1
|
Mampay M, Al‐Hity G, Rolle SO, Alzboon W, Stewart NA, Flint MS, Sheridan GK. Impact of Psychological Stress and Spontaneous Tumour Regression on the Hippocampal Proteome in a Mouse Model of Breast Cancer. J Neurochem 2025; 169:e70052. [PMID: 40172096 PMCID: PMC11963485 DOI: 10.1111/jnc.70052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/03/2025] [Accepted: 03/17/2025] [Indexed: 04/04/2025]
Abstract
Cognitive impairment is common in people diagnosed with breast cancer, but the molecular mechanisms that underlie maladaptive changes in the brain are unknown. The psychological stress of a cancer diagnosis is certainly a contributing factor. Here, we investigated alterations in the hippocampal proteome in response to both cancer and psychological stress using label-free quantitative mass spectrometry techniques. An orthotopic syngeneic model of triple-negative breast cancer (TNBC) was established by injecting Py230 cells into the mammary fat pads of female C57Bl/6 mice. Half of the mice were subjected to a daily restraint stress paradigm. Mice that experienced both cancer and restraint stress lost weight and displayed larger tumours compared to non-stressed mice. Their urinary corticosterone levels were also elevated, as measured by enzyme-linked immunosorbent assay. Non-stressed tumour-bearing mice displayed higher levels of TNFα in the prefrontal cortex (PFC) compared to stressed mice with cancer. Flow cytometry results suggested that the CD4+/CD8+ T cell ratios were also raised in non-stressed tumour-bearing mice compared to both controls and stressed mice with TNBC. Bioinformatic analysis of hippocampal proteomes indicated that cancer alone causes reduced mitochondrial respiration and ATP synthesis, as well as impaired glutamate recycling and synaptic plasticity. Moreover, daily stress in TNBC mice caused further mitochondrial dysfunction, increased oxidative phosphorylation, and altered lipid metabolism. Importantly, over half of the mammary tumours that initially developed spontaneously regressed after 7-9 weeks in these young immunocompetent mice. Tumour regression inhibited TNFα increases in the PFC. However, the hippocampal proteomes of tumour-bearing mice were largely similar to mice in which tumours regressed, suggesting that spontaneous regression of breast cancer confers lasting physiological dysregulations that impact hippocampal protein expression. This study in mice may help to identify molecular mechanisms responsible for long-term memory impairments in cancer survivors and reveal novel drug targets for cancer-related cognitive impairment.
Collapse
Affiliation(s)
- Myrthe Mampay
- School of Applied SciencesUniversity of BrightonBrightonUK
| | - Gheed Al‐Hity
- School of Applied SciencesUniversity of BrightonBrightonUK
| | | | - Walla Alzboon
- School of Life SciencesUniversity of NottinghamNottinghamUK
| | | | | | | |
Collapse
|
2
|
Rasooli Tehrani R, Asgarian-Omran H, Taghiloo S, Valadan R, Azizi S, Ajami A. Infiltration of innate and adoptive lymphoid cells in 4T1 and MC4-L2 breast cancer models. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2025; 28:63-71. [PMID: 39877637 PMCID: PMC11771339 DOI: 10.22038/ijbms.2024.80535.17434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 08/28/2024] [Indexed: 01/31/2025]
Abstract
Objectives Innate lymphoid cells (ILCs) are tissue-resident lymphocytes that have vital roles in activating further immune responses. However, due to their tumor-induced diversity, we decided to examine ILCs, T cells, and the associated cytokines in mouse models of breast cancer. Materials and Methods 4T1 and MC4-L2 cells were used to induce triple-negative and hormone-receptor-positive breast cancer, respectively. Tumor tissue was resected at early and late stages of tumor growth and used for further analysis. Total RNA was extracted and used in Real-Time PCR to analyze the expression of IFN-γ, IL-4, IL-10, IL-13, and IL-22. Tumor tissue was digested and used in a flow cytometric assay. H&E staining was used to examine the pathology of tumor progression. Results Both tumor models showed a notable increase in T-cell frequency at the early stage of tumor growth. However, as the tumors progressed, the frequency of T cells significantly decreased, while the ILC component exhibited a significant increase in tumor progression. Gene analysis indicated a significant increase in the inflammatory to anti-inflammatory cytokine ratio during tumor progression in the tumor model. In contrast, this ratio was considerably reduced in advanced MC4-L2 tumors. Both tumor models showed the development of invasive breast carcinoma and lung metastasis in advanced tumors. Conclusion Our study highlighted the expansion of ILCs during tumor progression in two distinct breast cancer models with different immunogenicity. These findings suggest that ILCs may actively modulate the tumor microenvironment during the advanced stage of tumor growth.
Collapse
Affiliation(s)
- Reihane Rasooli Tehrani
- Department of Medical Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hossein Asgarian-Omran
- Department of Medical Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Gastrointestinal Cancer Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saeid Taghiloo
- Department of Medical Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Reza Valadan
- Department of Medical Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Molecular and Cell Biology Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Soheil Azizi
- Department of Laboratory Sciences, Faculty of Paramedicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abolghasem Ajami
- Department of Medical Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Molecular and Cell Biology Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
3
|
Franzolin G, Brundu S, Cojocaru CF, Curatolo A, Ponzo M, Mastrantonio R, Mihara E, Kumanogoh A, Suga H, Takagi J, Tamagnone L, Giraudo E. PlexinB1 Inactivation Reprograms Immune Cells in the Tumor Microenvironment, Inhibiting Breast Cancer Growth and Metastatic Dissemination. Cancer Immunol Res 2024; 12:1286-1301. [PMID: 38874583 PMCID: PMC11369622 DOI: 10.1158/2326-6066.cir-23-0289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 03/15/2024] [Accepted: 06/12/2024] [Indexed: 06/15/2024]
Abstract
Semaphorin-plexin signaling plays a major role in the tumor microenvironment (TME). In particular, Semaphorin 4D (SEMA4D) has been shown to promote tumor growth and metastasis; however, the role of its high-affinity receptor Plexin-B1 (PLXNB1), which is expressed in the TME, is poorly understood. In this study, we directly targeted PLXNB1 in the TME of triple-negative murine breast carcinoma to elucidate its relevance in cancer progression. We found that primary tumor growth and metastatic dissemination were strongly reduced in PLXNB1-deficient mice, which showed longer survival. PLXNB1 loss in the TME induced a switch in the polarization of tumor-associated macrophages (TAM) toward a pro-inflammatory M1 phenotype and enhanced the infiltration of CD8+ T lymphocytes both in primary tumors and in distant metastases. Moreover, PLXNB1 deficiency promoted a shift in the Th1/Th2 balance of the T-cell population and an antitumor gene signature, with the upregulation of Icos, Perforin-1, Stat3, and Ccl5 in tumor-infiltrating lymphocytes (TILs). We thus tested the translational relevance of TME reprogramming driven by PLXNB1 inactivation for responsiveness to immunotherapy. Indeed, in the absence of PLXNB1, the efficacy of anti-PD-1 blockade was strongly enhanced, efficiently reducing tumor growth and distant metastasis. Consistent with this, pharmacological PLXNB1 blockade by systemic treatment with a specific inhibitor significantly hampered breast cancer growth and enhanced the antitumor activity of the anti-PD-1 treatment in a preclinical model. Altogether, these data indicate that PLXNB1 signaling controls the antitumor immune response in the TME and highlight this receptor as a promising immune therapeutic target for metastatic breast cancers.
Collapse
Affiliation(s)
- Giulia Franzolin
- Laboratory of Tumor Microenvironment, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
- Department of Science and Drug Technology, University of Torino, Torino, Italy.
| | - Serena Brundu
- Laboratory of Tumor Microenvironment, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
- Department of Science and Drug Technology, University of Torino, Torino, Italy.
| | - Carina F. Cojocaru
- Laboratory of Tumor Microenvironment, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
- Department of Science and Drug Technology, University of Torino, Torino, Italy.
| | - Aurora Curatolo
- Laboratory of Tumor Microenvironment, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
- Department of Science and Drug Technology, University of Torino, Torino, Italy.
| | - Matteo Ponzo
- Laboratory of Tumor Microenvironment, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
| | - Roberta Mastrantonio
- Department Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy.
- Fondazione Policlinico Gemelli–IRCCS, Rome, Italy.
| | - Emiko Mihara
- Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka, Japan.
| | - Atsushi Kumanogoh
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Department of Respiratory Medicine and Clinical Immunology, Osaka University, Osaka, Japan.
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan.
| | - Junichi Takagi
- Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka, Japan.
| | - Luca Tamagnone
- Department Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy.
- Fondazione Policlinico Gemelli–IRCCS, Rome, Italy.
| | - Enrico Giraudo
- Laboratory of Tumor Microenvironment, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
- Department of Science and Drug Technology, University of Torino, Torino, Italy.
| |
Collapse
|
4
|
Nanajian A, Scott M, Burcus NI, Ruedlinger BL, Oshin EA, Beebe SJ, Guo S. Nano-Pulse Treatment Overcomes the Immunosuppressive Tumor Microenvironment to Elicit In Situ Vaccination Protection against Breast Cancer. Vaccines (Basel) 2024; 12:633. [PMID: 38932362 PMCID: PMC11209453 DOI: 10.3390/vaccines12060633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
We previously reported that nano-pulse treatment (NPT), a pulsed power technology, resulted in 4T1-luc mammary tumor elimination and a strong in situ vaccination, thereby completely protecting tumor-free animals against a second live tumor challenge. The mechanism whereby NPT mounts effective antitumor immune responses in the 4T1 breast cancer predominantly immunosuppressive tumor microenvironment (TME) remains unanswered. In this study, orthotopic 4T1 mouse breast tumors were treated with NPT (100 ns, 50 kV/cm, 1000 pulses, 3 Hz). Blood, spleen, draining lymph nodes, and tumors were harvested at 4-h, 8-h, 1-day, 3-day, 7-day, and 3-month post-treatment intervals for the analysis of frequencies, death, and functional markers of various immune cells in addition to the suppressor function of regulatory T cells (Tregs). NPT was verified to elicit strong in situ vaccination (ISV) against breast cancer and promote both acute and long-term T cell memory. NPT abolished immunosuppressive dominance systemically and in the TME by substantially reducing Tregs, myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs). NPT induced apoptosis in Tregs and TAMs. It also functionally diminished the Treg suppression capacity, explained by the downregulation of activation markers, particularly 4-1BB and TGFβ, and a phenotypic shift from predominantly activated (CD44+CD62L-) to naïve (CD44-CD62L+) Tregs. Importantly, NPT selectively induced apoptosis in activated Tregs and spared effector CD4+ and CD8+ T cells. These changes were followed by a concomitant rise in CD8+CD103+ tissue-resident memory T cells and TAM M1 polarization. These findings indicate that NPT effectively switches the TME and secondary lymphatic systems from an immunosuppressive to an immunostimulatory state, allowing cytotoxic T cell function and immune memory formation to eliminate cancer cells and account for the NPT in situ vaccination.
Collapse
Affiliation(s)
- Anthony Nanajian
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA; (A.N.); (M.S.); (N.I.B.); (B.L.R.); (E.A.O.); (S.J.B.)
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Megan Scott
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA; (A.N.); (M.S.); (N.I.B.); (B.L.R.); (E.A.O.); (S.J.B.)
| | - Niculina I. Burcus
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA; (A.N.); (M.S.); (N.I.B.); (B.L.R.); (E.A.O.); (S.J.B.)
| | - Brittney L. Ruedlinger
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA; (A.N.); (M.S.); (N.I.B.); (B.L.R.); (E.A.O.); (S.J.B.)
| | - Edwin A. Oshin
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA; (A.N.); (M.S.); (N.I.B.); (B.L.R.); (E.A.O.); (S.J.B.)
- Department of Electrical & Computer Engineering, Old Dominion University, Norfolk, VA 23529, USA
| | - Stephen J. Beebe
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA; (A.N.); (M.S.); (N.I.B.); (B.L.R.); (E.A.O.); (S.J.B.)
| | - Siqi Guo
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA; (A.N.); (M.S.); (N.I.B.); (B.L.R.); (E.A.O.); (S.J.B.)
| |
Collapse
|
5
|
Salembier R, De Haes C, Bellemans J, Demeyere K, Van Den Broeck W, Sanders NN, Van Laere S, Lyons TR, Meyer E, Steenbrugge J. Chitin-mediated blockade of chitinase-like proteins reduces tumor immunosuppression, inhibits lymphatic metastasis and enhances anti-PD-1 efficacy in complementary TNBC models. Breast Cancer Res 2024; 26:63. [PMID: 38605414 PMCID: PMC11007917 DOI: 10.1186/s13058-024-01815-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 03/23/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Chitinase-like proteins (CLPs) play a key role in immunosuppression under inflammatory conditions such as cancer. CLPs are enzymatically inactive and become neutralized upon binding of their natural ligand chitin, potentially reducing CLP-driven immunosuppression. We investigated the efficacy of chitin treatment in the context of triple-negative breast cancer (TNBC) using complementary mouse models. We also evaluated the immunomodulatory influence of chitin on immune checkpoint blockade (ICB) and compared its efficacy as general CLP blocker with blockade of a single CLP, i.e. chitinase 3-like 1 (CHI3L1). METHODS Female BALB/c mice were intraductally injected with luciferase-expressing 4T1 or 66cl4 cells and systemically treated with chitin in combination with or without anti-programmed death (PD)-1 ICB. For single CLP blockade, tumor-bearing mice were treated with anti-CHI3L1 antibodies. Metastatic progression was monitored through bioluminescence imaging. Immune cell changes in primary tumors and lymphoid organs (i.e. axillary lymph nodes and spleen) were investigated through flow cytometry, immunohistochemistry, cytokine profiling and RNA-sequencing. CHI3L1-stimulated RAW264.7 macrophages were subjected to 2D lymphatic endothelial cell adhesion and 3D lymphatic integration in vitro assays for studying macrophage-mediated lymphatic remodeling. RESULTS Chitin significantly reduced primary tumor progression in the 4T1-based model by decreasing the high production of CLPs that originate from tumor-associated neutrophils (TANs) and Stat3 signaling, prominently affecting the CHI3L1 and CHI3L3 primary tumor levels. It reduced immunosuppressive cell types and increased anti-tumorigenic T-cells in primary tumors as well as axillary lymph nodes. Chitin also significantly reduced CHI3L3 primary tumor levels and immunosuppression in the 66cl4-based model. Compared to anti-CHI3L1, chitin enhanced primary tumor growth reduction and anti-tumorigenicity. Both treatments equally inhibited lymphatic adhesion and integration of macrophages, thereby hampering lymphatic tumor cell spreading. Upon ICB combination therapy, chitin alleviated anti-PD-1 resistance in both TNBC models, providing a significant add-on reduction in primary tumor and lung metastatic growth compared to chitin monotherapy. These add-on effects occurred through additional increase in CD8α+ T-cell infiltration and activation in primary tumor and lymphoid organs. CONCLUSIONS Chitin, as a general CLP blocker, reduces CLP production, enhances anti-tumor immunity as well as ICB responses, supporting its potential clinical relevance in immunosuppressed TNBC patients.
Collapse
Affiliation(s)
- Robbe Salembier
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Caro De Haes
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Julie Bellemans
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Kristel Demeyere
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Wim Van Den Broeck
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Niek N Sanders
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Steven Van Laere
- Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Traci R Lyons
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, CO, USA
| | - Evelyne Meyer
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Jonas Steenbrugge
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
- Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium.
| |
Collapse
|
6
|
Krishnamohan M, Kaplanov I, Maudi-Boker S, Yousef M, Machluf-Katz N, Cohen I, Elkabets M, Titus J, Bersudsky M, Apte RN, Voronov E, Braiman A. Tumor Cell-Associated IL-1α Affects Breast Cancer Progression and Metastasis in Mice through Manipulation of the Tumor Immune Microenvironment. Int J Mol Sci 2024; 25:3950. [PMID: 38612760 PMCID: PMC11011794 DOI: 10.3390/ijms25073950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/25/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024] Open
Abstract
IL-1α is a dual function cytokine that affects inflammatory and immune responses and plays a pivotal role in cancer. The effects of intracellular IL-1α on the development of triple negative breast cancer (TNBC) in mice were assessed using the CRISPR/Cas9 system to suppress IL-1α expression in 4T1 breast cancer cells. Knockout of IL-1α in 4T1 cells modified expression of multiple genes, including downregulation of cytokines and chemokines involved in the recruitment of tumor-associated pro-inflammatory cells. Orthotopical injection of IL-1α knockout (KO) 4T1 cells into BALB/c mice led to a significant decrease in local tumor growth and lung metastases, compared to injection of wild-type 4T1 (4T1/WT) cells. Neutrophils and myeloid-derived suppressor cells were abundant in tumors developing after injection of 4T1/WT cells, whereas more antigen-presenting cells were observed in the tumor microenvironment after injection of IL-1α KO 4T1 cells. This switch correlated with increased infiltration of CD3+CD8+ and NKp46+cells. Engraftment of IL-1α knockout 4T1 cells into immunodeficient NOD.SCID mice resulted in more rapid tumor growth, with increased lung metastasis in comparison to engraftment of 4T1/WT cells. Our results suggest that tumor-associated IL-1α is involved in TNBC progression in mice by modulating the interplay between immunosuppressive pro-inflammatory cells vs. antigen-presenting and cytotoxic cells.
Collapse
Affiliation(s)
- Mathumathi Krishnamohan
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Irena Kaplanov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Sapir Maudi-Boker
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Muhammad Yousef
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Noy Machluf-Katz
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Idan Cohen
- Cancer Center, Emek Medical Center, Afula 18101, Israel;
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Jaison Titus
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Marina Bersudsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Ron N. Apte
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Elena Voronov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| | - Alex Braiman
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (M.K.); (M.E.); (J.T.); (M.B.)
| |
Collapse
|
7
|
Zhou M, Liang S, Liu D, Ma K, Yun K, Yao J, Peng Y, Hai L, Zhang Q, Wang Z. Manganese-Enriched Zinc Peroxide Functional Nanoparticles for Potentiating Cancer Immunotherapy. NANO LETTERS 2023; 23:10350-10359. [PMID: 37930173 DOI: 10.1021/acs.nanolett.3c02941] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Immunotherapies have shown high clinical success, however, the therapeutical efficacy is largely restrained by insufficient immune activation and an immunosuppressive microenvironment. Herein, we report tumor microenvironment (TME)-responsive manganese-enriched zinc peroxide nanoparticles (MONPs) for synergistic cancer immunotherapy by inducing the immunogenic death (ICD) of cancer cells and activating the stimulator of the interferon gene (STING) pathway. MONPs especially disassociate upon exposure to acidic tumor tissue and in situ generate •OH for the ICD effect. Moreover, Mn2+ activated the STING and synergistically induced the secretion of type I interferon and inflammatory cytokines for specific T cell responses. Meanwhile, MONPs relieved the immunosuppression of TME through decreasing Tregs and polarizing M2 macrophages to the M1 type to unleash a cascade adaptive immune response. In combination with the anti-PD-1 antibody, MONPs showed superior efficacy in inhibiting tumor growth and preventing lung metastasis. Our study demonstrates the feasibility of functional nanoparticles to amplify STING innate stimulation, showing a prominent strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Mengli Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Shuang Liang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Dan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Kongshuo Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Kaiqing Yun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jianjun Yao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuxuan Peng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Linna Hai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Qiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhaohui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
8
|
Steenbrugge J, Pauwelyn G, Demeyere K, Devriendt N, de Rooster H, Sanders NN, Spaas JH, Meyer E. Xenogeneic equine stem cells activate anti-tumor adaptive immunity in a 4T1-based intraductal mouse model for triple-negative breast cancer: proof-of-principle. Front Immunol 2023; 14:1252374. [PMID: 37928528 PMCID: PMC10623058 DOI: 10.3389/fimmu.2023.1252374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/03/2023] [Indexed: 11/07/2023] Open
Abstract
Triple-negative breast cancer (TNBC) remains difficult to treat, especially due to ineffective immune responses. Current treatments mainly aim at a cytotoxic effect, whereas (stem) cell therapies are being investigated for their immune stimulatory capacities to initiate the anti-tumor immunity. Here, a thoroughly characterized, homogenous and non-tumorigenic mixture of equine mesenchymal stem cells (eMSCs) harvested from horse peripheral blood as innovative xenogeneic immunomodulators were tested in a 4T1-based intraductal mouse model for TNBC. The eMSCs significantly reduced 4T1 progression upon systemic injection, with induction of inflammatory mediators and T-cell influx in primary tumors, already after a single dose. These xenogeneic anti-cancer effects were not restricted to MSCs as systemic treatment with alternative equine epithelial stem cells (eEpSCs) mimicked the reported disease reduction. Mechanistically, effective eMSC treatment did not rely on the spleen as systemic entrapment site, whereas CD4+ and CD8α+ T-cell infiltration and activation were critical. These results show that eMSCs and potentially also other equine stem cell types can be a valuable TNBC treatment strategy for further (pre)clinical evaluation.
Collapse
Affiliation(s)
- Jonas Steenbrugge
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Glenn Pauwelyn
- Boehringer Ingelheim Veterinary Medicine Belgium, Evergem, Belgium
| | - Kristel Demeyere
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Nausikaa Devriendt
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Hilde de Rooster
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Niek N. Sanders
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Jan H. Spaas
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Boehringer-Ingelheim Animal Health USA, Athens, GA, United States
| | - Evelyne Meyer
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
9
|
Morin SM, Gregory KJ, Medeiros B, Terefe T, Hoshyar R, Alhusseiny A, Chen S, Schwartz RC, Jerry DJ, Vandenberg LN, Schneider SS. Benzophenone-3 exposure alters composition of tumor infiltrating immune cells and increases lung seeding of 4T1 breast cancer cells. ADVANCES IN CANCER BIOLOGY - METASTASIS 2023; 7:100080. [PMID: 37593105 PMCID: PMC10434833 DOI: 10.1016/j.adcanc.2022.100080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
Environmental chemicals are a persistent and pervasive part of everyday life. A subset of environmental chemicals are xenoestrogens, compounds that bind to the estrogen receptor (ER) and drive estrogen-related processes. One such chemical, benzophenone-3 (BP3), is a common chemical in sunscreen. It is a potent UV protectant but also is quickly absorbed through the skin. While it has been approved by the FDA, there is a renewed interest in the safety of BP3, particularly in relation to breast cancer. The focus of this study was to examine the impact that BP3 has on triple negative breast cancer (TNBC) through alterations to cells in the immune microenvironment. In this study, we exposed female mice to one of two doses of BP3 before injecting them with a TNBC cell line. Several immune endpoints were examined both in the primary tissues and from in vitro studies of T cell behavior. Our studies revealed that in the lung tumor microenvironment, exposure to BP3 not only increased the number of metastases, but also the total area of tumor coverage. We also found that BP3 caused alterations in immune populations in a tissue-dependent manner, particularly in T cells. Taken together, our data suggest that while BP3 may not directly affect the proliferation of TNBC, growth and metastasis of TNBC-derived tumors can be altered by BP3 exposures via the alterations in the immune populations of the tumor microenvironment.
Collapse
Affiliation(s)
- Stephanie M. Morin
- Pioneer Valley Life Sciences Institute, Springfield, MA, 01199, USA
- Dept of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Kelly J. Gregory
- Pioneer Valley Life Sciences Institute, Springfield, MA, 01199, USA
| | - Brenda Medeiros
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Amherst, 01003, USA
| | - Tigist Terefe
- Pioneer Valley Life Sciences Institute, Springfield, MA, 01199, USA
| | - Reyhane Hoshyar
- Breast Cancer and the Environment Research Program, Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Ahmed Alhusseiny
- University of Massachusetts Chan Medical School-Baystate, Department of Pathology, Springfield, MA, 01199, USA
| | - Shiuan Chen
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Richard C. Schwartz
- Breast Cancer and the Environment Research Program, Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - D. Joseph Jerry
- Dept of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Laura N. Vandenberg
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Amherst, 01003, USA
| | - Sallie S. Schneider
- Pioneer Valley Life Sciences Institute, Springfield, MA, 01199, USA
- Dept of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA
- University of Massachusetts Chan Medical School-Baystate, Department of Surgery, Springfield, MA, 01199, USA
| |
Collapse
|
10
|
Bernhardt SM, Mitchell E, Stamnes S, Hoffmann RJ, Calhoun A, Klug A, Russell TD, Pennock ND, Walker JM, Schedin P. Isogenic Mammary Models of Intraductal Carcinoma Reveal Progression to Invasiveness in the Absence of a Non-Obligatory In Situ Stage. Cancers (Basel) 2023; 15:2257. [PMID: 37190184 PMCID: PMC10136757 DOI: 10.3390/cancers15082257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/04/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
In breast cancer, progression to invasive ductal carcinoma (IDC) involves interactions between immune, myoepithelial, and tumor cells. Development of IDC can proceed through ductal carcinoma in situ (DCIS), a non-obligate, non-invasive stage, or IDC can develop without evidence of DCIS and these cases associate with poorer prognosis. Tractable, immune-competent mouse models are needed to help delineate distinct mechanisms of local tumor cell invasion and prognostic implications. To address these gaps, we delivered murine mammary carcinoma cell lines directly into the main mammary lactiferous duct of immune-competent mice. Using two strains of immune-competent mice (BALB/c, C57BL/6), one immune-compromised (severe combined immunodeficiency; SCID) C57BL/6 strain, and six different murine mammary cancer cell lines (D2.OR, D2A1, 4T1, EMT6, EO771, Py230), we found early loss of ductal myoepithelial cell differentiation markers p63, α-smooth muscle actin, and calponin, and rapid formation of IDC in the absence of DCIS. Rapid IDC formation also occurred in the absence of adaptive immunity. Combined, these studies demonstrate that loss of myoepithelial barrier function does not require an intact immune system, and suggest that these isogenic murine models may prove a useful tool to study IDC in the absence of a non-obligatory DCIS stage-an under-investigated subset of poor prognostic human breast cancer.
Collapse
Affiliation(s)
- Sarah M. Bernhardt
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Elizabeth Mitchell
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Stephanie Stamnes
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Reuben J. Hoffmann
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Andrea Calhoun
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Alex Klug
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Tanya D. Russell
- Center for Advancing Professional Excellence, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nathan D. Pennock
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Joshua M. Walker
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Pepper Schedin
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Young Women’s Breast Cancer Translational Program, University of Colorado Cancer Center, Aurora, CO 80045, USA
| |
Collapse
|
11
|
Steenbrugge J, Bellemans J, Vander Elst N, Demeyere K, De Vliegher J, Perera T, De Wever O, Van Den Broeck W, De Spiegelaere W, Sanders NN, Meyer E. One cisplatin dose provides durable stimulation of anti-tumor immunity and alleviates anti-PD-1 resistance in an intraductal model for triple-negative breast cancer. Oncoimmunology 2022; 11:2103277. [PMID: 35898705 PMCID: PMC9311321 DOI: 10.1080/2162402x.2022.2103277] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Aggressive triple-negative breast cancer (TNBC) is classically treated with chemotherapy. Besides direct tumor cell killing, some chemotherapeutics such as cisplatin provide additional disease reduction through stimulation of anti-tumor immunity. The cisplatin-induced immunomodulation in TNBC was here investigated in-depth using immunocompetent intraductal mouse models. Upon primary tumor transition to invasive carcinoma, cisplatin was injected systemically and significantly reduced tumor progression. Flow cytometric immunophenotyping was corroborated by immunohistochemical analyses and revealed both differential immune cell compositions and positivity for their programmed death (PD)-1 and PD-ligand (L)1 markers across body compartments, including the primary tumor, axillary lymph nodes and spleen. As key findings, a significant decrease in immunosuppressive and a concomitant increase in anti-tumor lymphocytic cell numbers were observed in the axillary lymph nodes and spleen, highlighting their importance in cisplatin-stimulated anti-tumor immunity. These immunomodulatory effects were already established following the first cisplatin dose, indicating that early cisplatin-mediated events may determine (immuno)therapeutic outcome. Furthermore, a single cisplatin dose sufficed to alleviate anti-PD-1 resistance in a 4T1-based model, providing add-on disease reduction without toxic side effects as seen upon multiple cisplatin dosing. Overall, these results highlight cisplatin as immunotherapeutic ally in TNBC, providing durable immunostimulation, even after a single dose.
Collapse
Affiliation(s)
- Jonas Steenbrugge
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Julie Bellemans
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Niels Vander Elst
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Kristel Demeyere
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Josephine De Vliegher
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | | | - Olivier De Wever
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Wim Van Den Broeck
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Ward De Spiegelaere
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Niek N. Sanders
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Evelyne Meyer
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
12
|
Yang Y, Cao Y. The impact of VEGF on cancer metastasis and systemic disease. Semin Cancer Biol 2022; 86:251-261. [PMID: 35307547 DOI: 10.1016/j.semcancer.2022.03.011] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 01/27/2023]
Abstract
Metastasis is the leading cause of cancer-associated mortality and the underlying mechanisms of cancer metastasis remain elusive. Both blood and lymphatic vasculatures are essential structures for mediating distal metastasis. The vasculature plays multiple functions, including accelerating tumor growth, sustaining the tumor microenvironment, supplying growth and invasive signals, promoting metastasis, and causing cancer-associated systemic disease. VEGF is one of the key angiogenic factors in tumors and participates in the initial stage of tumor development, progression and metastasis. Consequently, VEGF and its receptor-mediated signaling pathways have become one of the most important therapeutic targets for treating various cancers. Today, anti-VEGF-based antiangiogenic drugs (AADs) are widely used in the clinic for treating different types of cancer in human patients. Despite nearly 20-year clinical experience with AADs, the impact of these drugs on cancer metastasis and systemic disease remains largely unknown. In this review article, we focus our discussion on tumor VEGF in cancer metastasis and systemic disease and mechanisms underlying AADs in clinical benefits.
Collapse
Affiliation(s)
- Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Biomedicum, Karolinska Institute, 171 77 Stockholm, Sweden.
| |
Collapse
|
13
|
Wu Y, Li X, Li Q, Cheng C, Zheng L. Adipose tissue-to-breast cancer crosstalk: Comprehensive insights. Biochim Biophys Acta Rev Cancer 2022; 1877:188800. [PMID: 36103907 DOI: 10.1016/j.bbcan.2022.188800] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/29/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022]
Abstract
The review focuses on mechanistic evidence for the link between obesity and breast cancer. According to the IARC study, there is sufficient evidence that obesity is closely related to a variety of cancers. Among them, breast cancer is particularly disturbed by adipose tissue due to the unique histological structure of the breast. The review introduces the relationship between obesity and breast cancer from two aspects, including factors that promote tumorigenesis or metastasis. We summarize alterations in adipokines and metabolic pathways that contribute to breast cancer development. Breast cancer metastasis is closely related to obesity-induced pro-inflammatory microenvironment, adipose stem cells, and miRNAs. Based on the mechanism by which obesity causes breast cancer, we list possible therapeutic directions, including reducing the risk of breast cancer and inhibiting the progression of breast cancer. We also discussed the risk of autologous breast remodeling and fat transplantation. Finally, the causes of the obesity paradox and the function of enhancing immunity are discussed. Evaluating the balance between obesity-induced inflammation and enhanced immunity warrants further study.
Collapse
Affiliation(s)
- Yuan Wu
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Xu Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - Qiong Li
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Chienshan Cheng
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Lan Zheng
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China.
| |
Collapse
|
14
|
Ma S, Zhao Y, Lee WC, Ong LT, Lee PL, Jiang Z, Oguz G, Niu Z, Liu M, Goh JY, Wang W, Bustos MA, Ehmsen S, Ramasamy A, Hoon DSB, Ditzel HJ, Tan EY, Chen Q, Yu Q. Hypoxia induces HIF1α-dependent epigenetic vulnerability in triple negative breast cancer to confer immune effector dysfunction and resistance to anti-PD-1 immunotherapy. Nat Commun 2022; 13:4118. [PMID: 35840558 PMCID: PMC9287350 DOI: 10.1038/s41467-022-31764-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 07/01/2022] [Indexed: 12/12/2022] Open
Abstract
The hypoxic tumor microenvironment has been implicated in immune escape, but the underlying mechanism remains elusive. Using an in vitro culture system modeling human T cell dysfunction and exhaustion in triple-negative breast cancer (TNBC), we find that hypoxia suppresses immune effector gene expression, including in T and NK cells, resulting in immune effector cell dysfunction and resistance to immunotherapy. We demonstrate that hypoxia-induced factor 1α (HIF1α) interaction with HDAC1 and concurrent PRC2 dependency causes chromatin remolding resulting in epigenetic suppression of effector genes and subsequent immune dysfunction. Targeting HIF1α and the associated epigenetic machinery can reverse the immune effector dysfunction and overcome resistance to PD-1 blockade, as demonstrated both in vitro and in vivo using syngeneic and humanized mice models. These findings identify a HIF1α-mediated epigenetic mechanism in immune dysfunction and provide a potential strategy to overcome immune resistance in TNBC. Hypoxia can promote tumor escape from immune surveillance and immunotherapy. Here, the authors show that hypoxia induces T and NK cell dysfunction through HIF1α-mediated epigenetic suppression of effector gene expression, conferring resistance to anti-PD1 blockade in triple negative breast cancer models.
Collapse
Affiliation(s)
- Shijun Ma
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Yue Zhao
- Institute of Molecular and Cellular Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
| | - Wee Chyan Lee
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Li-Teng Ong
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Puay Leng Lee
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Zemin Jiang
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Gokce Oguz
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Zhitong Niu
- The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510655, China
| | - Min Liu
- Institute of Molecular and Cellular Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
| | - Jian Yuan Goh
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Wenyu Wang
- The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510655, China
| | - Matias A Bustos
- Department of Translational Molecular Medicine, Saint John's Cancer Institute, Providence Health System, Santa Monica, CA, 90404, USA
| | - Sidse Ehmsen
- Department of Oncology, Odense University Hospital, Odense, 5230, Denmark
| | - Adaikalavan Ramasamy
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Dave S B Hoon
- Department of Translational Molecular Medicine, Saint John's Cancer Institute, Providence Health System, Santa Monica, CA, 90404, USA
| | - Henrik J Ditzel
- Department of Oncology, Odense University Hospital, Odense, 5230, Denmark.,Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, 5230, Denmark
| | - Ern Yu Tan
- Department of General Surgery, Tan Tock Seng Hospital, Singapore, 308433, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cellular Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore.
| | - Qiang Yu
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138672, Singapore. .,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore. .,Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore.
| |
Collapse
|
15
|
Khandekar D, Dahunsi DO, Manzanera Esteve IV, Reid S, Rathmell JC, Titze J, Tiriveedhi V. Low-Salt Diet Reduces Anti-CTLA4 Mediated Systemic Immune-Related Adverse Events while Retaining Therapeutic Efficacy against Breast Cancer. BIOLOGY 2022; 11:810. [PMID: 35741331 PMCID: PMC9219826 DOI: 10.3390/biology11060810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/14/2022] [Accepted: 05/23/2022] [Indexed: 11/16/2022]
Abstract
Immune checkpoint inhibitor (ICI) therapy has revolutionized the breast cancer treatment landscape. However, ICI-induced systemic inflammatory immune-related adverse events (irAE) remain a major clinical challenge. Previous studies in our laboratory and others have demonstrated that a high-salt (HS) diet induces inflammatory activation of CD4+T cells leading to anti-tumor responses. In our current communication, we analyzed the impact of dietary salt modification on therapeutic and systemic outcomes in breast-tumor-bearing mice following anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA4) monoclonal antibody (mAb) based ICI therapy. As HS diet and anti-CTLA4 mAb both exert pro-inflammatory activation of CD4+T cells, we hypothesized that a combination of these would lead to enhanced irAE response, while low-salt (LS) diet through blunting peripheral inflammatory action of CD4+T cells would reduce irAE response. We utilized an orthotopic murine breast tumor model by injecting Py230 murine breast cancer cells into syngeneic C57Bl/6 mice. In an LS diet cohort, anti-CTLA4 mAb treatment significantly reduced tumor progression (day 35, 339 ± 121 mm3), as compared to isotype mAb (639 ± 163 mm3, p < 0.05). In an HS diet cohort, treatment with anti-CTLA4 reduced the survival rate (day 80, 2/15) compared to respective normal/regular salt (NS) diet cohort (8/15, p < 0.05). Further, HS plus anti-CTLA4 mAb caused an increased expression of inflammatory cytokines (IFNγ and IL-1β) in lung infiltrating and peripheral circulating CD4+T cells. This inflammatory activation of CD4+T cells in the HS plus anti-CTLA4 cohort was associated with the upregulation of inflammasome complex activity. However, an LS diet did not induce any significant irAE response in breast-tumor-bearing mice upon treatment with anti-CTLA4 mAb, thus suggesting the role of high-salt diet in irAE response. Importantly, CD4-specific knock out of osmosensitive transcription factor NFAT5 using CD4cre/creNFAT5flox/flox transgenic mice caused a downregulation of high-salt-mediated inflammatory activation of CD4+T cells and irAE response. Taken together, our data suggest that LS diet inhibits the anti-CTLA4 mAb-induced irAE response while retaining its anti-tumor efficacy.
Collapse
Affiliation(s)
- Durga Khandekar
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209, USA;
| | - Debolanle O. Dahunsi
- Department Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (D.O.D.); (J.C.R.)
| | | | - Sonya Reid
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Jeffrey C. Rathmell
- Department Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (D.O.D.); (J.C.R.)
| | - Jens Titze
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore 169857, Singapore;
- Division of Nephrology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Venkataswarup Tiriveedhi
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209, USA;
- Division of Pharmacology, Vanderbilt University, Nashville, TN 37240, USA
| |
Collapse
|
16
|
Lawther AJ, Phillips AJK, Chung NC, Chang A, Ziegler AI, Debs S, Sloan EK, Walker AK. Disrupting circadian rhythms promotes cancer-induced inflammation in mice. Brain Behav Immun Health 2022; 21:100428. [PMID: 35199050 PMCID: PMC8851215 DOI: 10.1016/j.bbih.2022.100428] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/07/2022] [Accepted: 02/07/2022] [Indexed: 02/09/2023] Open
Abstract
Disruption of circadian rhythms occurs in rotating shift-work, jetlag, and in individuals with irregular sleep schedules. Circadian disruption is known to alter inflammatory responses and impair immune function. However, there is limited understanding of how circadian disruption modulates cancer-induced inflammation. Inflammation is a hallmark of cancer and is linked to worse prognosis and impaired brain function in cancer patients. Here, we investigated the effect of circadian disruption on cancer-induced inflammation in an orthotopic breast cancer model. Using a validated chronic jetlag protocol that advances the light-cycle by 8 h every 2 days to disrupt circadian rhythms, we found that circadian disruption alters cancer-induced inflammation in a tissue-specific manner, increasing inflammation in the body and brain while decreasing inflammation within the tumor tissue. Circadian disruption did not affect inflammation in mice without tumors, suggesting that the impact of circadian disruption may be particularly detrimental in the context of underlying inflammatory conditions, such as cancer. Importantly, circadian disruption did not affect tumor burden, suggesting that increased inflammation was not a result of increased cancer progression. Overall, these findings identify the importance of healthy circadian rhythms for limiting cancer-induced inflammation. Circadian disruption enhances cancer-induced inflammation in the body and brain. The profile of inflammatory cytokines altered by circadian disruption is tissue specific. Changes in inflammatory profiles by circadian disruption are not due to enhanced tumor burden.
Collapse
Affiliation(s)
- Adam J Lawther
- Laboratory of ImmunoPsychiatry, Neuroscience Research Australia, Randwick, New South Wales, 2031, Australia
| | - Andrew J K Phillips
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Ni-Chun Chung
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Aeson Chang
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Alexandra I Ziegler
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Sophie Debs
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, New South Wales, 2031, Australia
| | - Erica K Sloan
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.,Division of Cancer Surgery, Peter MacCallum Cancer Centre, East Melbourne, VIC, 3002, Australia
| | - Adam K Walker
- Laboratory of ImmunoPsychiatry, Neuroscience Research Australia, Randwick, New South Wales, 2031, Australia.,Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.,School of Psychiatry, University of New South Wales, Kensington, NSW, 2033, Australia
| |
Collapse
|
17
|
CCL5 Deficiency Enhanced Cryo–Thermal-Triggered Long-Term Anti-Tumor Immunity in 4T1 Murine Breast Cancer. Biomedicines 2022; 10:biomedicines10030559. [PMID: 35327361 PMCID: PMC8945488 DOI: 10.3390/biomedicines10030559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 12/15/2022] Open
Abstract
Breast cancer remains one of the most common solid tumors. Tumor immunosuppressive factors mainly hinder the control of tumors. We previously developed an innovative cryo–thermal therapy that was shown to significantly suppress distal metastasis and improve long-term survival in murine B16F10 melanoma and 4T1 mammary carcinoma models. However, the effect of cryo–thermal therapy on the 4T1 model was not excellent. CCL5 has been reported to help the progression of breast cancer, so in this study, CCL5−/− was used to explore the role of host-derived CCL5 after cryo–thermal therapy. CCL5−/− could not completely resist tumor development, but it significantly improved survival rates when combined with cryo–thermal therapy. Mechanically, CCL5−/− mildly decreases the percentage of MDSCs, increases DC maturation and macrophage’s inflammatory function at an early stage after tumor inoculation, and later up-regulate the level of Th1 and down-regulate the level of Tregs. When combined with cryo–thermal therapy, CCL5−/− dramatically down-regulated the proportion of MDSCs and induced full M1 macrophage polarization, which further promoted Th1 differentiation and the cytotoxicity of CD8+ T cells. Our results indicated that CCL5−/− contributed to cryo–thermal-triggered, long-lasting anti-tumor memory immunity. The combination of cryo–thermal therapy and CCL5 blockades might extend the survival rates of patients with aggressive breast cancer.
Collapse
|
18
|
Gallyas F, Ramadan FHJ, Andreidesz K, Hocsak E, Szabo A, Tapodi A, Kiss GN, Fekete K, Bognar R, Szanto A, Bognar Z. Involvement of Mitochondrial Mechanisms and Cyclooxygenase-2 Activation in the Effect of Desethylamiodarone on 4T1 Triple-Negative Breast Cancer Line. Int J Mol Sci 2022; 23:ijms23031544. [PMID: 35163464 PMCID: PMC8836269 DOI: 10.3390/ijms23031544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/20/2022] [Accepted: 01/26/2022] [Indexed: 12/10/2022] Open
Abstract
Novel compounds significantly interfering with the mitochondrial energy production may have therapeutic value in triple-negative breast cancer (TNBC). This criterion is clearly fulfilled by desethylamiodarone (DEA), which is a major metabolite of amiodarone, a widely used antiarrhythmic drug, since the DEA previously demonstrated anti-neoplastic, anti-metastasizing, and direct mitochondrial effects in B16F10 melanoma cells. Additionally, the more than fifty years of clinical experience with amiodarone should answer most of the safety concerns about DEA. Accordingly, in the present study, we investigated DEA’s potential in TNBC by using a TN and a hormone receptor positive (HR+) BC cell line. DEA reduced the viability, colony formation, and invasive growth of the 4T1 cell line and led to a higher extent of the MCF-7 cell line. It lowered mitochondrial transmembrane potential and induced mitochondrial fragmentation. On the other hand, DEA failed to significantly affect various parameters of the cellular energy metabolism as determined by a Seahorse live cell respirometer. Cyclooxygenase 2 (COX-2), which was upregulated by DEA in the TNBC cell line only, accounted for most of 4T1’s DEA resistance, which was counteracted by the selective COX-2 inhibitor celecoxib. All these data indicate that DEA may have potentiality in the therapy of TNBC.
Collapse
Affiliation(s)
- Ferenc Gallyas
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
- Szentagothai Research Centre, University of Pecs, 7624 Pecs, Hungary
- LERN-UP Nuclear-Mitochondrial Interactions Research Group, 1245 Budapest, Hungary
| | - Fadi H. J. Ramadan
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
| | - Kitti Andreidesz
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
| | - Eniko Hocsak
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
| | - Aliz Szabo
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
| | - Antal Tapodi
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
| | - Gyongyi N. Kiss
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
| | - Katalin Fekete
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
| | - Rita Bognar
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
| | - Arpad Szanto
- Urology Clinic, UP Medical Center, University of Pecs Medical School, 7624 Pecs, Hungary;
| | - Zita Bognar
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary; (F.G.J.); (F.H.J.R.); (K.A.); (E.H.); (A.S.); (A.T.); (G.N.K.); (K.F.); (R.B.)
- Correspondence: ; Tel.: +36-72-536-276
| |
Collapse
|
19
|
Thomas SC, Madaan T, Kamble NS, Siddiqui NA, Pauletti GM, Kotagiri N. Engineered Bacteria Enhance Immunotherapy and Targeted Therapy through Stromal Remodeling of Tumors. Adv Healthc Mater 2022; 11:e2101487. [PMID: 34738725 PMCID: PMC8770579 DOI: 10.1002/adhm.202101487] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/28/2021] [Indexed: 01/03/2023]
Abstract
Desmoplastic solid tumors are characterized by the rapid build-up of extracellular matrix (ECM) macromolecules, such as hyaluronic acid (HA). The resulting physiological barrier prevents the infiltration of immune cells and also impedes the delivery of anticancer agents. The development of a hypervesiculating Escherichia coli Nissle (ΔECHy) based tumor targeting bacterial system capable of distributing a fusion peptide, cytolysin A (ClyA)-hyaluronidase (Hy) via outer membrane vesicles (OMVs) is reported. The capability of targeting hypoxic tumors, manufacturing recombinant proteins in situ and the added advantage of an on-site OMV based distribution system makes the engineered bacterial vector a unique candidate for peptide delivery. The HA degrading potential of Hy for stromal modulation is combined with the cytolytic activity of ClyA followed by testing it within syngeneic cancer models. ΔECHy is combined with immune checkpoint antibodies and tyrosine kinase inhibitors (TKIs) to demonstrate that remodeling the tumor stroma results in the improvement of immunotherapy outcomes and enhancing the efficacy of biological signaling inhibitors. The biocompatibility of ΔECHy is also investigated to show that the engineered bacteria are effectively cleared, elicit minimal inflammatory and immune responses, and therefore could be a reliable candidate as a live biotherapeutic.
Collapse
Affiliation(s)
- Shindu C. Thomas
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - Tushar Madaan
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - Nitin S. Kamble
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - Nabil A. Siddiqui
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - Giovanni M. Pauletti
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, 1 Pharmacy Place, St. Louis, MO 63110, USA
| | - Nalinikanth Kotagiri
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| |
Collapse
|
20
|
Liu W, Chakraborty B, Safi R, Kazmin D, Chang CY, McDonnell DP. Dysregulated cholesterol homeostasis results in resistance to ferroptosis increasing tumorigenicity and metastasis in cancer. Nat Commun 2021; 12:5103. [PMID: 34429409 PMCID: PMC8385107 DOI: 10.1038/s41467-021-25354-4] [Citation(s) in RCA: 179] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 08/04/2021] [Indexed: 12/21/2022] Open
Abstract
Hypercholesterolemia and dyslipidemia are associated with an increased risk for many cancer types and with poor outcomes in patients with established disease. Whereas the mechanisms by which this occurs are multifactorial we determine that chronic exposure of cells to 27-hydroxycholesterol (27HC), an abundant circulating cholesterol metabolite, selects for cells that exhibit increased cellular uptake and/or lipid biosynthesis. These cells exhibit substantially increased tumorigenic and metastatic capacity. Notably, the metabolic stress imposed upon cells by the accumulated lipids requires sustained expression of GPX4, a negative regulator of ferroptotic cell death. We show that resistance to ferroptosis is a feature of metastatic cells and further demonstrate that GPX4 knockdown attenuates the enhanced tumorigenic and metastatic activity of 27HC resistant cells. These findings highlight the general importance of ferroptosis in tumor growth and metastasis and suggest that dyslipidemia/hypercholesterolemia impacts cancer pathogenesis by selecting for cells that are resistant to ferroptotic cell death.
Collapse
Affiliation(s)
- Wen Liu
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Binita Chakraborty
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Rachid Safi
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Dmitri Kazmin
- Emory Vaccine Center, Emory University, Atlanta, GA, 30322, USA
| | - Ching-Yi Chang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
21
|
Hossain F, Majumder S, David J, Miele L. Precision Medicine and Triple-Negative Breast Cancer: Current Landscape and Future Directions. Cancers (Basel) 2021; 13:cancers13153739. [PMID: 34359640 PMCID: PMC8345034 DOI: 10.3390/cancers13153739] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/10/2021] [Accepted: 07/13/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary The implementation of precision medicine will revolutionize cancer treatment paradigms. Notably, this goal is not far from reality: genetically similar cancers can be treated similarly. The heterogeneous nature of triple-negative breast cancer (TNBC) made it a suitable candidate to practice precision medicine. Using TNBC molecular subtyping and genomic profiling, a precision medicine-based clinical trial is ongoing. This review summarizes the current landscape and future directions of precision medicine and TNBC. Abstract Triple-negative breast cancer (TNBC) is an aggressive and heterogeneous subtype of breast cancer associated with a high recurrence and metastasis rate that affects African-American women disproportionately. The recent approval of targeted therapies for small subgroups of TNBC patients by the US ‘Food and Drug Administration’ is a promising development. The advancement of next-generation sequencing, particularly somatic exome panels, has raised hopes for more individualized treatment plans. However, the use of precision medicine for TNBC is a work in progress. This review will discuss the potential benefits and challenges of precision medicine for TNBC. A recent clinical trial designed to target TNBC patients based on their subtype-specific classification shows promise. Yet, tumor heterogeneity and sub-clonal evolution in primary and metastatic TNBC remain a challenge for oncologists to design adaptive precision medicine-based treatment plans.
Collapse
Affiliation(s)
- Fokhrul Hossain
- Department of Genetics, Louisiana State University Health Sciences Center (LSUHSC), New Orleans, LA 70112, USA; (S.M.); (L.M.)
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center (LSUHSC), New Orleans, LA 70112, USA
- Correspondence:
| | - Samarpan Majumder
- Department of Genetics, Louisiana State University Health Sciences Center (LSUHSC), New Orleans, LA 70112, USA; (S.M.); (L.M.)
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center (LSUHSC), New Orleans, LA 70112, USA
| | - Justin David
- School of Medicine, Louisiana State University Health Sciences Center (LSUHSC), New Orleans, LA 70112, USA;
| | - Lucio Miele
- Department of Genetics, Louisiana State University Health Sciences Center (LSUHSC), New Orleans, LA 70112, USA; (S.M.); (L.M.)
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center (LSUHSC), New Orleans, LA 70112, USA
- School of Medicine, Louisiana State University Health Sciences Center (LSUHSC), New Orleans, LA 70112, USA;
| |
Collapse
|
22
|
Bencze N, Schvarcz C, Kriszta G, Danics L, Szőke É, Balogh P, Szállási Á, Hamar P, Helyes Z, Botz B. Desensitization of Capsaicin-Sensitive Afferents Accelerates Early Tumor Growth via Increased Vascular Leakage in a Murine Model of Triple Negative Breast Cancer. Front Oncol 2021; 11:685297. [PMID: 34336669 PMCID: PMC8317060 DOI: 10.3389/fonc.2021.685297] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/18/2021] [Indexed: 11/13/2022] Open
Abstract
There is growing interest in the role of nerve-driven mechanisms in tumorigenesis and tumor growth. Capsaicin-sensitive afferents have been previously shown to possess antitumoral and immune-regulatory properties, the mechanism of which is currently poorly understood. In this study, we have assessed the role of these terminals in the triple negative 4T1 orthotopic mouse model of breast cancer. The ultrapotent capsaicin-analogue resiniferatoxin (RTX) was used for the selective, systemic desensitization of capsaicin-sensitive afferents. Growth and viability of orthotopically implanted 4T1 tumors were measured by caliper, in vivo MRI, and bioluminescence imaging, while tumor vascularity and protease enzyme activity were assessed using fluorescent in vivo imaging. The levels of the neuropeptides Calcitonin Gene-Related Peptide (CGRP), Substance P (SP), and somatostatin were measured from tumor tissue homogenates using radioimmunoassay, while tumor structure and peritumoral inflammation were evaluated by conventional use of CD31, CD45 and CD3 immunohistology. RTX-pretreated mice demonstrated facilitated tumor growth in the early phase measured using a caliper, which was coupled with increased tumor vascular leakage demonstrated using fluorescent vascular imaging. The tumor size difference dissipated by day seven. The MRI tumor volume was similar, while the intratumoral protease enzyme activity measured by fluorescence imaging was also comparable in RTX-pretreated and non-pretreated animals. Tumor viability or immunohistopathological profile was measured using CD3, CD31, and CD45 stains and did not differ significantly from the non-pretreated control group. Intratumoral somatostatin, CGRP, and SP levels were similar in both groups. Our results underscore the beneficial, antitumoral properties of capsaicin sensitive nerve terminals in this aggressive model of breast cancer, which is presumed to be due to the inhibition of tumor vascular bed disruption. The absence of any difference in intratumoral neuropeptide levels indicates non-neural sources playing a substantial part in their expression.
Collapse
Affiliation(s)
- Noémi Bencze
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, Pécs, Hungary.,János Szentágothai Research Centre, Molecular Pharmacology Research Team and Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Csaba Schvarcz
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Gábor Kriszta
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, Pécs, Hungary.,János Szentágothai Research Centre, Molecular Pharmacology Research Team and Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Lea Danics
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Éva Szőke
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, Pécs, Hungary.,János Szentágothai Research Centre, Molecular Pharmacology Research Team and Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Péter Balogh
- Department of Immunology and Biotechnology, University of Pécs Medical School, Pécs, Hungary
| | - Árpád Szállási
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Péter Hamar
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary.,Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, Pécs, Hungary.,János Szentágothai Research Centre, Molecular Pharmacology Research Team and Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Bálint Botz
- János Szentágothai Research Centre, Molecular Pharmacology Research Team and Centre for Neuroscience, University of Pécs, Pécs, Hungary.,Department of Medical Imaging, University of Pécs, Medical School, Pécs, Hungary
| |
Collapse
|
23
|
Liu C, Wu P, Zhang A, Mao X. Advances in Rodent Models for Breast Cancer Formation, Progression, and Therapeutic Testing. Front Oncol 2021; 11:593337. [PMID: 33842308 PMCID: PMC8032937 DOI: 10.3389/fonc.2021.593337] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/27/2021] [Indexed: 01/01/2023] Open
Abstract
Breast cancer is a highly complicated disease. Advancement in the treatment and prevention of breast cancer lies in elucidation of the mechanism of carcinogenesis and progression. Rodent models of breast cancer have developed into premier tools for investigating the mechanisms and genetic pathways in breast cancer progression and metastasis and for developing and evaluating clinical therapeutics. Every rodent model has advantages and disadvantages, and the selection of appropriate rodent models with which to investigate breast cancer is a key decision in research. Design of a suitable rodent model for a specific research purpose is based on the integration of the advantages and disadvantages of different models. Our purpose in writing this review is to elaborate on various rodent models for breast cancer formation, progression, and therapeutic testing.
Collapse
Affiliation(s)
- Chong Liu
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Pei Wu
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ailin Zhang
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiaoyun Mao
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
24
|
Steenbrugge J, Vander Elst N, Demeyere K, De Wever O, Sanders NN, Van Den Broeck W, Ciamporcero E, Perera T, Meyer E. OMO-1 reduces progression and enhances cisplatin efficacy in a 4T1-based non-c-MET addicted intraductal mouse model for triple-negative breast cancer. NPJ Breast Cancer 2021; 7:27. [PMID: 33731699 PMCID: PMC7969607 DOI: 10.1038/s41523-021-00234-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 02/05/2021] [Indexed: 11/08/2022] Open
Abstract
c-MET is considered a driver of cancer progression, impacting tumor growth and tumor-supporting stroma. Here, we investigated the therapeutic efficacy of OMO-1, a potent and selective c-MET inhibitor, in an immunocompetent intraductal mouse model for triple-negative breast cancer (TNBC). OMO-1 reduced non-c-MET addicted 4T1 tumor progression dose dependently as monotherapeutic and provided additional disease reduction in combination with cisplatin. At the stromal level, OMO-1 significantly reduced neutrophil infiltration in 4T1 tumors, promoted immune activation, and enhanced cisplatin-mediated reduction of tumor-associated macrophages. OMO-1 treatment also reduced 4T1 tumor hypoxia and increased expression of pericyte markers, indicative for vascular maturation. Corroborating this finding, cisplatin delivery to the 4T1 primary tumor was enhanced upon OMO-1 treatment, increasing cisplatin DNA-adduct levels and tumor cell death. Although verification in additional cell lines is warranted, our findings provide initial evidence that TNBC patients may benefit from OMO-1 treatment, even in cases of non-c-MET addicted tumors.
Collapse
Affiliation(s)
- Jonas Steenbrugge
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - Niels Vander Elst
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Kristel Demeyere
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Olivier De Wever
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Niek N Sanders
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Wim Van Den Broeck
- Department of Morphology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | | | | | - Evelyne Meyer
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
25
|
The Shuganhuazheng Formula in Triple-Negative Breast Cancer: A Study Based on Network Pharmacology and In Vivo Experiments. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2020:8173147. [PMID: 33414839 PMCID: PMC7752265 DOI: 10.1155/2020/8173147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/18/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023]
Abstract
Breast cancer is the most common cancer in women. Among breast cancer subtypes, triple-negative breast cancer (TNBC) has the highest degree of malignancy and the worst prognosis. The Shuganhuazheng formula (SGHZF) is a traditional Chinese herbal formula for the treatment of TNBC, but the mechanism of SGHZF in the treatment of TNBC remains unclear. In this study, the therapeutic effect and mechanism of SGHZF against TNBC were preliminarily determined based on in vivo experimental verification and network pharmacology. In terms of therapeutic effects, the antitumour effect was verified by measuring and calculating tumour volume, and the expression of proto-oncogene c-Myc was verified by PCR. In terms of the mechanism, potential therapeutic targets were identified by overlapping the SGHZF-related and TNBC-related targets. After comprehensively analysing the results of the protein-protein interaction (PPI), gene ontology (GO) function, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses, Akt and HIF-1α were selected for verification by using immunohistochemical and Western blot analyses. The results of the study indicated that SGHZF can inhibit breast tumour growth in mice and that the mechanism may be related to the inhibition of Akt and HIF-1α expression.
Collapse
|
26
|
Meiyanto E, Zulfin U, Rahman A, Hanifa M, Utomo R, Haryanti S. Reactive oxygen species and senescence modulatory effects of rice bran extract on 4T1 and NIH-3T3 cells co-treatment with doxorubicin. Asian Pac J Trop Biomed 2021. [DOI: 10.4103/2221-1691.310204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
27
|
Tinganelli W, Durante M. Tumor Hypoxia and Circulating Tumor Cells. Int J Mol Sci 2020; 21:ijms21249592. [PMID: 33339353 PMCID: PMC7766826 DOI: 10.3390/ijms21249592] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Circulating tumor cells (CTCs) are a rare tumor cell subpopulation induced and selected by the tumor microenvironment's extreme conditions. Under hypoxia and starvation, these aggressive and invasive cells are able to invade the lymphatic and circulatory systems. Escaping from the primary tumor, CTCs enter into the bloodstream to form metastatic deposits or re-establish themselves in cancer's primary site. Although radiotherapy is widely used to cure solid malignancies, it can promote metastasis. Radiation can disrupt the primary tumor vasculature, increasing the dissemination of CTCs. Radiation also induces epithelial-mesenchymal transition (EMT) and eliminates suppressive signaling, causing the proliferation of existent, but previously dormant, disseminated tumor cells (DTCs). In this review, we collect the results and evidence underlying the molecular mechanisms of CTCs and DTCs and the effects of radiation and hypoxia in developing these cells.
Collapse
Affiliation(s)
- Walter Tinganelli
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291 Darmstadt, Germany;
| | - Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291 Darmstadt, Germany;
- Institut für Festkörperphysik, Technische Universität Darmstadt, 64291 Darmstadt, Germany
- Correspondence:
| |
Collapse
|
28
|
Meurer SK, Tezcan O, Lammers T, Weiskirchen R. Differential regulation of Lipocalin 2 (LCN2) in doxorubicin-resistant 4T1 triple negative breast cancer cells. Cell Signal 2020; 74:109731. [DOI: 10.1016/j.cellsig.2020.109731] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/25/2020] [Accepted: 07/27/2020] [Indexed: 12/22/2022]
|
29
|
Steenbrugge J, De Jaeghere EA, Meyer E, Denys H, De Wever O. Splenic Hematopoietic and Stromal Cells in Cancer Progression. Cancer Res 2020; 81:27-34. [PMID: 32998999 DOI: 10.1158/0008-5472.can-20-2339] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/31/2020] [Accepted: 09/24/2020] [Indexed: 11/16/2022]
Abstract
Tumor-derived secretory factors orchestrate splenic hematopoietic and stromal cells to fuel metastasis. The spleen acts as a reservoir site for hematopoietic stem and progenitor cells, which are rapidly exploited as myeloid-derived suppressor cells at the cost of tumor-reactive lymphoid cells. Splenic erythroid progenitor cells and mesenchymal stromal cells contribute directly and indirectly to both tumor immune escape and the metastatic cascade. Animal models provide valuable mechanistic insights, but their translation to a clinical setting highlights specific challenges and open issues. In this review, we envision the exploitation of the spleen as a source for novel biomarkers and therapeutic approaches.
Collapse
Affiliation(s)
- Jonas Steenbrugge
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Emiel A De Jaeghere
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Medical Oncology, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Gynecologic Pelvic Oncology Network Ghent (GYPON), Ghent, Belgium
| | - Evelyne Meyer
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Hannelore Denys
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Medical Oncology, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Gynecologic Pelvic Oncology Network Ghent (GYPON), Ghent, Belgium
| | - Olivier De Wever
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| |
Collapse
|
30
|
Zhang D, Lin Z, Zheng Y, Song J, Li J, Zeng Y, Liu X. Ultrasound-Driven Biomimetic Nanosystem Suppresses Tumor Growth and Metastasis through Sonodynamic Therapy, CO Therapy, and Indoleamine 2,3-Dioxygenase Inhibition. ACS NANO 2020; 14:8985-8999. [PMID: 32662971 DOI: 10.1021/acsnano.0c03833] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
The rational design of nanoplatforms to bypass reticuloendothelial system (RES) clearance, enhance spatiotemporal controllability, and boost host immune responses to achieve synergized tumor-targeted therapeutic purpose is highly desired. Herein, a biomimetic nanosystem is developed for tumor-targeted in situ delivery of singlet oxygen (1O2) and carbon monoxide (CO) in response to exogenous stimulus ultrasound (US) and endogenous stimulus hydrogen peroxide (H2O2) in tumor microenvironment, respectively. Taking advantages of tumor homing and RES evasion abilities of the macrophage membrane coating, our designed nanosystem shows excellent accumulation at the tumor site and effective suppression of tumor growth through US/H2O2-generated 1O2 and CO to induce cell apoptosis and mitochondrial dysfunction. Furthermore, our nanosystem can induce significant tumor immunogenic death by 1O2/CO therapy, then can achieve effective immune responses and long-term immune memory through the combination of indoleamin 2,3-dioxygenase (IDO) signal blocking to effectively against tumor rechallenge and prevent lung metastasis. Taken together, the here-presented therapeutic strategy based on sonodynamic/CO therapy and IDO signaling inhibition might provide a promising perspective for synergistically treating cancer in future clinical translations.
Collapse
Affiliation(s)
- Da Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, People's Republic of China
- The Key Lab of Analysis and Detection Technology for Food Safety of the MOE, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou 350002, People's Republic of China
| | - Ziguo Lin
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, People's Republic of China
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, People's Republic of China
| | - Youshi Zheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, People's Republic of China
| | - Jibin Song
- The Key Lab of Analysis and Detection Technology for Food Safety of the MOE, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou 350002, People's Republic of China
| | - Juan Li
- The Key Lab of Analysis and Detection Technology for Food Safety of the MOE, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou 350002, People's Republic of China
| | - Yongyi Zeng
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, People's Republic of China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, People's Republic of China
| |
Collapse
|