Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
2.
Blood ; 130(5): 606-618, 2017 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-28637663

RESUMO

Transfer of T-cell receptors (TCRs) specific for tumor-associated antigens is a promising approach for cancer immunotherapy. We developed the TCR gene editing technology that is based on the knockout of the endogenous TCR α and ß genes, followed by the introduction of tumor-specific TCR genes, and that proved safer and more effective than conventional TCR gene transfer. Although successful, complete editing requires extensive cell manipulation and 4 transduction procedures. Here we propose a novel and clinically feasible TCR "single editing" (SE) approach, based on the disruption of the endogenous TCR α chain only, followed by the transfer of genes encoding for a tumor-specific TCR. We validated SE with the clinical grade HLA-A2 restricted NY-ESO-1157-165-specific TCR. SE allowed the rapid production of high numbers of tumor-specific T cells, with optimal TCR expression and preferential stem memory and central memory phenotype. Similarly to unedited T cells redirected by TCR gene transfer (TCR transferred [TR]), SE T cells efficiently killed NY-ESO-1pos targets; however, although TR cells proved highly alloreactive, SE cells showed a favorable safety profile. Accordingly, when infused in NSG mice previously engrafted with myeloma, SE cells mediated tumor rejection without inducing xenogeneic graft-versus-host disease, thus resulting in significantly higher survival than that observed in mice treated with TR cells. Overall, single TCR gene editing represents a clinically feasible approach that is able to increase the safety and efficacy of cancer adoptive immunotherapy.


Assuntos
Transferência Adotiva , Edição de Genes/métodos , Memória Imunológica , Mieloma Múltiplo , Proteínas de Neoplasias , Fragmentos de Peptídeos , Receptores de Antígenos de Linfócitos T , Linfócitos T , Animais , Linhagem Celular Tumoral , Feminino , Técnicas de Transferência de Genes , Doença Enxerto-Hospedeiro , Camundongos , Mieloma Múltiplo/genética , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/terapia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Linfócitos T/transplante , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Mol Ther ; 26(9): 2107-2118, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-30017878

RESUMO

Extracellular vesicles (EVs) play a major role in cell-to-cell communication in physiological and pathological conditions, and their manipulation may represent a promising therapeutic strategy. Microglia, the parenchymal mononuclear phagocytes of the brain, modulate neighboring cells also through the release of EVs. The production of custom EVs filled with desired molecules, possibly targeted to make their uptake cell specific, and their administration in biological fluids may represent a valid approach for drug delivery. We engineered a murine microglia cell line, BV-2, to release EVs overexpressing the endogenous "eat me" signal Lactadherin (Mfg-e8) on the surface to target phagocytes and containing the anti-inflammatory cytokine IL-4. A single injection of 107 IL-4+Mfg-e8+ EVs into the cisterna magna modulated established neuroinflammation and significantly reduced clinical signs in the mouse model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). Injected IL-4+Mfg-e8+ EVs target mainly phagocytes (i.e., macrophages and microglia) surrounding liquoral spaces, and their cargo promote the upregulation of anti-inflammatory markers chitinase 3-like 3 (ym1) and arginase-1 (arg1), significantly reducing tissue damage. Engineered EVs may represent a biological drug delivery tool able to deliver multiple functional molecules simultaneously to treat neuroinflammatory diseases.


Assuntos
Vesículas Extracelulares/metabolismo , Interleucina-4/metabolismo , Esclerose Múltipla/metabolismo , Animais , Antígeno CD11b/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/ultraestrutura , Linhagem Celular , Células Cultivadas , Modelos Animais de Doenças , Vesículas Extracelulares/ultraestrutura , Feminino , Macrófagos/metabolismo , Macrófagos/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
J Nanobiotechnology ; 17(1): 49, 2019 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-30943991

RESUMO

BACKGROUND: The increasing use of gold nanoparticles (AuNPs) in the field of neuroscience instilled hope for their rapid translation to the clinical practice. AuNPs can be engineered to carry therapeutics or diagnostics in the diseased brain, possibly providing greater cell specificity and low toxicity. Although there is a general enthusiasm for these tools, we are in early stages of their development. Overall, their brain penetrance, stability and cell specificity are critical issues that must be addressed to drive AuNPs to the clinic. RESULTS: We studied the kinetic, distribution and stability of PEG-coated AuNPs in mice receiving a single injection into the cisterna magna of the 4th ventricle. AuNPs were conjugated with the fluorescent tag Cy5.5 (Cy5.5-AuNPs) to track their in vivo distribution. Fluorescence levels from such particles were detected in mice for weeks. In situ analysis of brains by immunofluorescence and electron microscopy revealed that Cy5.5-AuNPs penetrated the brain parenchyma, spreading in the CNS parenchyma beneath the 4th ventricle. Cy5.5-AuNPs were preferentially found in neurons, although a subset of resting microglia also entrapped these particles. CONCLUSIONS: Our results suggest that the ICM route for delivering gold particles allows the targeting of neurons. This approach might be pursued to carry therapeutics or diagnostics inside a diseased brain with a surgical procedure that is largely used in gene therapy approaches. Furthermore, this approach could be used for radiotherapy, enhancing the agent's efficacy to kill brain cancer cells.


Assuntos
Encéfalo/metabolismo , Ouro/química , Nanopartículas Metálicas/química , Polietilenoglicóis/química , Animais , Carbocianinas/química , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cisterna Magna , Estabilidade de Medicamentos , Corantes Fluorescentes/química , Humanos , Camundongos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Permeabilidade , Distribuição Tecidual
5.
Mol Ther ; 25(10): 2254-2269, 2017 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-28807569

RESUMO

T regulatory cells (Tregs) play a key role in modulating T cell responses. Clinical trials showed that Tregs modulate graft-versus-host disease (GvHD) after allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, their ability to mediate anti-leukemic activity (graft-versus-leukemia [GvL]) is largely unknown. Enforced interleukin-10 (IL-10) expression converts human CD4+ T cells into T regulatory type 1 (Tr1)-like (CD4IL-10) cells that suppress effector T cells in vitro and xenoGvHD in humanized mouse models. In the present study, we show that CD4IL-10 cells mediate anti-leukemic effects in vitro and in vivo in a human leukocyte antigen (HLA) class I-dependent but antigen-independent manner. The cytotoxicity mediated by CD4IL-10 cells is granzyme B (GzB) dependent, is specific for CD13+ target cells, and requires CD54 and CD112 expression on primary leukemic target blasts. CD4IL-10 cells adoptively transferred in humanized mouse models directly mediate anti-tumor and anti-leukemic effects. In addition, when co-transferred with peripheral blood mononuclear cells (PBMCs), CD4IL-10 cells contribute to the GvL activity but suppress xenoGvHD mediated by the PBMCs. These findings provide for the first time a strong rationale for CD4IL-10 cell immunotherapy to prevent GvHD and promote GvL in allo-HSCT for myeloid malignancies.


Assuntos
Interleucina-10/metabolismo , Leucemia Mieloide/terapia , Leucócitos Mononucleares/metabolismo , Linfócitos T Reguladores/metabolismo , Linfócitos T CD4-Positivos , Humanos , Imunoterapia , Leucemia Mieloide/imunologia , Leucemia Mieloide/metabolismo , Modelos Biológicos
6.
Sci Rep ; 14(1): 19878, 2024 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-39191985

RESUMO

In this work the effect of combining ultrasound (US) hyperthermia (HT) with radiotherapy (RT) was investigated. The treatment was applied to a GBM xenograft nude mouse model obtained by injecting 2 × 10 6 U87 luc+ cells. The combined treatment group received 6 Gy and HT at 43 ∘ for 8 min. The ultrasound field was generated by a closed-loop computationally controlled system, consisting of a High Intensity Focused Ultrasound (HIFU) transducer with centre frequency 3.57 MHz, a power amplifier, a function generator and a MATLAB controller. A mechanical cone adaptor has been designed to use the HIFU beam at a pre-defined post-focal distance. Two thermocouples were placed between the mechanical cone and the mice skin to measure and control the temperature during the HT treatment. Radiotherapy was carried out by using a dedicated small animal image guided radiotherapy system. Measurements of tumor volume performed with a caliper showed good tumor control for the RT-HT group with respect to the RT or control groups for up to 21 days after treatment. The mean value of the normalized (before therapy) tumor volume was almost equal to 0.5 for two weeks after treatment with an increase to 1.5 at sacrifice. The control and HT groups showed a higher value of about 1.5 during the first two weeks and 3.5 at the end of the follow-up period. We concluded that the use of HT as a radiosensitizer can improve the outcome for glioblastoma treatments.


Assuntos
Glioblastoma , Hipertermia Induzida , Camundongos Nus , Animais , Glioblastoma/radioterapia , Glioblastoma/patologia , Glioblastoma/terapia , Hipertermia Induzida/métodos , Camundongos , Humanos , Terapia Combinada , Linhagem Celular Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/patologia , Modelos Animais de Doenças , Carga Tumoral/efeitos da radiação
7.
Phys Med ; 124: 103420, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38970950

RESUMO

PURPOSE: The purpose of this study is to investigate the dosimetric characteristics of a collimator for minibeam radiotherapy (MBRT) with film dosimetry and Monte Carlo (MC) simulations. The outcome of MBRT with respect to conventional RT using a glioma preclinical model was also evaluated. METHODS: A multi-slit collimator was designed to be used with commercial small animal irradiator. The collimator was built by aligning 0.6 mm wide and 5 mm thick parallel lead leaves at 0.4 mm intervals. Dosimetry characteristics were evaluated by Gafchromic (CG) films and TOPAS Monte Carlo (MC) code. An in vivo experiment was performed using a glioma preclinical model by injecting two million GL261cells subcutaneously and treating with 25 Gy, single fraction, with MBRT and conventional RT. Survival curves and acute radiation damage were measured to compare both treatments. RESULTS: A satisfactory agreement between experimental results and MC simulations were obtained, the measured FWHM and distance between the peaks were respectively 0.431 and 1.098 mm. In vivo results show that MBRT can provide local tumor control for three weeks after RT treatment and a similar survival fraction of open beam radiotherapy. No severe acute effects were seen for the MBRT group. CONCLUSIONS: We developed a minibeam collimator and presented its dosimetric features. Satisfactory agreement between MC and GC films was found with differences consistent with uncertainties due to fabrication and set-up errors. The survival curves of MBRT and open field RT are similar while atoxicity is dramatically lower with MBRT, preliminarily confirming the expected effect.


Assuntos
Glioma , Método de Monte Carlo , Fótons , Glioma/radioterapia , Animais , Fótons/uso terapêutico , Camundongos , Radiometria , Linhagem Celular Tumoral , Dosagem Radioterapêutica , Dosimetria Fotográfica , Radioterapia/métodos , Radioterapia/instrumentação , Neoplasias Encefálicas/radioterapia
8.
Sci Rep ; 14(1): 484, 2024 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-38177637

RESUMO

Bladder mechanical properties are critical for organ function and tissue homeostasis. Therefore, alterations of tissue mechanics are linked to disease onset and progression. This study aims to characterize the tissue elasticity of the murine bladder wall considering its different anatomical components, both in healthy conditions and in actinic cystitis, a state characterized by tissue fibrosis. Here, we exploit Brillouin microscopy, an emerging technique in the mechanobiology field that allows mapping tissue mechanics at the microscale, in non-contact mode and free of labeling. We show that Brillouin imaging of bladder tissues is able to recognize the different anatomical components of the bladder wall, confirmed by histopathological analysis, showing different tissue mechanical properties of the physiological bladder, as well as a significant alteration in the presence of tissue fibrosis. Our results point out the potential use of Brillouin imaging on clinically relevant samples as a complementary technique to histopathological analysis, deciphering complex mechanical alteration of each tissue layer of an organ that strongly relies on mechanical properties to perform its function.


Assuntos
Cistite , Microscopia , Camundongos , Animais , Bexiga Urinária/diagnóstico por imagem , Elasticidade , Cistite/diagnóstico por imagem , Fibrose
9.
Front Immunol ; 15: 1315283, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38510235

RESUMO

Background: In adoptive T cell therapy, the long term therapeutic benefits in patients treated with engineered tumor specific T cells are limited by the lack of long term persistence of the infused cellular products and by the immunosuppressive mechanisms active in the tumor microenvironment. Exhausted T cells infiltrating the tumor are characterized by loss of effector functions triggered by multiple inhibitory receptors (IRs). In patients, IR blockade reverts T cell exhaustion but has low selectivity, potentially unleashing autoreactive clones and resulting in clinical autoimmune side effects. Furthermore, loss of long term protective immunity in cell therapy has been ascribed to the effector memory phenotype of the infused cells. Methods: We simultaneously redirected T cell specificity towards the NY-ESO-1 antigen via TCR gene editing (TCRED) and permanently disrupted LAG3, TIM-3 or 2B4 genes (IRKO) via CRISPR/Cas9 in a protocol to expand early differentiated long-living memory stem T cells. The effector functions of the TCRED-IRKO and IR competent (TCRED-IRCOMP) cells were tested in short-term co-culture assays and under a chronic stimulation setting in vitro. Finally, the therapeutic efficacy of the developed cellular products were evaluated in multiple myeloma xenograft models. Results: We show that upon chronic stimulation, TCRED-IRKO cells are superior to TCRED-IRCOMP cells in resisting functional exhaustion through different mechanisms and efficiently eliminate cancer cells upon tumor re-challenge in vivo. Our data indicate that TIM-3 and 2B4-disruption preserve T-cell degranulation capacity, while LAG-3 disruption prevents the upregulation of additional inhibitory receptors in T cells. Conclusion: These results highlight that TIM-3, LAG-3, and 2B4 disruptions increase the therapeutic benefit of tumor specific cellular products and suggest distinct, non-redundant roles for IRs in anti-tumor responses.


Assuntos
Linfócitos T CD8-Positivos , Mieloma Múltiplo , Humanos , Receptor Celular 2 do Vírus da Hepatite A/genética , Antígenos de Neoplasias/genética , Receptores de Antígenos de Linfócitos T/genética , Microambiente Tumoral
10.
Nanomaterials (Basel) ; 13(23)2023 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-38063740

RESUMO

Nanomedicine relies on the exploitation of nanoscale constructs for therapeutic and diagnostic functions. Gold and gold-iron alloy nanoparticles (NPs) are two examples of nanomaterials with favorable features for use in nanomedicine. While gold NPs have been studied extensively in the last decades, they are not biodegradable. Nonetheless, biodegradation was recently observed in gold alloys with iron obtained using laser ablation in liquid (LAL). Hence, there is a significant interest in the study of the biological effects of gold and gold-iron alloy nanoparticles, starting from their tolerability and cytotoxicity. In this study, these two classes of NPs, obtained via LAL and coated with biocompatible polymers such as polyethylene glycol, were investigated in terms of their cytotoxicity in fibroblasts, prostate cancer cells (PC3) and embryonic kidney cells (HEK). We also explored the effects of different synthetic procedures, stabilizing additives, and the possible mechanisms behind cell mortality such as the formation of reactive oxygen species (ROS) or ferroptosis. NPs larger than 200 nm were associated with lower cell tolerability. The most tolerable formulations were pure PEG-Au NPs, followed by PEG-Au-Fe NPs with a hydrodynamic size < 50 nm, which displayed a toxicity of only 20% in fibroblasts after 72 h of incubation. In addition, tumor cells and highly proliferating HEK cells are more sensitive to the NPs than fibroblasts. However, a protective effect of catalase was found for cells incubated with PEG-Au-Fe NPs, indicating an important role of hydrogen peroxide in alloy NP interactions with cells. These results are crucial for directing future synthetic efforts for the realization of biocompatible Au NPs and biodegradable and cytocompatible Au-Fe alloy NPs. Moreover, the correlation of the cytocompatibility of NPs with ROS and ferroptosis in cells is of general interest and applicability to other types of nanomaterials.

11.
ACS Appl Mater Interfaces ; 15(9): 12171-12188, 2023 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-36826830

RESUMO

Lanthanide-activated fluoride-based nanostructures are extremely interesting multifunctional tools for many modern applications in nanomedicine, e.g., bioimaging, sensing, drug delivery, and photodynamic therapy. Importantly, environmental-friendly preparations using a green chemistry approach, as hydrothermal synthesis route, are nowadays highly desirable to obtain colloidal nanoparticles, directly dispersible in hydrophilic media, as physiological solution. The nanomaterials under investigation are new KY3F10-based citrate-capped core@shell nanostructures activated with several lanthanide ions, namely, Er3+, Yb3+, Nd3+, and Gd3+, prepared as colloidal water dispersions. A new facile microwave-assisted synthesis has been exploited for their preparation, with significant reduction of the reaction times and a fine control of the nanoparticle size. These core@shell multifunctional architectures have been investigated for use as biocompatible and efficient contrast agents for optical, magnetic resonance imaging (MRI) and computerized tomography (CT) techniques. These multifunctional nanostructures are also efficient noninvasive optical nanothermometers. In fact, the lanthanide emission intensities have shown a relevant relative variation as a function of the temperature, in the visible and near-infrared optical ranges, efficiently exploiting ratiometric intensity methods for optical thermometry. Importantly, in contrast with other fluoride hosts, chemical dissolution of KY3F10 citrate-capped nanocrystals in aqueous environment is very limited, of paramount importance for applications in biological fluids. Furthermore, due to the strong paramagnetic properties of lanthanides (e.g., Gd3+), and X-ray absorption of both yttrium and lanthanides, the nanostructures under investigation are extremely useful for MRI and CT imaging. Biocompatibility studies of the nanomaterials have revealed very low cytotoxicity in dfferent human cell lines. All these features point to a successful use of these fluoride-based core@shell nanoarchitectures for simultaneous diagnostics and temperature sensing, ensuring an excellent biocompatibility.


Assuntos
Elementos da Série dos Lantanídeos , Nanoestruturas , Humanos , Elementos da Série dos Lantanídeos/química , Fluoretos/química , Nanomedicina , Luminescência , Nanoestruturas/química
12.
Cancer Res ; 83(2): 195-218, 2023 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-36409826

RESUMO

Glioblastoma (GBM) is a common and deadly form of brain tumor in adults. Dysregulated metabolism in GBM offers an opportunity to deploy metabolic interventions as precise therapeutic strategies. To identify the molecular drivers and the modalities by which different molecular subgroups of GBM exploit metabolic rewiring to sustain tumor progression, we interrogated the transcriptome, the metabolome, and the glycoproteome of human subgroup-specific GBM sphere-forming cells (GSC). L-fucose abundance and core fucosylation activation were elevated in mesenchymal (MES) compared with proneural GSCs; this pattern was retained in subgroup-specific xenografts and in subgroup-affiliated human patient samples. Genetic and pharmacological inhibition of core fucosylation significantly reduced tumor growth in MES GBM preclinical models. Liquid chromatography-mass spectrometry (LC-MS)-based glycoproteomic screening indicated that most MES-restricted core-fucosylated proteins are involved in therapeutically relevant GBM pathological processes, such as extracellular matrix interaction, cell adhesion, and integrin-mediated signaling. Selective L-fucose accumulation in MES GBMs was observed using preclinical minimally invasive PET, implicating this metabolite as a potential subgroup-restricted biomarker.Overall, these findings indicate that L-fucose pathway activation in MES GBM is a subgroup-specific dependency that could provide diagnostic markers and actionable therapeutic targets. SIGNIFICANCE: Metabolic characterization of subgroup-specific glioblastoma (GBM) sphere-forming cells identifies the L-fucose pathway as a vulnerability restricted to mesenchymal GBM, disclosing a potential precision medicine strategy for targeting cancer metabolism.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/patologia , Fucose/metabolismo , Transdução de Sinais , Neoplasias Encefálicas/patologia , Células-Tronco Neoplásicas/patologia , Linhagem Celular Tumoral
13.
Haematologica ; 102(4): e120-e124, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28034992
14.
Med Phys ; 49(10): 6588-6598, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35946490

RESUMO

PURPOSE: To investigate the feasibility of radioluminescence imaging (RLI) as a novel 2D quality assurance (QA) dosimetry system for CyberKnife®. METHODS: We developed a field size measurement system based on a commercial complementary metal oxide semiconductor (CMOS) camera facing a radioluminescence screen located at the isocenter normal to the beam axis. The radioluminescence light collected by a lens was used to measure 2D dose distributions. An image transformation procedure, based on two reference phantoms, was developed to correct for projective distortion due to the angle (15°) between the optical and beam axis. Dose profiles were measured for field sizes ranging from 5 mm to 60 mm using fixed circular and iris collimators and compared against gafchromic (GC) film. The corresponding full width at half maximum (FWHM) was measured using RLI and benchmarked against GC film. A small shift in the source-to-surface distance (SSD) of the measurement plane was intentionally introduced to test the sensitivity of the RLI system to field size variations. To assess reproducibility, the entire RLI procedure was tested by acquiring the 60 mm circle field three times on two consecutive days. RESULTS: The implemented procedure for perspective image distortion correction showed improvements of up to 1 mm using the star phantom against the square phantom. The FWHM measurements using the RLI system indicated a strong agreement with GC film with maximum absolute difference equal to 0.131 mm for fixed collimators and 0.056 mm for the iris. A 2D analysis of RLI with respect to GC film showed that the differences in the central region are negligible, while small discrepancies are in the penumbra region. Changes in field sizes of 0.2 mm were detectable by RLI. Repeatability measurements of the beam FWHM have shown a standard deviation equal to 0.11 mm. CONCLUSIONS: The first application of a RLI approach for CyberKnife® field size measurement was presented and tested. Results are in agreement with GC film measurements. Spatial resolution and immediate availability of the data indicate that RLI is a feasible technique for robotic radiosurgery QA.


Assuntos
Radiocirurgia , Procedimentos Cirúrgicos Robóticos , Estudos de Viabilidade , Óxidos , Radiometria/métodos , Radiocirurgia/métodos , Dosagem Radioterapêutica , Reprodutibilidade dos Testes
15.
Eur Radiol Exp ; 6(1): 7, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35132475

RESUMO

BACKGROUND: Malignant mesothelioma (MM) is an aggressive tumor, with a poor prognosis, usually unresectable due to late diagnosis, mainly treated with chemotherapy. BoxA, a truncated form of "high mobility group box 1" (HMGB1), acting as an HMGB1 antagonist, might exert a defensive action against MM. We investigated the potential of BoxA for MM treatment using experimental 40-MHz ultrasound and optical imaging (OI) in a murine model. METHODS: Murine MM cells infected with a lentiviral vector expressing the luciferase gene were injected into the peritoneum of 14 BALB/c mice (7 × 104 AB1-B/c-LUC cells). These mice were randomized to treatment with BoxA (n = 7) or phosphate-buffered saline (controls, n = 7). The experiment was repeated with 40 mice divided into two groups (n = 20 + 20) and treated as above to confirm the result and achieve greater statistical power. Tumor presence was investigated by experimental ultrasound and OI; suspected peritoneal masses underwent histopathology and immunohistochemistry examination. RESULTS: In the first experiment, none of the 7 controls survived beyond day 27, whereas 4/7 BoxA-treated mice (57.1%) survived up to day 70. In the second experiment, 6/20 controls (30.0%) and 16/20 BoxA-treated mice (80.0%) were still alive at day 34 (p = 0.004). In both experiments, histology confirmed the malignant nature of masses detected using experimental ultrasound and OI. CONCLUSION: In our preclinical experience on a murine model, BoxA seems to exert a protective role toward MM. Both experimental ultrasound and OI proved to be reliable techniques for detecting MM peritoneal masses.


Assuntos
Proteína HMGB1 , Mesotelioma Maligno , Animais , Modelos Animais de Doenças , Camundongos , Imagem Óptica , Ultrassonografia
16.
Sci Transl Med ; 14(653): eabl4106, 2022 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-35857642

RESUMO

Glioblastoma multiforme (GBM) is the most common and lethal brain tumor characterized by a strongly immunosuppressive tumor microenvironment (TME) that represents a barrier also for the development of effective immunotherapies. The possibility to revert this hostile TME by immunoactivating cytokines is hampered by the severe toxicity associated with their systemic administration. Here, we exploited a lentiviral vector-based platform to engineer hematopoietic stem cells ex vivo with the aim of releasing, via their tumor-infiltrating monocyte/macrophage progeny, interferon-α (IFN-α) or interleukin-12 (IL-12) at the tumor site with spatial and temporal selectivity. Taking advantage of a syngeneic GBM mouse model, we showed that inducible release of IFN-α within the TME achieved robust tumor inhibition up to eradication and outperformed systemic treatment with the recombinant protein in terms of efficacy, tolerability, and specificity. Single-cell RNA sequencing of the tumor immune infiltrate revealed reprogramming of the immune microenvironment toward a proinflammatory and antitumoral state associated with loss of a macrophage subpopulation shown to be associated with poor prognosis in human GBM. The spatial and temporal control of IL-12 release was critical to overcome an otherwise lethal hematopoietic toxicity while allowing to fully exploit its antitumor activity. Overall, our findings demonstrate a potential therapeutic approach for GBM and set the bases for a recently launched first-in-human clinical trial in patients with GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Animais , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Citocinas , Modelos Animais de Doenças , Glioblastoma/tratamento farmacológico , Interferon-alfa , Interleucina-12/uso terapêutico , Camundongos , Microambiente Tumoral
17.
Opt Express ; 19(13): 12605-18, 2011 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-21716501

RESUMO

Quite recently Cerenkov luminescence imaging (CLI) has been introduced as a novel pre-clinical imaging for the in vivo imaging of small animals such as mice. The CLI method is based on the detection of Cerenkov radiation (CR) generated by beta particles as they travel into the animal tissues with an energy such that Cerenkov emission condition is satisfied. This paper describes an image reconstruction method called multi spectral diffuse Cerenkov luminescence tomography (msCLT) in order to obtain 3D images from the detection of CR. The multispectral approach is based on a set of 2D planar images acquired using a number of narrow bandpass filters, and the distinctive information content at each wavelength is used in the 3D image reconstruction process. The proposed msCLT method was tested both in vitro and in vivo using 32P-ATP and all the images were acquired by using the IVIS 200 small animal optical imager (Caliper Life Sciences, Alameda USA). Source depth estimation and spatial resolution measurements were performed using a small capillary source placed between several slices of chicken breast. The theoretical Cerenkov emission spectrum and optical properties of chicken breast were used in the modelling of photon propagation. In vivo imaging was performed by injecting control nude mice with 10 MBq of 32P-ATP and the 3D tracer bio-distribution was reconstructed. Whole body MRI was acquired to provide an anatomical localization of the Cerenkov emission. The spatial resolution obtained from the msCLT reconstructed images of the capillary source showed that the FWHM is about 1.5 mm for a 6 mm depth. Co-registered MRI images showed that the Cerenkov emission regions matches fairly well with anatomical regions, such as the brain, heart and abdomen. Ex vivo imaging of the different organs such as intestine, brain, heart and ribs further confirms these findings. We conclude that in vivo 3D bio-distribution of a pure beta-minus emitting radiopharmaceutical such as 32P-ATP can be obtained using the msCLT reconstruction approach.


Assuntos
Processamento de Imagem Assistida por Computador/instrumentação , Processamento de Imagem Assistida por Computador/métodos , Músculo Esquelético/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/instrumentação , Tomografia por Emissão de Pósitrons/métodos , Trifosfato de Adenosina/farmacocinética , Algoritmos , Animais , Partículas beta , Capilares , Galinhas , Simulação por Computador , Elétrons , Luminescência , Imageamento por Ressonância Magnética , Camundongos , Camundongos Nus , Modelos Teóricos , Músculo Esquelético/irrigação sanguínea , Imagens de Fantasmas , Radioisótopos de Fósforo , Distribuição Tecidual
18.
Eur J Nucl Med Mol Imaging ; 38(1): 120-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20882278

RESUMO

PURPOSE: 2-[(18)F]Fluoro-2-deoxy-D-glucose ((18)F-FDG) is a widely used PET radiotracer for the in vivo diagnosis of several diseases such as tumours. The positrons emitted by (18)F-FDG, travelling into tissues faster than the speed of light in the same medium, are responsible for Cerenkov radiation (CR) emission which is prevalently in the visible range. The purpose of this study is to show that CR escaping from tumour tissues of small living animals injected with (18)F-FDG can be detected with optical imaging (OI) techniques using a commercial optical instrument equipped with charge-coupled detectors (CCD). METHODS: The theory behind the Cerenkov light emission and the source depth measurements using CR is first presented. Mice injected with (18)F-FDG or saline solution underwent dynamic OI acquisition and a comparison between images was performed. Multispectral analysis of the radiation was used to estimate the depth of the source of Cerenkov light. Small animal PET images were also acquired in order to compare the (18)F-FDG bio-distribution measured using OI and PET scanner. RESULTS: Cerenkov in vivo whole-body images of tumour-bearing mice and the measurements of the emission spectrum (560-660 nm range) are presented. Brain, kidneys and tumour were identified as a source of visible light in the animal body: the tissue time-activity curves reflected the physiological accumulation of (18)F-FDG in these organs. The identification is confirmed by the comparison between CR and (18)F-FDG images. CONCLUSION: These results will allow the use of conventional OI devices for the in vivo study of glucose metabolism in cancer and the assessment, for example, of anti-cancer drugs. Moreover, this demonstrates that (18)F-FDG can be employed as it is a bimodal tracer for PET and OI techniques.


Assuntos
Fluordesoxiglucose F18/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Animais , Transporte Biológico , Feminino , Fluordesoxiglucose F18/farmacocinética , Neoplasias Mamárias Experimentais/diagnóstico por imagem , Camundongos , Tomografia por Emissão de Pósitrons , Fatores de Tempo
19.
J Biophotonics ; 14(6): e202000445, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33704914

RESUMO

Laser scalpels used in medical surgery concentrate light energy, heating the tissues. Recently, we reported thermoluminescence emission from laser-treated soft tissues. Here we investigated the thermo-optical effects caused by a laser operating at 808 nm on animal bones (beef ribs) through luminescence and fluorescence imaging, thermal imaging and scanning electron microscopy. Laser-induced artificial lesions emitted luminescence peaking around 650 nm, with a half-life of almost 1 hour. As concerns fluorescence, 24 hours after laser treatment we observed an increase of the emission and a shift from 500 (untreated) to 580 nm (treated). Recrystallization observed by SEM indicates that the temperature in the artificial lesions is over 600°C. We can conclude that laser treatment induces specific luminescent and fluorescent emissions due to heating of the bone and modification of its components. Monitoring these emissions could help prevent tissue overheating and its potential damages during laser-assisted medical procedures.


Assuntos
Terapia a Laser , Fótons , Animais , Bovinos , Lasers , Luminescência , Imagem Óptica
20.
Int J Nanomedicine ; 16: 1943-1960, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33727808

RESUMO

INTRODUCTION: The overexpression of Human Epidermal Growth Factor Receptor 2 (HER2) is usually associated with aggressive and infiltrating breast cancer (BC) phenotype, and metastases. Functionalized silica-based nanocarriers (SiNPs) can be labeled for in vivo imaging applications and loaded with chemotherapy drugs, making possible the simultaneous noninvasive diagnosis and treatment (theranostic) for HER2-positive BC. METHODS: Firstly, FITC-filled SiNPs, were engineered with two different amounts of Hc-TZ (trastuzumab half-chain) per single nanoparticle (1:2 and 1:8, SiNPs to Hc-TZ ratio), which was 99mTc-radiolabeled at histidine residues for ex vivo and in vivo biodistribution evaluations. Secondly, nanoparticles were loaded with DOX and their in vitro and ex vivo/in vivo delivery was assessed, in comparison with liposomal Doxorubicin (Caelyx). Finally, the treatment efficacy of DOX-SiNPs-TZ (1:8 Hc-TZ) was evaluated in vivo by PET and supported by MS-based proteomics profiling of tumors. RESULTS: SiNPs-TZ (1:8 Hc-TZ) tumor uptake was significantly greater than that of SiNPs-TZ (1:2 Hc-TZ) at 6 hours post-injection (p.i.) in ex vivo biodistribution experiment. At 24 h p.i., radioactivity values remained steady. Fluorescence microscopy, confirmed the presence of radiolabeled SiNPs-TZ (1:8 Hc-TZ) within tumor even at later times. SiNPs-TZ (1:8 Hc-TZ) nanoparticles loaded with Doxorubicin (DOX-SiNPs-TZ) showed a similar DOX delivery capability than Caelyx (at 6 h p.i.), in in vitro and ex vivo assays. Nevertheless, at the end of treatment, tumor volume was significantly reduced by DOX-SiNPs-TZ (1:8 Hc-TZ), compared to Caelyx and DOX-SiNPs treatment. Proteomics study identified 88 high stringent differentially expressed proteins comparing the three treatment groups with controls. CONCLUSION: These findings demonstrated a promising detection specificity and treatment efficacy for our system (SiNPs-TZ, 1:8 Hc-TZ), encouraging its potential use as a new theranostic agent for HER2-positive BC lesions. In addition, proteomic profile confirmed that a set of proteins, related to tumor aggressiveness, were positively affected by targeted nanoparticles.


Assuntos
Neoplasias da Mama/diagnóstico , Portadores de Fármacos/química , Nanopartículas/química , Compostos Radiofarmacêuticos/química , Receptor ErbB-2/metabolismo , Dióxido de Silício/química , Tecnécio/química , Animais , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Doxorrubicina/análogos & derivados , Doxorrubicina/química , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Endocitose , Feminino , Fluoresceína-5-Isotiocianato/química , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia , Polietilenoglicóis/uso terapêutico , Proteoma/metabolismo , Proteômica , Compostos Radiofarmacêuticos/farmacocinética , Tecnécio/farmacocinética , Distribuição Tecidual/efeitos dos fármacos , Tomografia Computadorizada de Emissão de Fóton Único , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA