Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Med ; 17(11): e1003323, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33147277

RESUMEN

BACKGROUND: The tumor microenvironment (TME) is increasingly appreciated as an important determinant of cancer outcome, including in multiple myeloma (MM). However, most myeloma microenvironment studies have been based on bone marrow (BM) aspirates, which often do not fully reflect the cellular content of BM tissue itself. To address this limitation in myeloma research, we systematically characterized the whole bone marrow (WBM) microenvironment during premalignant, baseline, on treatment, and post-treatment phases. METHODS AND FINDINGS: Between 2004 and 2019, 998 BM samples were taken from 436 patients with newly diagnosed MM (NDMM) at the University of Arkansas for Medical Sciences in Little Rock, Arkansas, United States of America. These patients were 61% male and 39% female, 89% White, 8% Black, and 3% other/refused, with a mean age of 58 years. Using WBM and matched cluster of differentiation (CD)138-selected tumor gene expression to control for tumor burden, we identified a subgroup of patients with an adverse TME associated with 17 fewer months of progression-free survival (PFS) (95% confidence interval [CI] 5-29, 49-69 versus 70-82 months, χ2 p = 0.001) and 15 fewer months of overall survival (OS; 95% CI -1 to 31, 92-120 versus 113-129 months, χ2 p = 0.036). Using immunohistochemistry-validated computational tools that identify distinct cell types from bulk gene expression, we showed that the adverse outcome was correlated with elevated CD8+ T cell and reduced granulocytic cell proportions. This microenvironment develops during the progression of premalignant to malignant disease and becomes less prevalent after therapy, in which it is associated with improved outcomes. In patients with quantified International Staging System (ISS) stage and 70-gene Prognostic Risk Score (GEP-70) scores, taking the microenvironment into consideration would have identified an additional 40 out of 290 patients (14%, premutation p = 0.001) with significantly worse outcomes (PFS, 95% CI 6-36, 49-73 versus 74-90 months) who were not identified by existing clinical (ISS stage III) and tumor (GEP-70) criteria as high risk. The main limitations of this study are that it relies on computationally identified cell types and that patients were treated with thalidomide rather than current therapies. CONCLUSIONS: In this study, we observe that granulocyte signatures in the MM TME contribute to a more accurate prognosis. This implies that future researchers and clinicians treating patients should quantify TME components, in particular monocytes and granulocytes, which are often ignored in microenvironment studies.


Asunto(s)
Médula Ósea/patología , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/patología , Microambiente Tumoral , Adulto , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Pronóstico , Carga Tumoral
2.
Blood ; 132(6): 587-597, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-29884741

RESUMEN

Understanding the profile of oncogene and tumor suppressor gene mutations with their interactions and impact on the prognosis of multiple myeloma (MM) can improve the definition of disease subsets and identify pathways important in disease pathobiology. Using integrated genomics of 1273 newly diagnosed patients with MM, we identified 63 driver genes, some of which are novel, including IDH1, IDH2, HUWE1, KLHL6, and PTPN11 Oncogene mutations are significantly more clonal than tumor suppressor mutations, indicating they may exert a bigger selective pressure. Patients with more driver gene abnormalities are associated with worse outcomes, as are identified mechanisms of genomic instability. Oncogenic dependencies were identified between mutations in driver genes, common regions of copy number change, and primary translocation and hyperdiploidy events. These dependencies included associations with t(4;14) and mutations in FGFR3, DIS3, and PRKD2; t(11;14) with mutations in CCND1 and IRF4; t(14;16) with mutations in MAF, BRAF, DIS3, and ATM; and hyperdiploidy with gain 11q, mutations in FAM46C, and MYC rearrangements. These associations indicate that the genomic landscape of myeloma is predetermined by the primary events upon which further dependencies are built, giving rise to a nonrandom accumulation of genetic hits. Understanding these dependencies may elucidate potential evolutionary patterns and lead to better treatment regimens.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Mieloma Múltiple/genética , Mutagénesis , Oncogenes , Células Clonales , Análisis Mutacional de ADN , ADN de Neoplasias/genética , Conjuntos de Datos como Asunto , Dosificación de Gen , Estudio de Asociación del Genoma Completo , Inestabilidad Genómica , Genómica , Humanos , Pérdida de Heterocigocidad , Mieloma Múltiple/patología , Mutación , Pronóstico , Translocación Genética , Resultado del Tratamiento , Secuenciación del Exoma
3.
Haematologica ; 105(4): 1055-1066, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31221783

RESUMEN

MYC is a widely acting transcription factor and its deregulation is a crucial event in many human cancers. MYC is important biologically and clinically in multiple myeloma, but the mechanisms underlying its dysregulation are poorly understood. We show that MYC rearrangements are present in 36.0% of newly diagnosed myeloma patients, as detected in the largest set of next generation sequencing data to date (n=1,267). Rearrangements were complex and associated with increased expression of MYC and PVT1, but not other genes at 8q24. The highest effect on gene expression was detected in cases where the MYC locus is juxtaposed next to super-enhancers associated with genes such as IGH, IGK, IGL, TXNDC5/BMP6, FAM46C and FOXO3 We identified three hotspots of recombination at 8q24, one of which is enriched for IGH-MYC translocations. Breakpoint analysis indicates primary myeloma rearrangements involving the IGH locus occur through non-homologous end joining, whereas secondary MYC rearrangements occur through microhomology-mediated end joining. This mechanism is different to lymphomas, where non-homologous end joining generates MYC rearrangements. Rearrangements resulted in overexpression of key genes and chromatin immunoprecipitation-sequencing identified that HK2, a member of the glucose metabolism pathway, is directly over-expressed through binding of MYC at its promoter.


Asunto(s)
Genes myc , Mieloma Múltiple , ARN Largo no Codificante/genética , Genes de las Cadenas Pesadas de las Inmunoglobulinas , Humanos , Hibridación Fluorescente in Situ , Mieloma Múltiple/genética , Proteína Disulfuro Isomerasas , Translocación Genética
4.
Br J Haematol ; 184(4): 578-593, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30408155

RESUMEN

Recent studies suggest that multiple myeloma (MM) induces proliferation and expansion of bone marrow (BM) mesenchymal stem cells (MSCs), but others showed that MM cells induce MSC senescence. To clarify the interaction between MM and MSCs, we exploited our established MSC gene signature to identify gene expression changes in myeloma MSCs and associated functional differences. Single MSCs from patients with MM had changes in expression of genes associated with cellular proliferation and senescence and a higher proportion of senescent cells and lower proliferative potential than those from age-matched healthy donors. Single MSCs from both sources heterogeneously express MSC genes associated with adipogenesis and osteoblastogenesis. We identified the gene encoding insulin-like growth factor-binding protein 2 (IGFBP2), an MSC gene commonly altered in high risk MM, as under-expressed. Morphologically, IGFBP2+ cells are underrepresented in MM BM compared to smouldering MM. Strong IGFBP2 and adiponectin co-expression was detected in a subset of small adipocytes. Co-culturing normal MSCs with myeloma cells suppressed MSC differentiation to adipocytes and osteoblasts, and reduced expression of IGFBP2 and adiponectin. Recombinant IGFBP2 blocked IGF1-mediated myeloma cell growth. Our data demonstrate that myeloma MSCs are less proliferative and that IGFBP2+ small adipocytes are a distinct mesenchymal cell population suppressed by myeloma.


Asunto(s)
Adipocitos , Médula Ósea , Regulación Neoplásica de la Expresión Génica , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/biosíntesis , Células Madre Mesenquimatosas , Mieloma Múltiple , Proteínas de Neoplasias/biosíntesis , Adipocitos/metabolismo , Adipocitos/patología , Médula Ósea/metabolismo , Médula Ósea/patología , Diferenciación Celular , Femenino , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Factores de Riesgo
5.
Lancet ; 389(10068): 519-527, 2017 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-28017406

RESUMEN

BACKGROUND: Lenalidomide plus dexamethasone is a reference treatment for patients with newly diagnosed myeloma. The combination of the proteasome inhibitor bortezomib with lenalidomide and dexamethasone has shown significant efficacy in the setting of newly diagnosed myeloma. We aimed to study whether the addition of bortezomib to lenalidomide and dexamethasone would improve progression-free survival and provide better response rates in patients with previously untreated multiple myeloma who were not planned for immediate autologous stem-cell transplant. METHODS: In this randomised, open-label, phase 3 trial, we recruited patients with newly diagnosed multiple myeloma aged 18 years and older from participating Southwest Oncology Group (SWOG) and National Clinical Trial Network (NCTN) institutions (both inpatient and outpatient settings). Key inclusion criteria were presence of CRAB (C=calcium elevation; R=renal impairment; A=anaemia; B=bone involvement) criteria with measurable disease (measured by assessment of free light chains), Eastern Cooperative Oncology Group (ECOG) performance status of 0-3, haemoglobin concentration 9 g/dL or higher, absolute neutrophil count 1 × 103 cells per mm3 or higher, and a platelet count of 80 000/mm3 or higher. We randomly assigned (1:1) patients to receive either an initial treatment of bortezomib with lenalidomide and dexamethasone (VRd group) or lenalidomide and dexamethasone alone (Rd group). Randomisation was stratified based on International Staging System stage (I, II, or III) and intent to transplant (yes vs no). The VRd regimen was given as eight 21-day cycles. Bortezomib was given at 1·3 mg/m2 intravenously on days 1, 4, 8, and 11, combined with oral lenalidomide 25 mg daily on days 1-14 plus oral dexamethasone 20 mg daily on days 1, 2, 4, 5, 8, 9, 11, and 12. The Rd regimen was given as six 28-day cycles. The standard Rd regimen consisted of 25 mg oral lenalidomide once a day for days 1-21 plus 40 mg oral dexamethasone once a day on days 1, 8, 15, and 22. The primary endpoint was progression-free survival using a prespecified one-sided stratified log rank test at a significance level of 0·02. Analyses were intention to treat. This trial is registered with ClinicalTrials.gov, number NCT00644228. FINDINGS: Between April, 2008, and February, 2012, we randomly assigned 525 patients at 139 participating institutions (264 to VRd and 261 to Rd). In the randomly assigned patients, 21 patients in the VRd group and 31 in the Rd group were deemed ineligible based mainly on missing, insufficient, or early or late baseline laboratory data. Median progression-free survival was significantly improved in the VRd group (43 months vs 30 months in the Rd group; stratified hazard ratio [HR] 0·712, 96% CI 0·56-0·906; one-sided p value 0·0018). The median overall survival was also significantly improved in the VRd group (75 months vs 64 months in the Rd group, HR 0·709, 95% CI 0·524-0·959; two-sided p value 0·025). The rates of overall response (partial response or better) were 82% (176/216) in the VRd group and 72% (153/214) in the Rd group, and 16% (34/216) and 8% (18/214) of patients who were assessable for response in these respective groups had a complete response or better. Adverse events of grade 3 or higher were reported in 198 (82%) of 241 patients in the VRd group and 169 (75%) of 226 patients in the Rd group; 55 (23%) and 22 (10%) patients discontinued induction treatment because of adverse events, respectively. There were no treatment-related deaths in the Rd group, and two in the VRd group. INTERPRETATION: In patients with newly diagnosed myeloma, the addition of bortezomib to lenalidomide and dexamethasone resulted in significantly improved progression-free and overall survival and had an acceptable risk-benefit profile. FUNDING: NIH, NCI, NCTN, Millennium Pharmaceuticals, Takeda Oncology Company, and Celgene Corporation.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/uso terapéutico , Bortezomib/uso terapéutico , Dexametasona/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Talidomida/análogos & derivados , Adulto , Anciano , Supervivencia sin Enfermedad , Quimioterapia Combinada , Femenino , Trasplante de Células Madre Hematopoyéticas , Humanos , Lenalidomida , Masculino , Persona de Mediana Edad , Mieloma Múltiple/mortalidad , Tasa de Supervivencia , Talidomida/uso terapéutico , Resultado del Tratamiento
6.
Haematologica ; 103(6): 1047-1053, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29567784

RESUMEN

Fluorine-18 fluorodeoxyglucose positron emission tomography with computed tomography attenuation correction (PET-CT) in myeloma can detect and enumerate focal lesions by the quantitative characterization of metabolic activity. The aim of this study was to determine the prognostic significance of the suppression of PET-CT activity at a number of time points post therapy initiation: day 7, post induction, post transplant, and at maintenance therapy. As part of the TT4-6 trial series, 596 patients underwent baseline PET-CT and were evaluated serially during their disease course using peak standardized uptake values above background red marrow signal. We demonstrate that the presence of more than 3 focal lesions at presentation identifies a group of patients with an adverse progression-free survival and overall survival. At day 7 of therapy, patients with complete focal lesion signal suppression revert to the same prognosis as those with no lesions at diagnosis. At later time points, the continued suppression of signal remains prognostically important. We conclude that for newly diagnosed patients with focal lesions, treatment until these lesions are suppressed is an important therapeutic goal as the prognosis of these patients is the same as those without lesions at diagnosis. (clinicaltrials.gov identifiers: 00734877, 02128230, 00869232, 00871013).


Asunto(s)
Mieloma Múltiple/diagnóstico , Tomografía Computarizada por Tomografía de Emisión de Positrones , Anciano , Anciano de 80 o más Años , Terapia Combinada , Femenino , Fluorodesoxiglucosa F18 , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/mortalidad , Mieloma Múltiple/terapia , Neoplasia Residual/diagnóstico , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Pronóstico , Análisis de Supervivencia , Resultado del Tratamiento
7.
Future Oncol ; 13(13): 1181-1193, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28395525

RESUMEN

Unlike conventional cancer treatment, immuno-oncology therapies are commonly associated with delayed clinical benefit and durable responses, as seen with immuno-oncology therapies for multiple myeloma (MM). Therefore, a longer-term approach to immuno-oncology data assessment is required. Appropriate study designs, end points and statistical methods are essential for evaluating immuno-oncology therapies to assess treatment outcomes, and may better accommodate immuno-oncology clinical trial data. In addition to conventional end points including median progression-free survival (PFS) and overall survival (OS), end points such as hazard ratios for PFS and OS over time, PFS and OS landmark analyses beyond the median, and immune-response end points might provide better indications of the efficacy of immuno-oncology therapies. Long-term data with these agents will allow better prediction of outcomes in MM.


Asunto(s)
Determinación de Punto Final , Inmunoterapia , Mieloma Múltiple/inmunología , Mieloma Múltiple/terapia , Ensayos Clínicos como Asunto , Supervivencia sin Enfermedad , Humanos , Oncología Médica/tendencias , Mieloma Múltiple/epidemiología , Resultado del Tratamiento
9.
Blood ; 123(1): 78-85, 2014 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-24144643

RESUMEN

All cases of clinical myeloma (CMM) are preceded by an asymptomatic monoclonal gammopathy (AMG), classified as either monoclonal gammopathy of undetermined significance (MGUS) or asymptomatic multiple myeloma (AMM). We analyzed data from AMG patients (n = 331) enrolled in a prospective, observational clinical trial (S0120). Baseline data from clinical variables, gene expression profiles (GEP) of purified tumor cells, and findings of magnetic resonance imaging (MRI) were correlated with the risk of progression to CMM requiring therapy. GEP of purified tumor cells revealed that all molecular subtypes of CMM are also represented in the AMG phase. An increased risk score (>-0.26) (based on a 70-gene signature, GEP70) was an independent predictor of the risk of progression to CMM. Combination of elevated serum free light chain, M-spike, and GEP70 risk score identified a subset with high risk (67% at 2 years) of progression to CMM requiring therapy. Importantly, absence of these factors in AMM patients predicted low risk similar to MGUS. Detection of multiple (>1) focal lesions by MRI also conferred an increased risk of progression. These data demonstrate that signatures associated with high-risk CMM impact disease risk and support inclusion of genomic analysis in the clinical management of AMGs.


Asunto(s)
Gammopatía Monoclonal de Relevancia Indeterminada/diagnóstico , Gammopatía Monoclonal de Relevancia Indeterminada/genética , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/genética , Anciano , Estudios de Cohortes , Progresión de la Enfermedad , Femenino , Humanos , Cadenas Ligeras de Inmunoglobulina/sangre , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Gammopatía Monoclonal de Relevancia Indeterminada/terapia , Mieloma Múltiple/terapia , Lesiones Precancerosas/diagnóstico , Lesiones Precancerosas/genética , Lesiones Precancerosas/terapia , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Riesgo , Sindecano-1/metabolismo , Factores de Tiempo
10.
Blood ; 121(10): 1819-23, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23305732

RESUMEN

Prognostic implications of 3 imaging tools, metastatic bone survey, magnetic resonance imaging, and positron emission tomography (PET), were evaluated in 2 consecutive Total Therapy 3 trials for newly diagnosed myeloma. Data including PET at baseline and on day 7 of induction as well as standard prognostic factors were available in 302 patients of whom 277 also had gene expression profiling (GEP)-derived risk information. According to multivariate analysis, more than 3 focal lesions on day 7 imparted inferior overall survival and progression-free survival, overall and in the subset with GEP-risk data. GEP high-risk designation retained independent significance for all 3 end points examined. Thus, the presence of > 3 focal lesions on day 7 PET follow-up may be exploited toward early therapy change, especially for the 15% of patients with GEP-defined high-risk disease with a median overall survival expectation of 2 years. This trial was registered at www.clinicaltrials.gov as #NCT00081939 and # NCT00572169.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias Óseas/secundario , Fluorodesoxiglucosa F18 , Mieloma Múltiple/patología , Recurrencia Local de Neoplasia/patología , Tomografía de Emisión de Positrones , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/mortalidad , Ensayos Clínicos como Asunto , Estudios de Seguimiento , Perfilación de la Expresión Génica , Humanos , Imagen por Resonancia Magnética , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/mortalidad , Análisis Multivariante , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/mortalidad , Pronóstico , Radiofármacos , Tasa de Supervivencia
11.
Blood ; 121(23): 4753-7, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23603914

RESUMEN

Lenalidomide has been linked to myelodysplastic syndrome (MDS) after autotransplants for myeloma. Total therapy trials (TT; TT2(-/+) thalidomide) and TT3 (TT3a with bortezomib, thalidomide; TT3b with additional lenalidomide) offered the opportunity to examine the contribution of these immune-modulatory agents to MDS-associated cytogenetic abnormalities (MDS-CA) and clinical MDS or acute leukemia ("clinical MDS/AL"). Of 1080 patients with serial cytogenetic studies, MDS-CA occurred in 11% and clinical MDS/AL in 3%. Risk features of MDS-CA included TT3b, age ≥65 years, male gender, levels of ß-2-microglobulin >5.5 mg/L, and multiple myeloma relapse. Clinical MDS/AL occurred less frequently in the control arm of TT2 and more often with TT3a and TT3b. Since MDS-CA often antedated clinical disease, periodic cytogenetic monitoring is recommended. Larger CD34 quantities should be collected upfront as the risk of MDS could be reduced by applying higher CD34 doses with transplant. Thus, treatment, host, and myeloma features could be linked to MDS development after therapy for this malignancy. This trial was registered at www.clinicaltrials.gov: TT3A: NCT00081939, TT3B: NCT00572169.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Aberraciones Cromosómicas , Leucemia/epidemiología , Mieloma Múltiple/tratamiento farmacológico , Síndromes Mielodisplásicos/epidemiología , Recurrencia Local de Neoplasia/epidemiología , Enfermedad Aguda , Adolescente , Adulto , Anciano , Ácidos Borónicos/administración & dosificación , Bortezomib , Femenino , Estudios de Seguimiento , Humanos , Incidencia , Lenalidomida , Leucemia/inducido químicamente , Leucemia/genética , Masculino , Persona de Mediana Edad , Mieloma Múltiple/complicaciones , Síndromes Mielodisplásicos/inducido químicamente , Síndromes Mielodisplásicos/genética , Recurrencia Local de Neoplasia/inducido químicamente , Recurrencia Local de Neoplasia/genética , Pronóstico , Pirazinas/administración & dosificación , Factores de Riesgo , Talidomida/administración & dosificación , Talidomida/análogos & derivados , Adulto Joven
12.
Haematologica ; 100(9): 1214-21, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26022710

RESUMEN

Multiple myeloma is preceded by an asymptomatic phase, comprising monoclonal gammopathy of uncertain significance and smoldering myeloma. Compared to the former, smoldering myeloma has a higher and non-uniform rate of progression to clinical myeloma, reflecting a subset of patients with higher risk. We evaluated the gene expression profile of smoldering myeloma plasma cells among 105 patients enrolled in a prospective observational trial at our institution, with a view to identifying a high-risk signature. Baseline clinical, bone marrow, cytogenetic and radiologic data were evaluated for their potential to predict time to therapy for symptomatic myeloma. A gene signature derived from four genes, at an optimal binary cut-point of 9.28, identified 14 patients (13%) with a 2-year therapy risk of 85.7%. Conversely, a low four-gene score (< 9.28) combined with baseline monoclonal protein < 3 g/dL and albumin ≥ 3.5 g/dL identified 61 patients with low-risk smoldering myeloma with a 5.0% chance of progression at 2 years. The top 40 probe sets showed concordance with indices of chromosome instability. These data demonstrate high discriminatory power of a gene-based assay and suggest a role for dysregulation of mitotic checkpoints in the context of genomic instability as a hallmark of high-risk smoldering myeloma.


Asunto(s)
Genes Relacionados con las Neoplasias , Inestabilidad Genómica , Mieloma Múltiple/genética , Proteínas de Neoplasias/genética , Anciano , Anciano de 80 o más Años , Médula Ósea/patología , Femenino , Humanos , Masculino , Mieloma Múltiple/patología , Estudios Prospectivos
13.
Blood ; 120(8): 1597-600, 2012 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-22674807

RESUMEN

Thalidomide and lenalidomide constitute an important part of effective myeloma therapy. Recent data from the Intergroup Francophone du Myélome, Cancer and Leukemia Group B, and Gruppo Italiano Malattie Ematologiche dell Adulto MM-015 trials suggest that lenalidomide maintenance therapy is associated with a higher incidence of second primary malignancies (SPMs), including both hematologic and solid malignancies. In the present study, we analyzed data from the Total Therapy 2 (TT2) trial, along with the 2 Total Therapy 3 (TT3) trials. TT2 patients were assigned randomly to either a control group (no thalidomide) or to the experimental group (thalidomide during induction, between transplantations, and during consolidation and maintenance). The 2 TT3 trials used thalidomide and bortezomib during induction, before and in consolidation after tandem melphalan-based transplantation; TT3A applied VTD (bortezomib, thalidomide, dexamethasone) in the first year of maintenance and TD for 2 more years, whereas TT3B used VRD (bortezomib, lenalidomide, dexamethasone) maintenance for 3 years. The cumulative incidence of SPMs did not differ significantly among the TT trial components when measured from enrollment (P = .78) or from initiation of maintenance (P = .82). However, a pairwise comparison of the TT2 arms suggested a lower incidence of hematologic SPMs in the thalidomide maintenance arm (hazard ratio = 0.38; P = .09). These trials are registered at www.clinicaltrials.gov as NCT00573391 (TT2), NCT00081939 (TT3A), and NCT00572169 (TT3B).


Asunto(s)
Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Quimioterapia de Mantención/métodos , Mieloma Múltiple/tratamiento farmacológico , Neoplasias Primarias Secundarias/tratamiento farmacológico , Talidomida/análogos & derivados , Talidomida/uso terapéutico , Anciano , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Ácidos Borónicos/administración & dosificación , Ácidos Borónicos/uso terapéutico , Bortezomib , Dexametasona/administración & dosificación , Dexametasona/uso terapéutico , Femenino , Humanos , Incidencia , Quimioterapia de Inducción , Lenalidomida , Masculino , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/epidemiología , Neoplasias Primarias Secundarias/diagnóstico , Neoplasias Primarias Secundarias/epidemiología , Pirazinas/administración & dosificación , Pirazinas/uso terapéutico , Talidomida/administración & dosificación
14.
Haematologica ; 98(7): 1147-53, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23716540

RESUMEN

Relapsed/refractory multiple myeloma represents a major challenge in multiple myeloma therapy. For patients with relapsed/refractory multiple myeloma, we developed a treatment schema of metronomically scheduled drug therapy. We identified 186 patients who had been treated with metronomic therapy between March 2004 and January 2012 with a median follow up of 24.2 months. Median age was 61 years (range 36-83). Median number of prior therapies was 14 (range 1-51). Median number of completed metronomic therapy cycles was 1 (range 1-5), while 45 of 186 (25%) received 2 or more cycles. Responses included complete remission in 11 of 186 patients (6%), very good partial remission in 12 of 186 (7%), partial remission in 65 of 179 (36%), and minimal response in 29 of 186 (16%), for an overall response rate of 63% (117 of 186). Median overall survival and progression-free survival were 11.2 and 3.6 months, respectively. Hematologic toxicity grading was problematic as 146 of 186 (78%) of patients presented with at least grade 2 thrombocytopenia within 90 days prior to starting metronomic therapy. Grade 4 leukopenia, anemia, and/or thrombocytopenia following metronomic therapy occurred in 108 of 186 (58%), 12 of 186 (6%), and 147 of 186 (79%) patients, respectively. Incidence of grade 3-4 neutropenic fever was 4 of 186 (2%). Most patients (177 of 186, 95%) were treated in an outpatient unit and secondary admissions due to regimen-related toxicity occurred in 37 of 186 (20%). Treatment-related mortality was evident in 2 of 186 (1%). In conclusion, metronomic therapy is an effective late salvage treatment in relapsed/refractory multiple myeloma, with a high overall response rate and a favorable toxicity profile.


Asunto(s)
Administración Metronómica , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/mortalidad , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/mortalidad , Terapia Recuperativa/métodos , Adulto , Anciano , Anciano de 80 o más Años , Cisplatino/administración & dosificación , Dexametasona/administración & dosificación , Doxorrubicina/administración & dosificación , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Tasa de Supervivencia/tendencias , Talidomida/administración & dosificación , Resultado del Tratamiento
15.
Clin Trials ; 10(3): 422-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23529697

RESUMEN

BACKGROUND: Traditional phase I trials are designed to be conservative. Many times a traditional phase I trial design stops at a dose level below the maximal tolerated dose (MTD), thus potentially treating patients at a suboptimal level in all subsequent trials. This has been confirmed by our recent simulation studies. PURPOSE: We propose a phase I/II trial design to determine the most promising dose level in terms of toxicity and efficacy for cytostatic or targeted agents. This design evaluates three dose levels for efficacy and toxicity using a modified phase II selection design. The dose levels include the phase I recommended dose (RD) in addition to the dose levels immediately below and above that level. METHODS: This phase I/II trial design uses a two-step approach. In the first step, a traditional phase I trial design is used to get close to a good dose level. The second step consists of a modified selection design, randomizing patients to three dose levels: the phase I RD level and the dose levels immediately below and above the phase I RD level. Both efficacy and toxicity are used to determine a good or best dose level. Appropriate toxicity stopping rules in the phase II portion of the trial are implemented as part of such a trial. We perform simulation studies for a variety of toxicity and efficacy scenarios to determine the operating characteristics of this design and compare those to our originally proposed trial where we only explore dose levels at and below the phase I RD in the second phase of the trial, as well as to the traditional setting where a phase I trial is followed by a single-arm phase II trial at the phase I RD. RESULTS: The 3-arm modified selection design exploring the dose levels immediately above and below as well as the RD performs as well or better than the 2-arm modified selection design or the single-arm design for almost all toxicity and efficacy scenario combinations tested. CONCLUSION: We demonstrate that this design has a higher success rate at identifying a good or best dose level when exploring dose levels immediately above and below the RD in the early phase II setting, in most cases without needing larger sample sizes.


Asunto(s)
Ensayos Clínicos Fase I como Asunto/métodos , Ensayos Clínicos Fase II como Asunto/métodos , Citostáticos/administración & dosificación , Relación Dosis-Respuesta a Droga , Dosis Máxima Tolerada , Terapia Molecular Dirigida , Simulación por Computador , Citostáticos/toxicidad , Humanos , Modelos Logísticos , Ensayos Clínicos Controlados Aleatorios como Asunto , Proyectos de Investigación
16.
J Thorac Oncol ; 18(5): 564-575, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36773775

RESUMEN

In the past 20 years, the International Association for the Study of Lung Cancer (IASLC) has been working on a global project to revise the TNM classification of lung cancer. The first and second phases of the staging projects proposed recommendations for revision of the TNM classification, which were adopted by the Union for International Cancer Control and the American Joint Committee on Cancer as their seventh and eighth editions of the TNM classifications of lung cancer. For the third phase of the IASLC Staging Project, a new database of lung cancer cases diagnosed between January 2011 and December 2019 has been established. The Staging and Prognostic Factors Committee of the IASLC is in charge of the process of proposing new recommendations. The newly established database consisted of 124,581 cases. The data were obtained from Asia and Australia (56.0%), Europe (24.7%), North America (15.7%), South/Central America (3.4%), and Africa and the Middle East (0.1%). After cases with incomplete data are excluded, 87,043 cases were enrolled in the analysis, of which 52,069 (59.8%) were invasive adenocarcinoma and 15,872 (18.2%) were squamous cell carcinoma. Both clinical and pathologic stages were available in 44,831 (51.5%) cases. Analyses of this database are expected to provide proposals for changing the TNM classification toward the ninth edition, which is scheduled to be in use in January 2024. This newly established global database on lung cancer is described to provide fundamental elements for revisions of the TNM rules for staging lung cancer.


Asunto(s)
Adenocarcinoma , Carcinoma de Células Escamosas , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/patología , Estadificación de Neoplasias , Adenocarcinoma/patología , Carcinoma de Células Escamosas/patología , Pulmón/patología , Pronóstico
17.
Blood ; 115(21): 4168-73, 2010 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-20124509

RESUMEN

The Total Therapy 3 trial 2003-33 enrolled 303 newly diagnosed multiple myeloma patients and was noted to provide superior clinical outcomes compared with predecessor trial Total Therapy 2, especially in gene expression profiling (GEP)-defined low-risk disease. We report here on the results of successor trial 2006-66 with 177 patients, using bortezomib, lenalidomide, and dexamethasone maintenance for 3 years versus bortezomib, thalidomide, and dexamethasone in year 1 and thalidomide/dexamethasone in years 2 and 3 in the 2003-33 protocol. Overall survival (OS) and event-free survival (EFS) plots were super-imposable for the 2 trials, as were onset of complete response and complete response duration (CRD), regardless of GEP risk. GEP-defined high-risk designation, pertinent to 17% of patients, imparted inferior OS, EFS, and CRD in both protocols and, on multivariate analysis, was the sole adverse feature affecting OS, EFS, and CRD. Mathematical modeling of CRD in low-risk myeloma predicted a 55% cure fraction (P < .001). Despite more rapid onset and higher rate of CR than in other molecular subgroups, CRD was inferior in CCND1 without CD20 myeloma, resembling outcomes in MAF/MAFB and proliferation entities. The robustness of the GEP risk model should be exploited in clinical trials aimed at improving the notoriously poor outcome in high-risk disease.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Perfilación de la Expresión Génica , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Anciano , Ácidos Borónicos/administración & dosificación , Bortezomib , Dexametasona/administración & dosificación , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Lenalidomida , Masculino , Persona de Mediana Edad , Análisis Multivariante , Pirazinas/administración & dosificación , Factores de Riesgo , Talidomida/administración & dosificación , Talidomida/análogos & derivados
18.
Blood ; 116(8): 1220-7, 2010 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-20501894

RESUMEN

The impact of cumulative dosing and premature drug discontinuation (PMDD) of bortezomib (V), thalidomide (T), and dexamethasone (D) on overall survival (OS), event-free survival (EFS), time to next therapy, and post-relapse survival in Total Therapy 3 were examined, using time-dependent methodology, relevant to induction, peritransplantation, consolidation, and maintenance phases. Univariately, OS and EFS were longer in case higher doses were used of all agents during induction, consolidation (except T), and maintenance (except V and T). The favorable OS and EFS impact of D induction dosing provided the rationale for examining the expression of glucocorticoid receptor NR3C1, top-tertile levels of which significantly prolonged OS and EFS and rendered outcomes independent of D and T dosing, whereas T and D, but not V, dosing was critical to outcome improvement in the bottom-tertile NR3C1 setting. PMDD of V was an independent highly adverse feature for OS (hazard ratio = 6.44; P < .001), whereas PMDD of both T and D independently imparted shorter time to next therapy. The absence of adverse effects on postrelapse survival of dosing of any VTD components and indeed a benefit from V supports the use up-front of all active agents in a dose-dense and dose-intense fashion, as practiced in Total Therapy 3, toward maximizing myeloma survival.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Biomarcadores de Tumor/genética , Ácidos Borónicos/administración & dosificación , Bortezomib , Dexametasona/administración & dosificación , Relación Dosis-Respuesta a Droga , Estudios de Seguimiento , Humanos , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Reacción en Cadena de la Polimerasa , Pirazinas/administración & dosificación , Receptores de Glucocorticoides/genética , Tasa de Supervivencia , Talidomida/administración & dosificación , Resultado del Tratamiento
19.
Haematologica ; 97(11): 1761-7, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22689675

RESUMEN

BACKGROUND: Extramedullary disease is an uncommon manifestation in multiple myeloma and can either accompany newly diagnosed disease or develop with disease progression or relapse. We evaluated the impact of this disease feature on patients' outcome in the context of novel agents. DESIGN AND METHODS: We analyzed clinical and biological features of extramedullary disease in 936 patients with multiple myeloma enrolled in Total Therapy protocols, 240 patients in non-Total Therapy protocols, and 789 non-protocol patients, all of whom had baseline positron emission tomography scans to document extramedullary disease at diagnosis and its subsequent development at the time of disease progression or relapse. RESULTS: The most common sites for extramedullary disease at diagnosis were skin and soft tissue whereas liver involvement was the striking feature in extramedullary disease at disease relapse or progression. Regardless of therapy, extramedullary disease was associated with shorter progression-free and overall survival, as well as the presence of anemia, thrombocytopenia, elevated serum lactate dehydrogenase, cytogenetic abnormalities, and high-risk features in 70-and 80-gene risk models in univariate analysis. Multivariate analysis with logistic regression revealed that this disease feature was more prevalent in patients with an elevated centrosome index, as determined by gene expression profiling, as well as in myeloma molecular subtypes that are more prone to relapse. These include the MF subtype (also called the "MAF" subtype, associated with over-expression of the MAF gene seen with chromosome translocation 14;16 or 14;20) and the PR subtype (also called the "Proliferation" subtype, associated with overexpression of pro-proliferative genes). CONCLUSIONS: These data show that extramedullary disease is more prevalent in genomically defined high-risk multiple myeloma and is associated with shorter progression-free survival and overall survival, even in the era of novel agents. All clinical trials included in the analyses were registered with www.clinicaltrials.gov (NCT00083551, NCT00083876, NCT00081939, NCT00572169, NCT00644228,NCT00002548,NCT00734877).


Asunto(s)
Neoplasias Hepáticas , Mieloma Múltiple , Tomografía de Emisión de Positrones , Neoplasias Cutáneas , Neoplasias de los Tejidos Blandos , Femenino , Humanos , Neoplasias Hepáticas/diagnóstico por imagen , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/terapia , Masculino , Mieloma Múltiple/diagnóstico por imagen , Mieloma Múltiple/mortalidad , Mieloma Múltiple/terapia , Radiografía , Estudios Retrospectivos , Neoplasias Cutáneas/diagnóstico por imagen , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/terapia , Neoplasias de los Tejidos Blandos/diagnóstico por imagen , Neoplasias de los Tejidos Blandos/mortalidad , Neoplasias de los Tejidos Blandos/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA