Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 141
Filtrar
1.
J Cancer Res Clin Oncol ; 149(11): 9277-9284, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37202579

RESUMO

PURPOSE: Unfortunately, cure of multi-drug resistant (MDR) hematologic malignancies remains an unmet need. Donor lymphocyte infusion (DLI) following allogeneic stem cell transplantation (SCT) can sometimes eliminate multi-drug resistant leukemia but at a risk of acute and chronic graft-vs-host disease (GVHD) and procedure-related toxicity. Supported by pre-clinical experiments in animal models, we hypothesized that immunotherapy induced by non-engrafting intentionally mismatched IL-2 activated killers (IMAK) including both T & NK cells could induce safer, faster and much more effective immunotherapy while avoiding the need for SCT and the risks of GVHD. METHODS: IMAK treatment was applied in 33 patients with MDR hematologic malignancies conditioned with cyclophosphamide 1000 mg/m2 based protocol. Haploidentical or unrelated donor lymphocytes were preactivated with IL-2 6000 IU/ml for 4 days. IMAK was combined with Rituximab in 12/23 patients with CD20+ B cells. RESULTS: A total of 23/33 patients with MDR (4 failing SCT) achieved complete remission (CR). First patient currently 30 years with no further treatment and 6 observed for > 5 years (2 AML; 2 multiple myeloma, 1 ALL & 1 NHL) can be considered cured. No patient developed > grade 3 toxicity or GVHD. No residual male cells were detectable among six females treated with male cells beyond day + 6, confirming that GVHD was prevented by consistent early rejection of donor lymphocytes. CONCLUSIONS: We hypothesize that safe and superior immunotherapy of MDR with cure potential may be accomplished by IMAK, most probably in patients with low tumor burden, but that remains to be confirmed by future clinical trials.


Assuntos
Doença Enxerto-Hospedeiro , Neoplasias Hematológicas , Transplante de Células-Tronco Hematopoéticas , Feminino , Animais , Masculino , Interleucina-2/uso terapêutico , Neoplasias Hematológicas/terapia , Linfócitos , Transplante de Células-Tronco Hematopoéticas/métodos , Imunoterapia/métodos , Doença Enxerto-Hospedeiro/prevenção & controle
2.
Infect Disord Drug Targets ; 22(7): 1-6, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35440336

RESUMO

More than 200 viruses infect humans, but treatments are available for less than ten of them. To narrow the gap between 'bugs and drugs,' a paradigm shift is required. The "one drug, one bug" approach can be expanded to a "one drug, multiple bugs" strategy such that the host's defense system is targeted rather than the virus. Viral superinfection therapy (SIT) activates interferon genes' natural, antiviral defense system of host cells following exposure to viral infection, e.g., superinfection with an attenuated infectious bursal disease virus (IBDV) with the release of its double-stranded RNA (dsRNA) cargo inside host cells. An attenuated IBDV therapeutic vaccine has successfully treated hepatitis A virus infection (HAV) in marmoset monkeys as well as acute hepatitis B and hepatitis C virus infections (HBV/HCV) in 42 patients. SIT has also been shown to be safe and effective in four patients with chronic HBV or HCV infection with hepatic decompensation. The proof-of-principle of SIT has also been demonstrated in a 43-year-old male patient with COVID-19. Three doses of orally administered IBDV (3x106 IU) alleviated most of his COVID-19 symptoms; even his sense of smell returned within a week. Two additional COVID-19 patients responded similarly to oral treatment with IBDV. Furthermore, a severe herpes zoster ophthalmicus outbreak with orbital edema responded to a combination of acyclovir and 7 doses of IBDV (7x106 IU) within a few days. IBDV is simple to manufacture and affordable, even in resource-limited settings. Acid-resistant IBDV can be orally administered in an outpatient setting, providing simple dosing and high medication adherence. Under an Emergency Use Authorization, the broad-spectrum IBDV drug candidate could be tested immediately in clinical trials and rapidly distributed to millions of early-stage patients with COVID-19. The German Paul Ehrlich Institute is currently supporting a phase I safety study for persons acutely infected with SARS­CoV-2. An expert team of the US National Institutes of Health-sponsored ACTIV public-private partnership came to the conclusion that the IBDV drug candidate shows merit as a potential treatment for COVID19, and an FDA-approved clinical trial is in the pipelines in Los Angeles.

3.
Cancer Immunol Immunother ; 69(5): 683-687, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32152702

RESUMO

More than 2000 immuno-oncology agents are being tested or are in use as a result of the cancer immunotherapy revolution. Manipulation of co-inhibitory receptors has achieved tumor eradication in a minority of patients, but widespread immune-related adverse events (irAEs) compromised tolerance to healthy self-tissues in the majority. We have proposed that a major mechanism of irAEs is similar to a graft-versus-malignancy effect of graft-versus-host disease. To verify our hypothesis, we retrieved post-marketing data of adverse events from the U.S. Food and Drug Administration Adverse Event Reporting System. A significant positive correlation was revealed in 7677 patients between the reporting odds ratio of irAEs during immune checkpoint inhibitor therapy and the corresponding tumor mutational burden across 19 cancer types. These results can be interpreted to mean that the ICI drugs unleashed T cells against "altered-self," self, and tumors resulting in better overall survival.


Assuntos
Antineoplásicos Imunológicos/efeitos adversos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/genética , Neoplasias/tratamento farmacológico , Tolerância a Antígenos Próprios/genética , Linfócitos T/efeitos dos fármacos , Sistemas de Notificação de Reações Adversas a Medicamentos/estatística & dados numéricos , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/imunologia , Humanos , Mutação , Neoplasias/genética , Neoplasias/imunologia , Tolerância a Antígenos Próprios/efeitos dos fármacos , Linfócitos T/imunologia
4.
PLoS One ; 14(9): e0222354, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31513644

RESUMO

INTRODUCTION: The primary rational for using mesenchymal stromal cells (MSCs) to rejuvenate damaged tissue is mostly based on their capacity to trans-differentiate and repair injured organs. However, previous studies have demonstrated that MSCs are beneficial even at very early stages, before differentiation and proliferation can be expected. The aim of the current study was to investigate the multifaceted immunological effects of systemically administrating MSCs in the setting of acute kidney injury (AKI) induced by ischemic-reperfusion (I/R). METHODS: A rat model of I/R induced AKI was used. The rats underwent a unilateral nephrectomy with simultaneously clamping the contralateral kidney for 60 minutes. Four treatment groups received intravenously, increasing doses of human MSCs and after 48 hours, the rats were sacrificed. Blood was taken to evaluate renal functions and to measure systemic inflammatory markers. Kidneys were taken for histopathologic examinations and evaluations of intra-renal complement activation and inflammatory mediators. RESULTS: Renal functions improved in U shaped dose dependent manner. Mean serum creatinine levels were 4.5, 2.9, 2.6, 1.7 and 4.1 mg/dL in I/R + placebo, I/R + 150x103 cells, I/R + 250x103 cells, I/R + 500x103 cells and I/R + 1,000x103 cells respectfully (p-values<0.05). Urea demonstrated consistent results with the same U shape improvement manner. The extensive activation of the complement system was ameliorated in the MSCs treatment groups. In addition, MSCs significantly decreased intra-renal levels of IL-1ß and TNF-α. It should be noted that the highest doses of MSCs induced renal hypoxia, marked by the Hypoxy-probe staining. CONCLUSIONS: The early beneficial effect of MSCs in the setting of AKI may be attributed to their immunomodulatory effects. Safe treatment with MSCs can block the deleterious activation of the complement cascade and alleviate the hazardous inflammatory mediator-related cascade.


Assuntos
Injúria Renal Aguda/terapia , Células-Tronco Mesenquimais/imunologia , Traumatismo por Reperfusão/terapia , Injúria Renal Aguda/patologia , Animais , Ativação do Complemento/imunologia , Modelos Animais de Doenças , Humanos , Inflamação/metabolismo , Rim/metabolismo , Rim/patologia , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/patologia
5.
Cell Transplant ; 27(3): 520-530, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29737200

RESUMO

Acute kidney injury (AKI) is a major clinical problem that still has no established treatment. We investigated the efficacy of cultured human peripheral blood mononuclear cells (PBMNCs) for AKI. Ischemia/reperfusion injury (IRI) was used to induce AKI in male nonobese diabetic (NOD/severe combined immunodeficiency) mice aged 7 to 8 wk. PBMNCs were isolated from healthy volunteers and were subjected to quality and quantity controlled (QQc) culture for 7 d in medium containing stem cell factor, thrombopoietin, Flt-3 ligand, vascular endothelial growth factor, and interleukin 6. IRI-induced mice were divided into 3 groups and administered (1) 1 × 106 PBMNCs after QQc culture (QQc PBMNCs group), (2) 1 × 106 PBMNCs without QQc culture (non-QQc PBMNCs group), or (3) vehicle without PBMNCs (IRI control group). PBMNCs were injected via the tail vein 24 h after induction of IRI, followed by assessment of renal function, histological changes, and homing of injected cells. Blood urea nitrogen and serum creatinine (Cr) 72 h after induction of IRI in the QQc PBMNCs group dramatically improved compared with those in the IRI control and the non-QQc PBMNCs groups, accompanied by the improvement of tubular damages. Interstitial fibrosis 14 d after induction of IRI was also significantly improved in the QQc PBMNCs group compared with the other groups. The renoprotective effect noted in the QQc PBMNCs group was accompanied by reduction of peritubular capillary loss. The change of PBMNCs' population (increase of CD34+ cells, CD133+ cells, and CD206+ cells) and increased endothelial progenitor cell colony-forming potential by QQc culture might be one of the beneficial mechanisms for restoring AKI. In conclusion, an injection of human QQc PBMNCs 24 h after induction of IRI dramatically improved AKI in mice.


Assuntos
Injúria Renal Aguda/terapia , Meios de Cultivo Condicionados/farmacologia , Leucócitos Mononucleares/fisiologia , Traumatismo por Reperfusão/terapia , Animais , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Trombopoetina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
Stem Cell Res Ther ; 7(1): 149, 2016 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-27724977

RESUMO

BACKGROUND: Newcastle disease virus (NDV) is an avian paramyxovirus, which selectively exerts oncolytic effects in cancer cells. Mesenchymal stem cells (MSCs) have been reported to affect tumor growth and deliver anti-tumor agents to experimental glioblastoma (GBM). Here, we explored the effects of NDV-infected MSCs derived from different sources, on glioma cells and glioma stem cells (GSCs) and the mechanisms involved in their effects. METHODS: The glioma cell lines (A172 and U87) and primary GSCs that were generated from GBM tumors were used in this study. MSCs derived from bone marrow, adipose tissue or umbilical cord were infected with NDV (MTH-68/H). The ability of these cells to deliver the virus to glioma cell lines and GSCs and the effects of NDV-infected MSCs on cell death and on the stemness and self-renewal of GSCs were examined. The mechanisms involved in the cytotoxic effects of the NDV-infected MSCs and their influence on the radiation sensitivity of GSCs were examined as well. RESULTS: NDV induced a dose-dependent cell death in glioma cells and a low level of apoptosis and inhibition of self-renewal in GSCs. MSCs derived from bone marrow, adipose and umbilical cord that were infected with NDV delivered the virus to co-cultured glioma cells and GSCs. Conditioned medium of NDV-infected MSCs induced higher level of apoptosis in the tumor cells compared with the apoptosis induced by their direct infection with similar virus titers. These results suggest that factor(s) secreted by the infected MSCs sensitized the glioma cells to the cytotoxic effects of NDV. We identified TRAIL as a mediator of the cytotoxic effects of the infected MSCs and demonstrated that TRAIL synergized with NDV in the induction of cell death in glioma cells and GSCs. Moreover, conditioned medium of infected MSCs enhanced the sensitivity of GSCs to γ-radiation. CONCLUSIONS: NDV-infected umbilical cord-derived MSCs may provide a novel effective therapeutic approach for targeting GSCs and GBM and for sensitizing these tumors to γ-radiation.


Assuntos
Glioma/terapia , Glioma/virologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Neoplásicas/citologia , Vírus da Doença de Newcastle/fisiologia , Vírus Oncolíticos/fisiologia , Apoptose/fisiologia , Morte Celular/fisiologia , Linhagem Celular Tumoral , Células Cultivadas , Técnicas de Cocultura/métodos , Glioblastoma/metabolismo , Glioblastoma/terapia , Glioblastoma/virologia , Glioma/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/virologia , Células-Tronco Neoplásicas/metabolismo , Terapia Viral Oncolítica/métodos , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Cordão Umbilical/citologia , Cordão Umbilical/metabolismo
7.
Blood ; 127(7): 938-47, 2016 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-26670632

RESUMO

We evaluated 917 adult lymphoma patients who received haploidentical (n = 185) or HLA-matched unrelated donor (URD) transplantation either with (n = 241) or without antithymocyte globulin (ATG; n = 491) following reduced-intensity conditioning regimens. Haploidentical recipients received posttransplant cyclophosphamide-based graft-versus-host disease (GVHD) prophylaxis, whereas URD recipients received calcineurin inhibitor-based prophylaxis. Median follow-up of survivors was 3 years. The 100-day cumulative incidence of grade III-IV acute GVHD on univariate analysis was 8%, 12%, and 17% in the haploidentical, URD without ATG, and URD with ATG groups, respectively (P = .44). Corresponding 1-year rates of chronic GVHD on univariate analysis were 13%, 51%, and 33%, respectively (P < .001). On multivariate analysis, grade III-IV acute GVHD was higher in URD without ATG (P = .001), as well as URD with ATG (P = .01), relative to haploidentical transplants. Similarly, relative to haploidentical transplants, risk of chronic GVHD was higher in URD without ATG and URD with ATG (P < .0001). Cumulative incidence of relapse/progression at 3 years was 36%, 28%, and 36% in the haploidentical, URD without ATG, and URD with ATG groups, respectively (P = .07). Corresponding 3-year overall survival (OS) was 60%, 62%, and 50% in the 3 groups, respectively, with multivariate analysis showing no survival difference between URD without ATG (P = .21) or URD with ATG (P = .16), relative to haploidentical transplants. Multivariate analysis showed no difference between the 3 groups in terms of nonrelapse mortality (NRM), relapse/progression, and progression-free survival (PFS). These data suggest that reduced-intensity conditioning haploidentical transplantation with posttransplant cyclophosphamide does not compromise early survival outcomes compared with matched URD transplantation, and is associated with significantly reduced risk of chronic GVHD.


Assuntos
Soro Antilinfocitário/administração & dosagem , Ciclofosfamida/administração & dosagem , Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Linfoma , Condicionamento Pré-Transplante , Doadores não Relacionados , Adolescente , Adulto , Idoso , Aloenxertos , Doença Crônica , Intervalo Livre de Doença , Feminino , Doença Enxerto-Hospedeiro/mortalidade , Doença Enxerto-Hospedeiro/prevenção & controle , Teste de Histocompatibilidade , Humanos , Linfoma/mortalidade , Linfoma/terapia , Masculino , Pessoa de Meia-Idade , Taxa de Sobrevida
8.
Biol Blood Marrow Transplant ; 21(12): 2091-2099, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26253007

RESUMO

This study was conducted to compare long-term outcomes in patients with refractory/relapsed grades 1 and 2 follicular lymphoma (FL) after allogeneic (allo) versus autologous (auto) hematopoietic cell transplantation (HCT) in the rituximab era. Adult patients with relapsed/refractory grades 1 and 2 FL undergoing first reduced-intensity allo-HCT or first autograft during 2000 to 2012 were evaluated. A total of 518 rituximab-treated patients were included. Allo-HCT patients were younger and more heavily pretreated, and more patients had advanced stage and chemoresistant disease. The 5-year adjusted probabilities, comparing auto-HCT versus allo-HCT groups for nonrelapse mortality (NRM) were 5% versus 26% (P < .0001); relapse/progression: 54% versus 20% (P < .0001); progression-free survival (PFS): 41% versus 58% (P < .001), and overall survival (OS): 74% versus 66% (P = .05). Auto-HCT was associated with a higher risk of relapse/progression beyond 5 months after HCT (relative risk [RR], 4.4; P < .0001) and worse PFS (RR, 2.9; P < .0001) beyond 11 months after HCT. In the first 24 months after HCT, auto-HCT was associated with improved OS (RR, .41; P < .0001), but beyond 24 months, it was associated with inferior OS (RR, 2.2; P = .006). A landmark analysis of patients alive and progression-free at 2 years after HCT confirmed these observations, showing no difference in further NRM between both groups, but there was significantly higher risk of relapse/progression (RR, 7.3; P < .0001) and inferior PFS (RR, 3.2; P < .0001) and OS (RR, 2.1; P = .04) after auto-HCT. The 10-year cumulative incidences of second hematological malignancies after allo-HCT and auto-HCT were 0% and 7%, respectively. Auto-HCT and reduced-intensity-conditioned allo-HCT as first transplantation approach can provide durable disease control in grades 1 and 2 FL patients. Continued disease relapse risk after auto-HCT translates into improved PFS and OS after allo-HCT in long-term survivors.


Assuntos
Antineoplásicos/uso terapêutico , Transplante de Células-Tronco Hematopoéticas/métodos , Linfoma Folicular/terapia , Agonistas Mieloablativos/uso terapêutico , Rituximab/uso terapêutico , Condicionamento Pré-Transplante/métodos , Adulto , Idoso , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Estudos Longitudinais , Linfoma Folicular/imunologia , Linfoma Folicular/mortalidade , Linfoma Folicular/patologia , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Recidiva , Análise de Sobrevida , Sobreviventes , Transplante Autólogo , Transplante Homólogo , Resultado do Tratamento
9.
Leuk Lymphoma ; 56(12): 3387-92, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26025300

RESUMO

Interleukin-7 (IL-7) is a critical cytokine in early B and T cell development. Peripheral T cell expansion and thymopoiesis and is a result of the ongoing reconstitution from uncommitted stem cells after transplant. We investigated the efficacy of ex vivo incubated bone marrow cells treated with recombinant human IL-7 (rIL-7) on subsequent in vivo murine models of syngeneic bone marrow (BM) transplant. After ex vivo culture with rIL-7, we observed a 1½-fold increase in BM cellularity; this increase was associated with an enhanced reconstitution of bone marrow cells and thymocytes at 45 days post-transplant. In addition to increased cellularity, lymphocytes from mice transplanted with cultured rIL-7 showed enhanced proliferative responses to mitogenic stimulation. These findings suggest rIL-7 to be a promising agent for the clinical application of treating immune deficiency and enhancing immuno-hematopoietic reconstitution of the stem cell auto/allograft.


Assuntos
Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Transplante de Medula Óssea , Diferenciação Celular , Hematopoese/efeitos dos fármacos , Hematopoese/fisiologia , Imunomodulação , Interleucina-7/farmacologia , Animais , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea/métodos , Técnicas de Cultura de Células , Feminino , Expressão Gênica , Interleucina-2/farmacologia , Camundongos , Modelos Animais , RNA Mensageiro , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Baço/citologia , Baço/efeitos dos fármacos , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/metabolismo , Timócitos/efeitos dos fármacos , Timócitos/metabolismo , Condicionamento Pré-Transplante
10.
Reprod Biomed Online ; 29(3): 340-51, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25066893

RESUMO

Haematopoietic stem cell transplantation (HSCT) remains the best therapeutic option for many acquired and inherited paediatric haematological disorders. Unfortunately, the probability of finding an HLA matched donor is limited. An alternative technique is PGD combined with HLA matching, which offers the possibility of selecting unaffected embryos that are HLA compatible with the sick child, with the aim of possible use of stem cells from the resulting baby in future. Since the first successful report for Fanconi anaemia a decade ago, the therapeutic success of this technique was reported in a few cases and for a limited number of disorders. Here, we report full recovery of 44 sick children who received HSCT from healthy infants conceived after pre-implantation HLA matching for the following 10 indications; beta-thalassaemia, Wiskott-Aldrich syndrome, Fanconi anaemia, sickle cell anaemia, acute myeloid leukaemia, acute lymphoblastic leukaemia, Glanzmann's thrombasthaenia, Diamond-Blackfan anaemia, X-linked adrenoleukodystrophy and mucopolysaccharidosis type I. No serious complications were observed among recipients and donors. Graft failure occurred in four children with beta-thalassaemia where a second HSCT was planned. Preimplantation HLA matching is a reliable technique and provides a realistic option for couples seeking treatment for an affected child when no HLA-matched donor is available.


Assuntos
Encefalopatias Metabólicas Congênitas/terapia , Antígenos HLA , Doenças Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas , Encefalopatias Metabólicas Congênitas/diagnóstico , Doenças Hematológicas/congênito , Doenças Hematológicas/diagnóstico , Teste de Histocompatibilidade , Humanos , Diagnóstico Pré-Implantação , Irmãos
11.
Immunotherapy ; 6(4): 409-17, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24815781

RESUMO

BACKGROUND: Relapse of the original disease remains the most common cause of treatment failure after allogeneic hematopoietic stem cell transplantation (allo-SCT). Patients who relapse post-allo-SCT can achieve prolonged remission after donor lymphocyte infusion. Donor lymphocyte infusion as well as other immunotherapeutic strategies are usually complicated by severe graft versus host disease. AIM: In the present study, we examined the effect of irradiation on the cytotoxic activity of mononuclear cells (MNCs). MATERIALS & METHODS: Cytotoxic activity of fresh and irradiated MNCs from healthy donors was tested against the leukemic cell line K562 and against fresh leukemic cells from patients with acute myeloid leukemia. Cytotoxicity was assessed by using a flow-cytometry assay. RESULTS & DISCUSSION: Interestingly, we observed that 25 Gy irradiated MNCs retain significant cytotoxic activity against K562. Based on these in vitro data, the safety and efficacy of irradiated haploidentical, IL-2-activated lymphocytes were tested in six patients after allo-SCT. Acute skin graft versus host disease developed in two patients and was resolved after a short course of steroids. One patient with mixed chimera converted to full donor chimera after infusion of irradiated donor cells. CONCLUSION: The efficacy of irradiated haploidentical lymphocytes should be further tested in a larger number of patients.


Assuntos
Raios gama , Transplante de Células-Tronco Hematopoéticas , Células Matadoras Ativadas por Linfocina/transplante , Leucemia Mieloide Aguda/terapia , Transfusão de Linfócitos , Prevenção Secundária/métodos , Doadores não Relacionados , Adulto , Aloenxertos , Feminino , Humanos , Imunidade Celular/imunologia , Imunidade Celular/efeitos da radiação , Células K562 , Células Matadoras Ativadas por Linfocina/imunologia , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/patologia , Masculino , Recidiva
12.
Pharmacol Res ; 79: 9-12, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24200897

RESUMO

In this perspective article, we address the controversy regarding the safety-efficacy issue in ipilimumab trials. While the CTLA-4 blockade interrupted T-cell pathways responsible for immune down-regulation and mediated regression of established malignant tumors in a minority of patients, this has to be weighed against the immune related adverse events (irAEs) suffered by the majority. Based on two groundbreaking but neglected proof-of-principle papers that demonstrated augmented graft-vs.-malignancy (GVM) effect that reversed the relapse of malignancy without worsening the graft-vs.-host disease (GVHD) by a CTLA-4 blockade, here we suggest a therapeutic paradigm shift, which may help break the impasse and resolve this timely issue in oncology.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Animais , Anticorpos Monoclonais/efeitos adversos , Antineoplásicos/efeitos adversos , Autoimunidade , Antígeno CTLA-4/antagonistas & inibidores , Doença Enxerto-Hospedeiro , Humanos , Ipilimumab , Melanoma/tratamento farmacológico , Melanoma/imunologia , Camundongos , Neoplasia Residual/imunologia , Neoplasia Residual/terapia , Transplante de Células-Tronco
13.
Oncotarget ; 4(5): 665-76, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23714687

RESUMO

Glioblastomas (GBM), the most common and aggressive malignant astrocytic tumors, contain a small subpopulation of cancer stem cells (GSCs) that are implicated in therapeutic resistance and tumor recurrence. Here, we study the expression and function of miR-137, a putative suppressor miRNA, in GBM and GSCs. We found that the expression of miR-137 was significantly lower in GBM and GSCs compared to normal brains and neural stem cells (NSCs) and that the miR-137 promoter was hypermethylated in the GBM specimens. The expression of miR-137 was increased in differentiated NSCs and GSCs and overexpression of miR-137 promoted the neural differentiation of both cell types. Moreover, pre-miR-137 significantly decreased the self-renewal of GSCs and the stem cell markers Oct4, Nanog, Sox2 and Shh. We identified RTVP-1 as a novel target of miR-137 in GSCs; transfection of the cells with miR-137 decreased the expression of RTVP-1 and the luciferase activity of RTVP-1 3'-UTR reporter plasmid. Furthermore, overexpression of RTVP-1 plasmid lacking its 3'-UTR abrogated the inhibitory effect of miR-137 on the self-renewal of GSCs. Silencing of RTVP-1 decreased the self-renewal of GSCs and the expression of CXCR4 and overexpression of CXCR4 abrogated the inhibitory effect of RTVP-1 silencing on GSC self-renewal. These results demonstrate that miR-137 is downregulated in GBM probably due to promoter hypermethylation. miR-137 inhibits GSC self-renewal and promotes their differentiation by targeting RTVP-1 which downregulates CXCR4. Thus, miR-137 and RTVP-1 are attractive therapeutic targets for the eradication of GSCs and for the treatment of GBM.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , MicroRNAs/metabolismo , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/citologia , Proteínas do Tecido Nervoso/metabolismo , Receptores CXCR4/biossíntese , Encéfalo/metabolismo , Neoplasias Encefálicas/genética , Diferenciação Celular , Movimento Celular/genética , Proliferação de Células , Metilação de DNA , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Proteínas Hedgehog/biossíntese , Proteínas de Homeodomínio/biossíntese , Humanos , Proteínas de Membrana , MicroRNAs/biossíntese , MicroRNAs/genética , Proteína Homeobox Nanog , Proteínas de Neoplasias/genética , Proteínas do Tecido Nervoso/genética , Células-Tronco Neurais/metabolismo , Fator 3 de Transcrição de Octâmero/biossíntese , Regiões Promotoras Genéticas/genética , Fatores de Transcrição SOXB1/biossíntese , Transdução de Sinais/genética , Células Tumorais Cultivadas
14.
Oncotarget ; 4(2): 346-61, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23548312

RESUMO

MicroRNAs (miRNAs) have emerged as potential cancer therapeutics; however, their clinical use is hindered by lack of effective delivery mechanisms to tumor sites. Mesenchymal stem cells (MSCs) have been shown to migrate to experimental glioma and to exert anti-tumor effects by delivering cytotoxic compounds. Here, we examined the ability of MSCs derived from bone marrow, adipose tissue, placenta and umbilical cord to deliver synthetic miRNA mimics to glioma cells and glioma stem cells (GSCs). We examined the delivery of miR-124 and miR-145 mimics as glioma cells and GSCs express very low levels of these miRNAs. Using fluorescently labeled miRNA mimics and in situ hybridization, we demonstrated that all the MSCs examined delivered miR-124 and miR-145 mimics to co-cultured glioma cells and GSCs via gap junction- dependent and independent processes. The delivered miR-124 and miR-145 mimics significantly decreased the luciferase activity of their respected reporter target genes, SCP-1 and Sox2, and decreased the migration of glioma cells and the self-renewal of GSCs. Moreover, MSCs delivered Cy3-miR-124 mimic to glioma xenografts when administered intracranially. These results suggest that MSCs can deliver synthetic exogenous miRNA mimics to glioma cells and GSCs and may provide an efficient route of therapeutic miRNA delivery in vivo.


Assuntos
Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Movimento Celular/genética , Glioma/patologia , Glioma/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , MicroRNAs/administração & dosagem , Células-Tronco Neoplásicas/patologia , Animais , Neoplasias Encefálicas/genética , Diferenciação Celular/genética , Linhagem Celular Tumoral , Técnicas de Cocultura , Feminino , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Humanos , Camundongos , Camundongos Nus , MicroRNAs/genética , Ensaios Antitumorais Modelo de Xenoenxerto
15.
J Clin Oncol ; 31(13): 1669-76, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23547077

RESUMO

PURPOSE: Allogeneic hematopoietic cell transplantation (HCT) can cure bone marrow failure in patients with Fanconi anemia (FA). Data on outcomes in patients with pretransplantation cytogenetic abnormalities, myelodysplastic syndrome (MDS), or acute leukemia have not been separately analyzed. PATIENTS AND METHODS: We analyzed data on 113 patients with FA with cytogenetic abnormalities (n = 54), MDS (n = 45), or acute leukemia (n = 14) who were reported to the Center for International Blood and Marrow Transplant Research from 1985 to 2007. RESULTS: Neutrophil recovery occurred in 78% and 85% of patients at days 28 and 100, respectively. Day 100 cumulative incidences of acute graft-versus-host disease grades B to D and C to D were 26% (95% CI, 19% to 35%) and 12% (95% CI, 7% to 19%), respectively. Survival probabilities at 1, 3, and 5 years were 64% (95% CI, 55% to 73%), 58% (95% CI, 48% to 67%), and 55% (95% CI, 45% to 64%), respectively. In univariate analysis, younger age was associated with superior 5-year survival (≤ v > 14 years: 69% [95% CI, 57% to 80%] v 39% [95% CI, 26% to 53%], respectively; P = .001). In transplantations from HLA-matched related donors (n = 82), younger patients (≤ v > 14 years: 78% [95% CI, 64% to 90%] v 34% [95% CI, 20% to 50%], respectively; P < .001) and patients with cytogenetic abnormalities only versus MDS/acute leukemia (67% [95% CI, 52% to 81%] v 43% [95% CI, 27% to 59%], respectively; P = .03) had superior 5-year survival. CONCLUSION: Our analysis indicates that long-term survival for patients with FA with cytogenetic abnormalities, MDS, or acute leukemia is achievable. Younger patients and recipients of HLA-matched related donor transplantations who have cytogenetic abnormalities only have the best survival.


Assuntos
Anemia de Fanconi/cirurgia , Transplante de Células-Tronco Hematopoéticas/métodos , Síndromes Mielodisplásicas/cirurgia , Leucemia-Linfoma Linfoblástico de Células Precursoras/cirurgia , Adolescente , Adulto , Criança , Pré-Escolar , Análise Citogenética , Anemia de Fanconi/genética , Anemia de Fanconi/terapia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/sangue , Síndromes Mielodisplásicas/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/sangue , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Fatores de Risco , Análise de Sobrevida , Condicionamento Pré-Transplante/métodos , Transplante Homólogo , Resultado do Tratamento , Adulto Jovem
16.
PLoS One ; 8(2): e54652, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23390502

RESUMO

Glioblastomas (GBM), the most common and aggressive type of malignant glioma, are characterized by increased invasion into the surrounding brain tissues. Despite intensive therapeutic strategies, the median survival of GBM patients has remained dismal over the last decades. In this study we examined the expression of miR-145 in glial tumors and its function in glioma cells. Using TCGA analysis and real-time PCR we found that the expression of miR-145/143 cluster was downregulated in astrocytic tumors compared to normal brain specimens and in glioma cells and glioma stem cells (GSCs) compared to normal astrocytes and neural stem cells. Moreover, the low expression of both miR-145 and miR-143 in GBM was correlated with poor patient prognosis. Transfection of glioma cells with miR-145 mimic or transduction with a lentivirus vector expressing pre-miR 145 significantly decreased the migration and invasion of glioma cells. We identified connective tissue growth factor (CTGF) as a novel target of miR-145 in glioma cells; transfection of the cells with this miRNA decreased the expression of CTGF as determined by Western blot analysis and the expression of its 3'-UTR fused to luciferase. Overexpression of a CTGF plasmid lacking the 3'-UTR and administration of recombinant CTGF protein abrogated the inhibitory effect of miR-145 on glioma cell migration. Similarly, we found that silencing of CTGF decreased the migration of glioma cells. CTGF silencing also decreased the expression of SPARC, phospho-FAK and FAK and overexpression of SPARC abrogated the inhibitory effect of CTGF silencing on cell migration. These results demonstrate that miR-145 is downregulated in glial tumors and its low expression in GBM predicts poor patient prognosis. In addition miR-145 regulates glioma cell migration by targeting CTGF which downregulates SPARC expression. Therefore, miR-145 is an attractive therapeutic target for anti-invasive treatment of astrocytic tumors.


Assuntos
Neoplasias Encefálicas/genética , Fator de Crescimento do Tecido Conjuntivo/genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , MicroRNAs/genética , Proteínas Supressoras de Tumor/genética , Regiões 3' não Traduzidas , Astrócitos/citologia , Astrócitos/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Quinase 1 de Adesão Focal/genética , Quinase 1 de Adesão Focal/metabolismo , Inativação Gênica , Genes Reporter , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Luciferases , MicroRNAs/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Osteonectina , RNA Interferente Pequeno/genética , Transdução de Sinais , Transfecção , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/metabolismo
17.
EPMA J ; 3(1): 16, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-23140237

RESUMO

In the present state of healthcare, usual medical care is generally given to the already diseased person, while the key link-personal health monitoring underlain by predictive, preventive, and personalised medicine (PPPM) techniques that are being intensively elaborated worldwide-is simply missing. It is this link, based on the recognition of subclinical conditions, prediction, and further preventive measures, that is capable of regulating morbidity and diminishing the rates of disability among able-bodied population, thus significantly cutting the traditionally high costs of treating the already diseased people. To achieve the above-mentioned goal-the elaboration of the PPPM concept and its practical implementation-it is necessary to create a fundamentally new strategy based upon the subclinical recognition of the signs-bioindicators of cryptic abnormalities long before the disease clinically manifests itself. The implementation of PPPM programme requires an adjusted technology for the proper interpretation of diagnostic data, which would allow for the current 'physician-patient' model to be gradually replaced by a novel model, 'medical advisor-healthy men-at-risk'. This is the reason for an additional need in organising combinatorial scientific, clinical, training and educational projects in the area of PPPM to elicit the content of this new branch of medicine.

18.
Blood ; 120(20): 4256-62, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23007405

RESUMO

The best conditioning regimen before allogeneic transplantation for high-risk diffuse large B-cell lymphoma (DLBCL) remains to be clarified. We analyzed data from 396 recipients of allotransplants for DLBCL receiving myeloablative (MAC; n = 165), reduced intensity (RIC; n = 143), or nonmyeloablative conditioning (NMAC; n = 88) regimens. Acute and chronic GVHD rates were similar across the groups. Five-year nonrelapse mortality (NRM) was higher in MAC than RIC and NMAC (56% vs 47% vs 36%; P = .007). Five-year relapse/progression was lower in MAC than in RIC/NMAC (26% vs 38% vs 40%; P = .031). Five-year progression-free survival (15%-25%) and overall survival (18%-26%) did not differ significantly between the cohorts. In multivariate analysis, NMAC and more recent transplant year were associated with lower NRM, whereas a lower Karnofsky performance score (< 90), prior relapse resistant to therapy, and use of unrelated donors were associated with higher NRM. NMAC transplants, no prior use of rituximab, and prior relapse resistant to therapy were associated with a greater risk of relapse/progression. In conclusion, allotransplantation with RIC or NMAC induces long-term progression-free survival in selected DLBCL patients with a lower risk of NRM but with higher risk of lymphoma progression or relapse.


Assuntos
Linfoma Difuso de Grandes Células B/cirurgia , Agonistas Mieloablativos/administração & dosagem , Condicionamento Pré-Transplante/métodos , Irradiação Corporal Total , Doença Aguda , Adolescente , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Doença Crônica , Terapia Combinada , Progressão da Doença , Intervalo Livre de Doença , Feminino , Doença Enxerto-Hospedeiro/epidemiologia , Doença Enxerto-Hospedeiro/etiologia , Doença Enxerto-Hospedeiro/prevenção & controle , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Histocompatibilidade , Humanos , Estimativa de Kaplan-Meier , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Agonistas Mieloablativos/efeitos adversos , Recidiva , Estudos Retrospectivos , Transplante Homólogo/efeitos adversos , Resultado do Tratamento , Irradiação Corporal Total/efeitos adversos , Adulto Jovem
19.
Pediatr Transplant ; 16(8): E342-7, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22553996

RESUMO

The present case report represents a successful attempt to induce transplantation tolerance to organ allograft by combined administration of donor hematopoietic cells and kidney based on in vivo deletion of alloreactive host-vs-graft and graft-vs-host alloreactive T cells following non-myeloablative conditioning. We were able to induce mixed and eventually full donor chimerism and tolerance of kidney allograft in a 15-yr-old male with ESRD after cisplatin treatment and autologous HSCT for mediastinal germ cell tumor. Our approach to induce tolerance was based on preferential depletion of alloreactive T cells induced by exposure to donor's alloantigens and administration of cyclophosphamide at day 2 and day 3 after stem cell infusion. Additional non-specific immunosuppression as part of the conditioning included exposure to two fractions of TLI, treatment with alemtuzumab (monoclonal anti-CD52) and short-term conventional IS treatment to avoid early graft loss, because of request of IRB. Using this approach, with rapid tapering of all conventional IS treatment, the patient maintains good renal functions without evidence of both acute and chronic rejection for 32 months off all medications.


Assuntos
Transplante de Células-Tronco Hematopoéticas/métodos , Transplante de Rim/métodos , Linfócitos T/imunologia , Adolescente , Alemtuzumab , Anticorpos Monoclonais Humanizados/farmacologia , Antígenos CD/biossíntese , Antígenos de Neoplasias/biossíntese , Antígeno CD52 , Cisplatino/farmacologia , Ciclofosfamida/farmacologia , Glicoproteínas/biossíntese , Rejeição de Enxerto , Humanos , Tolerância Imunológica , Imunossupressores/farmacologia , Falência Renal Crônica/terapia , Masculino , Neoplasias Embrionárias de Células Germinativas/metabolismo , Nefrite Intersticial/imunologia , Nefrite Intersticial/terapia , Condicionamento Pré-Transplante/métodos , Transplante Homólogo
20.
EMBO Mol Med ; 4(4): 313-29, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22351600

RESUMO

Telomerase is expressed in the neonatal brain, in distinct regions of adult brain, and was shown to protect developing neurons from apoptosis. Telomerase reactivation by gene manipulation reverses neurodegeneration in aged telomerase-deficient mice. Hence, we and others hypothesized that increasing telomerase expression by pharmaceutical compounds may protect brain cells from death caused by damaging agents. In this study, we demonstrate for the first time that the novel compound AGS-499 increases telomerase activity and expression in the mouse brain and spinal cord (SC). It exerts neuroprotective effects in NMDA-injected CD-1 mice, delays the onset and progression of the amyotrophic lateral sclerosis (ALS) disease in SOD1 transgenic mice, and, after the onset of ALS, it increases the survival of motor neurons in the SC by 60%. The survival of telomerase-expressing cells (i.e. motor neurons), but not telomerase-deficient cells, exposed to oxidative stress was increased by AGS-499 treatment, suggesting that the AGS-499 effects are telomerase-mediated. Therefore, a controlled and transient increase in telomerase expression and activity in the brain by AGS-499 may exert neuroprotective effects.


Assuntos
Esclerose Lateral Amiotrófica/enzimologia , Esclerose Lateral Amiotrófica/prevenção & controle , Encéfalo/enzimologia , Fármacos Neuroprotetores/uso terapêutico , Fenóis/uso terapêutico , Medula Espinal/enzimologia , Telomerase/metabolismo , Compostos de Tritil/uso terapêutico , Esclerose Lateral Amiotrófica/genética , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Masculino , Camundongos , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Fenóis/farmacologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Telomerase/genética , Compostos de Tritil/farmacologia , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA