Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 326
Filter
Add more filters

Country/Region as subject
Publication year range
1.
EMBO J ; 42(15): e113126, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37345898

ABSTRACT

N6 -methyladenosine (m6 A) in messenger RNA (mRNA) regulates immune cells in homeostasis and in response to infection and inflammation. The function of the m6 A reader YTHDF2 in the tumor microenvironment (TME) in these contexts has not been explored. We discovered that the loss of YTHDF2 in regulatory T (Treg) cells reduces tumor growth in mice. Deletion of Ythdf2 in Tregs does not affect peripheral immune homeostasis but leads to increased apoptosis and impaired suppressive function of Treg cells in the TME. Elevated tumor necrosis factor (TNF) signaling in the TME promotes YTHDF2 expression, which in turn regulates NF-κB signaling by accelerating the degradation of m6 A-modified transcripts that encode NF-κB-negative regulators. This TME-specific regulation of Treg by YTHDF2 points to YTHDF2 as a potential target for anti-cancer immunotherapy, where intratumoral Treg cells can be targeted to enhance anti-tumor immune response while avoiding Treg cells in the periphery to minimize undesired inflammations.


Subject(s)
NF-kappa B , Neoplasms , Mice , Animals , NF-kappa B/genetics , Neoplasms/genetics , Signal Transduction , Immunotherapy , Inflammation , Tumor Microenvironment
2.
Immunity ; 49(3): 490-503.e4, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30170810

ABSTRACT

The NF-κB pathway plays a crucial role in supporting tumor initiation, progression, and radioresistance of tumor cells. However, the role of the NF-κB pathway in radiation-induced anti-tumor host immunity remains unclear. Here we demonstrated that inhibiting the canonical NF-κB pathway dampened the therapeutic effect of ionizing radiation (IR), whereas non-canonical NF-κB deficiency promoted IR-induced anti-tumor immunity. Mechanistic studies revealed that non-canonical NF-κB signaling in dendritic cells (DCs) was activated by the STING sensor-dependent DNA-sensing pathway. By suppressing recruitment of the transcription factor RelA onto the Ifnb promoter, activation of the non-canonical NF-κB pathway resulted in decreased type I IFN expression. Administration of a specific inhibitor of the non-canonical NF-κB pathway enhanced the anti-tumor effect of IR in murine models. These findings reveal the potentially interactive roles for canonical and non-canonical NF-κB pathways in IR-induced STING-IFN production and provide an alternative strategy to improve cancer radiotherapy.


Subject(s)
Colonic Neoplasms/radiotherapy , Dendritic Cells/immunology , Melanoma/radiotherapy , NF-kappa B/metabolism , Neoplasms, Experimental/radiotherapy , Radiotherapy/methods , Receptors, Pattern Recognition/metabolism , Animals , Colonic Neoplasms/immunology , DNA/immunology , Disease Models, Animal , Humans , Immunity, Cellular , Melanoma/immunology , Melanoma, Experimental , Membrane Proteins/metabolism , Mice , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Radiation Tolerance , Radiation, Ionizing , Signal Transduction , Transcription Factor RelA/metabolism , Xenograft Model Antitumor Assays
3.
Immunity ; 47(2): 363-373.e5, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28801234

ABSTRACT

Inhibition of cytosolic DNA sensing represents a strategy that tumor cells use for immune evasion, but the underlying mechanisms are unclear. Here we have shown that CD47-signal regulatory protein α (SIRPα) axis dictates the fate of ingested DNA in DCs for immune evasion. Although macrophages were more potent in uptaking tumor DNA, increase of DNA sensing by blocking the interaction of SIRPα with CD47 preferentially occurred in dendritic cells (DCs) but not in macrophages. Mechanistically, CD47 blockade enabled the activation of NADPH oxidase NOX2 in DCs, which in turn inhibited phagosomal acidification and reduced the degradation of tumor mitochondrial DNA (mtDNA) in DCs. mtDNA was recognized by cyclic-GMP-AMP synthase (cGAS) in the DC cytosol, contributing to type I interferon (IFN) production and antitumor adaptive immunity. Thus, our findings have demonstrated how tumor cells inhibit innate sensing in DCs and suggested that the CD47-SIRPα axis is critical for DC-driven antitumor immunity.


Subject(s)
Antigens, Differentiation/metabolism , Colonic Neoplasms/immunology , DNA, Mitochondrial/immunology , Dendritic Cells/immunology , Membrane Proteins/metabolism , Receptors, Immunologic/metabolism , Animals , Antibodies, Blocking/therapeutic use , CD47 Antigen/immunology , CD47 Antigen/metabolism , Cells, Cultured , Colonic Neoplasms/genetics , Colonic Neoplasms/therapy , Cross-Priming , Disease Models, Animal , Humans , Interferon Type I/metabolism , Macrophages/immunology , Membrane Glycoproteins/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Nucleotidyltransferases/metabolism , Signal Transduction , Tumor Escape
4.
Nature ; 566(7743): 270-274, 2019 02.
Article in English | MEDLINE | ID: mdl-30728504

ABSTRACT

There is growing evidence that tumour neoantigens have important roles in generating spontaneous antitumour immune responses and predicting clinical responses to immunotherapies1,2. Despite the presence of numerous neoantigens in patients, complete tumour elimination is rare, owing to failures in mounting a sufficient and lasting antitumour immune response3,4. Here we show that durable neoantigen-specific immunity is regulated by mRNA N6-methyadenosine (m6A) methylation through the m6A-binding protein YTHDF15. In contrast to wild-type mice, Ythdf1-deficient mice show an elevated antigen-specific CD8+ T cell antitumour response. Loss of YTHDF1 in classical dendritic cells enhanced the cross-presentation of tumour antigens and the cross-priming of CD8+ T cells in vivo. Mechanistically, transcripts encoding lysosomal proteases are marked by m6A and recognized by YTHDF1. Binding of YTHDF1 to these transcripts increases the translation of lysosomal cathepsins in dendritic cells, and inhibition of cathepsins markedly enhances cross-presentation of wild-type dendritic cells. Furthermore, the therapeutic efficacy of PD-L1 checkpoint blockade is enhanced in Ythdf1-/- mice, implicating YTHDF1 as a potential therapeutic target in anticancer immunotherapy.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/metabolism , Dendritic Cells/immunology , Neoplasms/immunology , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , B7-H1 Antigen/metabolism , Binding Sites , CD8-Positive T-Lymphocytes/immunology , Cathepsins/antagonists & inhibitors , Cathepsins/biosynthesis , Cathepsins/genetics , Cross-Priming/immunology , Dendritic Cells/enzymology , Female , Humans , Methylation , Mice , Mice, Inbred C57BL , Neoplasms/therapy , Protein Biosynthesis , Proteins/genetics , RNA, Messenger/chemistry , RNA-Binding Proteins/genetics , Transcriptome/genetics
5.
Nature ; 568(7751): E3, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30911170

ABSTRACT

In this Letter, a citation to 'Fig. 1e' has been corrected to 'Fig. 1d' in the sentence starting "By contrast, the anti-tumour response…". This has been corrected online.

6.
Lancet Oncol ; 25(8): e352-e362, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39089313

ABSTRACT

Radiotherapy exerts immunostimulatory and immunosuppressive effects, both locally, within the irradiated tumour microenvironment, and systemically, outside the radiation field. Inspired by preclinical data that showed synergy between radiotherapy and immune checkpoint inhibitors, multiple clinical trials were initiated with the hypothesis that combined treatment with radiotherapy and immune checkpoint inhibitors could stimulate a robust systemic immune response and improve clinical outcomes. However, despite early optimism, radioimmunotherapy trials in the curative and metastatic settings have met with little success. In this Review, we summarise the immunostimulatory effects of radiotherapy that provided the theoretical basis for trials of combination radiotherapy and immune checkpoint inhibitors. We also discuss findings from clinical trials incorporating radiotherapy and immune checkpoint inhibitors and examine the success of these trials in the context of the immunosuppressive effects of radiotherapy. We conclude by highlighting targets for relieving radiotherapy-induced immunosuppression with the goal of enhancing the combined effects of radiotherapy and immune checkpoint inhibitors.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Tumor Microenvironment , Humans , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/immunology , Neoplasms/radiotherapy , Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects , Animals , Radioimmunotherapy , Combined Modality Therapy
7.
J Am Chem Soc ; 146(1): 849-857, 2024 01 10.
Article in English | MEDLINE | ID: mdl-38134050

ABSTRACT

Phthalocyanine photosensitizers (PSs) have shown promise in fluorescence imaging and photodynamic therapy (PDT) of malignant tumors, but their practical application is limited by the aggregation-induced quenching (AIQ) and inherent photobleaching of PSs. Herein, we report the synthesis of a two-dimensional nanoscale covalent organic framework (nCOF) with staggered (AB) stacking of zinc-phthalocyanines (ZnPc), ZnPc-PI, for fluorescence imaging and mitochondria-targeted PDT. ZnPc-PI isolates and confines ZnPc PSs in the rigid nCOF to reduce AIQ, improve photostability, enhance cellular uptake, and increase the level of reactive oxygen species (ROS) generation via mitochondrial targeting. ZnPc-PI shows efficient tumor accumulation, which allowed precise tumor imaging and nanoparticle tracking. With high cellular uptake and tumor accumulation, intrinsic mitochondrial targeting, and enhanced ROS generation, ZnPc-PI exhibits potent PDT efficacy with >95% tumor growth inhibition on two murine colon cancer models without causing side effects.


Subject(s)
Metal-Organic Frameworks , Neoplasms , Organometallic Compounds , Photochemotherapy , Zinc Compounds , Mice , Humans , Animals , Photochemotherapy/methods , Metal-Organic Frameworks/therapeutic use , Reactive Oxygen Species , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Isoindoles , Neoplasms/drug therapy , Organometallic Compounds/pharmacology , Organometallic Compounds/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Mitochondria , Cell Line, Tumor
8.
J Am Chem Soc ; 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38837955

ABSTRACT

Covalent organic frameworks (COFs) have been explored for photodynamic therapy (PDT) of cancer, but their antitumor efficacy is limited by excited state quenching and low reactive oxygen species generation efficiency. Herein, we report a simultaneous protonation and metalation strategy to significantly enhance the PDT efficacy of a nanoscale two-dimensional imine-linked porphyrin-COF. The neutral and unmetalated porphyrin-COF (Ptp) and the protonated and metalated porphyrin-COF (Ptp-Fe) were synthesized via imine condensation between 5,10,15,20-tetrakis(4-aminophenyl)porphyrin and terephthalaldehyde in the absence and presence of ferric chloride, respectively. The presence of ferric chloride generated both doubly protonated and Fe3+-coordinated porphyrin units, which red-shifted and increased the Q-band absorption and disrupted exciton migration to prevent excited state quenching, respectively. Under light irradiation, rapid energy transfer from protonated porphyrins to Fe3+-coordinated porphyrins in Ptp-Fe enabled 1O2 and hydroxyl radical generation via type II and type I PDT processes. Ptp-Fe also catalyzed the conversion of hydrogen peroxide to hydroxy radical through a photoenhanced Fenton-like reaction under slightly acidic conditions and light illumination. As a result, Ptp-Fe-mediated PDT exhibited much higher cytotoxicity than Ptp-mediated PDT on CT26 and 4T1 cancer cells. Ptp-Fe-mediated PDT afforded potent antitumor efficacy in subcutaneous CT26 murine colon cancer and orthotopic 4T1 murine triple-negative breast tumors and prevented metastasis of 4T1 breast cancer to the lungs. This work underscores the role of fine-tuning the molecular structures of COFs in significantly enhancing their PDT efficacy.

9.
Angew Chem Int Ed Engl ; : e202412844, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39146242

ABSTRACT

Cholesterol is an essential membrane component, and the metabolites from cholesterol play important biological functions to intricately support cancer progression and dampen immune responses. Preclinical and clinical studies have demonstrated the role of cholesterol metabolism regulation on inhibiting tumor growth, remodeling the immunosuppressive tumor microenvironment (TME), and enhancing anti-tumor immunity. In this minireview, we discuss complex cholesterol metabolism in tumors, its important role in cancer progression, and its influences on immune cells in the TME. We provide an overview of recent advances in cancer treatment through regulating cholesterol metabolism. We discuss the design of cholesterol-altering multifunctional nanomaterials to regulate oxidative stress, modulate immune checkpoints, manipulate mechanical stress responses, and alter cholesterol metabolic pathways. Additionally, we examine the interactions between cholesterol metabolism regulation and established cancer treatments with the aim of identifying efficient strategies to disrupt cholesterol metabolism and synergistic combination therapies for effective cancer treatment.

10.
J Am Chem Soc ; 145(34): 18698-18704, 2023 08 30.
Article in English | MEDLINE | ID: mdl-37581644

ABSTRACT

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.


Subject(s)
Metal-Organic Frameworks , Neoplasms , Prodrugs , Radiation-Sensitizing Agents , Animals , Mice , Metal-Organic Frameworks/therapeutic use , Prodrugs/pharmacology , Prodrugs/therapeutic use , X-Rays , Reactive Oxygen Species , Neoplasms/drug therapy , Radiation-Sensitizing Agents/therapeutic use , Cell Line, Tumor
11.
Small ; 19(52): e2305440, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37635106

ABSTRACT

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.


Subject(s)
Metal-Organic Frameworks , Neoplasms , Humans , Metal-Organic Frameworks/pharmacology , Cholesterol Oxidase , Pyroptosis , T-Lymphocytes , Cholesterol , Tumor Microenvironment
12.
Immunity ; 41(5): 843-52, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25517616

ABSTRACT

Ionizing radiation-mediated tumor regression depends on type I interferon (IFN) and the adaptive immune response, but several pathways control I IFN induction. Here, we demonstrate that adaptor protein STING, but not MyD88, is required for type I IFN-dependent antitumor effects of radiation. In dendritic cells (DCs), STING was required for IFN-? induction in response to irradiated-tumor cells. The cytosolic DNA sensor cyclic GMP-AMP (cGAMP) synthase (cGAS) mediated sensing of irradiated-tumor cells in DCs. Moreover, STING was essential for radiation-induced adaptive immune responses, which relied on type I IFN signaling on DCs. Exogenous IFN-? treatment rescued the cross-priming by cGAS or STING-deficient DCs. Accordingly, activation of STING by a second messenger cGAMP administration enhanced antitumor immunity induced by radiation. Thus radiation-mediated antitumor immunity in immunogenic tumors requires a functional cytosolic DNA-sensing pathway and suggests that cGAMP treatment might provide a new strategy to improve radiotherapy.


Subject(s)
DNA/immunology , Membrane Proteins/genetics , Neoplasms/radiotherapy , Nucleotidyltransferases/immunology , Adaptive Immunity , Adaptor Proteins, Vesicular Transport/genetics , Animals , Antineoplastic Agents/pharmacology , Cells, Cultured , Cross-Priming/immunology , Dendritic Cells/immunology , Immunity, Innate , Interferon-beta/biosynthesis , Interferon-beta/immunology , Interferon-beta/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Neoplasms/immunology , Nucleotides, Cyclic/pharmacology , RNA Interference , RNA, Small Interfering , Radiation, Ionizing , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Signal Transduction/immunology , Xanthones/pharmacology
13.
Proc Natl Acad Sci U S A ; 117(36): 22423-22429, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32848073

ABSTRACT

Metastases are the cause of the vast majority of cancer deaths. In the metastatic process, cells migrate to the vasculature, intravasate, extravasate, and establish metastatic colonies. This pattern of spread requires the cancer cells to change shape and to navigate tissue barriers. Approaches that block this mechanical program represent new therapeutic avenues. We show that 4-hydroxyacetophenone (4-HAP) inhibits colon cancer cell adhesion, invasion, and migration in vitro and reduces the metastatic burden in an in vivo model of colon cancer metastasis to the liver. Treatment with 4-HAP activates nonmuscle myosin-2C (NM2C) (MYH14) to alter actin organization, inhibiting the mechanical program of metastasis. We identify NM2C as a specific therapeutic target. Pharmacological control of myosin isoforms is a promising approach to address metastatic disease, one that may be readily combined with other therapeutic strategies.


Subject(s)
Acetophenones/pharmacology , Actomyosin/metabolism , Cytoskeleton , Neoplasm Metastasis/physiopathology , Actins/metabolism , Animals , Cell Adhesion/drug effects , Cell Movement/drug effects , Colorectal Neoplasms/metabolism , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Female , HCT116 Cells , Humans , Mice , Mice, Nude
14.
Gut ; 71(3): 521-533, 2022 03.
Article in English | MEDLINE | ID: mdl-33685966

ABSTRACT

OBJECTIVE: Our goals were to evaluate the antitumour efficacy of Lactobacillus rhamnosus GG (LGG) in combination with immune checkpoint blockade (ICB) immunotherapies on tumour growth and to investigate the underlying mechanisms. DESIGN: We used murine models of colorectal cancer and melanoma to evaluate whether oral administration of LGG improves the efficacy of ICB therapies. We performed the whole genome shotgun metagenome sequencing of intestinal contents and RNA sequencing of dendritic cells (DCs). In a series of in vitro and in vivo experiments, we further defined the immunological and molecular mechanisms of LGG-mediated antitumour immunity. RESULTS: We demonstrate that oral administration of live LGG augmented the antitumour activity of anti-programmed cell death 1 (PD-1) immunotherapy by increasing tumour-infiltrating DCs and T cells. Moreover, the combination treatment shifted the gut microbial community towards enrichment in Lactobacillus murinus and Bacteroides uniformis, that are known to increase DC activation and CD8+tumour recruitment. Mechanistically, treatment with live LGG alone or in combination with anti-PD-1 antibody triggered type I interferon (IFN) production in DCs, enhancing the cross-priming of antitumour CD8+ T cells. In DCs, cyclic GMP-AMP synthase (cGAS)/stimulator of IFN genes (STING) was required for IFN-ß induction in response to LGG, as evidenced by the significant decrease in IFN-ß levels in cGAS or STING-deficient DCs. LGG induces IFN-ß production via the cGAS/STING/TANK binding kinase 1/interferon regulatory factor 7 axis in DCs. CONCLUSION: Our findings have offered valuable insight into the molecular mechanisms of live LGG-mediated antitumour immunity and establish an empirical basis for developing oral administration of live LGG as a combination agent with ICB for cancer therapies.


Subject(s)
Colorectal Neoplasms/therapy , Immune Checkpoint Inhibitors/therapeutic use , Lacticaseibacillus rhamnosus , Melanoma/therapy , Probiotics/therapeutic use , Administration, Oral , Animals , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Disease Models, Animal , Interferon Type I/metabolism , Melanoma/etiology , Melanoma/pathology , Mice
15.
Ann Surg Oncol ; 28(13): 8532-8543, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34091777

ABSTRACT

BACKGROUND: Pancreatic neuroendocrine tumors (PanNETs) are increasingly common. Experts debate whether small tumors should be resected. Tumor destruction via injection of cytotoxic agents could offer a minimal invasive approach to this controversy. We hypothesize that a new drug delivery system comprising chondroitin sulfate (CS) hydrogels loaded with sunitinib (SUN) suppresses tumor growth in PanNET cells. METHODS: Injectable hydrogels composed of CS modified with methacrylate groups (MA) were fabricated and loaded with SUN. Loading target was either 200 µg (SUN200-G) or 500 µg (SUN500-G) as well as sham hydrogel with no drug loading (SUN0-G). SUN release from hydrogels was monitored in vitro over time and cytotoxicity induced by the released SUN was evaluated using QGP-1 and BON1 PanNET cell lines. QGP-1 xenografts were developed in 35 mice and directly injected with 25 µL of either SUN200-G, SUN500-G, SUN0-G, 100 µL of Sunitinib Malate (SUN-inj), or given 40 mg/kg/day oral sunitinib (SUN-oral). RESULTS: SUN-loaded CSMA hydrogel retained complete in vitro cytotoxicity toward the QGP-1 PanNET and BON-1 PanNET cell lines for 21 days. Mouse xenograft models with QGP-1 PanNETs showed a significant delay in tumor growth in the SUN200/500-G, SUN-inj and SUN-oral groups compared with SUN0-G (p = 0.0014). SUN500-G hydrogels induced significantly more tumor necrosis than SUN0-G (p = 0.04). There was no difference in tumor growth delay between SUN200/500G, SUN-inj, and SUN-oral. CONCLUSIONS: This study demonstrates that CSMA hydrogels loaded with SUN suppress PanNETs growth. This drug delivery could approach represents a novel way to treat PanNETs and other neoplasms via intratumoral injection.


Subject(s)
Neuroendocrine Tumors , Pancreatic Neoplasms , Animals , Cell Line, Tumor , Chondroitin Sulfates/therapeutic use , Drug Delivery Systems , Hydrogels , Mice , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Sunitinib/therapeutic use
16.
BMC Biol ; 18(1): 32, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32209106

ABSTRACT

BACKGROUND: Nuclear factor-κB (NF-κB) plays a prominent role in promoting inflammation and resistance to DNA damaging therapy. We searched for proteins that modulate the NF-κB response as a prerequisite to identifying novel factors that affect sensitivity to DNA damaging chemotherapy. RESULTS: Using streptavidin-agarose pull-down, we identified the DExD/H-box RNA helicase, DDX39B, as a factor that differentially interacts with κB DNA probes. Subsequently, using both RNA interference and CRISPR/Cas9 technology, we demonstrated that DDX39B inhibits NF-κB activity by a general mechanism involving inhibition of p65 phosphorylation. Mechanistically, DDX39B mediates this effect by interacting with the pattern recognition receptor (PRR), LGP2, a pathway that required the cellular response to cytoplasmic double-stranded RNA (dsRNA). From a functional standpoint, loss of DDX39B promoted resistance to alkylating chemotherapy in glioblastoma cells. Further examination of DDX39B demonstrated that its protein abundance was regulated by site-specific sumoylation that promoted its poly-ubiquitination and degradation. These post-translational modifications required the presence of the SUMO E3 ligase, PIASx-ß. Finally, genome-wide analysis demonstrated that despite the link to the PRR system, DDX39B did not generally inhibit interferon-stimulated gene expression, but rather acted to attenuate expression of factors associated with the extracellular matrix, cellular migration, and angiogenesis. CONCLUSIONS: These results identify DDX39B, a factor with known functions in mRNA splicing and nuclear export, as an RNA-binding protein that blocks a subset of the inflammatory response. While these findings identify a pathway by which DDX39B promotes sensitization to DNA damaging therapy, the data also reveal a mechanism by which this helicase may act to mitigate autoimmune disease.


Subject(s)
DEAD-box RNA Helicases/genetics , NF-kappa B/metabolism , Receptors, Pattern Recognition/genetics , Signal Transduction , Alkylation , Animals , DEAD-box RNA Helicases/metabolism , DNA Probes , Drug Therapy , Humans , Mice , Receptors, Pattern Recognition/metabolism
17.
Int J Mol Sci ; 22(17)2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34502479

ABSTRACT

Radiotherapy and immunotherapy are most effective as cancer therapies in the setting of low-volume disease. Although initial studies of radio-immunotherapy in patients with metastatic cancer have not confirmed the efficacy of this approach, the role of radio-immunotherapy in patients with limited metastatic burden is unclear. We propose that further investigation of radio-immunotherapy in metastatic patients should focus upon patients with oligometastatic disease.


Subject(s)
Neoplasms/radiotherapy , Radioimmunotherapy , Humans , Neoplasm Metastasis , Neoplasms/immunology , Neoplasms/pathology
18.
Carcinogenesis ; 41(11): 1605-1615, 2020 11 13.
Article in English | MEDLINE | ID: mdl-32221533

ABSTRACT

Human papillomavirus (HPV) infection is necessary but insufficient for progression of epithelial cells from dysplasia to carcinoma-in situ (CIS) to invasive cancer. The combination of mutant cellular and viral oncogenes that regulate progression of cervical cancer (CC) remains unclear. Using combinations of HPV16 E6/E7 (E+), mutant Kras (mKras) (K+) and/or loss of Pten (P-/-), we generated autochthonous models of CC without exogenous estrogen, carcinogen or promoters. Furthermore, intravaginal instillation of adenoCre virus enabled focal activation of the oncogenes/inactivation of the tumor suppressor gene. In P+/+ mice, E6/E7 alone (P+/+E+K-) failed to cause premalignant changes, while mKras alone (P+/+E-K+) caused persistent mucosal abnormalities in about one-third of mice, but no cancers. To develop cancer, P+/+ mice needed both E6/E7 and mKras expression. Longitudinal endoscopies of P+/+E+K+ mice predicted carcinoma development by detection of mucosal lesions, found on an average of 23 weeks prior to death, unlike longitudinal quantitative PCRs of vaginal lavage samples from the same mice. Endoscopy revealed that individual mice differed widely in the time required for mucosal lesions to appear after adenoCre and in the time required for these lesions to progress to cancer. These cancers developed in the transition zone that extends, unlike in women, from the murine cervix to the distal vagina. The P-/-E+K+ genotype led to precipitous cancer development within a few weeks and E6/E7-independent cancer development occurred in the P-/-E-K+ genotype. In the P-/-E+K- genotype, mice only developed CIS. Thus, distinct combinations of viral and cellular oncogenes are involved in distinct steps in cervical carcinogenesis.


Subject(s)
Carcinogens/toxicity , Endoscopy/methods , Estrogens/toxicity , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Repressor Proteins/metabolism , Uterine Cervical Neoplasms/pathology , Vaginal Neoplasms/pathology , Animals , Carcinogenesis , Female , Human papillomavirus 16/isolation & purification , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , PTEN Phosphohydrolase/physiology , Papillomavirus Infections/complications , Papillomavirus Infections/virology , Proto-Oncogene Proteins p21(ras)/genetics , Uterine Cervical Neoplasms/diagnostic imaging , Uterine Cervical Neoplasms/etiology , Uterine Cervical Neoplasms/metabolism , Vaginal Neoplasms/diagnostic imaging , Vaginal Neoplasms/etiology , Vaginal Neoplasms/metabolism
19.
Lancet Oncol ; 20(8): e434-e442, 2019 08.
Article in English | MEDLINE | ID: mdl-31364595

ABSTRACT

Metastasis is the leading cause of cancer-related mortality and remains one of the prevailing challenges in cancer treatment. Most patients with metastatic disease are treated with systemic agents, which prolong survival and improve symptoms but are typically not curative. The oligometastatic hypothesis challenges the perspective that metastasis is an invariably disseminated process, and proposes a biological spectrum of metastatic virulence. Mounting evidence supports the idea that patients with numerically and spatially restricted sites of metastases, termed oligometastases, can achieve prolonged survival following metastasis-directed therapies, such as surgery or radiotherapy. Improvements in clinical and molecular staging of metastatic disease, as well as integration of effective systemic therapies with localised interventions, might achieve better outcomes for patients with diverse metastatic states. In this Series paper, we propose a rationale for the integration of immune checkpoint inhibitors with radiotherapy to advance the potential for effective treatment along the spectrum of disease, with emphasis on how immunotherapy can potentiate radiotherapy treatment in the oligometastatic setting.


Subject(s)
Immunotherapy/methods , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/radiotherapy , Radiotherapy/methods , Animals , Combined Modality Therapy/methods , Humans
20.
Cancer ; 125(3): 340-352, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30521067

ABSTRACT

The term "oligometastatic prostate cancer" refers to a heterogeneous group of disease states currently defined solely on the basis of clinical features. Oligorecurrent disease, de novo oligometastases, and oligoprogressive disease likely have unique biologic underpinnings and natural histories. Evidence suggesting the existence of a subset of patients who harbor prostate cancer with limited metastatic potential currently includes disparate and overwhelmingly retrospective reports. Nevertheless, emerging prospective data have corroborated the "better-than-expected," retrospectively observed outcomes, particularly in the setting of oligorecurrent prostate cancer. Improved functional imaging with prostate-specific membrane antigen-targeted strategies may enhance the identification of patients with oligometastatic prostate cancer in the short term. In the long term, refinement of the oligometastatic case definition likely will require biologic risk-stratification schemes. To determine optimal treatment strategies and identify patients most likely to benefit from metastasis-directed therapy, future efforts should focus on conducting high-quality, prospective trials with much-needed molecular correlative studies.


Subject(s)
Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/pathology , Humans , Male , Neoplasm Metastasis , Prognosis , Prostatic Neoplasms/classification , Prostatic Neoplasms/therapy , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL