Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.123
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Nanobiotechnology ; 22(1): 461, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39090622

RESUMEN

BACKGROUND: The combination of programmed cell death ligand-1 (PD-L1) immune checkpoint blockade (ICB) and immunogenic cell death (ICD)-inducing chemotherapy has shown promise in cancer immunotherapy. However, triple-negative breast cancer (TNBC) patients undergoing this treatment often face obstacles such as systemic toxicity and low response rates, primarily attributed to the immunosuppressive tumor microenvironment (TME). METHODS AND RESULTS: In this study, PD-L1-targeted theranostic systems were developed utilizing anti-PD-L1 peptide (APP) conjugated with a bio-orthogonal click chemistry group. Initially, TNBC was treated with azide-modified sugar to introduce azide groups onto tumor cell surfaces through metabolic glycoengineering. A PD-L1-targeted probe was developed to evaluate the PD-L1 status of TNBC using magnetic resonance/near-infrared fluorescence imaging. Subsequently, an acidic pH-responsive prodrug was employed to enhance tumor accumulation via bio-orthogonal click chemistry, which enhances PD-L1-targeted ICB, the pH-responsive DOX release and induction of pyroptosis-mediated ICD of TNBC. Combined PD-L1-targeted chemo-immunotherapy effectively reversed the immune-tolerant TME and elicited robust tumor-specific immune responses, resulting in significant inhibition of tumor progression. CONCLUSIONS: Our study has successfully engineered a bio-orthogonal multifunctional theranostic system, which employs bio-orthogonal click chemistry in conjunction with a PD-L1 targeting strategy. This innovative approach has been demonstrated to exhibit significant promise for both the targeted imaging and therapeutic intervention of TNBC.


Asunto(s)
Antígeno B7-H1 , Química Clic , Inmunoterapia , Piroptosis , Neoplasias de la Mama Triple Negativas , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Antígeno B7-H1/metabolismo , Animales , Femenino , Inmunoterapia/métodos , Ratones , Piroptosis/efectos de los fármacos , Humanos , Línea Celular Tumoral , Microambiente Tumoral/efectos de los fármacos , Ratones Endogámicos BALB C , Doxorrubicina/farmacología , Doxorrubicina/química , Doxorrubicina/uso terapéutico , Imagen Óptica/métodos , Profármacos/química , Profármacos/farmacología
2.
ACS Nano ; 18(28): 18412-18424, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38949962

RESUMEN

As a noninvasive treatment modality, high-intensity focused ultrasound (HIFU)-induced antitumor immune responses play a vital role in surgery prognosis. However, limited response intensity largely hinders postoperative immunotherapy. Herein, a hypoxia-specific metal-organic framework (MOF) nanosystem, coordinated by Fe3+, hypoxic-activated prodrug AQ4N, and IDO-1 signaling pathway inhibitor NLG919, is developed for the potentiating immunotherapy of HIFU surgery. The loaded AQ4N enhances the photoacoustic imaging effects to achieve accurate intraoperative navigation. Within the HIFU-established severe hypoxic environment, AQ4N is activated sequentially, following which it cooperates with Fe3+ to effectively provoke immunogenic cell death. In addition, potent NLG919 suppresses IDO-1 activity and degrades the immunosuppressive tumor microenvironment aggravated by postoperative hypoxia. In vivo studies demonstrate that the MOF-mediated immunotherapy greatly inhibits the growth of primary/distant tumors and eliminates lung metastasis. This work establishes a robust delivery platform to improve immunotherapy and the overall prognosis of HIFU surgery with high specificity and potency.


Asunto(s)
Inmunoterapia , Estructuras Metalorgánicas , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Animales , Ratones , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Microambiente Tumoral/efectos de los fármacos , Ultrasonido Enfocado de Alta Intensidad de Ablación , Línea Celular Tumoral , Ratones Endogámicos BALB C , Proliferación Celular/efectos de los fármacos , Profármacos/química , Profármacos/farmacología , Femenino , Neoplasias/terapia , Neoplasias/diagnóstico por imagen , Neoplasias/patología , Neoplasias/inmunología , Hipoxia
3.
Nano Lett ; 24(28): 8741-8751, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-38953486

RESUMEN

The degradation of oncoproteins mediated by proteolysis-targeting chimera (PROTAC) has emerged as a potent strategy in cancer therapy. However, the clinical application of PROTACs is hampered by challenges such as poor water solubility and off-target adverse effects. Herein, we present an ultrasound (US)-activatable PROTAC prodrug termed NPCe6+PRO for actuating efficient sono-immunotherapy in a spatiotemporally controllable manner. Specifically, US irradiation, which exhibits deep-tissue penetration capability, results in Ce6-mediated generation of ROS, facilitating sonodynamic therapy (SDT) and inducing immunogenic cell death (ICD). Simultaneously, the generated ROS cleaves the thioketal (TK) linker through a ROS-responsive mechanism, realizing the on-demand activation of the PROTAC prodrug in deep tissues. This prodrug activation results in the degradation of the target protein BRD4, while simultaneously reversing the upregulation of PD-L1 expression associated with the SDT process. In the orthotopic mouse model of pancreatic tumors, NPCe6+PRO effectively suppressed tumor growth in conjunction with US stimulation.


Asunto(s)
Inmunoterapia , Neoplasias Pancreáticas , Profármacos , Animales , Profármacos/farmacología , Profármacos/uso terapéutico , Profármacos/química , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/inmunología , Ratones , Humanos , Línea Celular Tumoral , Proteolisis/efectos de los fármacos , Terapia por Ultrasonido/métodos , Antígeno B7-H1 , Factores de Transcripción , Proteínas de Ciclo Celular , Especies Reactivas de Oxígeno/metabolismo , Proteínas que Contienen Bromodominio
4.
Theranostics ; 14(9): 3565-3582, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38948069

RESUMEN

Cancer therapy has moved from single agents to more mechanism-based targeted approaches. In recent years, the combination of HDAC inhibitors and other anticancer chemicals has produced exciting progress in cancer treatment. Herein, we developed a novel prodrug via the ligation of dichloroacetate to selenium-containing potent HDAC inhibitors. The effect and mechanism of this compound in the treatment of prostate cancer were also studied. Methods: The concerned prodrug SeSA-DCA was designed and synthesized under mild conditions. This compound's preclinical studies, including the pharmacokinetics, cell toxicity, and anti-tumor effect on prostate cancer cell lines, were thoroughly investigated, and its possible synergistic mechanism was also explored and discussed. Results: SeSA-DCA showed good stability in physiological conditions and could be rapidly decomposed into DCA and selenium analog of SAHA (SeSAHA) in the tumor microenvironment. CCK-8 experiments identified that SeSA-DCA could effectively inhibit the proliferation of a variety of tumor cell lines, especially in prostate cancer. In further studies, we found that SeSA-DCA could also inhibit the metastasis of prostate cancer cell lines and promote cell apoptosis. At the animal level, oral administration of SeSA-DCA led to significant tumor regression without obvious toxicity. Moreover, as a bimolecular coupling compound, SeSA-DCA exhibited vastly superior efficacy than the mixture with equimolar SeSAHA and DCA both in vitro and in vivo. Our findings provide an important theoretical basis for clinical prostate cancer treatment. Conclusions: Our in vivo and in vitro results showed that SeSA-DCA is a highly effective anti-tumor compound for PCa. It can effectively induce cell cycle arrest and growth suppression and inhibit the migration and metastasis of PCa cell lines compared with monotherapy. SeSA-DCA's ability to decrease the growth of xenografts is a little better than that of docetaxel without any apparent signs of toxicity. Our findings provide an important theoretical basis for clinical prostate cancer treatment.


Asunto(s)
Apoptosis , Puntos de Control del Ciclo Celular , Inhibidores de Histona Desacetilasas , Neoplasias de la Próstata , Fosfatasas cdc25 , Masculino , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Humanos , Animales , Apoptosis/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Inhibidores de Histona Desacetilasas/química , Línea Celular Tumoral , Puntos de Control del Ciclo Celular/efectos de los fármacos , Fosfatasas cdc25/metabolismo , Ratones , Antineoplásicos/farmacología , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Ratones Desnudos , Selenio/farmacología , Selenio/química , Selenio/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Profármacos/farmacología , Profármacos/química , Ratones Endogámicos BALB C
5.
Cells ; 13(13)2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38994978

RESUMEN

We report a three-pronged phenotypic evaluation of the bioprecursor prodrug 10ß,17ß-dihydroxyestra-1,4-dien-3-one (DHED) that selectively produces 17ß-estradiol (E2) in the retina after topical administration and halts glaucomatous neurodegeneration in a male rat model of the disease. Ocular hypertension (OHT) was induced by hyperosmotic saline injection into an episcleral vein of the eye. Animals received daily DHED eye drops for 12 weeks. Deterioration of visual acuity and contrast sensitivity by OHT in these animals were markedly prevented by the DHED-derived E2 with concomitant preservation of retinal ganglion cells and their axons. In addition, we utilized targeted retina proteomics and a previously established panel of proteins as preclinical biomarkers in the context of OHT-induced neurodegeneration as a characteristic process of the disease. The prodrug treatment provided retina-targeted remediation against the glaucomatous dysregulations of these surrogate endpoints without increasing circulating E2 levels. Collectively, the demonstrated significant neuroprotective effect by the DHED-derived E2 in the selected animal model of glaucoma supports the translational potential of our presented ocular neuroprotective approach owing to its inherent therapeutic safety and efficacy.


Asunto(s)
Modelos Animales de Enfermedad , Estradiol , Glaucoma , Profármacos , Células Ganglionares de la Retina , Animales , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/metabolismo , Glaucoma/tratamiento farmacológico , Glaucoma/patología , Glaucoma/metabolismo , Profármacos/farmacología , Estradiol/farmacología , Masculino , Ratas , Retina/efectos de los fármacos , Retina/patología , Retina/metabolismo , Visión Ocular/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico
6.
Sci Adv ; 10(28): eadn0960, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38996025

RESUMEN

Celastrol (CEL), an active compound isolated from the root of Tripterygium wilfordii, exhibits broad anticancer activities. However, its poor stability, narrow therapeutic window and numerous adverse effects limit its applications in vivo. In this study, an adenosine triphosphate (ATP) activatable CEL-Fe(III) chelate was designed, synthesized, and then encapsulated with a reactive oxygen species (ROS)-responsive polymer to obtain CEL-Fe nanoparticles (CEL-Fe NPs). In normal tissues, CEL-Fe NPs maintain structural stability and exhibit reduced systemic toxicity, while at the tumor site, an ATP-ROS-rich tumor microenvironment, drug release is triggered by ROS, and antitumor potency is restored by competitive binding of ATP. This intelligent CEL delivery system improves the biosafety and bioavailability of CEL for cancer therapy. Such a CEL-metal chelate strategy not only mitigates the challenges associated with CEL but also opens avenues for the generation of CEL derivatives, thereby expanding the therapeutic potential of CEL in clinical settings.


Asunto(s)
Adenosina Trifosfato , Triterpenos Pentacíclicos , Profármacos , Especies Reactivas de Oxígeno , Triterpenos Pentacíclicos/farmacología , Triterpenos Pentacíclicos/química , Profármacos/química , Profármacos/farmacología , Adenosina Trifosfato/metabolismo , Humanos , Animales , Especies Reactivas de Oxígeno/metabolismo , Ratones , Línea Celular Tumoral , Triterpenos/química , Triterpenos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Quelantes/química , Quelantes/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Microambiente Tumoral/efectos de los fármacos , Liberación de Fármacos , Nanopartículas/química , Ensayos Antitumor por Modelo de Xenoinjerto , Compuestos Férricos/química
7.
MAbs ; 16(1): 2373325, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38962811

RESUMEN

T-cell engaging (TCE) bispecific antibodies are potent drugs that trigger the immune system to eliminate cancer cells, but administration can be accompanied by toxic side effects that limit dosing. TCEs function by binding to cell surface receptors on T cells, frequently CD3, with one arm of the bispecific antibody while the other arm binds to cell surface antigens on cancer cells. On-target, off-tumor toxicity can arise when the target antigen is also present on healthy cells. The toxicity of TCEs may be ameliorated through the use of pro-drug forms of the TCE, which are not fully functional until recruited to the tumor microenvironment. This can be accomplished by masking the anti-CD3 arm of the TCE with an autoinhibitory motif that is released by tumor-enriched proteases. Here, we solve the crystal structure of the antigen-binding fragment of a novel anti-CD3 antibody, E10, in complex with its epitope from CD3 and use this information to engineer a masked form of the antibody that can activate by the tumor-enriched protease matrix metalloproteinase 2 (MMP-2). We demonstrate with binding experiments and in vitro T-cell activation and killing assays that our designed prodrug TCE is capable of tumor-selective T-cell activity that is dependent upon MMP-2. Furthermore, we demonstrate that a similar masking strategy can be used to create a pro-drug form of the frequently used anti-CD3 antibody SP34. This study showcases an approach to developing immune-modulating therapeutics that prioritizes safety and has the potential to advance cancer immunotherapy treatment strategies.


Asunto(s)
Anticuerpos Biespecíficos , Complejo CD3 , Inmunoterapia , Profármacos , Linfocitos T , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/farmacología , Humanos , Complejo CD3/inmunología , Inmunoterapia/métodos , Linfocitos T/inmunología , Profármacos/farmacología , Profármacos/química , Neoplasias/inmunología , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Ingeniería de Proteínas/métodos , Metaloproteinasa 2 de la Matriz/inmunología
8.
Langmuir ; 40(29): 14908-14921, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39001842

RESUMEN

It is crucial to use simple methods to prepare stable polymeric micelles with multiple functions for cancer treatment. Herein, via a "bottom-up" strategy, we reported the fabrication of ß-CD-(PEOSMA-PCPTMA-PPEGMA)21 (ßPECP) unimolecular micelles that could simultaneously treat tumors and bacteria with chemotherapy and photodynamic therapy (PDT). The unimolecular micelles consisted of a 21-arm ß-cyclodextrin (ß-CD) core as a macromolecular initiator, photosensitizer eosin Y (EOS-Y) monomer EOSMA, anticancer drug camptothecin (CPT) monomer, and a hydrophilic shell PEGMA. Camptothecin monomer (CPTMA) could achieve controlled release of the CPT due to the presence of responsively broken disulfide bonds. PEGMA enhanced the biocompatibility of micelles as a hydrophilic shell. Two ßPECP with different lengths were synthesized by modulating reaction conditions and the proportion of monomers, which both were self-assembled to unimolecular micelles in water. ßPECP unimolecular micelles with higher EOS-Y/CPT content exhibited more excellent 1O2 production, in vitro drug release efficiency, higher cytotoxicity, and superior antibacterial activity. Also, we carried out simulations of the self-assembly and CPT release process of micelles, which agreed with the experiments. This nanosystem, which combines antimicrobial and antitumor functions, provides new ideas for bacteria-mediated tumor clinical chemoresistance.


Asunto(s)
Antineoplásicos , Micelas , Fotoquimioterapia , Fármacos Fotosensibilizantes , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Humanos , Antineoplásicos/química , Antineoplásicos/farmacología , Profármacos/química , Profármacos/farmacología , Antibacterianos/química , Antibacterianos/farmacología , Animales , Ratones , beta-Ciclodextrinas/química , Camptotecina/química , Camptotecina/farmacología
9.
Adv Pharmacol ; 100: 157-180, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39034051

RESUMEN

The recognition that rapidly proliferating cancer cells rely heavily on glutamine for their survival and growth has renewed interest in the development of glutamine antagonists for cancer therapy. Glutamine plays a pivotal role as a carbon source for synthesizing lipids and metabolites through the TCA cycle, as well as a nitrogen source for synthesis of amino acid and nucleotides. Numerous studies have explored the significance of glutamine metabolism in cancer, providing a robust rationale for targeting this metabolic pathway in cancer treatment. The glutamine antagonist 6-diazo-5-oxo-l-norleucine (DON) has been explored as an anticancer therapeutic for nearly six decades. Initial investigations revealed remarkable efficacy in preclinical studies and promising outcomes in early clinical trials. However, further advancement of DON was hindered due to dose-limiting gastrointestinal (GI) toxicities as the GI system is highly dependent on glutamine for regulating growth and repair. In an effort to repurpose DON and mitigate gastrointestinal (GI) toxicity concerns, prodrug strategies were utilized. These strategies aimed to enhance the delivery of DON to specific target tissues, such as tumors and the central nervous system (CNS), while sparing DON delivery to normal tissues, particularly the GI tract. When administered at low daily doses, optimized for metabolic inhibition, these prodrugs exhibit remarkable effectiveness without inducing significant toxicity to normal tissues. This approach holds promise for overcoming past challenges associated with DON, offering an avenue for its successful utilization in cancer treatment.


Asunto(s)
Diazooxonorleucina , Neoplasias , Profármacos , Humanos , Profármacos/farmacología , Profármacos/uso terapéutico , Diazooxonorleucina/farmacología , Diazooxonorleucina/uso terapéutico , Animales , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Glutamina/metabolismo
10.
J Am Chem Soc ; 146(29): 20080-20085, 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39001844

RESUMEN

The utility of antibody therapeutics is hampered by potential cross-reactivity with healthy tissue. Over the past decade, significant advances have been made in the design of activatable antibodies, which increase, or create altogether, the therapeutic window of a parent antibody. Of these, antibody prodrugs (pro-antibodies) are masked antibodies that have advanced the most for therapeutic use. They are designed to reveal the active, parent antibody only when encountering proteases upregulated in the microenvironment of the targeted disease tissue, thereby minimizing off-target activity. However, current pro-antibody designs are relegated to fusion proteins that append masking groups restricted to the use of only canonical amino acids, offering excellent control of the site of introduction, but with no authority over where the masking group is installed other than the N-terminus of the antibody. Here, we present a palladium-based bioconjugation approach for the site-specific introduction of a masked tyrosine mimic in the complementary determining region of the FDA approved antibody therapeutic ipilimumab used as a model system. The approach enables the introduction of a protease cleavable group tethered to noncanonical polymers (polyethylene glycol (PEG)) resulting in 47-fold weaker binding to cells expressing CTLA-4, the target antigen of ipilimumab. Upon exposure to tumor-associated proteases, the masking group is cleaved, unveiling a tyrosine-mimic (dubbed hydroxyphenyl cysteine (HPC)) that restores (>90% restoration) binding affinity to its target antigen.


Asunto(s)
Profármacos , Tirosina , Profármacos/química , Profármacos/farmacología , Humanos , Tirosina/química , Paladio/química , Estructura Molecular , Inmunoconjugados/química
11.
Int J Pharm ; 661: 124422, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38977163

RESUMEN

The challenges posed by intractable relapse and metastasis in cancer treatment have led to the development of various forms of photodynamic therapy (PDT). However, traditional drug delivery systems, such as virus vectors, liposomes, and polymers, often suffer from issues like desynchronized drug release, carrier instability, and drug leakage during circulation. To address these problems, we have developed a dual-prodrug nanogel (PVBN) consisting of Pyro (Pyropheophorbide a) and SAHA (Vorinostat) bound to BSA (Bovine Serum Albumin), which facilitates synchronous and spontaneous drug release in situ within the lysosome. Detailed results indicate that PVBN-treated tumor cells exhibit elevated levels of ROS and Acetyl-H3, leading to necrosis, apoptosis, and cell cycle arrest, with PDT playing a dominant role in the synergistic therapeutic effect. Furthermore, the anti-tumor efficacy of PVBN was validated in melanoma-bearing mice, where it significantly inhibited tumor growth and pulmonary metastasis. Overall, our dual-prodrug nanogel, formed by the binding of SAHA and Pyro to BSA and releasing drugs within the lysosome, represents a novel and promising strategy for enhancing the clinical efficacy of photochemotherapy.


Asunto(s)
Clorofila , Nanogeles , Fotoquimioterapia , Profármacos , Albúmina Sérica Bovina , Vorinostat , Animales , Vorinostat/administración & dosificación , Vorinostat/farmacología , Vorinostat/química , Fotoquimioterapia/métodos , Clorofila/análogos & derivados , Clorofila/química , Clorofila/administración & dosificación , Clorofila/farmacología , Albúmina Sérica Bovina/química , Albúmina Sérica Bovina/administración & dosificación , Línea Celular Tumoral , Nanogeles/química , Profármacos/administración & dosificación , Profármacos/química , Ratones , Apoptosis/efectos de los fármacos , Liberación de Fármacos , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacología , Humanos , Especies Reactivas de Oxígeno/metabolismo , Fármacos Fotosensibilizantes/administración & dosificación , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/química , Ratones Endogámicos C57BL , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Melanoma Experimental/tratamiento farmacológico , Polietileneimina/química
12.
Sci Rep ; 14(1): 14175, 2024 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-38898077

RESUMEN

Central nervous system tumors have resisted effective chemotherapy because most therapeutics do not penetrate the blood-tumor-brain-barrier. Nanomedicines between ~ 10 and 100 nm accumulate in many solid tumors by the enhanced permeability and retention effect, but it is controversial whether the effect can be exploited for treatment of brain tumors. PLX038A is a long-acting prodrug of the topoisomerase 1 inhibitor SN-38. It is composed of a 15 nm 4-arm 40 kDa PEG tethered to four SN-38 moieties by linkers that slowly cleave to release the SN-38. The prodrug was remarkably effective at suppressing growth of intracranial breast cancer and glioblastoma (GBM), significantly increasing the life span of mice harboring them. We addressed the important issue of whether the prodrug releases SN-38 systemically and then penetrates the brain to exert anti-tumor effects, or whether it directly penetrates the blood-tumor-brain-barrier and releases the SN-38 cargo within the tumor. We argue that the amount of SN-38 formed systemically is insufficient to inhibit the tumors, and show by PET imaging that a close surrogate of the 40 kDa PEG carrier in PLX038A accumulates and is retained in the GBM. We conclude that the prodrug penetrates the blood-tumor-brain-barrier, accumulates in the tumor microenvironment and releases its SN-38 cargo from within. Based on our results, we pose the provocative question as to whether the 40 kDa nanomolecule PEG carrier might serve as a "Trojan horse" to carry other drugs past the blood-tumor-brain-barrier and release them into brain tumors.


Asunto(s)
Barrera Hematoencefálica , Neoplasias Encefálicas , Irinotecán , Profármacos , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Irinotecán/farmacocinética , Barrera Hematoencefálica/metabolismo , Ratones , Profármacos/farmacocinética , Profármacos/química , Profármacos/metabolismo , Humanos , Línea Celular Tumoral , Femenino , Ensayos Antitumor por Modelo de Xenoinjerto , Glioblastoma/metabolismo , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Camptotecina/análogos & derivados , Camptotecina/farmacocinética , Camptotecina/uso terapéutico
13.
J Nanobiotechnology ; 22(1): 358, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38907270

RESUMEN

BACKGROUND: Hypoxia-activated prodrug (HAP) is a promising candidate for highly tumor-specific chemotherapy. However, the oxygenation heterogeneity and dense extracellular matrix (ECM) of tumor, as well as the potential resistance to chemotherapy, have severely impeded the resulting overall efficacy of HAP. RESULTS: A HAP potentiating strategy is proposed based on ultrasound responsive nanodroplets (PTP@PLGA), which is composed of protoporphyrin (PpIX), perfluoropropane (PFP) and a typical HAP, tirapazamine (TPZ). The intense vaporization of PFP upon ultrasound irradiation can magnify the sonomechanical effect, which loosens the ECM to promote the penetration of TPZ into the deep hypoxic region. Meanwhile, the PpIX enabled sonodynamic effect can further reduce the oxygen level, thus activating the TPZ in the relatively normoxic region as well. Surprisingly, abovementioned ultrasound effect also results in the downregulation of the stemness of cancer cells, which is highly associated with drug-refractoriness. CONCLUSIONS: This work manifests an ideal example of ultrasound-based nanotechnology for potentiating HAP and also reveals the potential acoustic effect of intervening cancer stem-like cells.


Asunto(s)
Fluorocarburos , Nanopartículas , Profármacos , Protoporfirinas , Tirapazamina , Humanos , Tirapazamina/farmacología , Tirapazamina/química , Protoporfirinas/farmacología , Protoporfirinas/química , Fluorocarburos/química , Fluorocarburos/farmacología , Profármacos/farmacología , Profármacos/química , Línea Celular Tumoral , Nanopartículas/química , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/química , Ondas Ultrasónicas , Animales , Matriz Extracelular/metabolismo , Ratones , Neoplasias/tratamiento farmacológico
14.
Chem Commun (Camb) ; 60(51): 6524-6527, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38836387

RESUMEN

Synthesis and in vitro testing of a prodrug designed for the controlled delivery of the anticancer drug camptothecin within pancreatic cancer cells are reported. Our study reveals a non-conventional pharmacokinetic release characterized by an exponential pattern before reaching the half-life (t1/2) and a linear pattern thereafter. The release mechanism was triggered either by hydrolytic enzymes and/or by the acid microenvironment of cancer cells.


Asunto(s)
Camptotecina , Profármacos , Humanos , Camptotecina/química , Camptotecina/farmacología , Profármacos/química , Profármacos/farmacología , Profármacos/síntesis química , Línea Celular Tumoral , Liberación de Fármacos , Neoplasias Pancreáticas/tratamiento farmacológico , Antineoplásicos/química , Antineoplásicos/farmacología , Preparaciones de Acción Retardada/química , Estructura Molecular
15.
J Nanobiotechnology ; 22(1): 374, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38926723

RESUMEN

BACKGROUND: Hypoxia-activated prodrugs present new opportunities for safe and effective tumor drug resistance therapy due to their high selectivity for hypoxic cells. However, the uneven distribution of oxygen in solid tumor and insufficient hypoxia in the tumor microenvironment greatly limit its therapeutic efficacy. RESULTS: In this paper, a novel AQ4N-Mn(II)@PDA coordination nanoplatform was designed and functionalized with GMBP1 to target drug-resistant tumor cells. Its excellent photothermal conversion efficiency could achieve local high-temperature photothermal therapy in tumors, which could not only effectively exacerbate tumor hypoxia and thus improve the efficacy of hypoxia-activated chemotherapy of AQ4N but also significantly accelerate Mn2+-mediated Fenton-like activity to enhance chemodynamic therapy. Moreover, real-time monitoring of blood oxygen saturation through photoacoustic imaging could reflect the hypoxic status of tumors during treatment. Furthermore, synergistic treatment effectively inhibited tumor growth and improved the survival rate of mice bearing orthotopic drug-resistant tumors. CONCLUSIONS: This study not only provided a new idea for PTT combined with hypoxia-activated chemotherapy and CDT for drug-resistant tumors but also explored a vital theory for real-time monitoring of hypoxia during treatment.


Asunto(s)
Resistencia a Antineoplásicos , Terapia Fototérmica , Animales , Ratones , Resistencia a Antineoplásicos/efectos de los fármacos , Línea Celular Tumoral , Humanos , Terapia Fototérmica/métodos , Ratones Endogámicos BALB C , Nanopartículas/química , Antineoplásicos/farmacología , Antineoplásicos/química , Microambiente Tumoral/efectos de los fármacos , Ratones Desnudos , Profármacos/farmacología , Profármacos/química , Hipoxia Tumoral/efectos de los fármacos , Manganeso/química , Femenino , Neoplasias/tratamiento farmacológico , Antraquinonas
16.
Int J Mol Sci ; 25(12)2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38928299

RESUMEN

Bacterial nitroreductase enzymes capable of activating imaging probes and prodrugs are valuable tools for gene-directed enzyme prodrug therapies and targeted cell ablation models. We recently engineered a nitroreductase (E. coli NfsB F70A/F108Y) for the substantially enhanced reduction of the 5-nitroimidazole PET-capable probe, SN33623, which permits the theranostic imaging of vectors labeled with oxygen-insensitive bacterial nitroreductases. This mutant enzyme also shows improved activation of the DNA-alkylation prodrugs CB1954 and metronidazole. To elucidate the mechanism behind these enhancements, we resolved the crystal structure of the mutant enzyme to 1.98 Å and compared it to the wild-type enzyme. Structural analysis revealed an expanded substrate access channel and new hydrogen bonding interactions. Additionally, computational modeling of SN33623, CB1954, and metronidazole binding in the active sites of both the mutant and wild-type enzymes revealed key differences in substrate orientations and interactions, with improvements in activity being mirrored by reduced distances between the N5-H of isoalloxazine and the substrate nitro group oxygen in the mutant models. These findings deepen our understanding of nitroreductase substrate specificity and catalytic mechanisms and have potential implications for developing more effective theranostic imaging strategies in cancer treatment.


Asunto(s)
Metronidazol , Nitroimidazoles , Nitrorreductasas , Nitrorreductasas/metabolismo , Nitrorreductasas/química , Nitrorreductasas/genética , Nitroimidazoles/química , Nitroimidazoles/metabolismo , Metronidazol/química , Metronidazol/metabolismo , Metronidazol/farmacología , Profármacos/metabolismo , Profármacos/química , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Tomografía de Emisión de Positrones/métodos , Escherichia coli/genética , Escherichia coli/metabolismo , Dominio Catalítico , Ingeniería de Proteínas , Modelos Moleculares , Aziridinas/química , Aziridinas/metabolismo
17.
ACS Appl Bio Mater ; 7(6): 3991-3996, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38835291

RESUMEN

Mitigating the adverse effects of anticancer agents requires innovative prodrug engineering. In this study, we showcase the potential of our o-quinone methide-based trigger-release-conjugation platform as a versatile tool for constructing advanced prodrug systems. Using this platform, we achieved the light-triggered release of an anticancer drug mechlorethamine, targeting mitochondrial DNA. The entire process was adeptly tracked through the emission of fluorescence signals, revealing notable effects across various cancer cell lines compared to a normal cell line. Exploring alternative cancer-associated triggers, including enzymes, and incorporating cancer/tumor-specific targeting elements could lead to effective prodrugs with reduced cytotoxicity.


Asunto(s)
Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Luz , Mitocondrias , Profármacos , Profármacos/química , Profármacos/farmacología , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Ensayo de Materiales , Estructura Molecular , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Supervivencia Celular/efectos de los fármacos , Fluorescencia , Tamaño de la Partícula , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Liberación de Fármacos
18.
Chem Commun (Camb) ; 60(50): 6427-6430, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38829169

RESUMEN

We developed prodrug nanoparticles that release drugs through intracellular dissolution and a cancer-specific hydrogen peroxide response. To reveal the unclear mechanism regarding drug release from nanoparticles by reacting with hydrogen peroxide in cancer cells, this study demonstrates the in vitro evaluation of drug release kinetics under conditions simulated in cancer cells.


Asunto(s)
Antineoplásicos , Liberación de Fármacos , Peróxido de Hidrógeno , Nanopartículas , Profármacos , Peróxido de Hidrógeno/química , Humanos , Profármacos/química , Profármacos/farmacología , Nanopartículas/química , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Solubilidad , Portadores de Fármacos/química , Doxorrubicina/química , Doxorrubicina/farmacología
19.
Chem Commun (Camb) ; 60(57): 7335-7338, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38915280

RESUMEN

A nucleus-targeted nanocomposite was prepared by clickable amino acid-tuned one-step co-assembly of proteins and chemotherapeutics. The nanocomposite with favorable pharmacokinetic behavior can effectively accumulate in the nucleus, thereby significantly enhancing the anticancer therapeutic effect both in vitro and in vivo.


Asunto(s)
Aminoácidos , Antineoplásicos , Profármacos , Profármacos/química , Profármacos/farmacología , Aminoácidos/química , Aminoácidos/farmacología , Humanos , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/síntesis química , Animales , Núcleo Celular/metabolismo , Química Clic , Ratones , Línea Celular Tumoral , Nanocompuestos/química , Neoplasias/tratamiento farmacológico
20.
Int J Pharm ; 660: 124300, 2024 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-38851409

RESUMEN

Uveal melanoma is one of the most common and aggressive intraocular malignancies, and, due to its great capability of metastasize, it constitutes the most incident intraocular tumor in adults. However, to date there is no effective treatment since achieving the inner ocular tissues still constitutes one of the greatest challenges in actual medicine, because of the complex structure and barriers. Uncoated and PEGylated nanostructured lipid carriers were developed to achieve physico-chemical properties (mean particle size, homogeneity, zeta potential, pH and osmolality) compatible for the ophthalmic administration of (S)-(-)-MRJF22, a new custom-synthetized prodrug for the potential treatment of uveal melanoma. The colloidal physical stability was investigated at different temperatures by Turbiscan® Ageing Station. Morphology analysis and mucoadhesive studies highlighted the presence of small particles suitable to be topically administered on the ocular surface. In vitro release studies performed using Franz diffusion cells demonstrated that the systems were able to provide a slow and prolonged prodrug release. In vitro cytotoxicity test on Human Corneal Epithelium and Human Uveal Melanoma cell lines and Hen's egg-chorioallantoic membrane test showed a dose-dependent cytotoxic effect of the free prodrug on corneal cells, whose cytocompatibility improved when encapsulated into nanoparticles, as also confirmed by in vivo studies on New Zealand albino rabbits. Antiangiogenic capability and preventive anti-inflammatory properties were also investigated on embryonated eggs and rabbits, respectively. Furthermore, preliminary in vivo biodistribution images of fluorescent nanoparticles after topical instillation in rabbits' eyes, suggested their ability to reach the posterior segment of the eye, as a promising strategy for the treatment of choroidal uveal melanoma.


Asunto(s)
Administración Oftálmica , Membrana Corioalantoides , Portadores de Fármacos , Melanoma , Nanopartículas , Profármacos , Neoplasias de la Úvea , Neoplasias de la Úvea/tratamiento farmacológico , Neoplasias de la Úvea/patología , Melanoma/tratamiento farmacológico , Melanoma/patología , Animales , Humanos , Conejos , Línea Celular Tumoral , Membrana Corioalantoides/efectos de los fármacos , Portadores de Fármacos/química , Nanopartículas/química , Nanopartículas/administración & dosificación , Profármacos/administración & dosificación , Profármacos/química , Lípidos/química , Lípidos/administración & dosificación , Liberación de Fármacos , Supervivencia Celular/efectos de los fármacos , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Polietilenglicoles/química , Polietilenglicoles/administración & dosificación , Embrión de Pollo , Epitelio Corneal/efectos de los fármacos , Tamaño de la Partícula
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA