Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.203
Filtrar
1.
Int J Mol Sci ; 25(9)2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38731863

RESUMEN

The maturation of B cells is a complex, multi-step process. During B cell differentiation, errors can occur, leading to the emergence of aberrant versions of B cells that, finally, constitute a malignant tumor. These B cell malignancies are classified into three main groups: leukemias, myelomas, and lymphomas, the latter being the most heterogeneous type. Since their discovery, multiple biological studies have been performed to characterize these diseases, aiming to define their specific features and determine potential biomarkers for diagnosis, stratification, and prognosis. The rise of advanced -omics approaches has significantly contributed to this end. Notably, proteomics strategies appear as promising tools to comprehensively profile the final molecular effector of these cells. In this narrative review, we first introduce the main B cell malignancies together with the most relevant proteomics approaches. Then, we describe the core studies conducted in the field and their main findings and, finally, we evaluate the advantages and drawbacks of flow cytometry, mass cytometry, and mass spectrometry for the profiling of human B cell disorders.


Asunto(s)
Linfocitos B , Neoplasias Hematológicas , Proteómica , Humanos , Proteómica/métodos , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/diagnóstico , Neoplasias Hematológicas/patología , Linfocitos B/metabolismo , Biomarcadores de Tumor/metabolismo , Espectrometría de Masas/métodos , Citometría de Flujo/métodos
2.
Med Oncol ; 41(6): 128, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38656461

RESUMEN

Myeloid neoplasms are a group of bone marrow diseases distinguished by disruptions in the molecular pathways that regulate the balance between hematopoietic stem cell (HSC) self-renewal and the generation of specialized cells. Cytokines and chemokines, two important components of the inflammatory process, also influence hematological differentiation. In this scenario, immunological dysregulation plays a pivotal role in the pathogenesis of bone marrow neoplasms. The STING pathway recognizes DNA fragments in the cell cytoplasm and triggers an immune response by type I interferons. The role of STING in cancer has not yet been established; however, both actions, as an oncogene or tumor suppressor, have been documented in other types of cancer. Therefore, we performed a systematic review (registered in PROSPERO database #CRD42023407512) to discuss the role of STING pathway in the advancement of pathogenesis and/or prognosis for different myeloid neoplasms. In brief, scientific evidence supports investigations that primarily use cell lines from myeloid neoplasms, such as leukemia. More high-quality research and clinical trials are needed to understand the role of the STING pathway in the pathology of hematological malignancies. Finally, the STING pathway suggests being a promising therapeutic molecular target, particularly when combined with current drug therapies.


Asunto(s)
Neoplasias Hematológicas , Proteínas de la Membrana , Humanos , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patología , Neoplasias Hematológicas/inmunología , Proteínas de la Membrana/metabolismo , Trastornos Mieloproliferativos/metabolismo , Transducción de Señal
3.
Blood Rev ; 65: 101195, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38523032

RESUMEN

B-cell lymphoma-2 (BCL-2) family proteins are fundamental regulators of the intrinsic apoptotic pathway which modulate cellular fate. In many haematological malignancies, overexpression of anti-apoptotic factors (BCL-2, BCL-XL and MCL-1) circumvent apoptosis. To address this cancer hallmark, a concerted effort has been made to induce apoptosis by inhibiting BCL-2 family proteins. A series of highly selective BCL-2 homology 3 (BH3) domain mimetics are in clinical use and in ongoing clinical trials for acute myeloid leukaemia (AML), chronic myeloid leukaemia (CML), chronic lymphocytic leukaemia (CLL), and multiple myeloma (MM). These inhibitors serve as promising candidates, both as single agents or in combination therapy to improve patient outcomes. In other diseases such as follicular lymphoma, efficacy has been notably limited. There are also clinical problems with BCL-2 family inhibition, including drug resistance, disease relapse, tumour lysis syndrome, and clinically relevant cytopenias. Here, we provide a balanced view on both the clinical benefits of BCL-2 inhibition as well as the associated challenges.


Asunto(s)
Antineoplásicos , Neoplasias Hematológicas , Leucemia Linfocítica Crónica de Células B , Humanos , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Hematológicas/metabolismo , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Apoptosis , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
4.
Nat Commun ; 15(1): 2803, 2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38555305

RESUMEN

Myeloid derived suppressor cells (MDSCs) are key regulators of immune responses and correlate with poor outcomes in hematologic malignancies. Here, we identify that MDSC mitochondrial fitness controls the efficacy of doxorubicin chemotherapy in a preclinical lymphoma model. Mechanistically, we show that triggering STAT3 signaling via ß2-adrenergic receptor (ß2-AR) activation leads to improved MDSC function through metabolic reprograming, marked by sustained mitochondrial respiration and higher ATP generation which reduces AMPK signaling, altering energy metabolism. Furthermore, induced STAT3 signaling in MDSCs enhances glutamine consumption via the TCA cycle. Metabolized glutamine generates itaconate which downregulates mitochondrial reactive oxygen species via regulation of Nrf2 and the oxidative stress response, enhancing MDSC survival. Using ß2-AR blockade, we target the STAT3 pathway and ATP and itaconate metabolism, disrupting ATP generation by the electron transport chain and decreasing itaconate generation causing diminished MDSC mitochondrial fitness. This disruption increases the response to doxorubicin and could be tested clinically.


Asunto(s)
Neoplasias Hematológicas , Células Supresoras de Origen Mieloide , Succinatos , Humanos , Glutamina/metabolismo , Neoplasias Hematológicas/metabolismo , Adenosina Trifosfato/metabolismo , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Doxorrubicina/metabolismo
5.
Front Immunol ; 15: 1348852, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38464520

RESUMEN

CD47 is a cell-surface ligand that is overexpressed in various malignancies and that binds to SIRPα on macrophages to promote tumor cell evasion of phagocytosis. Blocking the CD47-SIRPα axis can increase the phagocytosis of macrophages to exert antitumor effects. CD47-based immunotherapy is a current research focus. The combination of anti-CD47 antibodies with other drugs has shown encouraging response rates in patients with hematological tumors, but side effects also occur. Bispecific antibodies and SIRPα/Fc fusion proteins appear to balance the efficacy and safety of treatment. We review the latest clinical research advances and discuss the opportunities and challenges associated with CD47-based immunotherapy for hematological malignancies.


Asunto(s)
Neoplasias Hematológicas , Neoplasias , Humanos , Antígeno CD47/metabolismo , Fagocitosis , Macrófagos , Neoplasias/terapia , Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/metabolismo
6.
Biomed Pharmacother ; 173: 116334, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38422658

RESUMEN

Neutrophil extracellular traps (NETs) have emerged as a critical factor in malignant hematologic disease pathogenesis. These structures, comprising DNA, histones, and cytoplasmic proteins, were initially recognized for their role in immune defense against microbial threats. Growing evidence suggests that NETs contribute to malignant cell progression and dissemination, representing a double-edged sword. However, there is a paucity of reports on its involvement in hematological disorders. A comprehensive understanding of the intricate relationship between malignant cells and NETs is necessary to explore effective therapeutic strategies. This review highlights NET formation and mechanisms underlying disease pathogenesis. Moreover, we discuss recent advancements in targeted inhibitor development for selective NET disruption, empowering precise design and efficacious therapeutic interventions for malignant hematologic diseases.


Asunto(s)
Trampas Extracelulares , Enfermedades Hematológicas , Neoplasias Hematológicas , Neoplasias , Humanos , Neutrófilos/metabolismo , Histonas/metabolismo , ADN/metabolismo , Neoplasias Hematológicas/metabolismo , Neoplasias/patología , Enfermedades Hematológicas/metabolismo
7.
Front Immunol ; 15: 1320689, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38318177

RESUMEN

During lymphocyte development, a diverse repertoire of lymphocyte antigen receptors is produced to battle against pathogens, which is the basis of adaptive immunity. The diversity of the lymphocyte antigen receptors arises primarily from recombination-activated gene (RAG) protein-mediated V(D)J rearrangement in early lymphocytes. Furthermore, transcription factors (TFs), such as early B cell factor 1 (EBF1), paired box gene 5 (PAX5), and proto-oncogene myelocytomatosis oncogene (MYC), play critical roles in regulating recombination and maintaining normal B cell development. Therefore, the aberrant expression of these TFs may lead to hematologic neoplasms.


Asunto(s)
Neoplasias Hematológicas , Neoplasias , Factor de Transcripción PAX5 , Proteínas Proto-Oncogénicas c-myc , Transactivadores , Humanos , Linfocitos B , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Neoplasias/metabolismo , Factor de Transcripción PAX5/genética , Factor de Transcripción PAX5/metabolismo , Receptores de Antígenos/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo
8.
Exp Hematol ; 132: 104173, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38309573

RESUMEN

In the intricate orchestration of the central dogma, pre-mRNA splicing plays a crucial role in the post-transcriptional process that transforms DNA into mature mRNA. Widely acknowledged as a pivotal RNA processing step, it significantly influences gene expression and alters the functionality of gene product proteins. Although U2-dependent spliceosomes efficiently manage the removal of over 99% of introns, a distinct subset of essential genes undergo splicing with a different intron type, denoted as minor introns, using U12-dependent spliceosomes. Mutations in spliceosome component genes are now recognized as prevalent genetic abnormalities in cancer patients, especially those with hematologic malignancies. Despite the relative rarity of minor introns, genes containing them are evolutionarily conserved and play crucial roles in functions such as the RAS-MAPK pathway. Disruptions in U12-type minor intron splicing caused by mutations in snRNA or its regulatory components significantly contribute to cancer progression. Notably, recurrent mutations associated with myelodysplastic syndrome (MDS) in the minor spliceosome component ZRSR2 underscore its significance. Examination of ZRSR2-mutated MDS cells has revealed that only a subset of minor spliceosome-dependent genes, such as LZTR1, consistently exhibit missplicing. Recent technological advancements have uncovered insights into minor introns, raising inquiries beyond current understanding. This review comprehensively explores the importance of minor intron regulation, the molecular implications of minor (U12-type) spliceosomal mutations and cis-regulatory regions, and the evolutionary progress of studies on minor, aiming to provide a sophisticated understanding of their intricate role in cancer biology.


Asunto(s)
Neoplasias Hematológicas , Síndromes Mielodisplásicos , Humanos , Intrones , Empalmosomas/genética , Empalmosomas/metabolismo , Empalme del ARN , ARN Mensajero/genética , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Factores de Transcripción/genética
9.
Cancer Res Commun ; 4(2): 505-515, 2024 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-38319147

RESUMEN

In normal cells, binding of the transmembrane protein CD47 to signal regulatory protein-α (SIRPα) on macrophages induces an antiphagocytic signal. Tumor cells hijack this pathway and overexpress CD47 to evade immune destruction. Macrophage antitumor activity can be restored by simultaneously blocking the CD47-SIRPα signaling axis and inducing a prophagocytic signal via tumor-opsonizing antibodies. We identified a novel, fully human mAb (BMS-986351) that binds SIRPα with high affinity. BMS-986351 demonstrated broad binding coverage across SIRPα polymorphisms and potently blocked CD47-SIRPα binding at the CD47 binding site in a dose-dependent manner. In vitro, BMS-986351 increased phagocytic activity against cell lines from solid tumors and hematologic malignancies, and this effect was markedly enhanced when BMS-986351 was combined with the opsonizing antibodies cetuximab and rituximab. A phase I dose-escalation/-expansion study of BMS-986351 for the treatment of advanced solid and hematologic malignancies is underway (NCT03783403). SIGNIFICANCE: Increasing the phagocytotic capabilities of tumor-associated macrophages by modulating macrophage-tumor cell surface signaling via the CD47-SIRPα axis is a novel strategy. Molecules targeting CD47 have potential but its ubiquitous expression necessitates higher therapeutic doses to overcome potential antigen sink effects. The restricted expression pattern of SIRPα may limit toxicities and lower doses of the SIRPα antibody BMS-986351 may overcome target mediated drug disposition while maintaining the desired pharmacology.


Asunto(s)
Neoplasias Hematológicas , Neoplasias , Humanos , Antígeno CD47/genética , Receptores Inmunológicos/genética , Fagocitosis , Macrófagos , Neoplasias/tratamiento farmacológico , Anticuerpos Antineoplásicos/metabolismo , Proteínas Opsoninas/metabolismo , Neoplasias Hematológicas/metabolismo
10.
Int J Lab Hematol ; 46(1): 10-19, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37926488

RESUMEN

Bone marrow adipose tissue (BMAT) has been histologically recognized for decades. In this study, we performed a bibliometric analysis to quantitatively analyze the clusters of keywords of BMAT and hematopoiesis to better understand BMAT and hematopoiesis. Starting with conclusive keywords, our results demonstrated that BMAds is distinct from extramedullary adipose tissues and maintains a routine but dynamic accumulation throughout an individual's life. Various pathophysiological factors take part in dysregulation of the adipose-osteogenic balance throughout life. Bone marrow adipocytes (BMAds) are also contradictorily involved in normal hematopoiesis, and positively participate in the occurrence and progression of hematologic malignancies, exerting a chemoprotective role in tumor treatment. Mechanically, metabolic reprogramming and abnormal secretory profile of BMAds and tumor cells play a critical role in the chemotherapy resistance. Overall, we hope that this work will provide new ideas for relevant future research on BMAds.


Asunto(s)
Médula Ósea , Neoplasias Hematológicas , Humanos , Médula Ósea/metabolismo , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Neoplasias Hematológicas/metabolismo , Biología
11.
Protoplasma ; 261(3): 543-551, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38135806

RESUMEN

The secretion of IL-8 has been found increasing for different reasons in human bone marrow stromal cells (BMSCs), resulting in poor prognosis in patients with hematologic neoplasms. Hypoxia, a typical feature of numerous hematologic neoplasms microenvironment, often produces hypoxia inducible factor-1α (HIF-1α) which stabilizes and promotes tumor progression. Besides, hypoxic conditions also induce IL-8 production in BMSCs. However, very little is known about the mechanism of increased IL-8 expression in BMSCs caused by hypoxia. In the present study, HIF-1α and IL-8 were found highly expressed in BMSC lines under hypoxic conditions. In addition, the expression and secretion of IL-8 were significantly inhibited by the knockdown of HIF-1α under hypoxic conditions. Furthermore, HIF-1α was found to transcriptionally regulate IL-8 by binding to the region of IL-8 promoter at - 147 to - 140. Collectively, these results demonstrate that IL-8's increase is partly due to the hypoxic microenvironment in hematologic neoplasms, and activation of HIF-1α in BMSCs contributes to the induction and transcriptional regulation of IL-8 expression.


Asunto(s)
Neoplasias Hematológicas , Células Madre Mesenquimatosas , Humanos , Hipoxia de la Célula/genética , Neoplasias Hematológicas/metabolismo , Hipoxia/metabolismo , Interleucina-8/metabolismo , Células Madre Mesenquimatosas/metabolismo , Microambiente Tumoral
12.
Med Res Rev ; 44(1): 5-22, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37265248

RESUMEN

Cancer treatment brings about a phenomenon not fully clarified yet, termed chemobrain. Its strong negative impact on patients' well-being makes it a trending topic in current research, interconnecting many disciplines from clinical oncology to neuroscience. Clinical and animal studies have often reported elevated concentrations of proinflammatory cytokines in various types of blood cancers. This inflammatory burst could be the background for chemotherapy-induced cognitive deficit in patients with blood cancers. Cancer environment is a dynamic interacting system. The review puts into close relationship the inflammatory dysbalance and oxidative/nitrosative stress with disruption of the blood-brain barrier (BBB). The BBB breakdown leads to neuroinflammation, followed by neurotoxicity and neurodegeneration. High levels of intracellular reactive oxygen species (ROS) induce the progression of cancer resulting in increased mutagenesis, conversion of protooncogenes to oncogenes, and inactivation of tumor suppression genes to trigger cancer cell growth. These cell alterations may change brain functionality, as well as morphology. Multidrug chemotherapy is not without consequences to healthy tissue and could even be toxic. Specific treatment impacts brain function and morphology, functions of the immune system, and metabolism in a unique mixture. In general, a chemo-drug's effects on cognition in cancer are not direct and/or in-direct, usually a combination of effects is more probable. Last but not least, chemotherapy strongly impacts the immune system and could contribute to BBB disruption. This review points out inflammation as a possible mechanism of brain damage during blood cancers and discusses chemotherapy-induced cognitive impairment.


Asunto(s)
Deterioro Cognitivo Relacionado con la Quimioterapia , Neoplasias Hematológicas , Neoplasias , Animales , Humanos , Deterioro Cognitivo Relacionado con la Quimioterapia/metabolismo , Deterioro Cognitivo Relacionado con la Quimioterapia/patología , Neoplasias/tratamiento farmacológico , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patología , Encéfalo/metabolismo , Sistema Inmunológico
13.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 31(6): 1885-1889, 2023 Dec.
Artículo en Chino | MEDLINE | ID: mdl-38071077

RESUMEN

Cancer-associated fibroblasts (CAF) are a key component of the tumor microenvironment, which can secrete a variety of cytokines, chemokines and growth factors, directly and indirectly support cancer cells, also alter the immune cellular environment by inhibiting the activity of immune effector cells and recruiting immunosuppressive cells, thereby allowing cancer cells to evade immune surveillance. CAF has been proven to be associated with the development, progression, and poor prognosis of solid tumors. However, the role of CAF in hematological malignancies is still unclear. This article reviews the research progress of CAF in hematological malignancies.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias Hematológicas , Neoplasias , Humanos , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Neoplasias/metabolismo , Neoplasias Hematológicas/metabolismo , Microambiente Tumoral , Fibroblastos/patología
14.
Cancer Treat Res ; 190: 181-207, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38113002

RESUMEN

Chemical modifications on macromolecules such as DNA, RNA and proteins play important roles in almost all biological processes. The revival of RNA modification research began with the discovery of RNA modification machineries, and with the development of better techniques for characterizing and profiling these modifications at the transcriptome-wide level. Hematopoietic system is maintained by hematopoietic stem cells that possess efficient self-renewal capacity and the potential of differentiation into all lineages of blood cells, and the imbalance of this homeostasis frequently causes hematologic malignancies such as leukemia. Recent studies reveal that dysregulated RNA modifications play essential roles in hematologic malignancies. Herein, we summarize recent advances in some major RNA modifications, the detection methods, roles and mechanisms of these RNA modifications in hematologic malignancies.


Asunto(s)
Neoplasias Hematológicas , Humanos , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patología , ARN/genética
15.
Oncoimmunology ; 12(1): 2265703, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37808405

RESUMEN

Chimeric antigen receptor (CAR) T cell immunotherapy has demonstrated success in the treatment of hematological malignancies; however, its efficacy and applications in solid tumors remain limited. Immunosuppressive factors, particularly inhibitory checkpoint molecules, restrict CAR T cell activity inside solid tumors. The modulation of checkpoint pathways has emerged as a promising approach to promote anti-tumor responses in CAR T cells. Programmed cell death protein 1 (PD1) and T cell immunoreceptor with Ig and ITIM domains (TIGIT) are two critical immune-checkpoint molecules that suppress anti-tumor activity in T cells. Simultaneous targeting of these two inhibitory molecules could be an efficient checkpoint modulation strategy. Here, we developed a PD1-TIGIT chimeric immune-checkpoint switch receptor (CISR) that enhances the efficacy of CAR T cell immunotherapy by reversing the inhibitory checkpoint signals of PD1/PDL1 and/or TIGIT/CD155. In addition to neutralizing PDL1 and CD155, this chimeric receptor is engineered with the transmembrane region and intracellular domain of CD28, thereby effectively enhancing T cell survival and tumor-targeting functions. Notably, under simultaneous stimulation of PDL1 and CD155, CISR-CAR T cells demonstrate superior performance in terms of cell survival, proliferation, cytokine release, and cytotoxicity in vitro, compared with conventional CAR T cells. Experiments utilizing both cell line- and patient-derived xenotransplantation tumor models showed that CISR-CAR T cells exhibit robust infiltration and anti-tumor efficiency in vivo. Our results highlight the potential for the CISR strategy to enhance T cell anti-tumor efficacy and provide an alternative approach for T cell-based immunotherapies.


Asunto(s)
Neoplasias Hematológicas , Neoplasias , Humanos , Linfocitos T , Receptor de Muerte Celular Programada 1 , Neoplasias/terapia , Inmunoterapia , Neoplasias Hematológicas/metabolismo , Receptores Inmunológicos/metabolismo
16.
Curr Hematol Malig Rep ; 18(6): 264-272, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37751103

RESUMEN

PURPOSE OF REVIEW: The current review focuses on the preclinical development and clinical advances of natural killer (NK) cell therapeutics for hematologic malignancies and offers perspective on the unmet challenges that will direct future discovery in the field. RECENT FINDINGS: Approaches to improve or re-direct NK cell anti-tumor functions against hematologic malignancies have included transgenic expression of chimeric antigen receptors (CARs), administration of NK cell engagers including BiKEs and TriKEs that enhance antibody-dependent cellular cytotoxicity (ADCC) by co-engaging NK cell CD16 and antigens on tumors, incorporation of a non-cleavable CD16 that results in enhanced ADCC, use of induced memory-like NK cells alone or in combination with CARs, and blockade of NK immune checkpoints to enhance NK cytotoxicity. Recently reported and ongoing clinical trials support the feasibility and safety of these approaches. NK cell-based therapeutic strategies hold great promise as cost-effective, off-the-shelf cell therapies for patients with relapsed and refractory hematologic diseases.


Asunto(s)
Neoplasias Hematológicas , Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Células Asesinas Naturales , Neoplasias/terapia , Receptores Quiméricos de Antígenos/metabolismo , Neoplasias Hematológicas/metabolismo
17.
Blood ; 142(23): 1945-1959, 2023 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-37595276

RESUMEN

Understanding the genetic alterations, disrupted signaling pathways, and hijacked mechanisms in oncogene-transformed hematologic cells is critical for the development of effective and durable treatment strategies against liquid tumors. In this review, we focus on the specific involvement of the Hedgehog (HH)/GLI pathway in the manifestation and initiation of various cancer features in hematologic malignancies, including multiple myeloma, T- and B-cell lymphomas, and lymphoid and myeloid leukemias. By reviewing canonical and noncanonical, Smoothened-independent HH/GLI signaling and summarizing preclinical in vitro and in vivo studies in hematologic malignancies, we elucidate common molecular mechanisms by which HH/GLI signaling controls key oncogenic processes and cancer hallmarks such as cell proliferation, cancer stem cell fate, genomic instability, microenvironment remodeling, and cell survival. We also summarize current clinical trials with HH inhibitors and discuss successes and challenges, as well as opportunities for future combined therapeutic approaches. By providing a bird's eye view of the role of HH/GLI signaling in liquid tumors, we suggest that a comprehensive understanding of the general oncogenic effects of HH/GLI signaling on the formation of cancer hallmarks is essential to identify critical vulnerabilities within tumor cells and their supporting remodeled microenvironment, paving the way for the development of novel and efficient personalized combination therapies for hematologic malignancies.


Asunto(s)
Neoplasias Hematológicas , Transducción de Señal , Humanos , Proteínas Hedgehog/metabolismo , Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Microambiente Tumoral , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo
18.
J Environ Pathol Toxicol Oncol ; 42(4): 15-29, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37522565

RESUMEN

According to the World Health Organization (WHO), cancer is the second-highest cause of mortality worldwide, killing nearly 9.6 million people annually. Despite the advances in diagnosis and treatment during the last couple of decades, it remains a serious concern due to the limitations of currently available cancer management strategies. Therefore, alternative strategies are highly required to overcome these glitches. In addition, many etiological factors such as environmental and genetic factors initiate the activation of the Janus kinase (JAK)-signal transducer and activator of the transcription (STAT) pathway. This aberrant activation of the JAK-STAT pathway has been reported in various disease states, including inflammatory conditions, hematologic malignancies, and cancer. For instance, many patients with myeloproliferative neoplasms carry the acquired gain-of-function JAK2 V617F somatic mutation. This knowledge has dramatically improved our understanding of pathogenesis and has facilitated the development of therapeutics capable of suppressing the constitutive activation of the JAK-STAT pathway. Our aim is not to be expansive but to highlight emerging ideas towards preventive therapy in a modern view of JAK-STAT inhibitors. A series of agents with different specificities against different members of the JAK family of proteins is currently undergoing evaluation in clinical trials. Here we give a summary of how JAK-STAT inhibitors function and a detailed review of current clinical drugs for managing cancer as a new therapeutic approach.


Asunto(s)
Neoplasias Hematológicas , Neoplasias , Humanos , Quinasas Janus/genética , Quinasas Janus/metabolismo , Transducción de Señal , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo
19.
Eur J Med Chem ; 258: 115597, 2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-37423126

RESUMEN

Spleen tyrosine kinase (Syk) is an important oncogene and signal transduction mediator that is mainly expressed in hematopoietic cells. Syk plays a key role in the B cell receptor (BCR) signaling pathway. Abnormal activation of Syk is closely related to the occurrence and development of hematological malignancies. Therefore, Syk is a potential target for the treatment of various hematologic cancers. Starting from compound 6(Syk, IC50 = 15.8 µM), we performed fragment-based rational drug design for structural optimization based on the specific solvent-accessible region, hydrophobic region, and ribose region of Syk. This resulted in the discovery of a series of novel 3-(1H-benzo [d]imidazole-2-yl)-1H-pyrazol-4-amine Syk inhibitors, which led to the identification of 19q, a highly potent Syk inhibitor that exhibited excellent inhibitory activity on Syk enzyme (IC50 = 0.52 nM) and showed potency against several other kinases. In addition, compound 19q effectively reduced phosphorylation of downstream PLCγ2 level in Romos cells. And it also exhibited antiproliferative activity in multiple hematological tumour cells. More gratifyingly, 19q showed impressive efficacy at a low dosage (1 mg/kg/day) in the MV4-11 mouse xenograft model without affecting the body weight of the mice. These findings suggest that 19q is a promising new Syk inhibitor for treating blood cancers.


Asunto(s)
Neoplasias Hematológicas , Transducción de Señal , Humanos , Ratones , Animales , Quinasa Syk , Fosforilación , Imidazoles/farmacología , Imidazoles/uso terapéutico , Modelos Animales de Enfermedad , Aminas/farmacología , Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/metabolismo , Inhibidores de Proteínas Quinasas/química
20.
Mol Med ; 29(1): 97, 2023 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-37460961

RESUMEN

Toll-like receptors play a significant role in the innate immune system and are also involved in the pathophysiology of many different diseases. Over the past 35 years, there have been a growing number of publications exploring the role of the orphan toll-like receptor, CD180. We therefore set out to provide a narrative review of the current evidence surrounding CD180 in both health and disease. We first explore the evidence surrounding the role of CD180 in physiology including its expression, function and signaling in antigen presenting cells (APCs) (dendritic cells, monocytes, and B cells). We particularly focus on the role of CD180 as a modulator of other TLRs including TLR2, TLR4, and TLR9. We then discuss the role of CD180 in inflammatory and autoimmune diseases, as well as in hematological malignancies of B cell origin, including chronic lymphocytic leukemia (CLL). Based on this evidence we produce a current model for CD180 in disease and explore the potential role for CD180 as both a prognostic biomarker and therapeutic target. Throughout, we highlight specific areas of research which should be addressed to further the understanding of CD180 biology and the translational potential of research into CD180 in various diseases.


Asunto(s)
Neoplasias Hematológicas , Leucemia Linfocítica Crónica de Células B , Humanos , Antígenos CD/metabolismo , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Linfocitos B , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patología , Monocitos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...