 of 5058] for those aged ≥50 years, and 18% [173 of 946] for those aged ≥70 years) and was higher in men (314 [12%] of 2570) compared with women (485 [10%] 5013; p=0·0002), whereas MGIP prevalence did not differ significantly by gender. In those aged 50 years or older, the prevalence of mass spectrometry was significantly higher in Black participants (224 [17%] of 1356) compared with the controls (p=0·0012) but not in those with family history (368 [13%] of 2851) compared with the controls (p=0·1008). Screen-detected monoclonal gammopathies correlated with increased all-cause mortality in MGBB participants (hazard ratio 1·55, 95% CI 1·16-2·08; p=0·0035). All monoclonal gammopathies were associated with an increased likelihood of comorbidities, including myocardial infarction (odds ratio 1·60, 95% CI 1·26-2·02; p=0·00016 for MGIP-high and 1·39, 1·07-1·80; p=0·015 for mass-spectrometry MGUS). INTERPRETATION: We detected a high prevalence of monoclonal gammopathies, including age-associated MGIP, and made more precise estimates of mass-spectrometry MGUS compared with conventional gel-based methods. The use of mass spectrometry also highlighted the potential hidden clinical significance of MGIP. Our study suggests the association of monoclonal gammopathies with a variety of clinical phenotypes and decreased overall survival. FUNDING: Stand Up To Cancer Dream Team, the Multiple Myeloma Research Foundation, and National Institutes of Health.


Asunto(s)
Gammopatía Monoclonal de Relevancia Indeterminada , Mieloma Múltiple , Paraproteinemias , Estudios de Cohortes , Femenino , Humanos , Masculino , Espectrometría de Masas , Gammopatía Monoclonal de Relevancia Indeterminada/epidemiología , Mieloma Múltiple/epidemiología , Paraproteinemias/diagnóstico , Paraproteinemias/epidemiología , Prevalencia
11.
Leuk Res ; 108: 106610, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34048999

RESUMEN

Gilteritinib is primarily metabolized via cytochrome P450 (CYP). Therefore, concomitant administration of strong CYP3A4 inducers or inhibitors is not recommended. We evaluated the incidence of gilteritinib-related adverse events (AEs) in 47 patients who received gilteritinib with or without antifungal triazoles which are known inhibitors of CYP3A4. Reasons for coadministration were antifungal prophylaxis or treatment of suspected or confirmed fungal diseases. Gilteritinib-related AEs were similar in the gilteritinib-triazole group compared to the gilteritinib without triazole group (75 % vs. 55.5 %, P = 0.23). Additionally, severity of AEs, gilteritinib dose reductions (15 % vs. 14.8 %) or discontinuation due to AEs (10 % vs. 22.2 %), and 90-day mortality (35 % vs. 11.1 %) were similar in both groups. Thus, concomitant gilteritinib and triazole therapy is feasible and is not associated with clinically meaningful increase in gilteritinib-related AEs.


Asunto(s)
Compuestos de Anilina/efectos adversos , Antifúngicos/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Micosis/tratamiento farmacológico , Pirazinas/efectos adversos , Triazoles/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Compuestos de Anilina/administración & dosificación , Femenino , Estudios de Seguimiento , Humanos , Leucemia Mieloide Aguda/patología , Masculino , Persona de Mediana Edad , Micosis/inducido químicamente , Micosis/patología , Pronóstico , Pirazinas/administración & dosificación , Estudios Retrospectivos , Tasa de Supervivencia , Adulto Joven
13.
Bone Marrow Transplant ; 53(7): 942-945, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29426830

RESUMEN

Attenuated live virus vaccinations are generally recommended 24 months following hematopoietic cell transplantation (HCT) in patients not receiving immunosuppressive therapy. To date, there are no data regarding the safety of live-attenuated herpes zoster or measles-mumps-rubella (MMR) vaccinations in multiple myeloma patients on maintenance lenalidomide or bortezomib following autologous HCT. One hundred thirty-seven multiple myeloma patients on maintenance lenalidomide or bortezomib post-auto-HCT who received either MMR or herpes zoster vaccine were analyzed and any adverse events documented in the medical record in the 42 days following vaccination were recorded. Patients were vaccinated a median of 25 months (range, 18-62) post transplant. The most common post-vaccination adverse event was upper respiratory tract infection (18/137 patients); no rash attributed to vaccine strains or other adverse outcomes potentially related to the vaccines were identified. MMR and herpes zoster vaccination were safe and well-tolerated in this cohort.


Asunto(s)
Bortezomib/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/métodos , Lenalidomida/uso terapéutico , Mieloma Múltiple/complicaciones , Acondicionamiento Pretrasplante/métodos , Trasplante Autólogo/métodos , Adulto , Anciano , Bortezomib/farmacología , Femenino , Vacuna contra el Herpes Zóster , Humanos , Lenalidomida/farmacología , Masculino , Sarampión , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Paperas , Rubéola (Sarampión Alemán)
14.
Expert Rev Hematol ; 7(1): 157-68, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24405328

RESUMEN

Waldenström macroglobulinemia (WM) is distinct B-cell lymphoproliferative disorder primarily characterized by bone marrow infiltration of lymphoplasmacytic cells along with production of a serum monoclonal (IgM). In this review, we describe the biology of WM, the diagnostic evaluation for WM with a discussion of other conditions that are in the differential diagnosis and clinical manifestations of the disease as well as current treatment options. Within the novel agents discussed are everolimus, perifosine, enzastaurin, panobinostat, bortezomib and carfilzomib, pomalidomide and ibrutinib. Many of the novel agents have shown good responses and have a better toxicity profile compared to traditional chemotherapeutic agents, which makes them good candidates to be used as primary therapies for WM in the future.


Asunto(s)
Macroglobulinemia de Waldenström/metabolismo , Anticuerpos Monoclonales de Origen Murino/uso terapéutico , Aberraciones Cromosómicas , Perfilación de la Expresión Génica , Humanos , Inmunoglobulina M/sangre , Factores Inmunológicos/uso terapéutico , Inhibidores de Proteasoma/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteómica , Rituximab , Macroglobulinemia de Waldenström/patología , Macroglobulinemia de Waldenström/terapia
15.
Clin Lymphoma Myeloma Leuk ; 13 Suppl 2: S310-6, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24290218

RESUMEN

Waldenström macroglobulinemia (WM), first described by Jan Waldenström in 1944, is a lymphoplasmacytic lymphoma characterized by the presence of an immunoglobulin M monoclonal gammopathy in the blood and monoclonal small lymphocytes and lymphoplasmacytoid cells in the bone marrow. WM is a rare and indolent disease but remains incurable. In this review we discuss the pathogenesis of WM and focus on novel treatment options that target pathways deregulated in this disease. Recent studies have helped us identify specific genetic mutations that are commonly seen in WM and might prove to be important therapeutic targets in the future. We discuss the role of epigenetics and the changes in the bone marrow microenvironment that are important in the pathogenesis of WM. The commonly used drugs are discussed with a focus on novel agents that are currently being used as single agents or in combination to treat WM. We finally focus on some agents that have shown preclinical efficacy and might be available in the near future.


Asunto(s)
Macroglobulinemia de Waldenström/tratamiento farmacológico , Animales , Humanos , Inhibidores de Proteasoma/uso terapéutico , Macroglobulinemia de Waldenström/etiología , Macroglobulinemia de Waldenström/genética , Macroglobulinemia de Waldenström/patología
16.
Leuk Res ; 37(7): 747-51, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23643326

RESUMEN

All-trans retinoic acid (ATRA) used for the treatment of APL can lead to the development of differentiation syndrome (DS), a potentially life threatening complication. Since ATRA is metabolized by cytochrome P450 (CYP) enzymes, we sought to identify drug interactions that might be associated with a higher risk for the development of DS in addition to other predictive factors related to the incidence of DS. We identified 60 consecutive patients with APL treated at our institution with ATRA from May 2004 until January 2010. Of the 60 patients identified, 29 (48%) developed DS within a median of 5 days (range 1-31) of ATRA initiation. We did not find any difference in overall incidence of DS whether patients were on concurrent CYP 2C8, 2C9 or 3A4 inhibitors, inducers or substrates. In multivariable analysis, higher peripheral blood blast counts on admission (p=0.04) as well as higher body mass index (p=0.003) were associated with developing DS. Out of the 29 patients with DS, there were 4 early deaths of which 2 were attributed to DS compared to no early deaths in the patients who did not develop DS (p=0.05). Regarding disease-related outcomes, only CR rate was different between patients developing DS versus those who did not develop DS.


Asunto(s)
Antineoplásicos/efectos adversos , Diferenciación Celular/efectos de los fármacos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Recurrencia Local de Neoplasia/diagnóstico , Tretinoina/efectos adversos , Adolescente , Adulto , Anciano , Índice de Masa Corporal , Femenino , Estudios de Seguimiento , Humanos , Leucemia Promielocítica Aguda/complicaciones , Leucemia Promielocítica Aguda/mortalidad , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/mortalidad , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia , Síndrome , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA