Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 265
Filter
Add more filters

Publication year range
1.
Cell ; 179(5): 1191-1206.e21, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31730857

ABSTRACT

This study identifies mechanisms mediating responses to immune checkpoint inhibitors using mouse models of triple-negative breast cancer. By creating new mammary tumor models, we find that tumor mutation burden and specific immune cells are associated with response. Further, we developed a rich resource of single-cell RNA-seq and bulk mRNA-seq data of immunotherapy-treated and non-treated tumors from sensitive and resistant murine models. Using this, we uncover that immune checkpoint therapy induces T follicular helper cell activation of B cells to facilitate the anti-tumor response in these models. We also show that B cell activation of T cells and the generation of antibody are key to immunotherapy response and propose a new biomarker for immune checkpoint therapy. In total, this work presents resources of new preclinical models of breast cancer with large mRNA-seq and single-cell RNA-seq datasets annotated for sensitivity to therapy and uncovers new components of response to immune checkpoint inhibitors.


Subject(s)
B-Lymphocytes/immunology , Immunotherapy , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/immunology , Mutation/genetics , T-Lymphocytes, Helper-Inducer/immunology , Animals , CTLA-4 Antigen/metabolism , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Genetic Engineering , Genome , Humans , Immunoglobulin G/metabolism , Lymphocyte Activation/immunology , Mammary Neoplasms, Animal/therapy , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, T-Cell/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/therapy
2.
J Immunol ; 211(2): 295-305, 2023 07 15.
Article in English | MEDLINE | ID: mdl-37256255

ABSTRACT

Spontaneous tumors that arise in genetically engineered mice recapitulate the natural tumor microenvironment and tumor-immune coevolution observed in human cancers, providing a more physiologically relevant preclinical model relative to implanted tumors. Similar to many cancer patients, oncogene-driven spontaneous tumors are often resistant to immunotherapy, and thus novel agents that can effectively promote antitumor immunity against these aggressive cancers show considerable promise for clinical translation, and their mechanistic assessment can broaden our understanding of tumor immunology. In this study, we performed extensive immune profiling experiments to investigate how tumor-targeted TLR9 stimulation remodels the microenvironment of spontaneously arising tumors during an effective antitumor immune response. To model the clinical scenario of multiple tumor sites, we used MMTV-PyMT transgenic mice, which spontaneously develop heterogeneous breast tumors throughout their 10 mammary glands. We found that i.v. administration of a tumor-targeting TLR9 agonist, referred to as PIP-CpG, induced a systemic T cell-mediated immune response that not only promoted regression of existing mammary tumors, but also elicited immune memory capable of delaying growth of independent newly arising tumors. Within the tumor microenvironment, PIP-CpG therapy initiated an inflammatory cascade that dramatically amplified chemokine and cytokine production, prompted robust infiltration and expansion of innate and adaptive immune cells, and led to diverse and unexpected changes in immune phenotypes. This study demonstrates that effective systemic treatment of an autochthonous multisite tumor model can be achieved using a tumor-targeted immunostimulant and provides immunological insights that will inform future therapeutic strategies.


Subject(s)
Breast Neoplasms , Mammary Neoplasms, Animal , Mice , Animals , Humans , Female , Toll-Like Receptor 9 , Mice, Transgenic , Adjuvants, Immunologic/pharmacology , Mammary Neoplasms, Animal/therapy , Breast Neoplasms/therapy , Tumor Microenvironment , Cell Line, Tumor
3.
J Immunol ; 207(5): 1298-1309, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34362833

ABSTRACT

Intralesional therapy is a promising approach for remodeling the immunosuppressive tumor microenvironment while minimizing systemic toxicities. A combinatorial in situ immunomodulation (ISIM) regimen with intratumoral administration of Fms-like tyrosine kinase 3 ligand (Flt3L), local irradiation, and TLR3/CD40 stimulation induces and activates conventional type 1 dendritic cells in the tumor microenvironment and elicits de novo adaptive T cell immunity in poorly T cell-inflamed tumors. However, the impact of ISIM on myeloid-derived suppressor cells (MDSCs), which may promote treatment resistance, remains unknown. In this study, we examined changes in the frequencies and heterogeneity of CD11b+Ly-6CloLy-6G+ polymorphonuclear (PMN)-MDSCs and CD11b+Ly-6ChiLy-6G- monocytic (M)-MDSCs in ISIM-treated tumors using mouse models of triple-negative breast cancer. We found that ISIM treatment decreased intratumoral PMN-MDSCs, but not M-MDSCs. Although the frequency of M-MDSCs remained unchanged, ISIM caused a substantial reduction of CX3CR1+ M-MDSCs that express F4/80. Importantly, these ISIM-induced changes in tumor-residing MDSCs were not observed in Batf3-/- mice. ISIM upregulated PD-L1 expression in both M-MDSCs and PMN-MDSCs and synergized with anti-PD-L1 therapy. Furthermore, ISIM increased the expression of IFN regulatory factor 8 (IRF8) in myeloid cells, a known negative regulator of MDSCs, indicating a potential mechanism by which ISIM decreases PMN-MDSC levels. Accordingly, ISIM-mediated reduction of PMN-MDSCs was not observed in mice with conditional deletion of IRF8 in myeloid cells. Altogether, these findings suggest that ISIM holds promise as a multimodal intralesional therapy to alter both lymphoid and myeloid compartments of highly aggressive poorly T cell-inflamed, myeloid-enriched tumors resistant to anti-PD-L1 therapy.


Subject(s)
Dendritic Cells/immunology , Immunotherapy/methods , Interferon Regulatory Factors/metabolism , Mammary Neoplasms, Animal/therapy , Membrane Proteins/therapeutic use , Myeloid-Derived Suppressor Cells/immunology , T-Lymphocytes/immunology , Animals , B7-H1 Antigen , Basic-Leucine Zipper Transcription Factors/genetics , CD40 Antigens/metabolism , Cell Line, Tumor , Combined Modality Therapy , Drug Resistance , Gene Expression Regulation , Humans , Injections, Intralesional , Interferon Regulatory Factors/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Radiotherapy , Repressor Proteins/genetics , Toll-Like Receptor 3/metabolism , Tumor Microenvironment
4.
Reprod Domest Anim ; 58 Suppl 2: 102-108, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37312625

ABSTRACT

The poor outcomes associated with mammary carcinomas (MCs) in dogs and cats in terms of locoregional recurrence, distant metastasis and survival, highlight the need for better management of mammary cancers in small animals. By contrast, the outcomes of women with breast cancer (BC) have dramatically improved during the last 10 years, notably thanks to new therapeutic strategies. The aim of this article was to imagine what could be the future of therapy for dogs and cats with MCs if it became inspired from current practices in human BC. This article focuses on the importance of taking into account cancer stage and cancer subtypes in therapeutic plans, on locoregional treatments (surgery, radiation therapy), new developments in endocrine therapy, chemotherapy, PARP inhibitors and immunotherapy. Ideally, multimodal treatment regimens would be chosen according to cancer stage and cancer subtypes, and according to predictive factors that are still to be defined.


Subject(s)
Breast Neoplasms , Carcinoma , Cat Diseases , Dog Diseases , Mammary Neoplasms, Animal , Humans , Animals , Female , Cats , Dogs , Cat Diseases/therapy , Dog Diseases/therapy , Dog Diseases/pathology , Neoplasm Recurrence, Local/veterinary , Breast Neoplasms/therapy , Breast Neoplasms/veterinary , Breast Neoplasms/pathology , Carcinoma/veterinary , Mammary Neoplasms, Animal/therapy
5.
PLoS Pathog ; 16(10): e1008660, 2020 10.
Article in English | MEDLINE | ID: mdl-33075093

ABSTRACT

Mammary carcinoma, including triple-negative breast carcinomas (TNBC) are tumor-types for which human and canine pathologies are closely related at the molecular level. The efficacy of an oncolytic vaccinia virus (VV) was compared in low-passage primary carcinoma cells from TNBC versus non-TNBC. Non-TNBC cells were 28 fold more sensitive to VV than TNBC cells in which VV replication is impaired. Single-cell RNA-seq performed on two different TNBC cell samples, infected or not with VV, highlighted three distinct populations: naïve cells, bystander cells, defined as cells exposed to the virus but not infected and infected cells. The transcriptomes of these three populations showed striking variations in the modulation of pathways regulated by cytokines and growth factors. We hypothesized that the pool of genes expressed in the bystander populations was enriched in antiviral genes. Bioinformatic analysis suggested that the reduced activity of the virus was associated with a higher mesenchymal status of the cells. In addition, we demonstrated experimentally that high expression of one gene, DDIT4, is detrimental to VV production. Considering that DDIT4 is associated with a poor prognosis in various cancers including TNBC, our data highlight DDIT4 as a candidate resistance marker for oncolytic poxvirus therapy. This information could be used to design new generations of oncolytic poxviruses. Beyond the field of gene therapy, this study demonstrates that single-cell transcriptomics can be used to identify cellular factors influencing viral replication.


Subject(s)
Mammary Neoplasms, Animal/metabolism , Oncolytic Virotherapy/methods , Transcription Factors/metabolism , Transcriptome , Vaccinia virus/genetics , Vaccinia/metabolism , Virus Replication , Animals , Computational Biology , Dogs , Female , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/therapy , Mammary Neoplasms, Animal/virology , Single-Cell Analysis , Transcription Factors/genetics , Vaccinia/genetics , Vaccinia/virology
6.
Bull Exp Biol Med ; 174(1): 104-108, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36437316

ABSTRACT

We studied the relationship between the level of cytokines in the lymph of the thoracic duct and the morphometric parameters of the mesenteric lymph nodes after surgical treatment of breast cancer, chemotherapy, and administration of fragmented (double-stranded, dsDNA) human DNA. In comparison with surgical treatment and with chemotherapy alone, administration of a human dsDNA has a stimulating effect on the T-cell link of the immune response. In the paracortical zone, the relationship between the chemokine MCP-1 and increased content of small lymphocytes in this zone was revealed. Interrelations of IL-2 cytokines with small lymphocytes and of IL-4 with medium lymphocytes were revealed in germinal centers. We also observed interrelations of IL-7 with small lymphocytes and IL-4 with macrophages in the medullary cords, chemokine MIP-1α with immature and mature plasma cells (the number of these cells is reduced), and of MCP-1 with immunoblasts (the number of which is also reduced) in the medullary sinuses.


Subject(s)
Adjuvants, Immunologic , Breast Neoplasms , Cytokines , DNA , Lymph Nodes , Mammary Neoplasms, Animal , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/surgery , Breast Neoplasms/therapy , Chemokines/metabolism , Cytokines/metabolism , DNA/administration & dosage , Interleukin-4/metabolism , Lymph/metabolism , Lymph Nodes/metabolism , Animals , Rats , Rats, Wistar , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/surgery , Mammary Neoplasms, Animal/therapy , Adjuvants, Immunologic/administration & dosage , T-Lymphocytes/immunology
7.
Int J Mol Sci ; 22(5)2021 Feb 26.
Article in English | MEDLINE | ID: mdl-33653008

ABSTRACT

Gold nanoparticles (AuNPs) are foreseen as a promising tool in nanomedicine, both as drug carriers and radiosensitizers. They have been also proposed as a potential anticancer drug due to the anti-angiogenic effect in tumor tissue. In this work we investigated the effect of citrate-coated AuNPs of nominal diameter 20 nm on the growth and metastatic potential of 4T1 cells originated from a mouse mammary gland tumor inoculated into the mammary fat pad of Balb/ccmdb mice. To evaluate whether AuNPs can prevent the tumor growth, one group of inoculated mice was intragastrically (i.g.) administered with 1 mg/kg of AuNPs daily from day 1 to day 14 after cancer cell implantation. To evaluate whether AuNPs can attenuate the tumor growth, the second group was intravenously (i.v.) administered with 1 or 5 mg/kg of AuNPs, twice on day 5 and day 14 after inoculation. We did not observe any anticancer activity of i.v. nor i.g. administered AuNPs, as they did not affect neither the primary tumor growth rate nor the number of lung metastases. Unexpectedly, both AuNP treatment regimens caused a marked vasodilating effect in the tumor tissue. As no change of potential angiogenic genes (Fgf2, Vegfa) nor inducible nitric oxygenase (Nos2) was observed, we proposed that the vasodilation was caused by AuNP-dependent decomposition of nitrosothiols and direct release of nitric oxide in the tumor tissue.


Subject(s)
Citric Acid/therapeutic use , Gold/therapeutic use , Mammary Neoplasms, Animal/blood supply , Metal Nanoparticles/therapeutic use , Animals , Cell Line, Tumor , Citric Acid/administration & dosage , Female , Gold/administration & dosage , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/therapy , Metal Nanoparticles/administration & dosage , Mice , Mice, Inbred BALB C , Nanomedicine , Particle Size , Vasodilation
8.
Nanomedicine ; 29: 102240, 2020 10.
Article in English | MEDLINE | ID: mdl-32553948

ABSTRACT

Overexpression of adenosine in the tumor region leads to suppression of various immune cells, particularly T cells through ligation with adenosine 2a receptor (A2aR). In this study, we intended to increase the efficacy of tumor lysate-loaded DC vaccine by silencing the expression of A2aR on T cells through the application of A2aR-specific siRNA-loaded PEG-chitosan-lactate (PCL) nanoparticles (NPs) in the 4T1 breast tumor-bearing mice. Combination therapy by DC vaccine and siRNA-loaded NPs markedly induced tumor regression and increased survival time of mice. These ameliorative effects were partly via downregulation of immunosuppressive cells, increased function of cytotoxic T lymphocytes, and induction of immune-stimulatory cytokines. Moreover, combination therapy could markedly suppress angiogenesis and metastasis processes. These results imply the efficacy of novel combination therapy for the treatment of breast cancer by using A2aR siRNA-loaded NPs and DC vaccine which can be translated into the initial phase of clinical trials in the near future.


Subject(s)
Breast Neoplasms/therapy , Mammary Neoplasms, Animal/therapy , Nanoparticles/chemistry , Receptor, Adenosine A2A/genetics , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/pharmacology , Cell Line, Tumor , Chitosan/chemistry , Chitosan/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunotherapy , Lactic Acid/chemistry , Lactic Acid/pharmacology , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology
9.
Nano Lett ; 19(8): 5587-5594, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31260628

ABSTRACT

In breast cancer chemophotothermal therapy, it is a great challenge for the development of multifunctional nanoagents for precision targeting and the effective treatment of tumors, especially for metastasis. Herein, we successfully design and synthesize a multifunctional black phosphorus (BP)-based nanoagent, BP/DTX@PLGA, to address this challenge. In this composite nanoagent, BP quantum dots (BPQDs) are loaded into poly(lactic-co-glycolic acid) (PLGA) with additional conjugation of a chemotherapeutic agent, docetaxel (DTX). The in vivo distribution results demonstrate that BP/DTX@PLGA shows striking tropism for targeting both primary tumors and lung metastatic tumors. Moreover, BP/DTX@PLGA exhibits outstanding controllable chemophotothermal combinatory therapeutics, which dramatically improves the efficacy of photothermal tumor ablation when combined with near-light irradiation. Mechanistically, accelerated DTX release from the nanocomplex upon heating and thermal treatment per se synergistically incurs apoptosis-dependent cell death, resulting in the elimination of lung metastasis. Meanwhile, in vitro and in vivo results further confirm that BP/DTX@PLGA possesses good biocompatibility. This study provides a promising BP-based multimodal nanoagent to constrain cancer metastasis.


Subject(s)
Antineoplastic Agents/therapeutic use , Docetaxel/therapeutic use , Mammary Neoplasms, Animal/therapy , Nanoconjugates/therapeutic use , Phosphorus/therapeutic use , Animals , Antineoplastic Agents/pharmacokinetics , Docetaxel/pharmacokinetics , Female , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mammary Neoplasms, Animal/pathology , Mice , Neoplasm Metastasis/pathology , Neoplasm Metastasis/therapy , Phosphorus/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/therapeutic use
10.
Nano Lett ; 19(2): 805-815, 2019 02 13.
Article in English | MEDLINE | ID: mdl-30592897

ABSTRACT

Amplification of intracellular oxidative stress has been found to be an effective strategy to induce cancer cell death. To this end, we prepare a unique type of ultrasmall gallic acid-ferrous (GA-Fe(II)) nanocomplexes as the catalyst of Fenton reaction to enable persistent conversion of H2O2 to highly cytotoxic hydroxyl radicals (•OH). Then, both GA-Fe(II) and l-buthionine sulfoximine (BSO), an inhibitor of glutathione (GSH) synthesis, are coencapsulated within a stealth liposomal nanocarrier. Interestingly, the obtained BSO/GA-Fe(II)@liposome is able to efficiently amplify intracellular oxidative stress via increasing •OH generation and reducing GSH biosynthesis. After chelating with 99mTc4+ radioisotope, such BSO/GA-Fe(II)@liposome could be tracked under in vivo single-photon-emission-computed-tomography (SPECT) imaging, which illustrates the time-dependent tumor homing of such liposomal nanoparticles after intravenous injection. With GA-Fe(II)-mediated •OH production and BSO-mediated GSH depletion, treatment with such BSO/GA-Fe(II)@liposome would lead to dramatically enhanced intratumoral oxidative stresses, which then result in remarkably improved therapeutic efficacies of concurrently applied chemotherapy or radiotherapy. This work thus presents the concise fabrication of biocompatible BSO/GA-Fe(II)@liposome as an effective adjuvant nanomedicine to promote clinically used conventional cancer chemotherapy and radiotherapy, by greatly amplifying the intratumoral oxidative stress.


Subject(s)
Buthionine Sulfoximine/therapeutic use , Ferrous Compounds/therapeutic use , Gallic Acid/therapeutic use , Glutathione/antagonists & inhibitors , Mammary Neoplasms, Animal/therapy , Oxidative Stress/drug effects , Animals , Buthionine Sulfoximine/administration & dosage , Cell Line, Tumor , Female , Ferrous Compounds/administration & dosage , Gallic Acid/administration & dosage , Glutathione/metabolism , Hydroxyl Radical/metabolism , Liposomes/chemistry , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/radiotherapy , Mice , Mice, Inbred BALB C , Tomography, Emission-Computed, Single-Photon
11.
J Cell Biochem ; 120(9): 15546-15552, 2019 09.
Article in English | MEDLINE | ID: mdl-31050374

ABSTRACT

Hypoxia is a condition of low oxygen level which poses a common feature of most cancers. In the current study, we investigated effect of water containing oxygen nanobubble (ONB) on tumor growth in breast cancer 4T1-bearing mice during 14-day treatment period. Tumor-bearing mice were randomly divided into three groups (six mice per group), including the ONB group drinking water containing ONB, the air nanobubble (ANB) group drinking water containing ANB, and control group drinking normal water. Tumor weight and size were measured in 2-day interval during 14-day treatment. mRNA expression of p53, vascular endothelial growth factor (VEGF), hypoxia-inducible factor (HIF), and cyclin D/Cdk2 genes were measured in the treated and control mice. After 8, 12, and 14 days of treatment, tumor size in ONB group was significantly decreased by 40.5%, 32.8%, and 28%, respectively, when compared with the control group. In addition, ANB group showed a significant reduction in tumor burden as well. The messenger RNA (mRNA) level of p53 in tumor cells of ONB and ANB group was found to be 36-fold (P = 0.0001) and 33-fold (P = 0.0001) higher than that in the control group, respectively. There was a ninefold increase in mRNA expression of VEGF gene in tumor cells of ANB mice than that in control mice; however, there was no significant changes in ONB group. Expression of HIF gene was significantly lower in tumor cells of ONB and ANB group than in the control group. It is concluded that drinking ONB water has potential to inhibit tumor growth, however more preclinical and proof-of-concept studies are needed to confirm its safety and therapeutic effect.


Subject(s)
Breast Neoplasms/therapy , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mammary Neoplasms, Animal/therapy , Oxygen/pharmacology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cyclin D/genetics , Cyclin-Dependent Kinase 2/genetics , Female , Fishes , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Oxygen/chemistry , RNA, Messenger/genetics , Tumor Hypoxia/drug effects , Vascular Endothelial Growth Factor A/genetics , Water/chemistry , Water/pharmacology
12.
Br J Cancer ; 120(2): 196-206, 2019 01.
Article in English | MEDLINE | ID: mdl-30498230

ABSTRACT

BACKGROUND: There are phase 3 clinical trials underway evaluating anti-PD-L1 antibodies as adjuvant (postoperative) monotherapies for resectable renal cell carcinoma (RCC) and triple-negative breast cancer (TNBC); in combination with antiangiogenic VEGF/VEGFR2 inhibitors (e.g., bevacizumab and sunitinib) for metastatic RCC; and in combination with chemotherapeutics as neoadjuvant (preoperative) therapies for resectable TNBC. METHODS: This study investigated these and similar clinically relevant drug combinations in highly translational preclinical models of micro- and macro-metastatic disease that spontaneously develop after surgical resection of primary kidney or breast tumours derived from orthotopic implantation of murine cancer cell lines (RENCAluc or EMT-6/CDDP, respectively). RESULTS: In the RENCAluc model, adjuvant sunitinib plus anti-PD-L1 improved overall survival compared to either drug alone, while the same combination was ineffective as early therapy for unresected primary tumours or late-stage therapy for advanced metastatic disease. In the EMT-6/CDDP model, anti-PD-L1 was highly effective as an adjuvant monotherapy, while its combination with paclitaxel chemotherapy (with or without anti-VEGF) was most effective as a neoadjuvant therapy. CONCLUSIONS: Our preclinical data suggest that anti-PD-L1 plus sunitinib may warrant further investigation as an adjuvant therapy for RCC, while anti-PD-L1 may be improved by combining with chemotherapy in the neoadjuvant but not the adjuvant setting of treating breast cancer.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Kidney Neoplasms/therapy , Mammary Neoplasms, Animal/therapy , Neovascularization, Pathologic/therapy , Animals , B7-H1 Antigen/immunology , Bevacizumab/administration & dosage , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Immunotherapy/methods , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Kidney Neoplasms/surgery , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/surgery , Mice , Neoadjuvant Therapy/methods , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Paclitaxel/administration & dosage , Sunitinib/administration & dosage
13.
Biochem Biophys Res Commun ; 511(3): 587-591, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30824183

ABSTRACT

BACKGROUND: Tungsten disulfide (WS2), which enjoyed a good potential to be a promising clinical theranostic agent for cancer treatment, is still subject to the tedious synthesis procedure. METHODS: Here, we reported a one-pot 'bottom-up' hydrothermal strategy for the fabrication of PEGylated WS2 nanoparticles (NPs). The WS2-PEG nanoparticles were characterized systematically. The CT imaging and photothermal therapy against tumor as well as biosafety in vitro and in vivo were also investigated. RESULTS: The obtained WS2-PEG NPs enjoyed obvious merits of good solubility and favorable photothermal performance. WS2-PEG NPs exhibited desirable photothermal ablation ability against cancer cells and cancer cell-bearing mice in vitro and in vivo. MTT assay and histological analysis demonstrated the low cytotoxicity and biotoxicity of WS2-PEG NPs, providing a valid biosafety guarantee for the coming biomedical applications. In addition, thanks to the obvious X-ray attenuation of W atom, the WS2-PEG NPs can also be served as a favorable contrast agent for CT imaging of tumors. CONCLUSION: WS2-PEG NPs has enjoyed a good potential to be a promising clinical CT-guided photothermal therapeutic agent against cancers.


Subject(s)
Mammary Neoplasms, Animal/therapy , Nanoparticles/therapeutic use , Polyethylene Glycols/therapeutic use , Tungsten Compounds/therapeutic use , Animals , Cell Line, Tumor , Female , Hyperthermia, Induced , Mammary Neoplasms, Animal/diagnostic imaging , Mice , Theranostic Nanomedicine , Tomography, X-Ray Computed
14.
Small ; 15(45): e1903462, 2019 11.
Article in English | MEDLINE | ID: mdl-31523920

ABSTRACT

The clinical use of interleukin-12 (IL12), a cytokine endowed with potent immunotherapeutic anticancer activity, is limited by systemic toxicity. The hypothesis is addressed that gold nanoparticles tagged with a tumor-homing peptide containing isoDGR, an αvß3-integrin binding motif, can be exploited for delivering IL12 to tumors and improving its therapeutic index. To this aim, gold nanospheres are functionalized with the head-to-tail cyclized-peptide CGisoDGRG (Iso1) and murine IL12. The resulting nanodrug (Iso1/Au/IL12) is monodispersed, stable, and bifunctional in terms of αvß3 and IL12-receptor recognition. Low-dose Iso1/Au/IL12, equivalent to 18-75 pg of IL12, induces antitumor effects in murine models of fibrosarcomas and mammary adenocarcinomas, with no evidence of toxicity. Equivalent doses of Au/IL12 (a nanodrug lacking Iso1) fail to delay tumor growth, whereas 15 000 pg of free IL12 is necessary to achieve similar effects. Iso1/Au/IL12 significantly increases tumor infiltration by innate immune cells, such as NK and iNKT cells, monocytes, and neutrophils. NK cell depletion completely inhibits its antitumor effects. Low-dose Iso1/Au/IL12 can also increase the therapeutic efficacy of adoptive T-cell therapy in mice with autochthonous prostate cancer. These findings indicate that coupling IL12 to isoDGR-tagged nanogold is a valid strategy for enhancing its therapeutic index and sustaining adoptive T-cell therapy.


Subject(s)
Gold/chemistry , Immunotherapy/methods , Interleukin-12/metabolism , Metal Nanoparticles/chemistry , Adenocarcinoma/therapy , Animals , Cells, Cultured , Female , Fibrosarcoma/therapy , Male , Mammary Neoplasms, Animal/therapy , Mice
15.
Proc Natl Acad Sci U S A ; 113(3): E319-27, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26729864

ABSTRACT

Immunotherapy is gathering momentum as a primary therapy for cancer patients. However, monotherapies have limited efficacy in improving outcomes and benefit only a subset of patients. Combination therapies targeting multiple pathways can augment an immune response to improve survival further. Here, we demonstrate that dual aOX40 (anti-CD134)/aCTLA-4 (anti-cytotoxic T-lymphocyte-associated protein 4) immunotherapy generated a potent antigen-specific CD8 T-cell response, enhancing expansion, effector function, and memory T-cell persistence. Importantly, OX40 and CTLA-4 expression on CD8 T cells was critical for promoting their maximal expansion following combination therapy. Animals treated with combination therapy and vaccination using anti-DEC-205 (dendritic and epithelial cells, 205 kDa)-HER2 (human epidermal growth factor receptor 2) had significantly improved survival in a mammary carcinoma model. Vaccination with combination therapy uniquely restricted Th2-cytokine production by CD4 cells, relative to combination therapy alone, and enhanced IFNγ production by CD8 and CD4 cells. We observed an increase in MIP-1α (macrophage inflammatory protein-1α)/CCL3 [chemokine (C-C motif) ligand 3], MIP-1ß/CCL4, RANTES (regulated on activation, normal T-cell expressed and excreted)/CCL5, and GM-CSF production by CD8 and CD4 T cells following treatment. Furthermore, this therapy was associated with extensive tumor destruction and T-cell infiltration into the tumor. Notably, in a spontaneous model of prostate adenocarcinoma, vaccination with combination therapy reversed anergy and enhanced the expansion and function of CD8 T cells recognizing a tumor-associated antigen. Collectively, these data demonstrate that the addition of a vaccine with combined aOX40/aCTLA-4 immunotherapy augmented antitumor CD8 T-cell function while limiting Th2 polarization in CD4 cells and improved overall survival.


Subject(s)
CTLA-4 Antigen/immunology , Clonal Anergy/immunology , Neoplasms/immunology , Neoplasms/therapy , Receptor, ErbB-2/immunology , Receptors, OX40/agonists , Vaccination , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cell Polarity , Cell Proliferation , Combined Modality Therapy , Female , Immunologic Memory , Immunotherapy , Male , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/therapy , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/pathology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Receptors, OX40/metabolism , Survival Analysis , Th2 Cells/cytology
16.
Nano Lett ; 18(11): 7330-7342, 2018 11 14.
Article in English | MEDLINE | ID: mdl-30339753

ABSTRACT

Without coordinated strategies to mitigate the immunosuppressive nature of the tumor microenvironment, cancer immunotherapy generally offers limited clinical benefit for established tumors. Tumor-associated macrophages (TAMs) are the critical driver of this immunosuppressive tumor microenvironment, which also promotes tumor metastasis. Here we successfully reprogrammed TAMs to an antitumor M1 phenotype using precision nanoparticle-based reactive oxygen species photogeneration, which demonstrated superior efficiency and efficacy over lipopolysaccharide stimulation. Meanwhile, antigen presentation and T-cell-priming by TAMs were enhanced by inhibiting lysosomal proton pump and proteolytic activity or by promoting tumor associated antigen release in the cytoplasm. The reprogrammed TAMs orchestrate cytotoxic lymphocyte (CTL) recruitment in the tumor and direct memory T-cells toward tumoricidal responses. This strategy could effectively eradicate tumors, inhibit metastasis, and further prevent their recurrence, which holds tremendous promise to realize potent cancer immunotherapy.


Subject(s)
Cellular Reprogramming , Immunotherapy , Macrophages/immunology , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/therapy , Nanoparticles/therapeutic use , Reactive Oxygen Species/immunology , Animals , Antigen Presentation/drug effects , Antigens, Neoplasm/immunology , Cellular Reprogramming/drug effects , Cellular Reprogramming/immunology , Female , Immunologic Memory , Macrophages/pathology , Mice , Mice, Inbred BALB C , T-Lymphocytes/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
17.
Nano Lett ; 18(11): 6804-6811, 2018 11 14.
Article in English | MEDLINE | ID: mdl-30350653

ABSTRACT

Free radicals have emerged as new-type and promising candidates for hypoxic tumor treatment, and further study of their therapeutic mechanism by real-time imaging is of great importance to explore their biomedical applications. Herein, we present a smart free-radical generator AuNC-V057-TPP for hypoxic tumor therapy; the AuNC-V057-TPP not only exhibits good therapeutic effect under both hypoxic and normoxic conditions but also can monitor the release of free radicals in real-time both in vitro and in vivo. What is more, with the mitochondria-targeting ability, the AuNC-V057-TPP is demonstrated with improved antitumor efficacy through enhanced free radical level in mitochondria, which leads to mitochondrial membrane damage and ATP production reduction and finally induces cancer cell apoptosis.


Subject(s)
Drug Delivery Systems/methods , Free Radicals/metabolism , Gold , Mammary Neoplasms, Animal , Metal Nanoparticles , Mitochondria , Molecular Imaging/methods , Tumor Hypoxia , Animals , Apoptosis/drug effects , Cell Line, Tumor , Female , Gold/chemistry , Gold/pharmacology , Mammary Neoplasms, Animal/diagnostic imaging , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/therapy , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice , Mitochondria/metabolism , Mitochondria/pathology
18.
Bioconjug Chem ; 29(4): 1283-1290, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29402074

ABSTRACT

Nanoagents of integrating multiple imaging and therapeutic modalities have attracted tremendous attention for biomedical applications. Herein, we synthesize porous hollow Fe3O4 as a theranostic agent for MRI and combined photothermal/chemo cancer therapy. The as-prepared porous iron oxide nanoagents allow for T2-weighted MR imaging. Interestingly, we demonstrate that the porous structure endows the nanoagents an outstanding photothermal property for cancer cell killing, in comparison with other types of iron oxide nanomaterials. Under the exposure of an NIR laser, the heat produced by porous Fe3O4 can accelerate the release of the loaded drug (e.g., DOX) to enhance chemotherapeutic efficacy, promoting the ablation of cancer cells with synergistic photothermal/chemotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Doxorubicin/therapeutic use , Ferric Compounds/therapeutic use , Mammary Neoplasms, Animal/therapy , Nanoshells/therapeutic use , Theranostic Nanomedicine/methods , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Combined Modality Therapy/methods , Doxorubicin/administration & dosage , Drug Carriers/administration & dosage , Drug Carriers/therapeutic use , Drug Liberation , Female , Ferric Compounds/administration & dosage , Hyperthermia, Induced/methods , Magnetic Resonance Imaging , Mammary Neoplasms, Animal/diagnostic imaging , Mice , Nanoshells/administration & dosage , Nanoshells/ultrastructure , Phototherapy/methods , Porosity
19.
Chemistry ; 24(49): 12827-12837, 2018 Sep 03.
Article in English | MEDLINE | ID: mdl-29978545

ABSTRACT

Indocyanine green (ICG) is an effective light absorber for laser-mediated photodynamic therapy. However, applications of ICG are limited due to its rapid degradation and poor photostability in water. Herein, we report the development of a multifunctional nanoplatform by coating ICG on the surface of single-walled carbon nanohorns (SWNHs) through π-π stacking, obtaining SWNH-ICGs with high solubility and stability under physiological conditions. The SWNH-ICGs could be used as a single nanoplatform to simultaneously produce satisfactory hyperthermia and reactive oxygen species under near-infrared (NIR) laser irradiation. In addition, the SWNH-ICGs not only improved the photostability of ICG in different media, but also protected it from light degradation. The SWNH-ICGs exhibited highly efficient thermal/photoacoustic (PA) imaging-guided photothermal therapy (PTT) and photodynamic therapy (PDT) effects, even under low-power laser irradiation (0.3 W cm-2 ) in vitro. Combined PTT and PDT effectively killed triple-negative breast cancer 4T1 cells, demonstrating a markedly improved and synergistic therapeutic effect compared to PTT or PDT alone. Furthermore, significant tumor growth inhibition as well as tumor cell death were observed following PTT/PDT at 808 nm laser irradiation, confirming the synergistic effects of SWNH-ICGs over free ICG in vivo. This facile and simple methodology for thermal/PA imaging-guided PTT/PDT suggests that SWNH-ICGs may serve as an effective nanoplatform for cancer therapy.


Subject(s)
Carbon/chemistry , Indocyanine Green/chemistry , Mammary Neoplasms, Animal/therapy , Nanoparticles/chemistry , Photochemotherapy/methods , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Combined Modality Therapy , Drug Carriers , Female , Hyperthermia, Induced , Indocyanine Green/therapeutic use , Infrared Rays , Low-Level Light Therapy , Mice , Reactive Oxygen Species/metabolism , Surface Properties , Tissue Distribution , Ultrasonic Waves
20.
Mol Pharm ; 15(9): 4049-4062, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30011996

ABSTRACT

Combination cancer therapy with various kinds of therapeutic approaches could improve the effectiveness of treatment while reducing side effects. Herein, we elaborately developed a theranostics nanoplatform based on magnetic polydopamine (MPDA) coated with hyaluronic acid-methotrexate conjugates (MPDA@HA-MTX) for chemo-photothermal treatment (PTT). In this nanoplatform, Fe3O4 served as the core was applied as contrast agent for T2-weighted magnetic resonance imaging (MRI) and early phase magnet targeting. Meanwhile, PDA was used as a versatile shell for effective loading of chemotherapeutic doxorubicin (DOX) to achieve controlled release and PTT simultaneously. Moreover, HA-MTX conjugates could offer later-phase specific cellular dual-targeting ability during the therapy. Both in vitro and in vivo studies demonstrated that DOX-loaded MPDA@HA-MTX (MPDA/DOX@HA-MTX) exhibited the preferential tumor accumulation, enhanced specificity to target tumor cells, pH-/laser-responsive release, and high tumor cell-killing efficiency. By combined chemo-PTT under the guidance of fluorescence/MR imaging, the tumors in mice were completely eliminated after treatment, indicating that MPDA@HA-MTX nanoparticles have great potential as a novel drug-loading platform for imaging-guided multistage targeted chemo-photothermal combination therapy.


Subject(s)
Hyaluronic Acid/chemistry , Indoles/chemistry , Methotrexate/chemistry , Nanoparticles/chemistry , Polymers/chemistry , Animals , Doxorubicin/chemistry , Doxorubicin/therapeutic use , Drug Delivery Systems/methods , Female , Flow Cytometry , HeLa Cells , Humans , Hyperthermia, Induced , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/therapy , Mice , Mice, Inbred BALB C , Mice, Nude , Multimodal Imaging , Phototherapy , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL