Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Cancers (Basel) ; 16(6)2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38539451

RESUMO

Autologous stem cell transplantation (ASCT) has been a mainstay in myeloma treatment for over three decades, but patient prognosis post-ASCT varies significantly. In a retrospective study of 5259 patients with multiple myeloma (MM) at the University of Arkansas for Medical Sciences undergoing ASCT with a median 57-month follow-up, we divided the dataset into training (70%) and validation (30%) subsets. Employing univariable and multivariable Cox analyses, we systematically assessed 29 clinical variables, identifying crucial adverse prognostic factors, such as extended duration between MM diagnosis and ASCT, elevated serum ferritin, and reduced transferrin levels. These factors could enhance existing prognostic models. Additionally, we pinpointed significant poor prognosis markers like high serum calcium and low platelet counts, though they are applicable to a smaller patient population. Utilizing seven easily accessible high-risk variables, we devised a four-stage system (ATM4S) with primary stage borders determined through K-adaptive partitioning. This staging system underwent validation in both the training dataset and an independent cohort of 514 ASCT-treated MM patients from the University of Iowa. We also explored cytogenetic risk factors within this staging system, emphasizing its potential clinical utility for refining prognostic assessments and guiding personalized treatment approaches.

2.
Blood Cancer J ; 14(1): 38, 2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38443358

RESUMO

Multiple myeloma (MM) is a heterogenous plasma cell malignancy, for which the established prognostic models exhibit limitations in capturing the full spectrum of outcome variability. Leveraging single-cell RNA-sequencing data, we developed a novel plasma cell gene signature. We evaluated and validated the associations of the resulting plasma cell malignancy (PBM) score with disease state, progression and clinical outcomes using data from five independent myeloma studies consisting of 2115 samples (1978 MM, 65 monoclonal gammopathy of undetermined significance, 35 smoldering MM, and 37 healthy controls). Overall, a higher PBM score was significantly associated with a more advanced stage within the spectrum of plasma cell dyscrasias (all p < 0.05) and a shorter overall survival in MM (hazard ratio, HR = 1.72; p < 0.001). Notably, the prognostic effect of the PBM score was independent of the International Staging System (ISS) and Revised ISS (R-ISS). The downstream analysis further linked higher PBM scores with the presence of cytogenetic abnormalities, TP53 mutations, and compositional changes in the myeloma tumor immune microenvironment. Our integrated analyses suggest the PBM score may provide an opportunity for refining risk stratification and guide decisions on therapeutic approaches to MM.


Assuntos
Mieloma Múltiplo , Paraproteinemias , Humanos , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/genética , Plasmócitos , Prognóstico , Análise de Sequência de RNA , Microambiente Tumoral
4.
Nat Commun ; 15(1): 615, 2024 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-38242888

RESUMO

Anti-multiple myeloma B cell maturation antigen (BCMA)-specific chimeric antigen receptor (CAR) T-cell therapies represent a promising treatment strategy with high response rates in myeloma. However, durable cures following anti-BCMA CAR-T cell treatment of myeloma are rare. One potential reason is that a small subset of minimal residual myeloma cells seeds relapse. Residual myeloma cells following BCMA-CAR-T-mediated treatment show less-differentiated features and express stem-like genes, including CD24. CD24-positive myeloma cells represent a large fraction of residual myeloma cells after BCMA-CAR-T therapy. In this work, we develop CD24-CAR-T cells and test their ability to eliminate myeloma cells. We find that CD24-CAR-T cells block the CD24-Siglec-10 pathway, thereby enhancing macrophage phagocytic clearance of myeloma cells. Additionally, CD24-CAR-T cells polarize macrophages to a M1-like phenotype. A dual-targeted BCMA-CD24-CAR-T exhibits improved efficacy compared to monospecific BCMA-CAR-T-cell therapy. This work presents an immunotherapeutic approach that targets myeloma cells and promotes tumor cell clearance by macrophages.


Assuntos
Mieloma Múltiplo , Receptores de Antígenos Quiméricos , Humanos , Mieloma Múltiplo/patologia , Linfócitos T , Antígeno de Maturação de Linfócitos B/genética , Recidiva Local de Neoplasia , Anticorpos , Antígeno CD24
5.
J Clin Invest ; 134(1)2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-37883186

RESUMO

We have previously demonstrated that cystatin E/M (CST6), which is elevated in a subset of patients with multiple myeloma (MM) lacking osteolytic lesions (OLs), suppresses MM bone disease by blocking osteoclast differentiation and function. CST6 is a secreted type 2 cystatin, a cysteine protease inhibitor that regulates lysosomal cysteine proteases and the asparaginyl endopeptidase legumain. Here, we developed B cell maturation antigen (BCMA) CST6 chimeric antigen receptor T cells (CAR-T cells), which lysed MM cells and released CST6 proteins. Our in vitro studies show that these CAR-T cells suppressed the differentiation and formation of tartrate-resistant acid phosphatase-positive (TRAP+) osteoclasts. Using xenografted MM mice, bioluminescence images showed that both BCMA-CAR-T and BCMA-CST6-CAR-T cells inhibited MM growth to a similar extent. Reconstructed micro-computed tomography images revealed that BCMA-CST6-CAR-T cells, but not BCMA-CAR-T cells, prevented MM-induced bone damage and decreased osteoclast numbers. Our results provide a CAR-T strategy that targets tumor cells directly and delivers an inhibitor of bone resorption.


Assuntos
Mieloma Múltiplo , Receptores de Antígenos Quiméricos , Humanos , Animais , Camundongos , Mieloma Múltiplo/patologia , Imunoterapia Adotiva/métodos , Antígeno de Maturação de Linfócitos B , Linfócitos T , Microtomografia por Raio-X , Cistatina M
6.
Cell Rep Med ; 4(10): 101214, 2023 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-37794587

RESUMO

Multiple myeloma (MM) growth is supported by an immune-tolerant bone marrow microenvironment. Here, we find that loss of Never in mitosis gene A (NIMA)-related kinase 2 (NEK2) in tumor microenvironmental cells is associated with MM growth suppression. The absence of NEK2 leads to both fewer tumor-associated macrophages (TAMs) and inhibitory T cells. NEK2 expression in myeloid progenitor cells promotes the generation of functional TAMs when stimulated with MM conditional medium. Clinically, high NEK2 expression in MM cells is associated with increased CD8+ T effector memory cells, while low NEK2 is associated with an IFN-γ gene signature and activated T cell response. Inhibition of NEK2 upregulates PD-L1 expression in MM cells and myeloid cells. In a mouse model, the combination of NEK2 inhibitor INH154 with PD-L1 blockade effectively eliminates MM cells and prolongs survival. Our results provide strong evidence that NEK2 inhibition may overcome tumor immune escape and support its further clinical development.


Assuntos
Mieloma Múltiplo , Camundongos , Animais , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Antígeno B7-H1/genética , Linfócitos T/metabolismo , Linhagem Celular Tumoral , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patologia , Microambiente Tumoral
7.
Haematologica ; 108(12): 3372-3383, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37439384

RESUMO

Multiple Myeloma (MM) is a plasma cell neoplasm originating in the bone marrow and is the second most common blood cancer in the United States. One challenge in understanding the pathogenesis of MM and improving treatment is a lack of immunocompetent mouse models. We previously developed the IL6Myc mouse that generates plasmacytomas at 100% penetrance that phenotypically resemble aggressive MM. Using comprehensive genomic analysis, we found that the IL6Myc tumors resemble aggressive MM by RNA and protein expression. We also found that IL6Myc tumors accumulated fusions and missense mutations in genes that overlap significantly with human myeloma, indicating that the mouse is good model for studying disease etiology. Lastly, we derived cell lines from IL6Myc tumors that express cell surface markers typical of MM and readily engraft into mice, home to the bone marrow, and induce osteolytic disease. The cell lines may be useful in developing immunotherapies directed against BAFF-R and TACI, though not BCMA, and may also be a good model for studying dexamethasone resistance. These data indicate that the IL6Myc model is useful for studying development of aggressive MM and for developing new treatments against such forms of the disease.


Assuntos
Mieloma Múltiplo , Camundongos , Humanos , Animais , Mieloma Múltiplo/patologia , Medula Óssea/patologia
8.
J Hematol Oncol ; 16(1): 70, 2023 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-37386588

RESUMO

Multiple myeloma is preceded by monoclonal gammopathy of undetermined significance (MGUS). Serum markers are currently used to stratify MGUS patients into clinical risk groups. A molecular signature predicting MGUS progression has not been produced. We have explored the use of gene expression profiling to risk-stratify MGUS and developed an optimized signature based on large samples with long-term follow-up. Microarrays of plasma cell mRNA from 334 MGUS with stable disease and 40 MGUS that progressed to MM within 10 years, was used to define a molecular signature of MGUS risk. After a three-fold cross-validation analysis, the top thirty-six genes that appeared in each validation and maximized the concordance between risk score and MGUS progression were included in the gene signature (GS36). The GS36 accurately predicted MGUS progression (C-statistic is 0.928). An optimal cut-point for risk of progression by the GS36 score was found to be 0.7, which identified a subset of 61 patients with a 10-year progression probability of 54.1%. The remainder of the 313 patients had a probability of progression of only 2.2%. The sensitivity and specificity were 82.5% and 91.6%. Furthermore, combination of GS36, free light chain ratio and immunoparesis identified a subset of MGUS patients with 82.4% risk of progression to MM within 10 years. A gene expression signature combined with serum markers created a highly robust model for predicting risk of MGUS progression. These findings strongly support the inclusion of genomic analysis in the management of MGUS to identify patients who may benefit from more frequent monitoring.


Assuntos
Gamopatia Monoclonal de Significância Indeterminada , Mieloma Múltiplo , Humanos , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/genética , Gamopatia Monoclonal de Significância Indeterminada/diagnóstico , Gamopatia Monoclonal de Significância Indeterminada/genética , Plasmócitos , Perfilação da Expressão Gênica , Genômica
9.
Cancer Immunol Immunother ; 72(4): 969-984, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36227341

RESUMO

Antiangiogenic therapy has shown significant clinical benefits in gastric cancer (GC) and non-small cell lung cancer (NSCLC). However, their effectiveness is limited by the immunosuppressive tumor microenvironment. The MHC class I chain-related molecules A and B (MICA/B) are expressed in many human cancers, enabling elimination of cancer cells by cytotoxic lymphocytes through natural killer group 2D (NKG2D) receptor activation. To improve antiangiogenic therapy and prolong its efficacy, we generated a bi-specific fusion protein (mAb04-MICA). This was comprised of an antibody targeting VEGFR2 fused to a MICA α1-α2 ectodomain. mAb04-MICA inhibited proliferation of GC and NSCLC cells through specific binding to VEGFR2 and had superior anti-tumor efficacy in both GC and NSCLC-bearing mouse models compared with ramucirumab. Further investigation revealed that the mAb04-MICA promoted NKG2D+ NK cell activation and induced the tumor-associated macrophage (TAM) polarization from M2 type to M1 type both in vitro and in vivo. The polarization of TAMs upon NKG2D and MICA mediated activation has not yet been reported. Moreover, given the up-regulation of PD-L1 in tumors during anti-angiogenesis therapy, anti-PD-1 antibody enhanced the anti-tumoral activity of mAb04-MICA through stimulating infiltration and activation of NKs and CD8+T cells in responding tumors. Our findings demonstrate that dual targeting of angiogenesis and NKG2D, or in combination with the PD-1/PD-L1 blockade, is a promising anti-tumor therapeutic strategy. This is accomplished through maintaining or reinstating tumor immunosurveillance during treatment, which expands the repertoire of anti-angiogenesis-based cancer immunotherapies.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Animais , Humanos , Camundongos , Anticorpos/farmacologia , Antígeno B7-H1/metabolismo , Carcinoma Pulmonar de Células não Pequenas/terapia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Antígenos de Histocompatibilidade Classe I , Imunoterapia , Células Matadoras Naturais , Neoplasias Pulmonares/terapia , Neoplasias Pulmonares/metabolismo , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Microambiente Tumoral , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
10.
Oncogene ; 41(32): 3899-3911, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35794249

RESUMO

The transcription factor, forkhead box M1 (FOXM1), has been implicated in the natural history and outcome of newly diagnosed high-risk myeloma (HRMM) and relapsed/refractory myeloma (RRMM), but the mechanism with which FOXM1 promotes the growth of neoplastic plasma cells is poorly understood. Here we show that FOXM1 is a positive regulator of myeloma metabolism that greatly impacts the bioenergetic pathways of glycolysis and oxidative phosphorylation (OxPhos). Using FOXM1-deficient myeloma cells as principal experimental model system, we find that FOXM1 increases glucose uptake, lactate output, and oxygen consumption in myeloma. We demonstrate that the novel 1,1-diarylethylene small-compound FOXM1 inhibitor, NB73, suppresses myeloma in cell culture and human-in-mouse xenografts using a mechanism that includes enhanced proteasomal FOXM1 degradation. Consistent with the FOXM1-stabilizing chaperone function of heat shock protein 90 (HSP90), the HSP90 inhibitor, geldanamycin, collaborates with NB73 in slowing down myeloma. These findings define FOXM1 as a key driver of myeloma metabolism and underscore the feasibility of targeting FOXM1 for new approaches to myeloma therapy and prevention.


Assuntos
Mieloma Múltiplo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Proteína Forkhead Box M1/genética , Proteína Forkhead Box M1/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glicólise , Humanos , Camundongos , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Fatores de Transcrição/metabolismo
11.
J Clin Invest ; 132(18)2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-35881476

RESUMO

Osteolytic bone disease is a hallmark of multiple myeloma (MM). A significant fraction (~20%) of MM patients do not develop osteolytic lesions (OLs). The molecular basis for the absence of bone disease in MM is not understood. We combined PET-CT and gene expression profiling (GEP) of purified BM CD138+ MM cells from 512 newly diagnosed MM patients to reveal that elevated expression of cystatin M/E (CST6) was significantly associated with the absence of OL in MM. An enzyme-linked immunosorbent assay revealed a strong correlation between CST6 levels in BM serum/plasma and CST6 mRNA expression. Both recombinant CST6 protein and BM serum from patients with high CST6 significantly inhibited the activity of the osteoclast-specific protease cathepsin K and blocked osteoclast differentiation and function. Recombinant CST6 inhibited bone destruction in ex vivo and in vivo myeloma models. Single-cell RNA-Seq showed that CST6 attenuates polarization of monocytes to osteoclast precursors. Furthermore, CST6 protein blocks osteoclast differentiation by suppressing cathepsin-mediated cleavage of NF-κB/p100 and TRAF3 following RANKL stimulation. Secretion by MM cells of CST6, an inhibitor of osteoclast differentiation and function, suppresses osteolytic bone disease in MM and probably other diseases associated with osteoclast-mediated bone loss.


Assuntos
Reabsorção Óssea , Mieloma Múltiplo , Osteólise , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Diferenciação Celular/fisiologia , Cistatina M/metabolismo , Humanos , Mieloma Múltiplo/complicações , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Osteoclastos/metabolismo , Osteólise/genética , Osteólise/metabolismo , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Ligante RANK/genética , Ligante RANK/metabolismo , Fator 3 Associado a Receptor de TNF/metabolismo
12.
Sci Rep ; 12(1): 6551, 2022 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-35449393

RESUMO

Acute graft-versus-host disease (aGVHD) is the main complication of and cause of death after allogeneic hematopoietic stem cell transplantation. Baicalin can protect the small intestinal epithelial cells of rats against TNF-α-induced injury and alleviate enteritis-related diarrhea. To verify whether baicalin can protect the small intestinal mucosal barrier by regulating abnormal autophagy and interfering with intestinal aGVHD, a mouse model of aGVHD was established. CB6F1 micewere intravenously injected with a suspension of mononuclear cells derived from BALB/c donor mouse bone marrow and splenic tissue after treatment with 60Co X-rays. After treatment with different doses of baicalin for 15 days, the survival time, serum TNF-α and IL-10 levels, and autophagy markers levels in the intestine were assessed. A cell model of intestinal barrier dysfunction was also used to verify the effect of baicalin. The results showed that baicalin significantly prolonged the survival time, significantly reduced the aGVHD pathology score and clinical score by decreasing the TNF-α level with increasing the IL-10 level compared with the control. Transmission electron microscopy examination showed that baicalin treatment increased the number of autophagic vacuoles and led to the recovery of mitochondrial structures in the intestinal mucosal epithelial cells of mice and in Caco-2 cells. Western blotting results showed that baicalin treatment enhanced autophagy in vivo by regulating the AMPK/mTOR autophagy pathway. Similar results were observed in vitro in Caco-2 cells. Furthermore, the effect of baicalin was reduced after combination treatment with the autophagy inhibitor 3-methyladenine(3-MA). Baicalin can decrease the severity of small intestinal aGVHD by regulating autophagy by influencing imbalances in inflammatory cytokine levels and mucosal barrier damage, thus baicalin may have potential as a new treatment for aGVHD.


Assuntos
Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Doença Aguda , Animais , Autofagia , Células CACO-2 , Flavonoides , Doença Enxerto-Hospedeiro/patologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Interleucina-10 , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/uso terapêutico
13.
Mol Cancer Ther ; 21(4): 502-510, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35086951

RESUMO

Dysregulated c-myc is a determinant of multiple myeloma progression. Translation of c-myc can be achieved by an mTOR-mediated, cap-dependent mechanism or a cap-independent mechanism where a sequence in the 5'UTR of mRNA, termed the internal ribosome entry site (IRES), recruits the 40S ribosomal subunit. This mechanism requires the RNA-binding factor hnRNP A1 (A1) and becomes critical when cap-dependent translation is inhibited during endoplasmic reticulum (ER) stress. Thus, we studied the role of A1 and the myc IRES in myeloma biology. A1 expression correlated with enhanced c-myc expression in patient samples. Expression of A1 in multiple myeloma lines was mediated by c-myc itself, suggesting a positive feedback circuit where myc induces A1 and A1 enhances myc translation. We then deleted the A1 gene in a myc-driven murine myeloma model. A1-deleted multiple myeloma cells demonstrated downregulated myc expression and were inhibited in their growth in vivo. Decreased myc expression was due to reduced translational efficiency and depressed IRES activity. We also studied the J007 inhibitor, which prevents A1's interaction with the myc IRES. J007 inhibited myc translation and IRES activity and diminished myc expression in murine and human multiple myeloma lines as well as primary samples. J007 also inhibited tumor outgrowth in mice after subcutaneous or intravenous challenge and prevented osteolytic bone disease. When c-myc was ectopically reexpressed in A1-deleted multiple myeloma cells, tumor growth was reestablished. These results support the critical role of A1-dependent myc IRES translation in myeloma.


Assuntos
Ribonucleoproteína Nuclear Heterogênea A1 , Camundongos , Mieloma Múltiplo , Proteínas Proto-Oncogênicas c-myc , Animais , Genes myc , Ribonucleoproteína Nuclear Heterogênea A1/metabolismo , Humanos , Sítios Internos de Entrada Ribossomal , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Biossíntese de Proteínas , Proteínas Proto-Oncogênicas c-myc/metabolismo
14.
J Hematol Oncol ; 14(1): 203, 2021 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-34876184

RESUMO

Unbiased genetic forward screening using retroviral insertional mutagenesis in a genetically engineered mouse model of human multiple myeloma may further our understanding of the genetic pathways that govern neoplastic plasma cell development. To evaluate this hypothesis, we performed a tumor induction study in MYC-transgenic mice infected as neonates with the Moloney-derived murine leukemia virus, MOL4070LTR. Next-generation DNA sequencing of proviral genomic integration sites yielded rank-ordered candidate tumor progression genes that accelerated plasma cell neoplasia in mice. Rigorous clinical and biological validation of these genes led to the discovery of two novel myeloma genes: WDR26 (WD repeat-containing protein 26) and MTF2 (metal response element binding transcription factor 2). WDR26, a core component of the carboxy-terminal to LisH (CTLH) complex, is overexpressed or mutated in solid cancers. MTF2, an ancillary subunit of the polycomb repressive complex 2 (PRC2), is a close functional relative of PHD finger protein 19 (PHF19) which is currently emerging as an important driver of myeloma. These findings underline the utility of genetic forward screens in mice for uncovering novel blood cancer genes and suggest that WDR26-CTLH and MTF2-PRC2 are promising molecular targets for new approaches to myeloma treatment and prevention.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Mieloma Múltiplo/genética , Complexo Repressor Polycomb 2/genética , Animais , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Transgênicos , Mieloma Múltiplo/terapia
15.
Front Immunol ; 12: 667054, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34149703

RESUMO

Mouse models of human cancer provide an important research tool for elucidating the natural history of neoplastic growth and developing new treatment and prevention approaches. This is particularly true for multiple myeloma (MM), a common and largely incurable neoplasm of post-germinal center, immunoglobulin-producing B lymphocytes, called plasma cells, that reside in the hematopoietic bone marrow (BM) and cause osteolytic lesions and kidney failure among other forms of end-organ damage. The most widely used mouse models used to aid drug and immunotherapy development rely on in vivo propagation of human myeloma cells in immunodeficient hosts (xenografting) or myeloma-like mouse plasma cells in immunocompetent hosts (autografting). Both strategies have made and continue to make valuable contributions to preclinical myeloma, including immune research, yet are ill-suited for studies on tumor development (oncogenesis). Genetically engineered mouse models (GEMMs), such as the widely known Vκ*MYC, may overcome this shortcoming because plasma cell tumors (PCTs) develop de novo (spontaneously) in a highly predictable fashion and accurately recapitulate many hallmarks of human myeloma. Moreover, PCTs arise in an intact organism able to mount a complete innate and adaptive immune response and tumor development reproduces the natural course of human myelomagenesis, beginning with monoclonal gammopathy of undetermined significance (MGUS), progressing to smoldering myeloma (SMM), and eventually transitioning to frank neoplasia. Here we review the utility of transplantation-based and transgenic mouse models of human MM for research on immunopathology and -therapy of plasma cell malignancies, discuss strengths and weaknesses of different experimental approaches, and outline opportunities for closing knowledge gaps, improving the outcome of patients with myeloma, and working towards a cure.


Assuntos
Gamopatia Monoclonal de Significância Indeterminada/diagnóstico , Gamopatia Monoclonal de Significância Indeterminada/tratamento farmacológico , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/tratamento farmacológico , Animais , Progressão da Doença , Humanos , Imunoterapia , Camundongos , Gamopatia Monoclonal de Significância Indeterminada/patologia , Mieloma Múltiplo/patologia
16.
Blood Rev ; 46: 100741, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32807576

RESUMO

The autonomic nervous system (ANS), which consists of antagonistic sympathetic (adrenergic) and parasympathetic (cholinergic) arms, has emerged as an important regulator of neoplastic development, yet little is known about its role in multiple myeloma (MM). Clinical findings that anti-adrenergic ß-blocker intake reduces risk of disease-specific death and overall mortality in patients with MM have indicated that adrenergic input may worsen myeloma outcome. However, preclinical studies using ß-adrenergic receptor agonists or antagonists produced controversial results as to whether sympathetic pathways promote or inhibit myeloma. Retrospective outcome data demonstrating that high message levels of cholinergic receptor genes predict inferior survival in the Multiple Myeloma Research Foundation CoMMpass trial suggest that parasympathetic input may drive myeloma progression in a subset of patients. Here we review the ill-defined role of the ANS in MM, put myeloma in the context of other cancers, and discuss knowledge gaps that may afford exciting research opportunities going forward.


Assuntos
Sistema Nervoso Autônomo/metabolismo , Suscetibilidade a Doenças , Mieloma Múltiplo/etiologia , Mieloma Múltiplo/metabolismo , Antagonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/uso terapêutico , Animais , Sistema Nervoso Autônomo/efeitos dos fármacos , Sistema Nervoso Autônomo/fisiopatologia , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Medula Óssea/patologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Osso e Ossos/inervação , Osso e Ossos/metabolismo , Gerenciamento Clínico , Progressão da Doença , Sinergismo Farmacológico , Humanos , Mieloma Múltiplo/patologia , Mieloma Múltiplo/terapia , Prognóstico , Transdução de Sinais/efeitos dos fármacos , Nicho de Células-Tronco/efeitos dos fármacos , Resultado do Tratamento
17.
Cell Signal ; 78: 109872, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33290841

RESUMO

Peripheral neuropathy (PN) is an incurable complication of multiple myeloma (MM) which adversely affects patients' quality of life. The important roles that Circular RNAs (circRNAs) play in tumor progression, and exosome-mediated intracellular communication has been recognized as a crucial factor in the pathogenesis of MM. However, the role of exosome-derived circRNAs (exo-circRNAs) in MM and MM-induced PN remains elusive. In this study, we aimed to investigate the correlation between serum exo-circRNAs and MM to preliminarily explore the role of exo-circRNAs in MM-related PN. A cohort of 25 MM patients and 5 healthy control (HC) individuals were enrolled in the study. High-throughput sequencing and qRT PCR validation of serum exo-circRNAs were used to generate the aberrantly expressed exo-circRNAs profiles. Bioinformatics analysis was done using GO, KEGG, miRanda, Targetscan and Metascape. Correlation analysis was conducted between chr2:2744228-2,744,407+ and clinical characteristics of PN. ROC curve, univariate and multivariate COX regression models were conducted to identify the prognostic potential of chr2:2744228-2,744,407+ in the MM-related PN. 265 upregulated circRNAs and 787 downregulated circRNAs, with at least a two-fold difference in expression level in MM patients vs HC, were screened. Bioinformatics analysis indicated that upregulated circRNAs had the potential to facilitate MM-related PN. Furthermore, PCR validated the abundant expression of chr2:2744228-2,744,407+ in the serum exosomes of 25 MM patients. Bioinformatics analysis indicated that chr2:2744228-2,744,407+ might induce MM related PN via the downstream miRNA and GRIN2B axis. Overexpressed chr2:2744228-2,744,407+ in the serum exosomes of MM patients might lead to the downregulation of hsa-miR-6829-3p, elevation of GRIN2B in the serum and PC12 cells, and inhibited cell viability. The correlation analysis indicated that the expression of chr 2:2744228-2,744,407+ was positively correlated with the clinical characteristics of PN. ROC curve, univariate and multivariate COX regression analysis identified that chr2:2744228-2,744,407+ is an independent prognostic factor in the MM related PN. We identified that the abnormal expression of the serum exo-circRNA was correlated with MM-related PN, implying that exo-circRNA has potential as a novel therapeutic target for MM related PN.


Assuntos
Ácidos Nucleicos Livres , Exossomos , Mieloma Múltiplo , Doenças do Sistema Nervoso Periférico , RNA Circular , RNA Neoplásico , Adulto , Animais , Ácidos Nucleicos Livres/sangue , Ácidos Nucleicos Livres/genética , Exossomos/genética , Exossomos/metabolismo , Humanos , Mieloma Múltiplo/sangue , Mieloma Múltiplo/complicações , Mieloma Múltiplo/genética , Mieloma Múltiplo/terapia , Células PC12 , Doenças do Sistema Nervoso Periférico/sangue , Doenças do Sistema Nervoso Periférico/etiologia , Doenças do Sistema Nervoso Periférico/genética , Doenças do Sistema Nervoso Periférico/terapia , RNA Circular/sangue , RNA Circular/genética , RNA Neoplásico/sangue , RNA Neoplásico/genética , Ratos
19.
Biomed Pharmacother ; 117: 109121, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31252265

RESUMO

Breast cancer is a fairly common cancer with high mortality in women worldwide. Targeted nano-drug delivery system for breast cancer treatment has achieved encouraging results, because of increased drug concentration at the tumor site, thereby improving biocompatibility and blood half-life while reducing chemoresistance. However, the absence of available target on cancer cells is one of the major obstacles for triple-negative breast cancer (TNBC). Increasing studies have shown that heparanase (HPA) is highly expressed in many cancers, including TNBC. Thus paclitaxel(PTX) -encapsulated PEGylated PLGA nanoparticles were developed and further surface-functionalized with the HPA aptamers (Apt(S1.5)-PTX-NP). Moreover, targeting and cytotoxicity of Apt(S1.5)-PTX-NP to TNBC cells were evaluated with MDA-MB-231 as a model. These nanoparticles bonded to the HPA overexpressed on the surface of TNBC cells and were taken up by these cells, resulting in remarkably enhanced cellular toxicity compared with non-targeted PTX-NP that lack the HPA aptamer (P < 0.01). Furthermore, Apt(S1.5)-PTX-NP significantly exhibited enhanced anti-invasive and superior anti-angiogenesis activity compared with those of other experiment groups at low administration dosage. The Apt(S1.5)-PTX-NP demonstrated the most dramatic efficacy with the final mean tumor sizes of 157.30 ± 41.09 mm3 (mean ± SD; n = 10) in vivo treatment. Thus, the present study indicated that HPA is a promising target for drug delivery to TNBC cells, and nanoparticle-HPA-aptamer bioconjugates can provide new insights for TNBC treatment.


Assuntos
Antineoplásicos/farmacologia , Aptâmeros de Nucleotídeos/química , Glucuronidase/química , Terapia de Alvo Molecular , Nanopartículas/química , Paclitaxel/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Microambiente Tumoral/efeitos dos fármacos , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Endocitose , Fluorescência , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Camundongos , Nanopartículas/ultraestrutura , Invasividade Neoplásica , Neovascularização Fisiológica/efeitos dos fármacos
20.
Front Genet ; 10: 424, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31139207

RESUMO

Genomic instability, a well-established hallmark of human cancer, is also a driving force in the natural history of multiple myeloma (MM) - a difficult to treat and in most cases fatal neoplasm of immunoglobulin producing plasma cells that reside in the hematopoietic bone marrow. Long recognized manifestations of genomic instability in myeloma at the cytogenetic level include abnormal chromosome numbers (aneuploidy) caused by trisomy of odd-numbered chromosomes; recurrent oncogene-activating chromosomal translocations that involve immunoglobulin loci; and large-scale amplifications, inversions, and insertions/deletions (indels) of genetic material. Catastrophic genetic rearrangements that either shatter and illegitimately reassemble a single chromosome (chromotripsis) or lead to disordered segmental rearrangements of multiple chromosomes (chromoplexy) also occur. Genomic instability at the nucleotide level results in base substitution mutations and small indels that affect both the coding and non-coding genome. Sometimes this generates a distinctive signature of somatic mutations that can be attributed to defects in DNA repair pathways, the DNA damage response (DDR) or aberrant activity of mutator genes including members of the APOBEC family. In addition to myeloma development and progression, genomic instability promotes acquisition of drug resistance in patients with myeloma. Here we review recent findings on the genetic predisposition to myeloma, including newly identified candidate genes suggesting linkage of germline risk and compromised genomic stability control. The role of ethnic and familial risk factors for myeloma is highlighted. We address current research gaps that concern the lack of studies on the mechanism by which germline risk alleles promote genomic instability in myeloma, including the open question whether genetic modifiers of myeloma development act in tumor cells, the tumor microenvironment (TME), or in both. We conclude with a brief proposition for future research directions, which concentrate on the biological function of myeloma risk and genetic instability alleles, the potential links between the germline genome and somatic changes in myeloma, and the need to elucidate genetic modifiers in the TME.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...