Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters










Publication year range
1.
ACS Nano ; 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38952208

ABSTRACT

Immunotherapy can potentially suppress the highly aggressive glioblastoma (GBM) by promoting T lymphocyte infiltration. Nevertheless, the immune privilege phenomenon, coupled with the generally low immunogenicity of vaccines, frequently hampers the presence of lymphocytes within brain tumors, particularly in brain tumors. In this study, the membrane-disrupted polymer-wrapped CuS nanoflakes that can penetrate delivery to deep brain tumors via releasing the cell-cell interactions, facilitating the near-infrared II (NIR II) photothermal therapy, and detaining dendritic cells for a self-cascading immunotherapy are developed. By convection-enhanced delivery, membrane-disrupted amphiphilic polymer micelles (poly(methoxypoly(ethylene glycol)-benzoic imine-octadecane, mPEG-b-C18) with CuS nanoflakes enhances tumor permeability and resides in deep brain tumors. Under low-power NIR II irradiation (0.8 W/cm2), the intense heat generated by well-distributed CuS nanoflakes actuates the thermolytic efficacy, facilitating cell apoptosis and the subsequent antigen release. Then, the positively charged polymer after hydrolysis of the benzoic-imine bond serves as an antigen depot, detaining autologous tumor-associated antigens and presenting them to dendritic cells, ensuring sustained immune stimulation. This self-cascading penetrative immunotherapy amplifies the immune response to postoperative brain tumors but also enhances survival outcomes through effective brain immunotherapy.

2.
Sci Rep ; 14(1): 8532, 2024 06 03.
Article in English | MEDLINE | ID: mdl-38830912

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) poses challenges due to late-stage diagnosis and limited treatment response, often attributed to the hypoxic tumor microenvironment (TME). Sonoporation, combining ultrasound and microbubbles, holds promise for enhancing therapy. However, additional preclinical research utilizing commercially available ultrasound equipment for PDAC treatment while delving into the TME's intricacies is necessary. This study investigated the potential of using a clinically available ultrasound system and phase 2-proven microbubbles to relieve tumor hypoxia and enhance the efficacy of chemotherapy and immunotherapy in a murine PDAC model. This approach enables early PDAC detection and blood-flow-sensitive Power-Doppler sonoporation in combination with chemotherapy. It significantly extended treated mice's median survival compared to chemotherapy alone. Mechanistically, this combination therapy enhanced tumor perfusion and substantially reduced tumor hypoxia (77% and 67%, 1- and 3-days post-treatment). Additionally, cluster of differentiation 8 (CD8) T-cell infiltration increased four-fold afterward. The combined treatment demonstrated a strengthening of the anti-programmed death-ligand 1(αPDL1) therapy against PDAC. Our study illustrates the feasibility of using a clinically available ultrasound system with NH-002 microbubbles for early tumor detection, alleviating hypoxic TME, and improving chemotherapy and immunotherapy. It suggests the development of an adjuvant theragnostic protocol incorporating Power-Doppler sonoporation for pancreatic tumor treatment.


Subject(s)
Carcinoma, Pancreatic Ductal , Immunotherapy , Microbubbles , Pancreatic Neoplasms , Tumor Microenvironment , Animals , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Mice , Immunotherapy/methods , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Tumor Microenvironment/drug effects , Cell Line, Tumor , Tumor Hypoxia/drug effects , Combined Modality Therapy , Humans , Female
3.
Elife ; 132024 May 22.
Article in English | MEDLINE | ID: mdl-38775133

ABSTRACT

Tissue-clearing and labeling techniques have revolutionized brain-wide imaging and analysis, yet their application to clinical formalin-fixed paraffin-embedded (FFPE) blocks remains challenging. We introduce HIF-Clear, a novel method for efficiently clearing and labeling centimeter-thick FFPE specimens using elevated temperature and concentrated detergents. HIF-Clear with multi-round immunolabeling reveals neuron circuitry regulating multiple neurotransmitter systems in a whole FFPE mouse brain and is able to be used as the evaluation of disease treatment efficiency. HIF-Clear also supports expansion microscopy and can be performed on a non-sectioned 15-year-old FFPE specimen, as well as a 3-month formalin-fixed mouse brain. Thus, HIF-Clear represents a feasible approach for researching archived FFPE specimens for future neuroscientific and 3D neuropathological analyses.


Subject(s)
Brain , Formaldehyde , Neurons , Paraffin Embedding , Tissue Fixation , Animals , Paraffin Embedding/methods , Mice , Tissue Fixation/methods , Neurons/physiology , Fixatives/chemistry
4.
Cancer Cell Int ; 24(1): 35, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38238749

ABSTRACT

BACKGROUND: It is well known that tumor-associated macrophages (TAMs) play essential roles in brain tumor resistance to chemotherapy. However, the detailed mechanisms of how TAMs are involved in brain tumor resistance are still unclear and lack a suitable analysis model. METHODS: A BV2 microglial cells with ALTS1C1 astrocytoma cells in vitro co-culture system was used to mimic the microglia dominating tumor stroma in the tumor invasion microenvironment and explore the interaction between microglia and brain tumor cells. RESULTS: Our result suggested that microglia could form colonies with glioma cells under high-density culturing conditions and protect glioma cells from apoptosis induced by chemotherapeutic drugs. Moreover, this study demonstrates that microglia could hijack drug substances from the glioma cells and reduce the drug intensity of ALTS1C1 via direct contact. Inhibition of gap junction protein prevented microglial-glioma colony formation and microglia-mediated chemoresistance. CONCLUSIONS: This study provides novel insights into how glioma cells acquire chemoresistance via microglia-mediated drug substance transferring, providing a new option for treating chemo-resistant brain tumors.

5.
Front Oncol ; 13: 1263873, 2023.
Article in English | MEDLINE | ID: mdl-37886177

ABSTRACT

Purpose: Malignant head and neck squamous cell carcinoma (HNSCC) is characterized by a poor prognosis and resistance to conventional radiotherapy. Infiltrating myeloid-derived suppressive cells (MDSCs) is prominent in HNSCC and is linked to immune suppression and tumor aggressiveness. This study aimed to investigate the impact of boron neutron capture therapy (BNCT) on the MDSCs in the tumor microenvironment and peripheral blood and to explore the potential for MDSCs depletion combined with BNCT to reactivate antitumor immunity. Methods and materials: Carcinogen, 4-NQO, -induced oral tumors were irradiated with a total physical dose of 2 Gy BNCT in Tsing Hua Open Reactor (THOR). Flow cytometry and immunohistochemistry accessed the dynamics of peripheral MDSCs and infiltrated MDSCs within the tumor microenvironment. Mice were injected with an inhibitor of CSF-1 receptor (CSF-1R), PLX3397, to determine whether modulating M-MDSCs could affect mice survival after BNCT. Results: Peripheral CD11b+Ly6ChighLy6G- monocytic-MDSCs (M-MDSCs), but not CD11b+Ly6CloLy6Ghigh polymorphonuclear-MDSCs (PMN-MDSCs), increased as tumor progression. After BNCT treatment, there were temporarily decreased and persistent increases of M-MDSCs thereafter, either in peripheral blood or in tumors. The administration of PLX-3397 hindered BNCT-caused M-MDSCs infiltration, prolonged mice survival, and activated tumor immunity by decreasing tumor-associated macrophages (TAMs) and increasing CD8+ T cells. Conclusion: M-MDSCs were recruited into 4-NQO-induced tumors after BNCT, and their number was also increased in peripheral blood. Assessment of M-MDSCs levels in peripheral blood could be an index to determine the optimal intervention window. Their temporal alteration suggests an association with tumor recurrence after BNCT, making M-MDSCs a potential intervention target. Our preliminary results showed that PLX-3397 had strong M-MDSCs, TAMs, and TIL (tumor-infiltrating lymphocyte) modulating effects that could synergize tumor control when combined with BNCT.

6.
JACS Au ; 3(8): 2192-2205, 2023 Aug 28.
Article in English | MEDLINE | ID: mdl-37654578

ABSTRACT

Eradication of head-and-neck (H&N) tumors is very difficult and challenging because of the characteristic feature of frequent recurrence and the difficulty in killing cancer stem cells. Neutron capture therapy (NCT) is emerging as a noninvasive potential modality for treatments of various types of tumors. Herein, we report that 98.5% 10B-enriched anti-EGFR-Gd10B6 nanoparticles can not only deliver large doses of 158 µg 10B/g tumor tissues as well as 56.8 µg 157Gd/g tumor tissues with a very high tumor-to-blood (T/B) 10B ratio of 4.18, but also exert very effective CT/MRI image-guided combined GdBNCT effects on killing cancer stem cells and eradication of recurrent head-and-neck (H&N) tumors. This leads to a long average half-lifespan of 81 days for H&N tumor-bearing mice, which is a record-making result, and surpasses the best result reported in the literature using combined radiotherapy and T cell-mediated immunotherapy (70 d).

7.
Nanomedicine (Lond) ; 18(9): 743-754, 2023 04.
Article in English | MEDLINE | ID: mdl-37306216

ABSTRACT

Background: Boron neutron capture therapy (BNCT) is a promising cancer treatment that eliminates tumor cells by triggering high-energy radiation within cancer cells. Aim: In vivo evaluation of poly(vinyl alcohol)/boric acid crosslinked nanoparticles (PVA/BA NPs) for BNCT. Materials & methods: PVA/BA NPs were synthesized and intravenously injected into tumor-bearing mice for BNCT. Results: The in vitro boron uptake of PVA/BA NPs in tumor cells was 70-fold higher than the required boron uptake for successful BNCT. In an in vivo study, PVA/BA NPs showed a 44.29% reduction in tumor size compared with clinically used boronophenylalanine for oral cancer in a murine model. Conclusion: PVA/BA NPs exhibited effective therapeutic results for oral cancer treatments in BNCT.


Subject(s)
Boron Neutron Capture Therapy , Mouth Neoplasms , Nanoparticles , Animals , Mice , Boron Neutron Capture Therapy/methods , Mouth Neoplasms/radiotherapy , Disease Models, Animal , Chemical Engineering , Male
8.
J Colloid Interface Sci ; 631(Pt A): 1-16, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36368211

ABSTRACT

Nanomaterial mediated phototherapies are believed to be promising candidates to overcome the bacterial drug resistance crisis. However, due to the lack of nanomaterials able to absorb long NIR light, especially in the NIR-III (1500-1850 nm) and -IV (2100-2300 nm) regimes, it was never investigated the utilization of NIR-III and NIR-IV light for in vivo treatments of cancer or bacterial infections. To this end, plasmonic metal-doped transition metal oxides (TMO) are attracting a great attention due to their tunable surface plasmon resonance absorption to the NIR region. Unique features with extendable NIR light absorption of plasmonic metal-doped transition metal oxides make their applications very attractive in several fields, but their utilization for bacterial infection treatments was not yet reported. Moreover, up-to-date bacterial eradication was limited to phototherapies in the NIR-I (700-950 nm) and NIR-II (1000-1350 nm) biological windows (BWs) and has not yet been studied in the NIR-III (1500-1870 nm) BW. To overcome these literature limitations, we engineered NIR-III (1550 nm) light activatable multifunctional plasmonic CuBi2O4 bimetallic particles (i.e., CBO bMPs) with very high molar extinction coefficients (2.75 × 1011 M-1cm-1 @ 808 nm, 2.75 × 1011 M-1cm-1 @ 980 nm, and 3.5 × 1011 M-1cm-1 @1550 nm), able to absorb and convert long NIR (980 and 1550 nm) light energy to thermal heat and generate cytotoxic reactive oxygen species (ROS) for in vivo treatment of drug resistant bacterial infections. Our in vitro and in vivo results reveal that NIR-III (1550 nm) light irradiation of CBO bMPs exerts a remarkable in vivo antibacterial activity via NIR-III photothermal therapy (NIR-III PTT), which is superior than its corresponding NIR-I (808 nm) PTT and NIR-II photodynamic therapy (NIR-II PDT, 980 nm). We observed that hyperthermia-based photothermal therapy is more effective than ROS-based photodynamic therapy in killing multi-drug resistant bacteria. We also show that CBO bMPs also show an enzyme oxidase and peroxidase like activity, which is an additional asset to enhance the therapeutic efficiency.


Subject(s)
Photochemotherapy , Photothermal Therapy , Reactive Oxygen Species , Phototherapy , Photochemotherapy/methods , Oxides , Bacteria , Drug Resistance
9.
J Control Release ; 352: 920-930, 2022 12.
Article in English | MEDLINE | ID: mdl-36334859

ABSTRACT

While immunotherapy has emerged as a promising strategy to treat glioblastoma multiforme (GBM), the limited availability of immunotherapeutic agents in tumors due to the presence of the blood-brain barrier (BBB) and immunosuppressive tumor microenvironment dampens efficacy. Nitric oxide (NO) plays a role in modulating both the BBB and tumor vessels and could thus be delivered to disrupt the BBB and improve the delivery of immunotherapeutics into GBM tumors. Herein, we report an immunotherapeutic approach that utilizes CXCR4-targeted lipid­calcium-phosphate nanoparticles with NO donors (LCP-NO NPs). The delivery of NO resulted in enhanced BBB permeability and thus improved gene delivery across the BBB. CXCR4-targeted LCP-NO NPs delivered siRNA against the immune checkpoint ligand PD-L1 to GBM tumors, silenced PD-L1 expression, increased cytotoxic T cell infiltration and activation in GBM tumors, and suppressed GBM progression. Thus, the codelivery of NO and PD-L1 siRNA by these CXCR4-targeted NPs may serve as a potential immunotherapy for GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , Nanoparticles , Humans , Glioblastoma/drug therapy , B7-H1 Antigen , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Nitric Oxide/therapeutic use , Brain Neoplasms/drug therapy , Immunotherapy , Tumor Microenvironment , Cell Line, Tumor , Receptors, CXCR4/genetics
10.
Biomaterials ; 290: 121861, 2022 11.
Article in English | MEDLINE | ID: mdl-36302304

ABSTRACT

Recurrent head-and-neck (H&N) cancer is one of the most malignant cancers in the world. Various treatment modalities, such as radiation therapy, chemotherapy, and surgery were adopted to treat H&N cancer, but recurrence of H&N tumor always occurs again, leading to poor prognosis and low 5-year survival rate. Recently, boron neutron capture therapy (BNCT) emerges an alternative modality for curing recurrent tumors. Presently, boron phenylalanine-fructose (BPA-F) and sodium borocaptate (BSH) are the two best BNCT molecular drugs, which, however, have poor therapeutic efficacies and are lack of tumor-targeting ability. In this study, 10B-riched (98.5% 10B) boron phosphate nanoparticles (10BPO4 NPs) of ∼100 nm in size were prepared in a single step using a unique microwave arcing method. The 10B-enriched 10BPO4 NPs were surface-modified with anti-EGFR antibody to endow the targeting ability toward H&N cancer cells. In in-vivo xenograft mice model, a large amount (∼63 µg 10B/g cancer cells) of 10B atoms could be effectively accumulated at the H&N tumor sites using 10BPO4 NPs as BNCT reagents. In in-vitro neutron irradiation experiments, 72% cell deaths were observed from anti-EGFR-10BPO4 NPs-treated H&N cancer cells, which is ∼2.4 folds higher than that (30%) treated with the most effective molecular drug, BPA-F. We demonstrated that upon neutron irradiation, the anti-EGFR-10BPO4 NPs could exert a much higher extent of destruction of H&N tumor, as well as effective suppression of the probability of H&N tumor recurrence, as compared to the most effective molecular drug, BPA-F. The median survival of the BNCT treated mice with anti-EGFR-10BPO4 NPs extends beyond 75 days, which is far better than the mice treated with BPA-F (33 days), blank + NR mice (25), and blank mice (23 days).


Subject(s)
Boron Neutron Capture Therapy , Head and Neck Neoplasms , Nanoparticles , Humans , Animals , Mice , Boron , Neoplasm Recurrence, Local/drug therapy , Boron Neutron Capture Therapy/methods , Boron Compounds/therapeutic use , Neutrons , Head and Neck Neoplasms/radiotherapy
11.
Nanoscale Horiz ; 7(6): 589-606, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35527504

ABSTRACT

Plasmonic field-field coupling-induced enhancement of the optical properties of dye molecules in the nanogaps among metal nanoparticle clusters and thin films has attracted significant attention especially in disease-related theranostic applications. However, it is very challenging to synthesize plasmonic core-gap-shell nanostructures with a well-controlled nanogap, uniform shape, and distances to maximize the plasmonic field-field coupling between the core and the shell. Herein, we synthesized Au@gap@AuAg nanopeanut-shaped core-gap-shell nanostructures (Au NPN) and tuned their optical absorption from near-infrared region-I (NIR-I) to near-infrared region-II (NIR-II) by filling their nanogap with a high dielectric NaCl(aq) aqueous solution, which led to a dramatic redshift in the plasmonic absorption band by 320 nm from 660 to 980 nm and a 12.6-fold increase (at 1064 nm) in the extinction coefficient in the NIR region (1000-1300 nm). Upon filling the nanogap with NaCl(aq) aqueous solution, the Au NPN6.5(NaCl) (i.e., ∼6.5 nm nanogap)-mediated NIR-II photodynamic therapy effect was dramatically enhanced, resulting in a much longer average lifespan of >55 days for the mice bearing a murine colon tumor and treated with Au NPN6.5(NaCl) plus 1064 nm light irradiation compared to the mice treated with Au NPN6.5 + 1064 nm light irradiation (without nanogap filled with dielectric NaCl(aq), 40 d) and the doxorubicin-treated group (23 d). This study demonstrates a simple but effective method to tune and maximize the plasmonic field-field coupling between the metal shell and metal core of core-gap-shell nanostructures, the plasmonic field-lattice interactions, and biomedical applications for the treatment of tumors. Overall, our work presents a new way to enhance/maximize the plasmonic field-field and field-lattice coupling, and thus the performance/sensitivities in nanogap-based bioimaging, sensing, and theranostic nanomaterials and devices.


Subject(s)
Metal Nanoparticles , Nanostructures , Photochemotherapy , Animals , Gold/chemistry , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice , Nanostructures/chemistry , Nanostructures/therapeutic use , Sodium Chloride
13.
ACS Nano ; 16(3): 4014-4027, 2022 03 22.
Article in English | MEDLINE | ID: mdl-35225594

ABSTRACT

T lymphocyte infiltration with immunotherapy potentially suppresses most devastating brain tumors. However, local immune privilege and tumor heterogeneity usually limit the penetration of immune cells and therapeutic agents into brain tumors, leading to tumor recurrence after treatment. Here, a rabies virus glycoprotein (RVG)-camouflaged gold yarnball (RVG@GY) that can boost the targeting efficiency at a brain tumor via dual hierarchy- and RVG-mediated spinal cord transportation, facilitating the decrease of tumor heterogeneity for T cell infiltration, is developed. Upon magnetoelectric irradiation, the electron current generated on the GYs activates the electrolytic penetration of palbociclib-loaded dendrimer (Den[Pb]) deep into tumors. In addition, the high-density GYs at brain tumors also induces the disruption of cell-cell interactions and T cell infiltration. The integration of the electrolytic effects and T cell infiltration promoted by drug-loaded RVG@GYs deep in the brain tumor elicits sufficient T cell numbers and effectively prolongs the survival rate of mice with orthotopic brain tumors.


Subject(s)
Brain Neoplasms , Rabies virus , Animals , Brain Neoplasms/drug therapy , Glycoproteins , Gold/therapeutic use , Mice , T-Lymphocytes/pathology
14.
Nanomaterials (Basel) ; 11(11)2021 Nov 02.
Article in English | MEDLINE | ID: mdl-34835699

ABSTRACT

Boron neutron capture therapy (BNCT) is a powerful and selective anti-cancer therapy utilizing 10B-enriched boron drugs. However, clinical advancement of BCNT is hampered by the insufficient loading of B-10 drugs throughout the solid tumor. Furthermore, the preparation of boron drugs for BNCT relies on the use of the costly B-10 enriched precursor. To overcome these challenges, polymer-coated boron carbon oxynitride (BCNO) nanoparticles, with ~30% of boron, were developed with enhanced biocompatibility, cell uptake, and tumoricidal effect via BNCT. Using the ALTS1C1 cancer cell line, the IC50 of the PEG@BCNO, bare, PEI@BCNO were determined to be 0.3 mg/mL, 0.1 mg/mL, and 0.05 mg/mL, respectively. As a proof-of-concept, the engineered non-10B enriched polymer-coated BCNO exhibited excellent anti-tumor effect via BNCT due to their high boron content per nanoparticle and due to the enhanced cellular internalization and retention compared to small molecular 10B-BPA drug. The astrocytoma ALTS1C1 cells treated with bare, polyethyleneimine-, and polyethylene glycol-coated BCNO exhibited an acute cell death of 24, 37, and 43%, respectively, upon 30 min of neutron irradiation compared to the negligible cell death in PBS-treated and non-irradiated cells. The radical approach proposed in this study addresses the expensive and complex issues of B-10 isotope enrichment process; thus, enabling the preparation of boron drugs at a significantly lower cost, which will facilitate the development of boron drugs for BNCT.

15.
Int J Mol Sci ; 22(18)2021 Sep 17.
Article in English | MEDLINE | ID: mdl-34576217

ABSTRACT

Radiation therapy (RT) recruits myeloid cells, leading to an immunosuppressive microenvironment that impedes its efficacy against tumors. Combination of immunotherapy with RT is a potential approach to reversing the immunosuppressive condition and enhancing tumor control after RT. This study aimed to assess the effects of local interleukin-12 (IL-12) therapy on improving the efficacy of RT in a murine prostate cancer model. Combined treatment effectively shrunk the radioresistant tumors by inducing a T helper-1 immune response and influx of CD8+ T cells. It also delayed the radiation-induced vascular damage accompanied by increased α-smooth muscle actin-positive pericyte coverage and blood perfusion. Moreover, RT significantly reduced the IL-12-induced levels of alanine aminotransferase in blood. However, it did not further improve the IL-12-induced anti-tumor effect on distant tumors. Upregulated expression of T-cell exhaustion-associated genes was found in tumors treated with IL-12 only and combined treatment, suggesting that T-cell exhaustion is potentially correlated with tumor relapse in combined treatment. In conclusion, this study illustrated that combination of radiation and local IL-12 therapy enhanced the host immune response and promoted vascular maturation and function. Furthermore, combination treatment was associated with less systemic toxicity than IL-12 alone, providing a potential option for tumor therapy in clinical settings.


Subject(s)
Immune System/radiation effects , Interleukin-12 Subunit p35/metabolism , Radiotherapy/methods , Actins/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Immunohistochemistry , Immunosuppressive Agents/pharmacology , Immunotherapy , Interferon-gamma/metabolism , Liver/metabolism , Liver/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth/metabolism , Neoplasm Transplantation , Pericytes/metabolism , Prostatic Neoplasms/metabolism , Tumor Microenvironment/immunology
16.
ACS Nano ; 15(9): 14404-14418, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34428028

ABSTRACT

Despite the development of various therapeutic modalities to tackle cancer, multidrug resistance (MDR) and incomplete destruction of deep tissue-buried tumors remain as long-standing challenges responsible for tumor recurrence and low survival rates. In addition to the MDR and deep tissue photoactivation problems, most primary tumors metastasize to the lungs and lymph nodes to form secondary tumors. Therefore, it leaves a great challenge to develop theranostic approaches to combat both MDR and deep tissue photoactivation problems. Herein, we develop a versatile plasmonic CuO/Cu2O truncated nanocube-based theranostic nanomedicine to act as a triple modal near-infrared fluorescence (NIRF) imaging agent in the biological window II (1000-1500 nm)/photoacoustic imaging (PAI)/T1-weighted magnetic resonance (MR) imaging agents, sensitize the formation of singlet oxygen (1O2) to exert nanomaterial-mediated photodynamic therapeutic (NIR-II NmPDT), and absorb long NIR light (i.e., 1550 nm) in the biological window III (1500-1700 nm) to exert nanomaterial-mediated photothermal therapeutic (NIR-III NmPTT) effects for the effective destruction of multi-drug-resistant lung tumors. We found that H69AR lung cancer cells do not create drug resistance toward plasmonic CuO/Cu2O TNCs-based nanomedicines.


Subject(s)
Carcinoma , Lung Neoplasms , Copper , Drug Resistance, Multiple , Humans , Lung , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/drug therapy
17.
Adv Mater ; 33(34): e2100701, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34270814

ABSTRACT

Most orally administered drugs fail to reach the intracerebral regions because of the intestinal epithelial barrier (IEB) and the blood-brain barrier (BBB), which are located between the gut and the brain. Herein, an oral prodrug delivery system that can overcome both the IEB and the BBB noninvasively is developed for treating gliomas. The prodrug is prepared by conjugating an anticancer drug on ß-glucans using a disulfide-containing linker. Following oral administration in glioma-bearing mice, the as-prepared prodrug can specifically target intestinal M cells, transpass the IEB, and be phagocytosed/hitchhiked by local macrophages (Mϕ). The Mϕ-hitchhiked prodrug is transported to the circulatory system via the lymphatic system, crossing the BBB. The tumor-overexpressed glutathione then cleaves the disulfide bond within the prodrug, releasing the active drug, improving its therapeutic efficacy. These findings reveal that the developed prodrug may serve as a gut-to-brain oral drug delivery platform for the well-targeted treatment of gliomas.


Subject(s)
Administration, Oral , Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Drug Delivery Systems , Glioma/drug therapy , Intestines/drug effects , Prodrugs/chemistry , Temozolomide/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Blood-Brain Barrier/drug effects , Disulfides , Endocytosis , Lymphatic System , Macrophages/metabolism , Magnetic Resonance Spectroscopy , Mice , Neoplasm Transplantation , Temozolomide/pharmacokinetics , beta-Glucans/chemistry
18.
Int J Mol Sci ; 22(4)2021 Feb 19.
Article in English | MEDLINE | ID: mdl-33669885

ABSTRACT

The low overall survival rate of patients with pancreatic cancer has driven research to seek a new therapeutic protocol. Radiotherapy (RT) is frequently an option in the neoadjuvant or palliative settings for pancreatic cancer treatment. This study explored the effect of RT protocols on the tumor microenvironment (TME) and their consequent impact on anti-programmed cell death ligand-1 (PD-L1) therapy. Using a murine orthotopic pancreatic tumor model, UN-KC-6141, RT-disturbed TME was examined by immunohistochemical staining. The results showed that ablative RT is more effective than fractionated RT at recruiting T cells. On the other hand, fractionated RT induces more myeloid-derived suppressor cell infiltration than ablative RT. The RT-disturbed TME presents a higher perfusion rate per vessel. The increase in vessel perfusion is associated with a higher amount of anti-PD-L1 antibody being delivered to the tumor. Animal survival is increased by anti-PD-L1 therapy after ablative RT, with 67% of treated animals surviving more than 30 days after tumor inoculation compared to a median survival time of 16.5 days for the control group. Splenocytes isolated from surviving animals were specifically cytotoxic for UN-KC-6141 cells. We conclude that the ablative RT-induced TME is more suited than conventional RT-induced TME to combination therapy with immune checkpoint blockade.


Subject(s)
Immune Checkpoint Inhibitors/therapeutic use , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/radiotherapy , Tumor Microenvironment , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Immune Checkpoint Inhibitors/pharmacology , Mice, Inbred C57BL , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects
19.
Nano Lett ; 21(3): 1375-1383, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33562964

ABSTRACT

T lymphocytes infiltrate the most devastating metastatic tumors for immunotherapy, allowing the potential for tumor metastasis suppression. However, tumor heterogeneity often restricts the infiltration of immune cells and possesses immune privilege that leads to protection from the immune attack, especially for invading metastatic clusters. Here, an exosome-camouflaged nanoraspberry (RB@Exo) doubling as a metastases-targeting agent and T cell-infiltration inducer that delivers an anticancer drug and energy is reported. The RB@Exo integrated an exosome-derived margination effect, and density-mediated nanoparticle-induced extracellular leakiness (nanoEL) exhibited more than a 70% colocalization of the RB@Exo to metastatic tumors in the lung in vivo. The release of cancer cell-cell interactions at the metastasis via nanoEL also elicited the 10-fold infiltration of T lymphocytes. The synergy of the T cell infiltration and photolytic effects transported by the RB@Exo deep into the metastatic tumors effectively inhibited the tumor in 60 days when treated with a single alternating magnetic field (AMF).


Subject(s)
Lung Neoplasms , Nanoparticles , Biomimetics , Cell Line, Tumor , Humans , Immunotherapy , T-Lymphocytes
20.
Int J Radiat Oncol Biol Phys ; 109(5): 1547-1558, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33188861

ABSTRACT

PURPOSE: To investigate the temporal and spatial infiltration of TRAMP-C1 tumors by myeloid-derived suppressor cells (MDSCs) after high-dose radiation therapy (RT), and to explore their effect on tumor growth. METHODS AND MATERIALS: TRAMP-C1 intramuscularly tumors were irradiated with a single dose of 8 Gy or 25 Gy. The dynamics of infiltrated MDSCs and their intratumoral spatial distribution were assessed by immunohistochemistry and flow cytometry. Cytokine levels in the blood and tumor were analyzed by multiplex immunoassay. Mice were injected with anti-Gr-1 antibody to determine whether MDSCs affect tumor growth after RT. RESULTS: CD11b+Gr-1+ MDSCs infiltrated TRAMP-C1 tumors irradiated with 25 Gy, but not 8 Gy, within 4 hours and recruitment persisted for at least 2 weeks. Both CD11b+Ly6G+Ly6C+ polymorphonuclear-MDSCs (PMN-MDSCs) and CD11b+Ly6G-Ly6Chi monocytic-MDSCs (M-MDSCs) were involved. Tumor RT also increased the representation of both MDSC subpopulations in the spleen and peripheral blood. Levels of multiple cytokines were increased in the tumors at 2 weeks, including GM-CSF, G-CSF, CCL-3, CCL-5, CXCL-5, IL-6, IL-17α, and VEGF-a; while G-CSF, IL-6, and TNF-α levels increased in the blood. PMN-MDSCs aggregated in the central necrotic region of the irradiated tumors over time, where they were associated with avascular hypoxia (CD31-PIMO+). MDSCs expressed the proangiogenic factor, matrix metalloproteinase-9, and, within the necrotic area, high levels of arginase-1 and indoleamine 2,3-dioxygenase. Depletion of PMN-MDSCs by Gr-1 antibody increased the efficacy of high-dose RT. CONCLUSIONS: PMN-MDSCs infiltrate TRAMP-C1 tumors after high-dose RT. Their spatial distribution suggests they are involved in the evolution of an intratumoral state of necrosis associated with avascular hypoxia, and their phenotype is consistent with them being immunosuppressive. They appear to promote tumor growth after RT, making them a prime therapeutic target for therapeutic intervention. Assessment of MDSCs and cytokine levels in blood could be an index of the need for such an intervention.


Subject(s)
Myeloid-Derived Suppressor Cells/physiology , Prostatic Neoplasms/radiotherapy , Receptors, Tumor Necrosis Factor, Member 25 , Animals , CD11b Antigen , Cell Movement , Chemokines/analysis , Cytokines/analysis , Disease Models, Animal , Flow Cytometry , Immunoassay/methods , Male , Mice , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/cytology , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/radiation effects , Prostatic Neoplasms/blood , Prostatic Neoplasms/immunology , Radiotherapy Dosage , Receptors, Chemokine/immunology , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...