Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Adv Drug Deliv Rev ; 214: 115448, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39303823

RESUMEN

Hematological cancers encompass a diverse group of malignancies affecting the blood, bone marrow, lymph nodes, and spleen. These disorders present unique challenges due to their complex etiology and varied clinical manifestations. Despite significant advancements in understanding and treating hematological malignancies, innovative therapeutic approaches are continually sought to enhance patient outcomes. This review highlights the application of RNA nanoparticles (RNA-NPs) in the treatment of hematological cancers. We delve into detailed discussions on in vitro and preclinical studies involving RNA-NPs for adult patients, as well as the application of RNA-NPs in pediatric hematological cancer. The review also addresses ongoing clinical trials involving RNA-NPs and explores the emerging field of CAR-T therapy engineered by RNA-NPs. Finally, we discuss the challenges still faced in translating RNA-NP research to clinics.

2.
Front Oncol ; 14: 1288501, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38559562

RESUMEN

SKP2 (S-phase kinase-associated protein 2) is a member of the F-box family of substrate-recognition subunits in the SCF ubiquitin-protein ligase complexes. It is associated with ubiquitin-mediated degradation in the mammalian cell cycle components and other target proteins involved in cell cycle progression, signal transduction, and transcription. Being an oncogene in solid tumors and hematological malignancies, it is frequently associated with drug resistance and poor disease outcomes. In the current review, we discussed the novel role of SKP2 in different hematological malignancies. Further, we performed a limited in-silico analysis to establish the involvement of SKP2 in a few publicly available cancer datasets. Interestingly, our study identified Skp2 expression to be altered in a cancer-specific manner. While it was found to be overexpressed in several cancer types, few cancer showed a down-regulation in SKP2. Our review provides evidence for developing novel SKP2 inhibitors in hematological malignancies. We also investigated the effect of SKP2 status on survival and disease progression. In addition, the role of miRNA and its associated families in regulating Skp2 expression was explored. Subsequently, we predicted common miRNAs against Skp2 genes by using miRNA-predication tools. Finally, we discussed current approaches and future prospective approaches to target the Skp2 gene by using different drugs and miRNA-based therapeutics applications in translational research.

3.
J Tradit Complement Med ; 14(2): 121-134, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38481552

RESUMEN

Hematological cancers include leukemia, myeloma and lymphoma and up to 178.000 new cases are diagnosed with these tumors each year. Different kinds of treatment including radiotherapy, chemotherapy, immunotherapy and stem cell transplantation have been employed in the therapy of hematological cancers. However, they are still causing death among patients. On the other hand, curcumin as an anti-cancer agent for the suppression of human cancers has been introduced. The treatment of hematological cancers using curcumin has been followed. Curcumin diminishes viability and survival rate of leukemia, myeloma and lymphoma cells. Curcumin stimulates apoptosis and G2/M arrest to impair progression of tumor. Curcumin decreases levels of matrix metalloproteinases in suppressing cancer metastasis. A number of downstream targets including VEGF, Akt and STAT3 undergo suppression by curcumin in suppressing progression of hematological cancers. Curcumin stimulates DNA damage and reduces resistance of cancer cells to irradiation. Furthermore, curcumin causes drug sensitivity of hematological tumors, especially myeloma. For targeted delivery of curcumin and improving its pharmacokinetic and anti-cancer features, nanostructures containing curcumin and other anti-cancer agents have been developed.

4.
Cancers (Basel) ; 15(24)2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-38136292

RESUMEN

Although chimeric antigen receptor (CAR)-T cell therapies are typically administered in the inpatient setting, outpatient administration is rapidly expanding. However, there is limited summarized evidence comparing outcomes between outpatient and inpatient administration. This systematic literature review aims to compare the safety, efficacy, quality of life (QoL), costs, and healthcare resource utilization (HCRU) outcomes in patients with hematological cancer who are administered CAR-T therapy in an outpatient versus an inpatient setting. Publications (2016 or later) that reported the outcomes of interest in patients treated with a CAR-T therapy in both outpatient and inpatient settings, or only the outpatient setting, were reviewed. In total, 38 publications based on 21 studies were included. Safety findings suggested the comparable frequency of adverse events in the two settings. Eleven studies that reported data in both settings showed comparable response rates (80-82% in outpatient and 72-80% in inpatient). Improvements in the QoL were observed in both settings while costs associated with CAR-T therapy were lower in the outpatient setting. Although unplanned hospitalizations were higher in the outpatient cohort, overall HCRU was lower. Outpatient administration of CAR-T therapy appears to have comparable outcomes in safety, efficacy, and QoL to inpatient administration while reducing the economic burden.

5.
Biomolecules ; 13(12)2023 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-38136573

RESUMEN

Mesenchymal stromal cells (MSCs) are a subset of heterogeneous, non-hematopoietic fibroblast-like cells which play important roles in tissue repair, inflammation, and immune modulation. MSCs residing in the bone marrow microenvironment (BMME) functionally interact with hematopoietic stem progenitor cells regulating hematopoiesis. However, MSCs have also emerged in recent years as key regulators of the tumor microenvironment. Indeed, they are now considered active players in the pathophysiology of hematologic malignancies rather than passive bystanders in the hematopoietic microenvironment. Once a malignant event occurs, the BMME acquires cellular, molecular, and epigenetic abnormalities affecting tumor growth and progression. In this context, MSC behavior is affected by signals coming from cancer cells. Furthermore, it has been shown that stromal cells themselves play a major role in several hematological malignancies' pathogenesis. This bidirectional crosstalk creates a functional tumor niche unit wherein tumor cells acquire a selective advantage over their normal counterparts and are protected from drug treatment. It is therefore of critical importance to unveil the underlying mechanisms which activate a protumor phenotype of MSCs for defining the unmasked vulnerabilities of hematological cancer cells which could be pharmacologically exploited to disrupt tumor/MSC coupling. The present review focuses on the current knowledge about MSC dysfunction mechanisms in the BMME of hematological cancers, sustaining tumor growth, immune escape, and cancer progression.


Asunto(s)
Neoplasias Hematológicas , Células Madre Mesenquimatosas , Neoplasias , Humanos , Médula Ósea , Células Madre Hematopoyéticas , Neoplasias/patología , Microambiente Tumoral , Células de la Médula Ósea/patología , Células Madre Mesenquimatosas/fisiología
6.
Eur J Med Chem ; 258: 115566, 2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-37354740

RESUMEN

New amide derivatives of the natural product 5,6,7-trimethoxyflavanone were designed as multifunctional antiproliferative molecules against blood cancer and the associated inflammatory conditions. The targeted compounds were synthesized efficiently in three linear steps employing known chalcone starting materials. Compounds 2h, 2i, 2l, 2t, 2v and 2x having bromo or nitro substituted-phenyl rings elicited potential inhibitory effects on macrophages production of nitric oxide, PGE2 and TNF-α which are proinflammatory mediators involved in tumorigenesis and progression of blood cancer. Additionally, evaluation of direct inhibitory effects on the growth of diverse blood cancers including leukemia, lymphoma, and myeloma cell lines unveiled compound 2v as the most potential molecules eliciting at least five-folds the potency of the standard imatinib drug over the used diverse blood cancers. Furthermore, compound 2v showed good selectivity to blood cancer cells rather than normal MRC5 cells. Moreover, compound 2v triggered death of HL60 leukemia cells via apoptosis induction. In conclusion, the natural product-derived compound 2v might serve as a multifunctional lead compound for further development of agents for treatment of blood cancers.


Asunto(s)
Antineoplásicos , Neoplasias Hematológicas , Leucemia , Neoplasias , Humanos , Estructura Molecular , Relación Estructura-Actividad , Antiinflamatorios/farmacología , Neoplasias Hematológicas/tratamiento farmacológico , Antineoplásicos/farmacología , Proliferación Celular , Ensayos de Selección de Medicamentos Antitumorales
7.
Biomark Res ; 11(1): 31, 2023 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-36927800

RESUMEN

BACKGROUND: Studies have not systematically compared the ability to verify performance of prognostic transcripts in paired bulk mononuclear cells versus viable CD34-expressing leukemic blasts from patients with acute myeloid leukemia. We hypothesized that examining the homogenous leukemic blasts will yield different biological information and may improve prognostic performance of expression biomarkers. METHODS: To assess the impact of cellular heterogeneity on expression biomarkers in acute myeloid leukemia, we systematically examined paired mononuclear cells and viable CD34-expressing leukemic blasts from SWOG diagnostic specimens. After enrichment, patients were assigned into discovery and validation cohorts based on availability of extracted RNA. Analyses of RNA sequencing data examined how enrichment impacted differentially expressed genes associated with pre-analytic variables, patient characteristics, and clinical outcomes. RESULTS: Blast enrichment yielded significantly different expression profiles and biological pathways associated with clinical characteristics (e.g., cytogenetics). Although numerous differentially expressed genes were associated with clinical outcomes, most lost their prognostic significance in the mononuclear cells and blasts after adjusting for age and ELN risk, with only 11 genes remaining significant for overall survival in both cell populations (CEP70, COMMD7, DNMT3B, ECE1, LNX2, NEGR1, PIK3C2B, SEMA4D, SMAD2, TAF8, ZNF444). To examine the impact of enrichment on biomarker verification, these 11 candidate biomarkers were examined by quantitative RT/PCR in the validation cohort. After adjusting for ELN risk and age, expression of 4 genes (CEP70, DNMT3B, ECE1, and PIK3CB) remained significantly associated with overall survival in the blasts, while none met statistical significance in mononuclear cells. CONCLUSIONS: This study provides insights into biological information gained/lost by examining viable CD34-expressing leukemic blasts versus mononuclear cells from the same patient and shows an improved verification rate for expression biomarkers in blasts.

8.
Cancers (Basel) ; 15(6)2023 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-36980677

RESUMEN

B cell-activating factor (BAFF; also known as CD257, TNFSF13B, BLyS) and a proliferation-inducing ligand (APRIL; also known as CD256, TNFSF13) belong to the tumor necrosis factor (TNF) family. BAFF was initially discovered as a B-cell survival factor, whereas APRIL was first identified as a protein highly expressed in various cancers. These discoveries were followed by over two decades of extensive research effort, which identified overlapping signaling cascades between BAFF and APRIL, controlling immune homeostasis in health and driving pathogenesis in autoimmunity and cancer, the latter being the focus of this review. High levels of BAFF, APRIL, and their receptors have been detected in different cancers and found to be associated with disease severity and treatment response. Here, we have summarized the role of the BAFF-APRIL system in immune cell differentiation and immune tolerance and detailed its pathogenic functions in hematological and solid cancers. We also highlight the emerging therapeutics targeting the BAFF-APRIL system in different cancer types.

9.
Cells ; 12(3)2023 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-36766776

RESUMEN

Primarily identified as an important regulator of cytoskeletal dynamics, the small GTPase Ras homolog gene family member A (RHOA) has been implicated in the transduction of signals regulating a broad range of cellular functions such as cell survival, migration, adhesion and proliferation. Deregulated activity of RHOA has been linked to the growth, progression and metastasis of various cancer types. Recent cancer genome-wide sequencing studies have unveiled both RHOA gain and loss-of-function mutations in primary leukemia/lymphoma, suggesting that this GTPase may exert tumor-promoting or tumor-suppressive functions depending on the cellular context. Based on these observations, RHOA signaling represents an attractive therapeutic target for the development of selective anticancer strategies. In this review, we will summarize the molecular mechanisms underlying RHOA GTPase functions in immune regulation and in the development of hematological neoplasms and will discuss the current strategies aimed at modulating RHOA functions in these diseases.


Asunto(s)
Neoplasias Hematológicas , Neoplasias , Humanos , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo , Transducción de Señal , Mutación , Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/genética
10.
Front Immunol ; 13: 1016059, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36304465

RESUMEN

The success of immunotherapeutic approaches in hematological cancers is partially hampered by the presence of an immunosuppressive microenvironment. Myeloid-derived suppressor cells (MDSC) are key components of this suppressive environment and are frequently associated with tumor cell survival and drug resistance. Based on their morphology and phenotype, MDSC are commonly subdivided into polymorphonuclear MDSC (PMN-MDSC or G-MDSC) and monocytic MDSC (M-MDSC), both characterized by their immunosuppressive function. The phenotype, function and prognostic value of MDSC in hematological cancers has been intensively studied; however, the therapeutic targeting of this cell population remains challenging and needs further investigation. In this review, we will summarize the prognostic value of MDSC and the different attempts to target MDSC (or subtypes of MDSC) in hematological cancers. We will discuss the benefits, challenges and opportunities of using MDSC-targeting approaches, aiming to enhance anti-tumor immune responses of currently used cellular and non-cellular immunotherapies.


Asunto(s)
Neoplasias Hematológicas , Células Supresoras de Origen Mieloide , Neoplasias , Humanos , Pronóstico , Monocitos , Neoplasias Hematológicas/terapia , Neoplasias Hematológicas/patología , Microambiente Tumoral
11.
JMIR Res Protoc ; 11(7): e39007, 2022 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-35776489

RESUMEN

BACKGROUND: To address the need for long-term, accessible, nonpharmacologic interventions targeting sleep in patients with chronic hematological cancer, we propose the first randomized controlled trial to determine the effects of a consumer-based mobile meditation app, Calm, on sleep disturbance in this population. OBJECTIVE: This study aims to test the efficacy of daily meditation delivered via Calm compared with a health education podcast control group in improving the primary outcome of self-reported sleep disturbance, as well as secondary sleep outcomes, including sleep impairment and sleep efficiency; test the efficacy of daily meditation delivered via Calm compared with a health education podcast control group on inflammatory markers, fatigue, and emotional distress; and explore free-living use during a 12-week follow-up period and the sustained effects of Calm in patients with chronic hematological cancer. METHODS: In a double-blinded randomized controlled trial, we will recruit 276 patients with chronic hematological cancer to an 8-week app-based wellness intervention-the active, daily, app-based meditation intervention or the health education podcast app control group, followed by a 12-week follow-up period. Participants will be asked to use their assigned app for at least 10 minutes per day during the 8-week intervention period; complete web-based surveys assessing self-reported sleep disturbance, fatigue, and emotional distress at baseline, 8 weeks, and 20 weeks; complete sleep diaries and wear an actigraphy device during the 8-week intervention period and at 20 weeks; and complete blood draws to assess inflammatory markers (tumor necrosis factor-α, interleukin-6, interleukin-8, and C-reactive protein) at baseline, 8 weeks, and 20 weeks. RESULTS: This project was funded by the National Institutes of Health National Cancer Institute (R01CA262041). The projects began in April 2022, and study recruitment is scheduled to begin in October 2022, with a total project duration of 5 years. We anticipate that we will be able to achieve our enrollment goal of 276 patients with chronic hematological cancers within the allotted project time frame. CONCLUSIONS: This research will contribute to broader public health efforts by providing researchers and clinicians with an evidence-based commercial product to improve sleep in the long term in an underserved and understudied cancer population with a high incidence of sleep disturbance. TRIAL REGISTRATION: ClinicalTrials.gov NCT05294991; https://clinicaltrials.gov/ct2/show/NCT05294991. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID): PRR1-10.2196/39007.

12.
Gene ; 838: 146735, 2022 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-35835403

RESUMEN

Lymphoma is one of the most prevalent hematological cancers, accounting for 15-20 % of new cancer diagnoses in dogs. Therefore, this study aims to explore the important genes and pathways involved in canine lymphoma progression and understand the underlying molecular mechanisms using RNA sequencing. In this study, RNAs acquired from seven pairs of lymphoma and non-lymphoma blood samples were sequenced from different breeds of dogs. Sequencing reads were preprocessed, aligned with the reference genome, assembled and expressions were estimated through bioinformatics approaches. At a false discovery rate (FDR) < 0.05 and fold change (FC) ≥ 1.5, a total of 625 differentially expressed genes (DEGs) were identified between lymphoma and non-lymphoma samples, including 347 up-regulated DEGs such as SLC38A11, SCN3A, ZIC5 etc. and 278 down-regulated DEGs such as LOC475937, CSMD1, KRT14 etc. GO enrichment analysis showed that these DEGs were highly enriched for molecular function of ATP binding and calcium ion binding, cellular process of focal adhesion, and biological process of immune response, and defense response to virus. Similarly, KEGG pathways analysis revealed 11 significantly enriched pathways such as ECM-receptor interaction, cell cycle, PI3K-Akt signaling pathway, ABC transporters etc. In the protein-protein interaction (PPI) network, CDK1 was found to be a top hub gene with highest degree of connectivity. Three modules selected from the PPI network showed that canine lymphoma was highly associated with cell cycle, ECM-receptor interaction, hypertrophic cardiomyopathy, dilated cardiomyopathy and RIG-I-like receptor signaling pathway. Overall, our findings highlighted new candidate therapeutic targets for further testing in canine lymphoma and facilitate the understanding of molecular mechanism of lymphoma's progression in dogs.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Fosfatidilinositol 3-Quinasas , Animales , Biología Computacional , Perros , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Fosfatidilinositol 3-Quinasas/genética , Mapas de Interacción de Proteínas/genética , Transducción de Señal/genética , Transcriptoma
13.
Support Care Cancer ; 30(11): 8733-8744, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35689676

RESUMEN

PURPOSE: To systematically synthesize the effect of music therapy interventions applied to patients with hematological cancer on fatigue. METHODS: The searches were conducted on PubMed, Web of Science, EBSCOhost/CINAHL Complete, Science Direct, Scopus, Cochrane Library, Ovid, ProQuest, and Springer Link databases until August 2021 without any year limitation. The review covered the period between 2003 and 2020. Comprehensive Meta-Analysis 3 software was used in the analysis of meta-analysis data. The meta-analysis was carried out following the PRISMA checklist. Risks of bias were examined by two independent researchers using the Cochrane Collaboration tool. RESULTS: Six randomized controlled trials consisting of 279 participants were included in the systematic review and meta-analysis. The count of music therapy interventions in the included studies ranged between 1 and 8 sessions per participant, each of which was 20 to 45 min long. The music therapy interventions applied to patients with hematological cancers were found to be effective in reducing the severity of fatigue (95% Cl = 0.10 ~ 0.57; Hedge's g = 0.03; p = 0.006). CONCLUSIONS: The findings of the meta-analysis indicated that music therapy interventions made important and positive contributions to reducing fatigue in patients with hematological cancer. Music therapy interventions are a convenient method to reduce fatigue because they are comfortable and non-invasive. It will be beneficial to increase the awareness of nurses about the implementation of music therapy interventions. It is recommended that music therapy interventions applied to patients with a diagnosis of hematological cancer should be considered interventions that can be used together with other non-pharmacological or pharmacological methods to reduce fatigue.


Asunto(s)
Fatiga , Neoplasias Hematológicas , Musicoterapia , Humanos , Fatiga/etiología , Fatiga/prevención & control , Neoplasias Hematológicas/complicaciones , Neoplasias Hematológicas/terapia , Musicoterapia/métodos , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento
14.
J Nippon Med Sch ; 89(1): 95-101, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35283406

RESUMEN

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel, highly pathogenic coronavirus that has spread rapidly worldwide and caused an international public health emergency. Patients with hematological cancers are regarded as a high-risk group for coronavirus disease 2019 (COVID-19). However, few reports have investigated factors that might account for the differential severity of COVID-19 disease in these patients. METHODS: Gene expression of SARS-CoV-2 entry-promoting factors and entry-restricting factors and the associated effects on myeloid malignancies were evaluated. Gene expression levels of 11 SARS-CoV-2 entry-promoting factors and 4 SARS-CoV-2 entry-restricting factors were analyzed in patients with myelodysplastic syndromes (MDS), chronic myeloid leukemia (CML), and acute myeloid leukemia and its subtypes. RESULTS: Expression levels of promoting and restricting factors were most affected in MDS. Specifically, 4 of the 11 analyzed SARS-CoV-2 entry-promoting factors were significantly increased (TMPRSS4, CD209, CLEC4G, and CTSB), and 2 of the 4 analyzed SARS-CoV-2 entry-restricting factors were significantly decreased (IFITM1 and IFITM2) in MDS. Patients with CML also exhibited a pattern of significant changes in SARS-CoV-2 entry-promoting and entry-restricting factors. Five of the 11 analyzed SARS-CoV-2 entry-promoting factors were significantly increased (ACE2, TMPRSS2, TMPRSS4, ANPEP, CD209), and 1 of the 4 analyzed SARS-CoV-2 entry-restricting factors was significantly decreased (LY6E) in CML. CONCLUSIONS: The present and past results highlight the importance of investigating SARS-CoV-2 entry-promoting factors and entry-restricting factors, because of their crucial role in determining the differential severity of COVID-19 disease.


Asunto(s)
COVID-19 , Neoplasias , Enzima Convertidora de Angiotensina 2 , Línea Celular , Humanos , Proteínas de la Membrana , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , SARS-CoV-2 , Serina Endopeptidasas/genética
15.
Front Oncol ; 12: 823115, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35280767

RESUMEN

Background: Increased success in the treatment of hematological cancers contributed to the increase of 5-year survival for most adolescent and young adults (AYAs) with these tumours. However, as 5-year survival increased, it became clear that AYA long-term survivors were at increased risk for severe late effects. Moreover, limited information on long-term cancer impact is available for AYAs, since most studies focused on children and adolescents. We aimed to assess various long-term outcomes on AYA survivors of hematological cancers. Methods: We selected patients diagnosed with a first primary hematological cancer between 1997 and 2006, in the Italian nationwide population-based cohort of AYA cancer survivors (i.e. alive at least 5 years after cancer diagnosis). Long-term outcomes of interest were: second malignant neoplasms (SMNs), hospitalizations and overall mortality. We calculated standardized incidence ratios (SIRs), standardized hospitalization rate ratios (SHRs) and standardized mortality rate ratios (SMRs). To study morbidity patterns over time, we modeled observed incidence rates by fitting flexible parametric models for nonlinear patterns and we used linear regression for linear patterns. Results: The study cohort included 5,042 AYA hematological cancer survivors of which 1,237 and 3,805 had a leukaemia and lymphoma diagnosis, respectively. AYA survivors were at substantially increased risk for SMN (SIR=2.1; 95%CI=1.7; 2.6), hospitalisation (SHR=1.5; 95%CI=1.5; 1.6), and mortality (SMR=1.4; 95%CI=1.2; 1.6) with differences between leukaemia and lymphoma survivors. The highest excess risks of hospitalisations were for infectious diseases, respiratory diseases, and diseases of blood and blood-forming organs. The morbidity pattern differs over time by morbidity type. Conclusions: Our results support the need for strict follow-up plans for survivors, and call for further study to better personalised follow-up plans for AYA cancer survivors.

16.
Clin Infect Dis ; 75(6): 1046-1053, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-35195716

RESUMEN

BACKGROUND: Patients with hematological cancers (HC) are at high risk of infections, in particular community-acquired pneumonia (CAP). Recent data on incidence and predictors of CAP among patients with HC are scarce. METHODS: We performed a cohort study (2016-2019) in 2 hospitals in the Netherlands among adults with HC to calculate incidence rates (IRs) of CAP. In addition, we performed a nested case-control study to identify predictors of CAP. RESULTS: We identified 275 CAP cases during 6264 patient-years of follow-up. The IR of CAP was 4390/100 000 patient-years of follow-up. Compared with the general population, IR ratios ranged from 5.4 to 55.3 for the different HCs. The case fatality and intensive care unit (ICU) admission rates were 5.5% and 9.8%, respectively. Predictors for CAP in patients with HC were male sex, anemia, lymphocytopenia, chronic kidney disease, cardiovascular disease, autologous and allogeneic stem cell transplantation, treatment with immunosuppressive medication for graft-vs-host disease, treatment with rituximab in the past year, and treatment with immunomodulators (lenalidomide, thalidomide, pomalidomide and/or methotrexate) in the past month. Independent predictors of a severe disease course (death or ICU admission) included neutropenia (odds ratio, 4.14 [95% confidence interval, 1.63-10.2]), pneumococcal pneumonia (10.24 [3.48-30.1]), chronic obstructive pulmonary disease (6.90 [2.07-23.0]), and the use of antibacterial prophylaxis (2.53 [1.05-6.08]). CONCLUSIONS: The burden of CAP in patients with HC is high, with significant morbidity and mortality rates. Therefore, vaccination against respiratory pathogens early in the disease course is recommended, in particular before starting certain immunosuppressive therapies.


Asunto(s)
Infecciones Comunitarias Adquiridas , Neoplasias Hematológicas , Neumonía Neumocócica , Neumonía , Adulto , Antibacterianos , Estudios de Casos y Controles , Estudios de Cohortes , Infecciones Comunitarias Adquiridas/epidemiología , Progresión de la Enfermedad , Femenino , Neoplasias Hematológicas/complicaciones , Neoplasias Hematológicas/epidemiología , Hospitalización , Humanos , Incidencia , Lenalidomida , Masculino , Metotrexato , Neumonía Neumocócica/epidemiología , Rituximab , Talidomida
17.
J Hematol Oncol ; 14(1): 179, 2021 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-34717692

RESUMEN

Ibrutinib plus venetoclax, given with an ibrutinib lead-in, has shown encouraging clinical activity in early phase studies in mantle cell lymphoma (MCL). The ongoing phase 3 SYMPATICO study evaluates the safety and efficacy of concurrently administered, once-daily, all-oral ibrutinib plus venetoclax in patients with relapsed/refractory MCL. A safety run-in (SRI) cohort was conducted to inform whether an ibrutinib lead-in should be implemented for the randomized portion. Patients received concurrent ibrutinib 560 mg continuously plus venetoclax in a 5-week ramp-up to venetoclax 400 mg for up to 2 years. The primary endpoint was occurrence of tumor lysis syndrome (TLS) and dose-limiting toxicities (DLTs). The SRI cohort enrolled 21 patients; six and 15 were in low- or increased-risk categories for TLS, respectively. During the 5-week venetoclax ramp-up, three patients had DLTs, and one patient at increased risk for TLS had a laboratory TLS; no additional TLS events occurred during follow-up. With a median follow-up of 31 months, the overall response rate was 81% (17/21); 62% (13/21) of patients had a complete response. SRI data informed that the randomized portion should proceed with concurrent ibrutinib plus venetoclax, with no ibrutinib lead-in. Ibrutinib plus venetoclax demonstrated promising efficacy; no new safety signals were observed.Trial registration: ClinicalTrials.gov, NCT03112174. Registered 13 April 2017, https://clinicaltrials.gov/ct2/show/NCT03112174 .


Asunto(s)
Adenina/análogos & derivados , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Linfoma de Células del Manto/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Piperidinas/uso terapéutico , Sulfonamidas/uso terapéutico , Adenina/efectos adversos , Adenina/uso terapéutico , Anciano , Antineoplásicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Compuestos Bicíclicos Heterocíclicos con Puentes/efectos adversos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Piperidinas/efectos adversos , Sulfonamidas/efectos adversos , Resultado del Tratamiento
18.
Biomark Res ; 9(1): 34, 2021 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-33958003

RESUMEN

The myristoylated alanine-rich C-kinase substrate (MARCKS) protein has been at the crossroads of multiple signaling pathways that govern several critical operations in normal and malignant cellular physiology. Functioning as a target of protein kinase C, MARCKS shuttles between the phosphorylated cytosolic form and the unphosphorylated plasma membrane-bound states whilst regulating several molecular partners including, but not limited to calmodulin, actin, phosphatidylinositol-4,5-bisphosphate, and phosphoinositide-3-kinase. As a result of these interactions, MARCKS directly or indirectly modulates a host of cellular functions, primarily including cytoskeletal reorganization, membrane trafficking, cell secretion, inflammatory response, cell migration, and mitosis. Recent evidence indicates that dysregulated expression of MARCKS is associated with the development and progression of hematological cancers. While it is understood that MARCKS impacts the overall carcinogenesis as well as plays a part in determining the disease outcome in blood cancers, we are still at an early stage of interpreting the pathophysiological roles of MARCKS in neoplastic disease. The situation is further complicated by contradictory reports regarding the role of phosphorylated versus an unphosphorylated form of MARCKS as an oncogene versus tumor suppressor in blood cancers. In this review, we will investigate the current body of knowledge and evolving concepts of the physical properties, molecular network, functional attributes, and the likely pathogenic roles of MARCKS in hematological malignancies. Key emphasis will also be laid upon understanding the novel mechanisms by which MARCKS determines the overall disease prognosis by playing a vital role in the induction of therapeutic resistance. Additionally, we will highlight the importance of MARCKS as a valuable therapeutic target in blood cancers and will discuss the potential of existing strategies available to tackle MARCKS-driven blood cancers.

19.
Asia Pac J Oncol Nurs ; 8(1): 5-17, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33426184

RESUMEN

Hematological malignancies require intensive and long-term treatment, which brings a significant burden on patients, leading to unmet supportive care needs. The purpose of this review was to investigate the unmet supportive care needs of patients with hematological malignancies during and after active treatment as well as the factors that affect them. A systematic bibliographic search was carried out in the PubMed database for English articles published between 2009 and 2020 according to the Preferred Reporting Items for Systematic Reviews guidelines and under the terms: "unmet needs", "supportive care", "hematological malignancy" and "hematological cancer." Twenty studies were evaluated and reviewed. Hierarchical frequently reported unmet supportive care needs were informational, emotional, physical, daily living/practical (accessibility, transportation, and financial problems), and family life/relational needs. In particular, patients with multiple myeloma most frequently reported unmet needs at the informational, physical, emotional, and daily living/practical domain. Patients with myelodysplastic syndromes reported physical, emotional, practical, and relational needs. Patients with leukemia and lymphoma rated their needs as informational, physical, psychological, daily living, and sexual. Sexual and spiritual unmet needs were reported at a low level. Predictive indicators for increased unmet supportive care needs were the type of the hematological malignancy, younger age, marital status, female gender, monthly income, coexistence of anxiety and depression, and altered quality of life. To conclude with, the literature reports a significant number of unmet supportive care needs in patients with hematological malignancies, whose frequency and intensity were influenced by a variety of factors. However, the large heterogeneity of studies (design, sample, and needs assessment tools) makes the generalization of the results difficult.

20.
Am J Epidemiol ; 190(1): 59-75, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-32706884

RESUMEN

Recently, we introduced a novel measure of "average life span shortened" (ALSS) to improve comparability of premature mortality over time. In this study, we applied this novel measure to examine trends in premature mortality caused by hematological cancers in Canada from 1980 to 2015. Mortality data for Hodgkin lymphoma, non-Hodgkin lymphoma, multiple myeloma, and leukemia were obtained from the World Health Organization mortality database. Years of life lost was calculated according to Canadian life tables. ALSS was defined as the ratio between years of life lost and expected life span. Over the study period, age-standardized rates of mortality decreased for all types of hematological cancers. Our new ALSS measure showed favorable trends in premature mortality for all types of hematological cancers among both sexes. For instance, men with non-Hodgkin lymphoma lost an average of 23.7% of their life span in 1980 versus 16.1% in 2015, while women with non-Hodgkin lymphoma lost an average of 21.7% of their life span in 1980 versus 15.5% in 2015. Results from this study showed that patients with hematological cancers experienced prolonged survival over a 35-year period although the magnitude of these life span gains varied by types of hematological cancers.


Asunto(s)
Enfermedad de Hodgkin/mortalidad , Leucemia/mortalidad , Linfoma no Hodgkin/mortalidad , Mortalidad Prematura/tendencias , Mieloma Múltiple/mortalidad , Adolescente , Adulto , Anciano , Canadá/epidemiología , Femenino , Humanos , Esperanza de Vida , Tablas de Vida , Masculino , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA