Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.878
Filtrar
Mais filtros

Eixos temáticos
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(20): e2318119121, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38709930

RESUMO

Brain metastasis of advanced breast cancer often results in deleterious consequences. Metastases to the brain lead to significant challenges in treatment options, as the blood-brain barrier (BBB) prevents conventional therapy. Thus, we hypothesized that creation of a nanoparticle (NP) that distributes to both primary tumor site and across the BBB for secondary brain tumor can be extremely beneficial. Here, we report a simple targeting strategy to attack both the primary breast and secondary brain tumors utilizing a single NP platform. The nature of these mitochondrion-targeted, BBB-penetrating NPs allow for simultaneous targeting and drug delivery to the hyperpolarized mitochondrial membrane of the extracranial primary tumor site in addition to tumors at the brain. By utilizing a combination of such dual anatomical distributing NPs loaded with therapeutics, we demonstrate a proof-of-concept idea to combat the increased metabolic plasticity of brain metastases by lowering two major energy sources, oxidative phosphorylation (OXPHOS) and glycolysis. By utilizing complementary studies and genomic analyses, we demonstrate the utility of a chemotherapeutic prodrug to decrease OXPHOS and glycolysis by pairing with a NP loaded with pyruvate dehydrogenase kinase 1 inhibitor. Decreasing glycolysis aims to combat the metabolic flexibility of both primary and secondary tumors for therapeutic outcome. We also address the in vivo safety parameters by addressing peripheral neuropathy and neurobehavior outcomes. Our results also demonstrate that this combination therapeutic approach utilizes mitochondrial genome targeting strategy to overcome DNA repair-based chemoresistance mechanisms.


Assuntos
Barreira Hematoencefálica , Neoplasias Encefálicas , Neoplasias da Mama , Nanopartículas , Fosforilação Oxidativa , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundário , Neoplasias Encefálicas/patologia , Animais , Humanos , Feminino , Nanopartículas/química , Camundongos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Fosforilação Oxidativa/efeitos dos fármacos , Linhagem Celular Tumoral , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Sistemas de Liberação de Medicamentos/métodos , Glicólise/efeitos dos fármacos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico
2.
Proc Natl Acad Sci U S A ; 120(21): e2304081120, 2023 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-37186828

RESUMO

Chemotherapy typically destroys the tumor mass but rarely eradicates the cancer stem cells (CSCs) that can drive metastatic recurrence. A key current challenge is finding ways to eradicate CSCs and suppress their characteristics. Here, we report a prodrug, Nic-A, created by combining a carbonic anhydrase IX (CAIX) inhibitor, acetazolamide, with a signal transducer and transcriptional activator 3 (STAT3) inhibitor, niclosamide. Nic-A was designed to target triple-negative breast cancer (TNBC) CSCs and was found to inhibit both proliferating TNBC cells and CSCs via STAT3 dysregulation and suppression of CSC-like properties. Its use leads to a decrease in aldehyde dehydrogenase 1 activity, CD44high/CD24low stem-like subpopulations, and tumor spheroid-forming ability. TNBC xenograft tumors treated with Nic-A exhibited decreased angiogenesis and tumor growth, as well as decreased Ki-67 expression and increased apoptosis. In addition, distant metastases were suppressed in TNBC allografts derived from a CSC-enriched population. This study thus highlights a potential strategy for addressing CSC-based cancer recurrence.


Assuntos
Pró-Fármacos , Neoplasias de Mama Triplo Negativas , Humanos , Linhagem Celular Tumoral , Neoplasias de Mama Triplo Negativas/metabolismo , Niclosamida/farmacologia , Niclosamida/metabolismo , Niclosamida/uso terapêutico , Pró-Fármacos/uso terapêutico , Recidiva Local de Neoplasia/patologia , Fatores de Transcrição/metabolismo , Células-Tronco Neoplásicas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Nature ; 569(7757): 509-513, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31068699

RESUMO

A universal gain-of-function approach for selective and temporal control of protein activity in living systems is crucial to understanding dynamic cellular processes. Here we report development of a computationally aided and genetically encoded proximal decaging (hereafter, CAGE-prox) strategy that enables time-resolved activation of a broad range of proteins in living cells and mice. Temporal blockage of protein activity was computationally designed and realized by genetic incorporation of a photo-caged amino acid in proximity to the functional site of the protein, which can be rapidly removed upon decaging, resulting in protein re-activation. We demonstrate the wide applicability of our method on diverse protein families, which enabled orthogonal tuning of cell signalling and immune responses, temporal profiling of proteolytic substrates upon caspase activation as well as the development of protein-based pro-drug therapy. We envision that CAGE-prox will open opportunities for the gain-of-function study of proteins and dynamic biological processes with high precision and temporal resolution.


Assuntos
Sobrevivência Celular , Proteínas/metabolismo , Animais , Caspase 3/genética , Caspase 3/metabolismo , Linhagem Celular , Ativação Enzimática , Mutação com Ganho de Função , Humanos , Masculino , Camundongos , Neoplasias/tratamento farmacológico , Fosfotransferases/metabolismo , Pró-Fármacos/metabolismo , Pró-Fármacos/uso terapêutico , Proteínas/genética , Proteínas/imunologia , Proteínas/uso terapêutico , Proteólise , Proteômica , Transdução de Sinais , Fatores de Tempo
4.
Proc Natl Acad Sci U S A ; 119(28): e2201423119, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35867758

RESUMO

Treatments for advanced and recurrent ovarian cancer remain a challenge due to a lack of potent, selective, and effective therapeutics. Here, we developed the basis for a transformative anticancer strategy based on anthrax toxin that has been engineered to be selectively activated by the catalytic power of zymogen-activating proteases on the surface of malignant tumor cells to induce cell death. Exposure to the engineered toxin is cytotoxic to ovarian tumor cell lines and ovarian tumor spheroids derived from patient ascites. Preclinical studies demonstrate that toxin treatment induces tumor regression in several in vivo ovarian cancer models, including patient-derived xenografts, without adverse side effects, supportive of progression toward clinical evaluation. These data lay the groundwork for developing therapeutics for treating women with late-stage and recurrent ovarian cancers, utilizing a mechanism distinct from current anticancer therapies.


Assuntos
Antígenos de Bactérias , Antineoplásicos , Toxinas Bacterianas , Neoplasias Ovarianas , Pró-Fármacos , Serina Proteases , Antígenos de Bactérias/farmacologia , Antígenos de Bactérias/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Toxinas Bacterianas/farmacologia , Toxinas Bacterianas/uso terapêutico , Linhagem Celular Tumoral , Precursores Enzimáticos/metabolismo , Feminino , Humanos , Recidiva Local de Neoplasia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Serina Proteases/metabolismo , Esferoides Celulares , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Nano Lett ; 24(28): 8741-8751, 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-38953486

RESUMO

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.


Assuntos
Imunoterapia , Neoplasias Pancreáticas , Pró-Fármacos , Animais , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Pró-Fármacos/química , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/imunologia , Camundongos , Humanos , Linhagem Celular Tumoral , Proteólise/efeitos dos fármacos , Terapia por Ultrassom/métodos , Antígeno B7-H1 , Fatores de Transcrição , Proteínas de Ciclo Celular , Espécies Reativas de Oxigênio/metabolismo , Proteínas que Contêm Bromodomínio
6.
Antimicrob Agents Chemother ; 68(7): e0048924, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38775484

RESUMO

Human adenoviruses can cause serious, disseminated infections in immunocompromised patients. For pediatric allogeneic stem cell transplant patients, the case fatality rate can reach 80%. Still, there is no available antiviral drug that is specifically approved by the Food and Drug Administration for the treatment of adenovirus infections. To fill this pressing medical need, we have developed NPP-669, a prodrug of cidofovir with broad activity against double-stranded DNA viruses, including adenoviruses. Here, we report on the in vivo anti-adenoviral efficacy of NPP-669. Using the immunosuppressed Syrian hamster as the model, we show that NPP-669 is highly efficacious when dosed orally at 1 mg/kg and 3 mg/kg. In a delayed administration experiment, NPP-669 was more effective than brincidofovir, a similar compound that reached Phase III clinical trials. Furthermore, parenteral administration of NPP-669 increased its efficacy approximately 10-fold compared to oral dosing without apparent toxicity, suggesting that this route may be preferable in a hospital setting. Based on these findings, we believe that NPP-669 is a promising new compound that needs to be further investigated.


Assuntos
Antivirais , Cidofovir , Citosina , Mesocricetus , Organofosfonatos , Pró-Fármacos , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Humanos , Cidofovir/farmacologia , Cidofovir/uso terapêutico , Organofosfonatos/farmacologia , Organofosfonatos/uso terapêutico , Citosina/análogos & derivados , Citosina/farmacologia , Citosina/uso terapêutico , Adenovírus Humanos/efeitos dos fármacos , Infecções por Adenovirus Humanos/tratamento farmacológico , Infecções por Adenovirus Humanos/virologia , Modelos Animais de Doenças , Cricetinae , Administração Oral
7.
Anal Chem ; 96(12): 5006-5013, 2024 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-38484040

RESUMO

The development of new imaging and treatment nanoprodrug systems is highly demanded for diagnosis and therapy of liver cancer, a severe disease characterized by a high recurrence rate. Currently, available small molecule drugs are not possible for cancer diagnosis because of the fast diffusion of imaging agents and low efficacy in treatment due to poor water solubility and significant toxic side effects. In this study, we report the development of a tumor microenvironment activatable nanoprodrug system for the diagnosis and treatment of liver cancer. This nanoprodrug system can accumulate in the tumor site and be selectively activated by an excess of hydrogen peroxide (H2O2) in the tumor microenvironment, releasing near-infrared solid-state organic fluorescent probe (HPQCY-1) and phenylboronic acid-modified camptothecin (CPT) prodrug. Both HPQCY-1 and CPT prodrugs can be further activated in tumor sites for achieving more precise in situ near-infrared (NIR) fluorescence imaging and treatment while reducing the toxic effects of drugs on normal tissues. Additionally, the incorporation of hydrophilic multivalent chitosan as a carrier effectively improved the water solubility of the system. This research thus provides a practical new approach for the diagnosis and treatment of liver cancer.


Assuntos
Neoplasias Hepáticas , Nanopartículas , Pró-Fármacos , Humanos , Microambiente Tumoral , Peróxido de Hidrogênio , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Neoplasias Hepáticas/diagnóstico por imagem , Neoplasias Hepáticas/tratamento farmacológico , Imagem Óptica , Água , Linhagem Celular Tumoral , Camptotecina/farmacologia
8.
Small ; 20(24): e2309482, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38150668

RESUMO

Developing intelligently targeted drugs with low side effects is urgent for cancer treatment. Toward this goal, a tumor-specific cascade-activating smart prodrug system consisting of a G-quadruplex(G4)-modulated tumor-targeted DNA vehicle and a well-designed cellular stimuli-responsive ligand-drug conjugates (LDCs) is proposed. An original "donor-acceptor" binary fluorescent ligand, with ultrahigh affinity, brightness, and photostability, is engineered to tightly bind G4 structures and significantly improve the nuclease resistance of the DNA vehicle, which serves as a bridge contributing to the construction of the prodrug system, named ApG4/LDCs. Sodium nitroprusside and doxorubicin are loaded into ApG4/LDCs in one pot and generate nitric oxide and superoxide anion in response to cancer cellular environments, which in cascade generates peroxynitrite to cause DNA damage while promoting the self-monitored drug release to achieve enhanced targeted therapy. Such a cascade activation and self-reinforcement process is executed only when the prodrug system targets the tumor tissue followed by cell uptake, showing significant antitumor efficacy and greatly weakening the damage to normal tissues. Given the unique features, the innovative strategy for prodrug design may open a new door to precision disease treatment.


Assuntos
Doxorrubicina , Pró-Fármacos , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Humanos , Doxorrubicina/farmacologia , Doxorrubicina/química , Animais , Linhagem Celular Tumoral , Neoplasias/tratamento farmacológico , Quadruplex G , Ligantes
9.
Chemistry ; 30(48): e202401593, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-38923644

RESUMO

Bioinspired molecular engineering strategies have emerged as powerful tools that significantly enhance the development of novel therapeutics, improving efficacy, specificity, and safety in disease treatment. Recent advancements have focused on identifying and utilizing disease-associated biomarkers to optimize drug activity and address challenges inherent in traditional therapeutics, such as frequent drug administrations, poor patient adherence, and increased risk of adverse effects. In this review, we provide a comprehensive overview of the latest developments in bioinspired artificial systems (BAS) that use molecular engineering to tailor therapeutic responses to drugs in the presence of disease-specific biomarkers. We examine the transition from open-loop systems, which rely on external cues, to closed-loop feedback systems capable of autonomous self-regulation in response to disease-associated biomarkers. We detail various BAS modalities designed to achieve biomarker-driven therapy, including activatable prodrug molecules, smart drug delivery platforms, autonomous artificial cells, and synthetic receptor-based cell therapies, elucidating their operational principles and practical in vivo applications. Finally, we discuss the current challenges and future perspectives in the advancement of BAS-enabled technology and envision that ongoing advancements toward more programmable and customizable BAS-based therapeutics will significantly enhance precision medicine.


Assuntos
Biomarcadores , Sistemas de Liberação de Medicamentos , Humanos , Sistemas de Liberação de Medicamentos/métodos , Pró-Fármacos/química , Pró-Fármacos/uso terapêutico , Medicina de Precisão/métodos , Células Artificiais/química , Animais
10.
Mol Pharm ; 21(4): 1838-1847, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38413029

RESUMO

The extensive use of opioids for chronic pain management has contributed significantly to the current opioid epidemic. While many alternative nonopioid analgesics are available, opioids remain the most potent analgesics for moderate to severe pain management. In addition to the implementation of multimodal analgesia, there is a pressing need for the development of more effective and safer opioids. In this study, we developed a thermoresponsive N-(2-hydroxypropyl) methacrylamide (HPMA) copolymer-based hydromorphone (HMP) prodrug (ProGel-HMP, HMP content = 16.2 wt %, in base form). The aqueous solution of ProGel-HMP was free-flowing at 4 °C but became a hydrogel when the temperature was raised to ≥37 °C, allowing sustained local retention when administered in vivo. When tested in the destabilization of the medial meniscus (DMM) mouse model of osteoarthritis (OA), ProGel-HMP was retained after intra-articular injection in the OA knee joint for at least 2 weeks postinjection, with low extra-articular distribution. ProGel-HMP was not detected in the central nervous system (CNS). A single dose of ProGel-HMP produced rapid and sustained joint pain resolution for greater than 14 days when compared to saline and dose-equivalent HMP controls, likely mediated through peripheral µ-opioid receptors in the knee joint. Systemic analgesia effect was absent in the DMM mice treated with ProGel-HMP, as evident in the lack of difference in tail flick response between the ProGel-HMP-treated mice and the controls (i.e., Healthy, Saline, and Sham). Repeated dosing of ProGel-HMP did not induce tolerance. Collectively, these data support the further development of ProGel-HMP as a potent, safe, long-acting and nonaddictive analgesic for better clinical pain management.


Assuntos
Analgesia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Osteoartrite , Pró-Fármacos , Camundongos , Animais , Hidromorfona , Manejo da Dor , Pró-Fármacos/uso terapêutico , Dor/tratamento farmacológico , Analgésicos Opioides/efeitos adversos , Analgésicos/uso terapêutico
11.
Mol Pharm ; 21(2): 688-703, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38243899

RESUMO

Persistent pain is a significant healthcare problem with limited treatment options. The high incidence of comorbid chronic pain and depression significantly reduces life quality and complicates the treatment of both conditions. Antidepressants are less effective for pain and depression than for depression alone and they induce severe side effects. Opioids are highly efficacious analgesics, but rapid development of tolerance, dependence, and debilitating side effects limit their efficacy and safe use. Leucine-enkephalin (Leu-ENK), the endogenous delta opioid receptor agonist, controls pain and mood and produces potent analgesia with reduced adverse effects compared to conventional opioids. High proteolytic instability, however, makes Leu-ENK ineffective after systemic administration and limits its clinical usefulness. KK-103, a Leu-ENK prodrug, was developed to overcome these limitations of Leu-ENK via markedly increased plasma stability in mice. We showed rapid and substantially increased systemic adsorption and blood plasma exposure of KK-103 compared to Leu-ENK. We also observed brain uptake of radiolabeled KK-103 after systemic administration, indicating a central effect of KK-103. We then established KK-103's prolonged antinociceptive efficacy in the ramped hot plate and formalin test. In both models, KK-103 produced a comparable dose to the maximum antinociceptive-effect relationship. The pain-alleviating effect of KK-103 primarily resulted from activating the delta opioid receptor after the likely conversion of KK-103 to Leu-ENK in vivo. Finally, KK-103 produced an antidepressant-like activity comparable to the antidepressant desipramine, but with minimal gastrointestinal inhibition and no incidence of sedation.


Assuntos
Encefalina Leucina , Pró-Fármacos , Camundongos , Animais , Receptores Opioides delta/uso terapêutico , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Dor/tratamento farmacológico , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico
12.
Bioorg Chem ; 144: 107150, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38309002

RESUMO

Nucleobases serve as essential molecular frameworks present in both natural and synthetic compounds that exhibit notable antiviral activity. Through molecular modifications, novel nucleobase-containing drugs (NCDs) have been developed, exhibiting enhanced antiviral activity against a wide range of viruses, including the recently emerged SARS­CoV­2. This article provides a detailed examination of the significant advancements in NCDs from 2015 till current, encompassing various aspects concerning their mechanisms of action, pharmacology and antiviral properties. Additionally, the article discusses antiviral prodrugs relevant to the scope of this review. It fills in the knowledge gap by examining the structure-activity relationship and trend of NCDs as therapeutics against a diverse range of viral diseases, either as approved drugs, clinical candidates or as early-stage development prospects. Moreover, the article highlights on the status of this field of study and addresses the prevailing limitations encountered.


Assuntos
Pró-Fármacos , Vírus , Antivirais/farmacologia , Antivirais/uso terapêutico , Relação Estrutura-Atividade , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico
13.
Bioorg Chem ; 144: 107161, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38306826

RESUMO

Hypoxia, as a prevalent feature of solid tumors, is correlated with tumorigenesis, proliferation, and invasion, playing an important role in mediating the drug resistance and affecting the cancer treatment outcomes. Due to the distinct oxygen levels between tumor and normal tissues, hypoxia-targeted therapy has attracted significant attention. The hypoxia-activated compounds mainly depend on reducible organic groups including azo, nitro, N-oxides, quinones and azide as well as some redox-active metal complex that are selectively converted into active species by the increased reduction potential under tumor hypoxia. In this review, we briefly summarized our current understanding on hypoxia-activated compounds with a particular highlight on the recently developed prodrugs and fluorescent probes for tumor treatment and diagnosis. We have also discussed the challenges and perspectives of small molecule-based hypoxia-activatable prodrug for future development.


Assuntos
Neoplasias , Pró-Fármacos , Humanos , Hipóxia/diagnóstico , Hipóxia/tratamento farmacológico , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Hipóxia Celular , Hipóxia Tumoral , Linhagem Celular Tumoral
14.
Acta Pharmacol Sin ; 45(8): 1740-1751, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38609561

RESUMO

Proteolysis targeting chimeras (PROTACs) have emerged as revolutionary anticancer therapeutics that degrade disease-causing proteins. However, the anticancer performance of PROTACs is often impaired by their insufficient bioavailability, unsatisfactory tumor specificity and ability to induce acquired drug resistance. Herein, we propose a polymer-conjugated PROTAC prodrug platform for the tumor-targeted delivery of the most prevalent von Hippel-Lindau (VHL)- and cereblon (CRBN)-based PROTACs, as well as for the precise codelivery of a degrader and conventional small-molecule drugs. The self-assembling PROTAC prodrug nanoparticles (NPs) can specifically target and be activated inside tumor cells to release the free PROTAC for precise protein degradation. The PROTAC prodrug NPs caused more efficient regression of MDA-MB-231 breast tumors in a mouse model by degrading bromodomain-containing protein 4 (BRD4) or cyclin-dependent kinase 9 (CDK9) with decreased systemic toxicity. In addition, we demonstrated that the PROTAC prodrug NPs can serve as a versatile platform for the codelivery of a PROTAC and chemotherapeutics for enhanced anticancer efficiency and combination benefits. This study paves the way for utilizing tumor-targeted protein degradation for precise anticancer therapy and the effective combination treatment of complex diseases.


Assuntos
Nanopartículas , Pró-Fármacos , Proteólise , Proteína Supressora de Tumor Von Hippel-Lindau , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Humanos , Animais , Proteólise/efeitos dos fármacos , Nanopartículas/química , Linhagem Celular Tumoral , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Feminino , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Proteínas de Ciclo Celular/metabolismo , Camundongos , Quinase 9 Dependente de Ciclina/antagonistas & inibidores , Quinase 9 Dependente de Ciclina/metabolismo , Fatores de Transcrição/metabolismo , Camundongos Nus , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Camundongos Endogâmicos BALB C , Sistemas de Liberação de Medicamentos , Proteínas que Contêm Bromodomínio , Ubiquitina-Proteína Ligases
15.
J Nanobiotechnology ; 22(1): 2, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38169390

RESUMO

BACKGROUND: Off-targeted distribution of chemotherapeutic drugs causes severe side effects, further leading to poor prognosis and patient compliance. Ligand/receptor-mediated targeted drug delivery can improve drug accumulation in the tumor but it always attenuated by protein corona barriers. RESULTS: To address these problems, a radically different strategy is proposed that can leave the off-targeted drugs inactive but activate the tumor-distributed drugs for cancer-targeting therapy in a tumor microenvironment-independent manner. The feasibility and effectiveness of this strategy is demonstrated by developing an ultrasound (US)-activated prodrug-loaded liposome (CPBSN38L) comprising the sonosensitizer chlorin e6 (Ce6)-modified lipids and the prodrug of pinacol boronic ester-conjugated SN38 (PBSN38). Once CPBSN38L is accumulated in the tumor and internalized into the cancer cells, under US irradiation, the sonosensitizer Ce6 rapidly induces extensive production of intracellular reactive oxygen species (ROS), thereby initiating a cascade amplified ROS-responsive activation of PBSN38 to release the active SN38 for inducing cell apoptosis. If some of the injected CPBSN38L is distributed into normal tissues, the inactive PBSN38 exerts no pharmacological activity on normal cells. CPBSN38L exhibited strong anticancer activity in multiple murine tumor models of colon adenocarcinoma and hepatocellular carcinoma with no chemotherapy-induced side effects, compared with the standard first-line anticancer drugs irinotecan and topotecan. CONCLUSIONS: This study established a side-effect-evitable, universal, and feasible strategy for cancer-targeting therapy.


Assuntos
Adenocarcinoma , Antineoplásicos , Neoplasias do Colo , Nanopartículas , Fotoquimioterapia , Pró-Fármacos , Humanos , Animais , Camundongos , Lipossomos , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Adenocarcinoma/tratamento farmacológico , Neoplasias do Colo/diagnóstico por imagem , Neoplasias do Colo/tratamento farmacológico , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Nanopartículas/metabolismo , Fármacos Fotossensibilizantes/uso terapêutico , Microambiente Tumoral
16.
J Nanobiotechnology ; 22(1): 622, 2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-39402673

RESUMO

Altering the mechanisms of tumor cell death and overcoming the limitations of traditional chemotherapy is pivotal to contemporary tumor treatment. Inducing ferroptosis, while circumventing safety concerns associated with ferrous vectors, through nonferrous ferroptosis is a promising but underexplored frontier in cancer therapy. Histidine phosphatase (LHPP) has emerged as a novel therapeutic target in treating hepatocellular carcinoma (HCC), but the precise mechanism of LHPP against HCC remains unclear. Herein, we explore the effects of upregulating LHPP expression on ferroptosis and tumor immunogenicity induction by simply delivering a miRNA-363-5p inhibitor (miR-363-5pi) via a previously optimized gemcitabine-oleic acid (GOA) prodrug. Efficient miRNA encapsulation was achieved through hydrogen bonding at an optimized GOA/miRNA molar feed ratio of 250:1, affording spherical nanoparticles with a uniform hydrodynamic size of 147.1 nm and a negative potential of -21.5 mV. The mechanism of this LHPP-ferroptosis crosstalk is disclosed to be an inhibited phosphorylation of the PI3K/Akt pathway, leading to a remarkable tumor inhibition rate of 88.2% in nude mice bearing Bel-7402 tumor xenografts via a combination of LHPP-triggered nonferrous ferroptosis and GOA-induced chemotherapy. The biocompatibility of GOA/miR-363-5pi is strongly supported by their non-hematologic toxicity and insignificant organ damage. In addition, the tumor immunogenic activation potential of GOA/miR-363-5pi was finally explored. Overall, this study is the first work that elucidates the precise mechanism of LHPP for treating HCC via ferroptosis induction and achieves the transformation of chemotherapy and gene therapy into ferroptosis activation with tumor cell immunogenicity, which lays a new therapeutic foundation for the clinical treatment of HCC.


Assuntos
Carcinoma Hepatocelular , Ferroptose , Neoplasias Hepáticas , Camundongos Nus , Animais , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Ferroptose/efeitos dos fármacos , Humanos , Camundongos , Linhagem Celular Tumoral , MicroRNAs/metabolismo , MicroRNAs/genética , Camundongos Endogâmicos BALB C , Nanopartículas/química , Pró-Fármacos/farmacologia , Pró-Fármacos/química , Pró-Fármacos/uso terapêutico , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Desoxicitidina/química , Gencitabina , Monoéster Fosfórico Hidrolases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Transdução de Sinais/efeitos dos fármacos , Pirofosfatase Inorgânica
17.
Neoplasma ; 71(2): 117-122, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38766855

RESUMO

The incidence of distant metastases is associated with most cancer-related mortalities. Extracellular vesicles (EVs), secreted from tumors and cancer-associated fibroblasts, are involved in the metastatic process mediating their organotropism through their involvement in the pre-metastatic niche formation. We have been developing suicide gene therapy mediated by EVs secreted from mesenchymal stem/ stromal cells, tumor cells, and cancer-associated fibroblasts. Suicide gene EVs conjugated with prodrug are tumor tropic, penetrate tumor cells, and kill them by intracellular conversion of nontoxic prodrug to an efficient anti-cancer drug. Here, we discuss findings regarding the possibility of using suicide gene EVs as a novel therapeutic approach for metastases, via pre-metastatic niche modification. The suicide gene EVs provide a future perspective for metastasis prevention.


Assuntos
Vesículas Extracelulares , Genes Transgênicos Suicidas , Metástase Neoplásica , Humanos , Terapia Genética , Neoplasias/patologia , Neoplasias/genética , Neoplasias/prevenção & controle , Pró-Fármacos/uso terapêutico , Animais , Células-Tronco Mesenquimais
18.
Nucleic Acids Res ; 50(D1): D1398-D1407, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-34718717

RESUMO

Drug discovery relies on the knowledge of not only drugs and targets, but also the comparative agents and targets. These include poor binders and non-binders for developing discovery tools, prodrugs for improved therapeutics, co-targets of therapeutic targets for multi-target strategies and off-target investigations, and the collective structure-activity and drug-likeness landscapes of enhanced drug feature. However, such valuable data are inadequately covered by the available databases. In this study, a major update of the Therapeutic Target Database, previously featured in NAR, was therefore introduced. This update includes (a) 34 861 poor binders and 12 683 non-binders of 1308 targets; (b) 534 prodrug-drug pairs for 121 targets; (c) 1127 co-targets of 672 targets regulated by 642 approved and 624 clinical trial drugs; (d) the collective structure-activity landscapes of 427 262 active agents of 1565 targets; (e) the profiles of drug-like properties of 33 598 agents of 1102 targets. Moreover, a variety of additional data and function are provided, which include the cross-links to the target structure in PDB and AlphaFold, 159 and 1658 newly emerged targets and drugs, and the advanced search function for multi-entry target sequences or drug structures. The database is accessible without login requirement at: https://idrblab.org/ttd/.


Assuntos
Bases de Dados Factuais , Descoberta de Drogas/tendências , Pró-Fármacos/classificação , Humanos , Terapia de Alvo Molecular , Pró-Fármacos/química , Pró-Fármacos/uso terapêutico , Relação Estrutura-Atividade
19.
Nanomedicine ; 57: 102735, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38295913

RESUMO

In this study, we aimed to assess the analgesic efficacy of a thermoresponsive polymeric dexamethasone (Dex) prodrug (ProGel-Dex) in a mouse model of osteoarthritis (OA). At 12 weeks post model establishment, the OA mice received a single intra-articular (IA) injection of ProGel-Dex, dose-equivalent Dex, or Saline. Comparing to Saline and Dex controls, ProGel-Dex provided complete and sustained pain relief for >15 weeks according to incapacitance tests. In vivo optical imaging confirmed the continuous presence of ProGel-Dex in joints for 15 weeks post-injection. According to micro-CT analysis, ProGel-Dex treated mice had significantly lower subchondral bone thickness and medial meniscus bone volume than Dex and Saline controls. Except for a transient delay of body weight increase and slightly lower endpoint liver and spleen weights, no other adverse effect was observed after ProGel-Dex treatment. These findings support ProGel-Dex's potential as a potent and safe analgesic candidate for management of OA pain.


Assuntos
Osteoartrite , Pró-Fármacos , Camundongos , Animais , Dexametasona/farmacologia , Dexametasona/uso terapêutico , Modelos Animais de Doenças , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Osteoartrite/tratamento farmacológico , Artralgia/induzido quimicamente , Artralgia/tratamento farmacológico , Analgésicos/farmacologia , Analgésicos/uso terapêutico
20.
Chem Soc Rev ; 52(3): 879-920, 2023 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-36637396

RESUMO

Cancer remains as one of the most significant health problems, with approximately 19 million people diagnosed worldwide each year. Chemotherapy is a routinely used method to treat cancer patients. However, current treatment options lack the appropriate selectivity for cancer cells, are prone to resistance mechanisms, and are plagued with dose-limiting toxicities. As such, researchers have devoted their attention to developing prodrug-based strategies that have the potential to overcome these limitations. This tutorial review highlights recently developed prodrug strategies for cancer therapy. Prodrug examples that provide an integrated diagnostic (fluorescent, photoacoustic, and magnetic resonance imaging) response, which are referred to as theranostics, are also discussed. Owing to the non-invasive nature of light (and X-rays), we have discussed external excitation prodrug strategies as well as examples of activatable photosensitizers that enhance the precision of photodynamic therapy/photothermal therapy. Activatable photosensitizers/photothermal agents can be seen as analogous to prodrugs, with their phototherapeutic properties at a specific wavelength activated in the presence of disease-related biomarkers. We discuss each design strategy and illustrate the importance of targeting biomarkers specific to the tumour microenvironment and biomarkers that are known to be overexpressed within cancer cells. Moreover, we discuss the advantages of each approach and highlight their inherent limitations. We hope in doing so, the reader will appreciate the current challenges and available opportunities in the field and inspire subsequent generations to pursue this crucial area of cancer research.


Assuntos
Neoplasias , Fotoquimioterapia , Pró-Fármacos , Humanos , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Fotoquimioterapia/métodos , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa