Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Angew Chem Int Ed Engl ; 63(16): e202319981, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38381713

RESUMEN

Chemoradiotherapy combines radiotherapy with concurrent chemotherapy to potentiate antitumor activity but exacerbates toxicities and causes debilitating side effects in cancer patients. Herein, we report the use of a nanoscale metal-organic layer (MOL) as a 2D nanoradiosensitizer and a reservoir for the slow release of chemotherapeutics to amplify the antitumor effects of radiotherapy. Coordination of phosphate-containing drugs to MOL secondary building units prolongs their intratumoral retention, allowing for continuous release of gemcitabine monophosphate (GMP) for effective localized chemotherapy. In the meantime, the MOL sensitizes cancer cells to X-ray irradiation and provides potent radiotherapeutic effects. GMP-loaded MOL (GMP/MOL) enhances cytotoxicity by 2-fold and improves radiotherapeutic effects over free GMP in vitro. In a colon cancer model, GMP/MOL retains GMP in tumors for more than four days and, when combined with low-dose radiotherapy, inhibits tumor growth by 98 %. The synergistic chemoradiotherapy enabled by GMP/MOL shows a cure rate of 50 %, improves survival, and ameliorates cancer-proliferation histological biomarkers.


Asunto(s)
Neoplasias , Fosfatos , Humanos , Gemcitabina , Quimioradioterapia , Neoplasias/tratamiento farmacológico
2.
Biomaterials ; 302: 122334, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37776767

RESUMEN

While activating antitumor immunity with toll-like receptor (TLR) agonists provides a promising approach toward cancer immunotherapy, existing TLR agonists, including resiquimod (R848), have shown poor tumor selectivity and ineffective TLR activation in tumors for optimal antitumor effects. We hypothesized that improved delivery of TLR agonists to tumors and their effective combination with tumor antigens could significantly enhance their antitumor efficacy. Here, we report a novel nanoscale coordination polymer, Ce6/R848, for the co-delivery of Ce6 photosensitizer to elicit immunogenic cell death via photodynamic therapy (PDT) and cholesterol-conjugated R848 (Chol-R848) for tumor-selective TLR7/8 activation. Upon light irradiation, Ce6-mediated PDT released tumor antigens while selectively delivered R848 activated TLR7/8 in the tumors to synergistically activate antigen-presenting cells and prime T cells for enhanced innate and adaptive antitumor immune responses. Ce6/R848 achieved a 50% cure rate and 99.4% inhibition of tumor growth in subcutaneous MC38 colorectal tumors with minimal systemic toxicity.


Asunto(s)
Neoplasias Colorrectales , Fotoquimioterapia , Humanos , Presentación de Antígeno , Polímeros , Receptor Toll-Like 7/agonistas , Adyuvantes Inmunológicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Antígenos de Neoplasias , Inmunoterapia
3.
J Am Chem Soc ; 145(34): 18698-18704, 2023 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-37581644

RESUMEN

As heavy-metal-based nanoscale metal-organic frameworks (nMOFs) are excellent radiosensitizers for radiotherapy via enhanced energy deposition and reactive oxygen species (ROS) generation, we hypothesize that nMOFs with covalently conjugated and X-ray triggerable prodrugs can harness the ROS for on-demand release of chemotherapeutics for chemoradiotherapy. Herein, we report the design of a novel nMOF, Hf-TP-SN, with an X-ray-triggerable 7-ethyl-10-hydroxycamptothecin (SN38) prodrug for synergistic radiotherapy and chemotherapy. Upon X-ray irradiation, electron-dense Hf12 secondary building units serve as radiosensitizers to enhance hydroxyl radical generation for the triggered release of SN38 via hydroxylation of the 3,5-dimethoxylbenzyl carbonate followed by 1,4-elimination, leading to 5-fold higher release of SN38 from Hf-TP-SN than its molecular counterpart. As a result, Hf-TP-SN plus radiation induces significant cytotoxicity to cancer cells and efficiently inhibits tumor growth in colon and breast cancer mouse models.


Asunto(s)
Estructuras Metalorgánicas , Neoplasias , Profármacos , Fármacos Sensibilizantes a Radiaciones , Animales , Ratones , Estructuras Metalorgánicas/uso terapéutico , Profármacos/farmacología , Profármacos/uso terapéutico , Rayos X , Especies Reactivas de Oxígeno , Neoplasias/tratamiento farmacológico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Línea Celular Tumoral
4.
Small ; 19(52): e2305440, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37635106

RESUMEN

Cancer cells alter mechanical tension in their cell membranes. New interventions to regulate cell membrane tension present a potential strategy for cancer therapy. Herein, the increase of cell membrane tension by cholesterol oxidase (COD) via cholesterol depletion in vitro and the design of a COD-functionalized nanoscale metal-organic framework, Hf-TBP/COD, for cholesterol depletion and mechanoregulation of tumors in vivo, are reported. COD is found to deplete cholesterol and disrupt the mechanical properties of lipid bilayers, leading to decreased cell proliferation, migration, and tolerance to oxidative stress. Hf-TBP/COD increases mechanical tension of plasma membranes and osmotic fragility of cancer cells, which induces influx of calcium ions, inhibits cell migration, increases rupturing propensity for effective caspase-1 mediated pyroptosis, and decreases tolerance to oxidative stress. In the tumor microenvironment, Hf-TBP/COD downregulates multiple immunosuppressive checkpoints to reinvigorate T cells and enhance T cell infiltration. Compared to Hf-TBP, Hf-TBP/COD improves anti-tumor immune response and tumor growth inhibition from 54.3% and 79.8% to 91.7% and 95% in a subcutaneous triple-negative breast cancer model and a colon cancer model, respectively.


Asunto(s)
Estructuras Metalorgánicas , Neoplasias , Humanos , Estructuras Metalorgánicas/farmacología , Colesterol Oxidasa , Piroptosis , Linfocitos T , Colesterol , Microambiente Tumoral
5.
Biomaterials ; 301: 122235, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37441902

RESUMEN

The combination chemotherapy regimen FOLFIRINOX comprising folinic acid, 5-fluorouracil, irinotecan, and oxaliplatin is the first-line treatment for patients with advanced pancreatic cancer, but its use remains prohibitive for the majority of patients due to severe side effects. Here, we report a core-shell nanoscale coordination polymer (NCP) nanoparticle co-delivering a potent and synergistic combination of oxaliplatin, gemcitabine, and SN38 (OGS), for the treatment of pancreatic cancer in mouse models. OGS contains key synergistic components of FOLFIRINOX in a controllable drug ratio., It exhibited particle stability in blood circulation and enhanced deposition of the drugs in acidic tumor environments. In vitro, OGS showed superior cytotoxicity over free drug combinations and robust cytotoxic synergism among its three components. In vivo, OGS improved drug circulation, increased tumor deposition, and exhibited superior antitumor efficacy over the free drug combination in both subcutaneous and orthotopic pancreatic tumor models. OGS treatment achieved 75-91% tumor growth inhibition and prolonged mouse survival by 1.6- to 2.8-folds while minimizing systemic toxicities such as neutropenia, hepatotoxicity, and renal toxicity. This work uncovers a novel and clinically relevant nanomedicine strategy to co-deliver synergistic combination chemotherapies for difficult-to-treat cancers.


Asunto(s)
Neoplasias Pancreáticas , Animales , Ratones , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Oxaliplatino/uso terapéutico , Polímeros/uso terapéutico , Desoxicitidina/uso terapéutico , Compuestos Organoplatinos/uso terapéutico , Fluorouracilo/uso terapéutico , Gemcitabina , Neoplasias Pancreáticas
6.
Chem Sci ; 14(19): 5106-5115, 2023 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-37206384

RESUMEN

We report the self-assembly of amphiphilic BDQ photosensitizers into lysosome-targeting nanophotosensitizer BDQ-NP for highly effective photodynamic therapy (PDT). Molecular dynamics simulation, live cell imaging, and subcellular colocalization studies showed that BDQ strongly incorporated into lysosome lipid bilayers to cause continuous lysosomal membrane permeabilization. Upon light irradiation, the BDQ-NP generated a high level of reactive oxygen species to disrupt lysosomal and mitochondrial functions, leading to exceptionally high cytotoxicity. The intravenously injected BDQ-NP accumulated in tumours to achieve excellent PDT efficacy on subcutaneous colorectal and orthotopic breast tumor models without causing systemic toxicity. BDQ-NP-mediated PDT also prevented metastasis of breast tumors to the lungs. This work shows that self-assembled nanoparticles from amphiphilic and organelle-specific photosensitizers provide an excellent strategy to enhance PDT.

7.
Angew Chem Int Ed Engl ; 62(22): e202301910, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-36997341

RESUMEN

The efficacy of photodynamic therapy (PDT) depends on the subcellular localization of photosensitizers. Herein, we report a dual-organelle-targeted nanoparticle platform for enhanced PDT of cancer. By grafting 5-aminolevulinic acid (ALA) to a Hf12 -based nanoscale metal-organic layer (Hf-MOL) via carboxylate coordination, ALA/Hf-MOL enhanced ALA delivery and protoporphyrin IX (PpIX) synthesis in mitochondria, and trapped the Hf-MOL comprising 5,15-di-p-benzoatoporphyrin (DBP) photosensitizers in lysosomes. Light irradiation at 630 nm simultaneously excited PpIX and DBP to generate singlet oxygen and rapidly damage both mitochondria and lysosomes, leading to synergistic enhancement of the PDT efficacy. The dual-organelle-targeted ALA/Hf-MOL outperformed Hf-MOL in preclinical PDT studies, with a 2.7-fold lower half maximal inhibitory concentration in cytotoxicity assays in vitro and a 3-fold higher cure rate in a colon cancer model in vivo.


Asunto(s)
Fotoquimioterapia , Porfirinas , Ácido Aminolevulínico/farmacología , Ácido Aminolevulínico/uso terapéutico , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Mitocondrias , Metales , Línea Celular Tumoral
8.
Biomaterials ; 295: 122037, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36773429

RESUMEN

Conventional chemotherapy targets proliferative cancer cells to halt tumor progression or regress tumors. However, the plasticity of tumor cells enables their phenotypical changes to acquire chemo-resistance, leading to treatment failure or tumor recurrence after a successful treatment course. Here, we report the use of high-dose pharmacologic ascorbate to potentiate treatment efficacy of nanoscale coordination polymers (NCPs) delivering two clinical combinations of chemotherapeutics, carboplatin/docetaxel and oxaliplatin/SN38, and to target metabolic plasticity of tumor cells. Combination treatments of high-dose ascorbate and NCPs overcome multi-drug resistance by significantly reducing the abundance of cancer stem cells (CSCs) in solid tumors, as evidenced by reduced expression of tumor pluripotency factors. The clearance of CSCs inhibits post-surgery recurrence and systemic metastasis in multiple mouse models of cancer.


Asunto(s)
Antineoplásicos , Neoplasias , Animales , Ratones , Nanomedicina , Antineoplásicos/farmacología , Docetaxel/uso terapéutico , Oxaliplatino/uso terapéutico , Neoplasias/tratamiento farmacológico , Células Madre Neoplásicas/patología , Línea Celular Tumoral
9.
Adv Mater ; 35(19): e2212069, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36840977

RESUMEN

Although sonodynamic therapy (SDT) has shown promise for cancer treatment, the lack of efficient sonosensitizers (SSs) has limited the clinical application of SDT. Here, a new strategy is reported for designing efficient nano-sonosensitizers based on 2D nanoscale metal-organic layers (MOLs). Composed of Hf-oxo secondary building units (SBUs) and iridium-based linkers, the MOL is anchored with 5,10,15,20-tetra(p-benzoato)porphyrin (TBP) sensitizers on the SBUs to afford TBP@MOL. TBP@MOL shows 14.1- and 7.4-fold higher singlet oxygen (1 O2 ) generation than free TBP ligands and Hf-TBP, a 3D nanoscale metal-organic framework, respectively. The 1 O2 generation of TBP@MOL is enhanced by isolating TBP SSs on the SBUs of the MOL, which prevents aggregation-induced quenching of the excited sensitizers, and by triplet-triplet Dexter energy transfer between excited iridium-based linkers and TBP SSs, which more efficiently harnesses broad-spectrum sonoluminescence. Anchoring TBP on the MOL surface also enhances the energy transfer between the excited sensitizer and ground-state triplet oxygen to increase 1 O2 generation efficacy. In mouse models of colorectal and breast cancer, TBP@MOL demonstrates significantly higher SDT efficacy than Hf-TBP and TBP. This work uncovers a new strategy to design effective nano-sonosensitizers by facilitating energy transfer to efficiently capture broad-spectrum sonoluminescence and enhance 1 O2 generation.


Asunto(s)
Neoplasias , Porfirinas , Terapia por Ultrasonido , Ratones , Animales , Iridio , Porfirinas/farmacología , Oxígeno Singlete , Oxígeno , Neoplasias/tratamiento farmacológico
10.
ACS Nano ; 16(12): 21417-21430, 2022 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-36382721

RESUMEN

Long-circulating nanomedicines efficiently deliver chemotherapies to tumors to reduce general toxicity. However, extended blood circulation of nanomedicines can increase drug exposure to leukocytes and lead to hematological toxicity. Here, we report a two-stage release strategy to enhance the drug deposition and antitumor efficacy of OxPt/SN38 core-shell nanoparticles with a hydrophilic oxaliplatin (OxPt) prodrug coordination polymer core and a lipid shell containing a hydrophobic cholesterol-conjugated SN38 prodrug (Chol-SN38). By conjugating cholesterol to the phenol group of SN38 via an acetal linkage and protecting the 20-hydroxy position with a trimethylsilyl (TMS) group, Chol-SN38 releases SN38 in two stages via esterase-catalyzed cleavage of the acetal linkage in the liver followed by acid-mediated hydrolysis of the TMS group to preferentially release SN38 in tumors. Compared to irinotecan, OxPt/SN38 reduces SN38 blood exposure by 9.0 times and increases SN38 tumor exposure by 4.7 times. As a result, OxPt/SN38 inhibits tumor growth on subcutaneous, spontaneous, and metastatic tumor models by causing apoptotic and immunogenic cell death. OxPt/SN38 exhibits strong synergy with the immune checkpoint blockade to regress subcutaneous colorectal and pancreatic tumors with 33-50% cure rates and greatly inhibits tumor growth and invasion in a spontaneous prostate cancer model and a liver metastasis model of colorectal cancer without causing side effects. Mechanistic studies revealed important roles of enhanced immunogenic cell death and upregulated PD-L1 expression by OxPt/SN38 in activating the tumor immune microenvironment to elicit potent antitumor immunity. This work highlights the potential of combining innovative prodrug design and nanomedicine formulation to address unmet needs in cancer therapy.


Asunto(s)
Nanopartículas , Neoplasias , Profármacos , Masculino , Humanos , Profármacos/farmacología , Profármacos/uso terapéutico , Profármacos/química , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Acetales/uso terapéutico , Neoplasias/tratamiento farmacológico , Nanopartículas/química , Oxaliplatino , Línea Celular Tumoral , Microambiente Tumoral
12.
Biomaterials ; 290: 121831, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36240687

RESUMEN

Immunotherapies including immune checkpoint blockade (ICB) have become integral to treatments for immunogenic tumors by reinvigorating host immune functions to attack tumor cells. However, the clinical applications of ICB are limited by relatively low response rates as well as inherent and acquired resistance in many types of cancer. A potential solution is to selectively deliver immune modulators to tumors to activate the tumor microenvironment (TME) and enhance the therapeutic effect of ICB. Here we report the design of polyinosinic: polycytidylic acid (pIC) nanoscale coordination polymer (NCP), pIC@NCP, and its ability to activate tumor-specific immune responses and synergize with ICB for potent antitumor effects. With prolonged blood circulation, facile cell uptake, and triggered release of pIC in acidic TMEs, pIC@NCP shows robust tumor growth inhibition via activation of the endosomal toll-like receptor 3 signaling pathway. The synergistic combination of pIC@NCP and ICB further controls tumor growth in syngeneic mouse models of colorectal cancer and a spontaneous mouse model of prostate cancer. This study highlights the potential of NCP delivery of nucleic acid therapeutics for immune activation and cancer therapy.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Neoplasias , Ratones , Masculino , Animales , Receptor Toll-Like 3/uso terapéutico , Polímeros/farmacología , Inmunoterapia , Neoplasias/tratamiento farmacológico , Microambiente Tumoral , Factores Inmunológicos/farmacología
13.
Angew Chem Int Ed Engl ; 61(46): e202208685, 2022 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-36149753

RESUMEN

High-Z metal-based nanoscale metal-organic frameworks (nMOFs) with photosensitizing ligands can enhance radiation damage to tumors via a unique radiotherapy-radiodynamic therapy (RT-RDT) process. Here we report Monte Carlo (MC) simulation-guided design of a Th-based nMOF built from Th6 -oxo secondary building units and 5,15-di(p-benzoato)porphyrin (DBP) ligands, Th-DBP, for enhanced RT-RDT. MC simulations revealed that the Th-lattice outperformed the Hf-lattice in radiation dose enhancement owing to its higher mass attenuation coefficient. Upon X-ray or γ-ray radiation, Th-DBP enhanced energy deposition, generated more reactive oxygen species, and induced significantly higher cytotoxicity to cancer cells over the previously reported Hf-DBP nMOF. With low-dose X-ray irradiation, Th-DBP suppressed tumor growth by 88 % in a colon cancer and 97 % in a pancreatic cancer mouse model.


Asunto(s)
Neoplasias del Colon , Estructuras Metalorgánicas , Nanoestructuras , Ratones , Animales , Estructuras Metalorgánicas/uso terapéutico , Estructuras Metalorgánicas/efectos de la radiación , Torio , Método de Montecarlo , Ligandos , Nanoestructuras/uso terapéutico , Neoplasias del Colon/tratamiento farmacológico
15.
Adv Mater ; 34(39): e2110588, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35952624

RESUMEN

Despite potent preclinical antitumor activity, activation of stimulator of interferon genes (STING) has shown modest therapeutic effects in clinical studies. Many STING agonists, including 2',3'-cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), show poor pharmacokinetic properties for sustaining STING activation in tumors and achieving optimal antitumor efficacy. Improved delivery of STING agonists and their effective combination with other treatments are needed to enhance their therapeutic effects. Herein, a 2D nanoplatform, cGAMP/MOL, is reported via conjugating cGAMP to a nanoscale metal-organic layer (MOL) for simultaneous STING activation and radiosensitization. The MOL not only exhibits strong radiosensitization effects for enhanced cancer killing and induction of immunogenic cell death, but also retains cGAMP in tumors for sustained STING activation. Compared to free cGAMP, cGAMP/MOL elicits stronger STING activation and regresses local tumors upon X-ray irradiation. Further combination with an immune checkpoint inhibitor bridges innate and adaptive immune systems by activating the tumor microenvironment to elicit systemic antitumor responses.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Neoplasias , Humanos , Inmunoterapia , Interferones , Proteínas de la Membrana/metabolismo , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
17.
Adv Ther (Weinh) ; 5(1)2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35812344

RESUMEN

Radiotherapy (RT) has demonstrated synergy with immune checkpoint blockade (ICB) in preclinical models. However, its potential as an immunoadjuvant is limited by low immunogenicity at low radiation doses and immunosuppression at high radiation doses. It is hypothesized that radiosensitizers can enhance both the anticancer and immunogenic effects of low-dose radiation. Herein the authors report the antitumor immunity of combined RT and immunotherapy with dimethylaminomicheliolide (DMAMCL), a prodrug of the anti-inflammatory sesquiterpene lactone micheliolide (MCL). DMAMCL sensitized cancer cells to a single fraction of RT in vitro by inducing apoptosis and DNA double-strand breaks. DMAMCL with 5 fractions of 2 Gy focal X-ray irradiation led to significant anticancer efficacy in subcutaneous and spontaneous models of murine cancer. DMAMCL-sensitized RT upregulated programmed death-ligand 1 (PD-L1) expression in the tumors. Combination of DMAMCL-sensitized RT with anti-PD-L1 ICB significantly enhanced antitumor efficacy by increasing tumor-infiltrating CD4+ and CD8+ T cells and establishing immune memory.

18.
Adv Sci (Weinh) ; 9(24): e2201614, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35748191

RESUMEN

The binding of plasma proteins to nanomedicines is widely considered detrimental to their delivery to tumors. Here, the design of OxPt/SN38 nanoparticle containing a hydrophilic oxaliplatin (OxPt) prodrug in a coordination polymer core and a hydrophobic cholesterol-conjugated SN38 prodrug on the lipid shell for active tumor targeting is reported. OxPt/SN38 hitchhikes on low-density lipoprotein (LDL) particles, concentrates in tumors via LDL receptor-mediated endocytosis, and selectively releases SN38 and OxPt in acidic, esterase-rich, and reducing tumor microenvironments, leading to 6.0- and 4.9-times higher accumulations in tumors over free drugs. By simultaneously crosslinking DNA and inhibiting topoisomerase I, OxPt/SN38 achieved 92-98% tumor growth inhibition in five colorectal cancer tumor models and prolonged mouse survival by 58-80 days compared to free drug controls in three human colorectal cancer tumor models without causing serious side effects. The study has uncovered a novel nanomedicine strategy to co-deliver combination chemotherapies to tumors via active targeting of the LDL receptor.


Asunto(s)
Neoplasias Colorrectales , Nanopartículas , Profármacos , Receptores de LDL , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Humanos , Lipoproteínas LDL/metabolismo , Ratones , Oxaliplatino/administración & dosificación , Profármacos/farmacología , Receptores de LDL/metabolismo , Microambiente Tumoral
19.
J Am Chem Soc ; 144(12): 5241-5246, 2022 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-35297640

RESUMEN

Herein we report that dimensional reduction from three-dimensional nanoscale metal-organic frameworks (nMOFs) to two-dimensional nanoscale metal-organic layers (nMOLs) increases the frequency of encounters between photosensitizers and oxygen and facilitates the diffusion of singlet oxygen from the nMOL to significantly enhance photodynamic therapy. The nMOFs and nMOLs share the same M12-oxo (M = Zr, Hf) secondary building units and 5,15-di-p-benzoatoporphyrin (DBP) ligands but exhibit three-dimensional and two-dimensional topologies, respectively. Molecular dynamics simulations and experimental studies revealed that the nMOLs with a monolayer morphology enhanced the generation of reactive oxygen species and exhibited over an order of magnitude higher cytotoxicity over the nMOFs. In a mouse model of triple-negative breast cancer, Hf-DBP nMOL showed 49.1% more tumor inhibition, an 80% higher cure rate, and 16.3-fold lower metastasis potential than Hf-DBP nMOF.


Asunto(s)
Estructuras Metalorgánicas , Nanoestructuras , Neoplasias , Fotoquimioterapia , Animales , Estructuras Metalorgánicas/farmacología , Estructuras Metalorgánicas/uso terapéutico , Ratones , Neoplasias/patología , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Oxígeno Singlete/uso terapéutico
20.
Nat Biomed Eng ; 6(2): 144-156, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35190678

RESUMEN

Checkpoint blockade elicits durable responses in immunogenic cancers, but it is largely ineffective in immunologically 'cold' tumours. Here we report the design, synthesis and performance of a bismuth-based nanoscale metal-organic framework that modulates the immunological and mechanical properties of the tumour microenvironment for enhanced radiotherapy-radiodynamic therapy. In mice with non-immunogenic prostate and pancreatic tumours irradiated with low X-ray doses, the intratumoural injection of the radiosensitizer mediated potent outcomes via the repolarization of immunosuppressive M2 macrophages into immunostimulatory M1 macrophages, the reduction of the concentration of intratumoural transforming growth factor beta (TGF-ß) and of collagen density, and the inactivation of cancer-associated fibroblasts. When intravenously injected in combination with checkpoint-blockade therapy, the radiosensitizer mediated the reversal of immunosuppression in primary and distant tumours via the systemic reduction of TGF-ß levels, which led to the downregulation of collagen expression, the stimulation of T-cell infiltration in the tumours and a robust abscopal effect. Nanoscale radiosensitizers that stimulate anti-tumour immunity and T-cell infiltration may enhance the therapeutic outcomes of checkpoint blockade in other tumour types.


Asunto(s)
Estructuras Metalorgánicas , Neoplasias , Animales , Inmunidad , Inmunoterapia , Masculino , Estructuras Metalorgánicas/farmacología , Ratones , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...