1
|
Bushnell GG, Sharma D, Wilmot HC, Zheng M, Fashina TD, Hutchens CM, Osipov S, Burness M, Wicha MS. Natural Killer Cell Regulation of Breast Cancer Stem Cells Mediates Metastatic Dormancy. Cancer Res 2024; 84:3337-3353. [PMID: 39106452 PMCID: PMC11474167 DOI: 10.1158/0008-5472.can-24-0030] [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: 01/04/2024] [Revised: 06/04/2024] [Accepted: 07/31/2024] [Indexed: 08/09/2024]
Abstract
Patients with breast cancer with estrogen receptor-positive tumors face a constant risk of disease recurrence for the remainder of their lives. Dormant tumor cells residing in tissues such as the bone marrow may generate clinically significant metastases many years after initial diagnosis. Previous studies suggest that dormant cancer cells display "stem-like" properties (cancer stem cell, CSC), which may be regulated by the immune system. To elucidate the role of the immune system in controlling dormancy and its escape, we studied dormancy in immunocompetent, syngeneic mouse breast cancer models. Three mouse breast cancer cell lines, PyMT, Met1, and D2.0R, contained CSCs that displayed short- and long-term metastatic dormancy in vivo, which was dependent on the host immune system. Each model was regulated by different components of the immune system. Natural killer (NK) cells were key for the metastatic dormancy phenotype in D2.0R cells. Quiescent D2.0R CSCs were resistant to NK cell cytotoxicity, whereas proliferative CSCs were sensitive. Resistance to NK cell cytotoxicity was mediated, in part, by the expression of BACH1 and SOX2 transcription factors. Expression of STING and STING targets was decreased in quiescent CSCs, and the STING agonist MSA-2 enhanced NK cell killing. Collectively, these findings demonstrate the role of immune regulation of breast tumor dormancy and highlight the importance of utilizing immunocompetent models to study this phenomenon. Significance: The immune system controls disseminated breast cancer cells during disease latency, highlighting the need to utilize immunocompetent models to identify strategies for targeting dormant cancer cells and reducing metastatic recurrence. See related commentary by Cackowski and Korkaya, p. 3319.
Collapse
Affiliation(s)
- Grace G. Bushnell
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Deeksha Sharma
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Henry C. Wilmot
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Michelle Zheng
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | | | - Chloe M. Hutchens
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Samuel Osipov
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Monika Burness
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Max S. Wicha
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
2
|
Balcioglu O, Gates BL, Freeman DW, Hagos BM, Mehrabad EM, Ayala-Talavera D, Spike BT. Mcam stabilizes a luminal progenitor-like breast cancer cell state via Ck2 control and Src/Akt/Stat3 attenuation. NPJ Breast Cancer 2024; 10:80. [PMID: 39277578 PMCID: PMC11401886 DOI: 10.1038/s41523-024-00687-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 08/28/2024] [Indexed: 09/17/2024] Open
Abstract
Cell state control is crucial for normal tissue development and cancer cell mimicry of stem/progenitor states, contributing to tumor heterogeneity, therapy resistance, and progression. Here, we demonstrate that the cell surface glycoprotein Mcam maintains the tumorigenic luminal progenitor (LP)-like epithelial cell state, leading to Basal-like mammary cancers. In the Py230 mouse mammary carcinoma model, Mcam knockdown (KD) destabilized the LP state by deregulating the Ck2/Stat3 axis, causing a switch to alveolar and basal states, loss of an estrogen-sensing subpopulation, and resistance to tamoxifen-an effect reversed by Ck2 and Stat3 inhibitors. In vivo, Mcam KD blocked generation of Basal-like tumors and Sox10+Krt14+ cells. In human tumors, MCAM loss was largely exclusive of the Basal-like subtype, linked instead to proliferative Luminal subtypes, including often endocrine-resistant Luminal B cancers. This study has implications for developing therapies targeting MCAM, CK2, and STAT3 and their likely effective contexts.
Collapse
Affiliation(s)
- Ozlen Balcioglu
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - Brooke L Gates
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - David W Freeman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - Berhane M Hagos
- Emergency Medicine, Oregon Health & Science University School of Medicine, Portland, OR, 97239, USA
| | | | - David Ayala-Talavera
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, 84112, USA
| | - Benjamin T Spike
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA.
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, 84112, USA.
- School of Computing, University of Utah, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
3
|
Hu M, Liu R, Castro N, Loza Sanchez L, Rueankham L, Learn JA, Huang R, Lam KS, Carraway KL. A novel lipophilic amiloride derivative efficiently kills chemoresistant breast cancer cells. Sci Rep 2024; 14:20263. [PMID: 39217266 PMCID: PMC11365969 DOI: 10.1038/s41598-024-71181-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
Derivatives of the potassium-sparing diuretic amiloride are preferentially cytotoxic toward tumor cells relative to normal cells, and have the capacity to target tumor cell populations resistant to currently employed therapeutic agents. However, a major barrier to clinical translation of the amilorides is their modest cytotoxic potency, with estimated IC50 values in the high micromolar range. Here we report the synthesis of ten novel amiloride derivatives and the characterization of their cytotoxic potency toward MCF7 (ER/PR-positive), SKBR3 (HER2-positive) and MDA-MB-231 (triple negative) cell line models of breast cancer. Comparisons of derivative structure with cytotoxic potency toward these cell lines underscore the importance of an intact guanidine group, and uncover a strong link between drug-induced cytotoxicity and drug lipophilicity. We demonstrate that our most potent derivative called LLC1 is preferentially cytotoxic toward mouse mammary tumor over normal epithelial organoids, acts in the single digit micromolar range on breast cancer cell line models representing all major subtypes, acts on cell lines that exhibit both transient and sustained resistance to chemotherapeutic agents, but exhibits limited anti-tumor effects in a mouse model of metastatic breast cancer. Nonetheless, our observations offer a roadmap for the future optimization of amiloride-based compounds with preferential cytotoxicity toward breast tumor cells.
Collapse
Affiliation(s)
- Michelle Hu
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA
| | - Noemi Castro
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA
| | - Liliana Loza Sanchez
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA
| | - Lapamas Rueankham
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA
| | - Julie A Learn
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA
| | - Ruiqi Huang
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA
| | - Kermit L Carraway
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA.
- UC Davis School of Medicine, 4645 2nd Avenue, Room 1100B, Sacramento, CA, 95817, USA.
| |
Collapse
|
4
|
Rosenbaum SR, Hughes CJ, Fields KM, Purdy SC, Gustafson A, Wolin A, Hampton D, Turner N, Ebmeier C, Costello JC, Ford HL. An EYA3/NF-κB/CCL2 signaling axis suppresses cytotoxic NK cells in the pre-metastatic niche to promote triple negative breast cancer metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.606072. [PMID: 39211066 PMCID: PMC11360953 DOI: 10.1101/2024.07.31.606072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Patients with Triple Negative Breast Cancer (TNBC) exhibit high rates of metastases and poor prognoses. The Eyes absent (EYA) family of proteins are developmental transcriptional cofactors/phosphatases that are re-expressed and/or upregulated in numerous cancers. Herein, we demonstrate that EYA3 correlates with decreased survival in breast cancer, and that it strongly, and specifically, regulates metastasis via a novel mechanism that involves NF-kB signaling and an altered innate immune profile at the pre-metastatic niche (PMN). Remarkably, restoration of NF-kB signaling downstream of Eya3 knockdown (KD) restores metastasis without restoring primary tumor growth, isolating EYA3/NF-kB effects to the metastatic site. We show that secreted CCL2, regulated downstream of EYA3/NF-kB, specifically decreases cytotoxic NK cells in the PMN and that re-expression of Ccl2 in Eya3 -KD cells is sufficient to rescue activation/levels of cytotoxic NK cells in vitro and at the PMN, where EYA3-mediated decreases in cytotoxic NK cells are required for metastatic outgrowth. Importantly, analysis of public breast cancer datasets uncovers a significant correlation of EYA3 with NF-kB/CCL2, underscoring the relevance of EYA3/NF-kB/CCL2 to human disease. Our findings suggest that inhibition of EYA3 could be a powerful means to re-activate the innate immune response at the PMN, inhibiting TNBC metastasis. Significance EYA3 promotes metastasis of TNBC cells by promoting NF-kB-mediated CCL2 expression and inhibiting cytotoxic NK cells at the pre-metastatic niche, highlighting a potential therapeutic target in this subset of breast cancer.
Collapse
|
5
|
Schmidt K, Thatcher A, Grobe A, Broussard P, Hicks L, Gu H, Ellies LG, Sears DD, Kalachev L, Kroll E. The combined treatment with ketogenic diet and metformin slows tumor growth in two mouse models of triple negative breast cancer. TRANSLATIONAL MEDICINE COMMUNICATIONS 2024; 9:21. [PMID: 39574543 PMCID: PMC11580796 DOI: 10.1186/s41231-024-00178-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 05/22/2024] [Indexed: 11/24/2024]
Abstract
Background Many tumors contain hypoxic microenvironments caused by inefficient tumor vascularization. Hypoxic tumors have been shown to resist conventional cancer therapies. Hypoxic cancer cells rely on glucose to meet their energetic and anabolic needs to fuel uncontrolled proliferation and metastasis. This glucose dependency is linked to a metabolic shift in response to hypoxic conditions. Methods To leverage the glucose dependency of hypoxic tumor cells, we assessed the effects of a mild reduction in systemic glucose by controlling both dietary carbohydrates with a ketogenic diet and endogenous glucose production by using metformin on two mouse models of triple-negative breast cancer (TNBC). Results Here, we showed that animals with TNBC treated with the combination regimen of ketogenic diet and metformin (a) had their tumor burden lowered by two-thirds, (b) displayed 38% slower tumor growth, and (c) showed 36% longer latency, compared to the animals treated with a ketogenic diet or metformin alone. As a result, lowering systemic glucose by this combined dietary and pharmacologic approach improved overall survival in our mouse TNBC models by 31 days, approximately equivalent to 3 years of life extension in human terms. Conclusion This preclinical study demonstrates that reducing systemic glucose by combining a ketogenic diet and metformin significantly inhibits tumor proliferation and increases overall survival. Our findings suggest a possible treatment for a broad range of hypoxic and glycolytic tumor types that can augment existing treatment options to improve patient outcomes.
Collapse
Affiliation(s)
- Karen Schmidt
- Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Amber Thatcher
- Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Albert Grobe
- Silverlake Research Corporation, Missoula, MT, USA
| | - Pamela Broussard
- College of Humanities and Sciences, University of Montana, Missoula, MT, USA
| | - Linda Hicks
- College of Humanities and Sciences, University of Montana, Missoula, MT, USA
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Lesley G Ellies
- Department of Pathology, University of California San Diego, San Diego, CA, USA
| | - Dorothy D. Sears
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Leonid Kalachev
- Department of Mathematical Sciences, University of Montana, Missoula, MT, USA
| | - Eugene Kroll
- Division of Biological Sciences, University of Montana, Missoula, MT, USA
- Present address: Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| |
Collapse
|
6
|
Hagan CE, Snyder AG, Headley M, Oberst A. Apoptotic cells promote circulating tumor cell survival and metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595217. [PMID: 38826267 PMCID: PMC11142129 DOI: 10.1101/2024.05.21.595217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
During tumor progression and especially following cytotoxic therapy, cell death of both tumor and stromal cells is widespread. Despite clinical observations that high levels of apoptotic cells correlate with poorer patient outcomes, the physiological effects of dying cells on tumor progression remain incompletely understood. Here, we report that circulating apoptotic cells robustly enhance tumor cell metastasis to the lungs. Using intravenous metastasis models, we observed that the presence of apoptotic cells, but not cells dying by other mechanisms, supports circulating tumor cell (CTC) survival following arrest in the lung vasculature. Apoptotic cells promote CTC survival by recruiting platelets to the forming metastatic niche. Apoptotic cells externalize the phospholipid phosphatidylserine to the outer leaflet of the plasma membrane, which we found increased the activity of the coagulation initiator Tissue Factor, thereby triggering the formation of platelet clots that protect proximal CTCs. Inhibiting the ability of apoptotic cells to induce coagulation by knocking out Tissue Factor, blocking phosphatidylserine, or administering the anticoagulant heparin abrogated the pro-metastatic effect of apoptotic cells. This work demonstrates a previously unappreciated role for apoptotic cells in facilitating metastasis by establishing CTC-supportive emboli, and suggests points of intervention that may reduce the pro-metastatic effect of apoptotic cells. GRAPHICAL ABSTRACT
Collapse
|
7
|
Balcioglu O, Gates BL, Freeman DW, Hagos BM, Mehrabad EM, Ayala-Talavera D, Spike BT. Mcam stabilizes luminal progenitor breast cancer phenotypes via Ck2 control and Src/Akt/Stat3 attenuation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.10.540211. [PMID: 38562809 PMCID: PMC10983870 DOI: 10.1101/2023.05.10.540211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Breast cancers are categorized into subtypes with distinctive therapeutic vulnerabilities and prognoses based on their expression of clinically targetable receptors and gene expression patterns mimicking different cell types of the normal gland. Here, we tested the role of Mcam in breast cancer cell state control and tumorigenicity in a luminal progenitor-like murine tumor cell line (Py230) that exhibits lineage and tumor subtype plasticity. Mcam knockdown Py230 cells show augmented Stat3 and Pi3K/Akt activation associated with a lineage state switch away from a hormone-sensing/luminal progenitor state toward alveolar and basal cell related phenotypes that were refractory to growth inhibition by the anti-estrogen therapeutic, tamoxifen. Inhibition of Stat3, or the upstream activator Ck2, reversed these cell state changes. Mcam binds Ck2 and acts as a regulator of Ck2 substrate utilization across multiple mammary tumor cell lines. In Py230 cells this activity manifests as increased mesenchymal morphology, migration, and Src/Fak/Mapk/Paxillin adhesion complex signaling in vitro, in contrast to Mcam's reported roles in promoting mesenchymal phenotypes. In vivo, Mcam knockdown reduced tumor growth and take rate and inhibited cell state transition to Sox10+/neural crest like cells previously been associated with tumor aggressiveness. This contrasts with human luminal breast cancers where MCAM copy number loss is highly coupled to Cyclin D amplification, increased proliferation, and the more aggressive Luminal B subtype. Together these data indicate a critical role for Mcam and its regulation of Ck2 in control of breast cancer cell state plasticity with implications for progression, evasion of targeted therapies and combination therapy design.
Collapse
Affiliation(s)
- Ozlen Balcioglu
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112 USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112 USA
| | - Brooke L. Gates
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112 USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112 USA
| | - David W. Freeman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112 USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112 USA
| | - Berhane M. Hagos
- Current Address: Emergency Medicine, Oregon Health & Science University School of Medicine, Portland, OR 97239 USA
| | | | - David Ayala-Talavera
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112 USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112 USA
| | - Benjamin T. Spike
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112 USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112 USA
- School of Computing, University of Utah, Salt Lake City, UT 84112 USA
| |
Collapse
|
8
|
Karlinsky KT, Bismuth M, Aronovich R, Ilovitsh T. Nonlinear Frequency Mixing Ultrasound Imaging of Nanoscale Contrast Agents. IEEE Trans Biomed Eng 2024; 71:866-875. [PMID: 37812544 DOI: 10.1109/tbme.2023.3321743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
OBJECTIVE Nanoscale ultrasound contrast agents show promise as alternatives for diagnostics and therapies due to their enhanced stability and ability to traverse blood vessels. Nonetheless, their reduced size limits echogenicity. This study introduces an enhanced nanobubble frequency mixing ultrasound imaging method, by capitalizing on their nonlinear acoustic response to dual-frequency excitation. METHODS A single broadband transducer (L12-3v) controlled by a programmable ultrasound system was used to transmit a dual-frequency single-cycle wavefront. The frequency mixing effect enabled simultaneous transducer capture of nanobubble-generated sum and difference frequencies in real time without the need for additional hardware or post-processing, by substituting the single-frequency wavefront in a standard contrast harmonic pulse inversion imaging protocol, with the dual-frequency wavefront. RESULTS Optimization experiments were conducted in tissue mimicking phantoms. Among the dual-frequency combinations that were tested, the highest contrast was obtained using 4&8 MHz. The nanobubble contrast improved with increased mechanical index, and achieved a maximal contrast improvement of 8.4 ± 0.5 dB compared to 4 MHz pulse inversion imaging. In imaging of a breast cancer tumor mouse model, after a systemic nanobubble injection, the contrast was improved by 3.4 ± 1.7, 4.8 ± 1.8, and 6.3 ± 1.6 dB for mechanical indices of 0.04, 0.08, and 0.1, respectively. CONCLUSION Nonlinear frequency mixing significantly improved the nanobubble contrast, which facilitated their imaging in-vivo. SIGNIFICANCE This study offers a new avenue to enhance ultrasound imaging utilizing nanobubbles, potentially leading to advancements in other diagnostic applications.
Collapse
|
9
|
Schmidt K, Thatcher A, Grobe A, Hicks L, Gu H, Sears DD, Ellies LG, Kalachev L, Kroll E. The Combined Treatment with Ketogenic Diet and Metformin Slows Tumor Growth in Two Mouse Models of Triple Negative Breast Cancer. RESEARCH SQUARE 2023:rs.3.rs-3664129. [PMID: 38196628 PMCID: PMC10775859 DOI: 10.21203/rs.3.rs-3664129/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
BACKGROUND Many tumors contain hypoxic microenvironments caused by inefficient tumor vascularization. Hypoxic tumors have been shown to resist conventional cancer therapies. Hypoxic cancer cells rely on glucose to meet their energetic and anabolic needs to fuel uncontrolled proliferation and metastasis. This glucose dependency is linked to a metabolic shift in response to hypoxic conditions. METHODS To leverage the glucose dependency of hypoxic tumor cells, we assessed the effects of a controlled reduction in systemic glucose by combining dietary carbohydrate restriction, using a ketogenic diet, with gluconeogenesis inhibition, using metformin, on two mouse models of triple-negative breast cancer (TNBC). RESULTS We confirmed that MET - 1 breast cancer cells require abnormally high glucose concentrations to survive in a hypoxic environment in vitro. Then, we showed that, compared to a ketogenic diet or metformin alone, animals treated with the combination regimen showed significantly lower tumor burden, higher tumor latency and slower tumor growth. As a result, lowering systemic glucose by this combined dietary and pharmacologic approach improved overall survival in our mouse model by 31 days, which is approximately equivalent to 3 human years. CONCLUSION This is the first preclinical study to demonstrate that reducing systemic glucose by combining a ketogenic diet and metformin significantly inhibits tumor proliferation and increases overall survival. Our findings suggest a possible treatment for a broad range of hypoxic and glycolytic tumor types, one that can also augment existing treatment options to improve patient outcomes.
Collapse
Affiliation(s)
- Karen Schmidt
- University of Montana Division of Biological Sciences
| | | | | | - Linda Hicks
- University of Montana Division of Biological Sciences
| | - Haiwei Gu
- Arizona State University School of Life Sciences
| | | | | | | | - Eugene Kroll
- University of Montana Missoula: University of Montana
| |
Collapse
|
10
|
Mihalik NE, Steinberger KJ, Stevens AM, Bobko AA, Hoblitzell EH, Tseytlin O, Akhter H, Dziadowicz SA, Wang L, O’Connell RC, Monaghan KL, Hu G, Mo X, Khramtsov VV, Tseytlin M, Driesschaert B, Wan EC, Eubank TD. Dose-Specific Intratumoral GM-CSF Modulates Breast Tumor Oxygenation and Antitumor Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1589-1604. [PMID: 37756529 PMCID: PMC10656117 DOI: 10.4049/jimmunol.2300326] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023]
Abstract
GM-CSF has been employed as an adjuvant to cancer immunotherapy with mixed results based on dosage. We previously showed that GM-CSF regulated tumor angiogenesis by stimulating soluble vascular endothelial growth factor (VEGF) receptor-1 from monocytes/macrophages in a dose-dependent manner that neutralized free VEGF, and intratumoral injections of high-dose GM-CSF ablated blood vessels and worsened hypoxia in orthotopic polyoma middle T Ag (PyMT) triple-negative breast cancer (TNBC). In this study, we assessed both immunoregulatory and oxygen-regulatory components of low-dose versus high-dose GM-CSF to compare effects on tumor oxygen, vasculature, and antitumor immunity. We performed intratumoral injections of low-dose GM-CSF or saline controls for 3 wk in FVB/N PyMT TNBC. Low-dose GM-CSF uniquely reduced tumor hypoxia and normalized tumor vasculature by increasing NG2+ pericyte coverage on CD31+ endothelial cells. Priming of "cold," anti-PD1-resistant PyMT tumors with low-dose GM-CSF (hypoxia reduced) sensitized tumors to anti-PD1, whereas high-dose GM-CSF (hypoxia exacerbated) did not. Low-dose GM-CSF reduced hypoxic and inflammatory tumor-associated macrophage (TAM) transcriptional profiles; however, no phenotypic modulation of TAMs or tumor-infiltrating lymphocytes were observed by flow cytometry. In contrast, high-dose GM-CSF priming increased infiltration of TAMs lacking the MHC class IIhi phenotype or immunostimulatory marker expression, indicating an immunosuppressive phenotype under hypoxia. However, in anti-PD1 (programmed cell death 1)-susceptible BALB/c 4T1 tumors (considered hot versus PyMT), high-dose GM-CSF increased MHC class IIhi TAMs and immunostimulatory molecules, suggesting disparate effects of high-dose GM-CSF across PyMT versus 4T1 TNBC models. Our data demonstrate a (to our knowledge) novel role for low-dose GM-CSF in reducing tumor hypoxia for synergy with anti-PD1 and highlight why dosage and setting of GM-CSF in cancer immunotherapy regimens require careful consideration.
Collapse
Affiliation(s)
- Nicole E. Mihalik
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506
- WVU Cancer Institute, West Virginia University, Morgantown, WV, 26505
| | - Kayla J. Steinberger
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506
- WVU Cancer Institute, West Virginia University, Morgantown, WV, 26505
| | - Alyson M. Stevens
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506
- WVU Cancer Institute, West Virginia University, Morgantown, WV, 26505
| | - Andrey A. Bobko
- WVU Cancer Institute, West Virginia University, Morgantown, WV, 26505
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506
- In vivo Multifunctional Magnetic Resonance (IMMR) center, West Virginia University, Morgantown, WV 26506
| | - E. Hannah Hoblitzell
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506
| | - Oxana Tseytlin
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506
- In vivo Multifunctional Magnetic Resonance (IMMR) center, West Virginia University, Morgantown, WV 26506
| | - Halima Akhter
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506
- Bioinformatics Core, West Virginia University, Morgantown, WV 26506
- Department of Computer Science and Electrical Engineering, West Virginia University, Morgantown, WV 26506
| | - Sebastian A. Dziadowicz
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506
- Bioinformatics Core, West Virginia University, Morgantown, WV 26506
| | - Lei Wang
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506
- Bioinformatics Core, West Virginia University, Morgantown, WV 26506
| | - Ryan C. O’Connell
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506
- In vivo Multifunctional Magnetic Resonance (IMMR) center, West Virginia University, Morgantown, WV 26506
| | - Kelly L. Monaghan
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506
| | - Gangqing Hu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506
- Bioinformatics Core, West Virginia University, Morgantown, WV 26506
| | - Xiaokui Mo
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, 1585 Neil Ave, Columbus, OH 43210, USA
| | - Valery V. Khramtsov
- West Virginia Clinical and Translational Science Institute, West Virginia University, Morgantown WV 26506
- WVU Cancer Institute, West Virginia University, Morgantown, WV, 26505
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506
- In vivo Multifunctional Magnetic Resonance (IMMR) center, West Virginia University, Morgantown, WV 26506
| | - Mark Tseytlin
- WVU Cancer Institute, West Virginia University, Morgantown, WV, 26505
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506
- In vivo Multifunctional Magnetic Resonance (IMMR) center, West Virginia University, Morgantown, WV 26506
| | - Benoit Driesschaert
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, 26506
- West Virginia Clinical and Translational Science Institute, West Virginia University, Morgantown WV 26506
- WVU Cancer Institute, West Virginia University, Morgantown, WV, 26505
- In vivo Multifunctional Magnetic Resonance (IMMR) center, West Virginia University, Morgantown, WV 26506
- C. Eugene Bennet Department of Chemistry, West Virginia University, Morgantown, WV, 26505, United States
| | - Edwin C.K. Wan
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506
- Department of Neuroscience, West Virginia University, Morgantown, WV, 26505
| | - Timothy D. Eubank
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV 26506
- West Virginia Clinical and Translational Science Institute, West Virginia University, Morgantown WV 26506
- WVU Cancer Institute, West Virginia University, Morgantown, WV, 26505
- In vivo Multifunctional Magnetic Resonance (IMMR) center, West Virginia University, Morgantown, WV 26506
| |
Collapse
|
11
|
Cohen N, Mundhe D, Deasy SK, Adler O, Ershaid N, Shami T, Levi-Galibov O, Wassermann R, Scherz-Shouval R, Erez N. Breast Cancer-Secreted Factors Promote Lung Metastasis by Signaling Systemically to Induce a Fibrotic Premetastatic Niche. Cancer Res 2023; 83:3354-3367. [PMID: 37548552 DOI: 10.1158/0008-5472.can-22-3707] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 06/12/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
Metastatic cancer is largely incurable and is the main cause of cancer-related deaths. The metastatic microenvironment facilitates formation of metastases. Cancer-associated fibroblasts (CAF) are crucial players in generating a hospitable metastatic niche by mediating an inflammatory microenvironment. Fibroblasts also play a central role in modifying the architecture and stiffness of the extracellular matrix (ECM). Resolving the early changes in the metastatic niche could help identify approaches to inhibit metastatic progression. Here, we demonstrate in mouse models of spontaneous breast cancer pulmonary metastasis that fibrotic changes and rewiring of lung fibroblasts occurred at premetastatic stages, suggesting systemic influence by the primary tumor. Activin A (ActA), a TGFβ superfamily member, was secreted from breast tumors and its levels in the blood were highly elevated in tumor-bearing mice. ActA upregulated the expression of profibrotic factors in lung fibroblasts, leading to enhanced collagen deposition in the lung premetastatic niche. ActA signaling was functionally important for lung metastasis, as genetic targeting of ActA in breast cancer cells significantly attenuated lung metastasis and improved survival. Moreover, high levels of ActA in human patients with breast cancer were associated with lung metastatic relapse and poor survival. This study uncovers a novel mechanism by which breast cancer cells systemically rewire the stromal microenvironment in the metastatic niche to facilitate pulmonary metastasis. SIGNIFICANCE ActA mediates cross-talk between breast cancer cells and cancer-associated fibroblasts in the lung metastatic niche that enhances fibrosis and metastasis, implicating ActA as a potential therapeutic target to inhibit metastatic relapse.
Collapse
Affiliation(s)
- Noam Cohen
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dhanashree Mundhe
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sarah K Deasy
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Omer Adler
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nour Ershaid
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tamar Shami
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Oshrat Levi-Galibov
- Department of Biomolecular Sciences, Weizmann Institute of Science, Tel Aviv, Israel
| | - Rina Wassermann
- Department of Biomolecular Sciences, Weizmann Institute of Science, Tel Aviv, Israel
| | - Ruth Scherz-Shouval
- Department of Biomolecular Sciences, Weizmann Institute of Science, Tel Aviv, Israel
| | - Neta Erez
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
12
|
Krisanits BA, Schuster R, Randise J, Nogueira LM, Lane JT, Panguluri GA, Li H, Helke K, Cuitiño MC, Koivisto C, Spruill L, Ostrowski MC, Anderson SM, Turner DP, Findlay VJ. Pubertal exposure to dietary advanced glycation end products disrupts ductal morphogenesis and induces atypical hyperplasia in the mammary gland. Breast Cancer Res 2023; 25:118. [PMID: 37803429 PMCID: PMC10559657 DOI: 10.1186/s13058-023-01714-4] [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: 04/27/2023] [Accepted: 09/18/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND Advanced glycation end products (AGEs) are reactive metabolites intrinsically linked with modern dietary patterns. Processed foods, and those high in sugar, protein and fat, often contain high levels of AGEs. Increased AGE levels are associated with increased breast cancer risk, however their significance has been largely overlooked due to a lack of direct cause-and-effect relationship. METHODS To address this knowledge gap, FVB/n mice were fed regular, low AGE, and high AGE diets from 3 weeks of age and mammary glands harvested during puberty (7 weeks) or adulthood (12 weeks and 7 months) to determine the effects upon mammary gland development. At endpoint mammary glands were harvested and assessed histologically (n ≥ 4). Immunohistochemistry and immunofluorescence were used to assess cellular proliferation and stromal fibroblast and macrophage recruitment. The Kruskal-Wallis test were used to compare continuous outcomes among groups. Mammary epithelial cell migration and invasion in response to AGE-mediated fibroblast activation was determined in two-compartment co-culture models. In vitro experiments were performed in triplicate. The nonparametric Wilcoxon rank sum test was used to compare differences between groups. RESULTS Histological analysis revealed the high AGE diet delayed ductal elongation, increased primary branching, as well as increased terminal end bud number and size. The high AGE diet also led to increased recruitment and proliferation of stromal cells to abnormal structures that persisted into adulthood. Atypical hyperplasia was observed in the high AGE fed mice. Ex vivo fibroblasts from mice fed dietary-AGEs retain an activated phenotype and promoted epithelial migration and invasion of non-transformed immortalized and tumor-derived mammary epithelial cells. Mechanistically, we found that the receptor for AGE (RAGE) is required for AGE-mediated increases in epithelial cell migration and invasion. CONCLUSIONS We observed a disruption in mammary gland development when mice were fed a diet high in AGEs. Further, both epithelial and stromal cell populations were impacted by the high AGE diet in the mammary gland. Educational, interventional, and pharmacological strategies to reduce AGEs associated with diet may be viewed as novel disease preventive and/or therapeutic initiatives during puberty.
Collapse
Affiliation(s)
- Bradley A Krisanits
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Reid Schuster
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Jaime Randise
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Lourdes M Nogueira
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Jackson T Lane
- Department of Surgery and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Gowtami A Panguluri
- Department of Surgery and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Hong Li
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
- Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Kristi Helke
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Maria C Cuitiño
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- College of Health Sciences, Comprehensive Cancer Center, Ohio State University, Columbus, OH, USA
| | - Christopher Koivisto
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Laura Spruill
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Michael C Ostrowski
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Steven M Anderson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - David P Turner
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA.
- Department of Surgery and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| | - Victoria J Findlay
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA.
- Department of Surgery and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
13
|
Bushnell GG, Sharma D, Wilmot HC, Zheng M, Fashina TD, Hutchens CM, Osipov S, Wicha MS. Natural killer cell regulation of breast cancer stem cells mediates metastatic dormancy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.02.560493. [PMID: 37873211 PMCID: PMC10592904 DOI: 10.1101/2023.10.02.560493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Breast cancer patients with estrogen receptor positive tumors face a constant risk of disease recurrence for the remainder of their lives. Dormant tumor cells residing in tissues such as the bone marrow may generate clinically significant metastases many years after initial diagnosis. Previous studies suggest that dormant cells display "stem like" properties (CSCs), which may be regulated by the immune system. Although many studies have examined tumor cell intrinsic characteristics of dormancy, the role of the immune system in controlling dormancy and its escape is not well understood. This scientific gap is due, in part, to a lack of immunocompetent mouse models of breast cancer dormancy with many studies involving human xenografts in immunodeficient mice. To overcome this limitation, we studied dormancy in immunocompetent, syngeneic mouse breast cancer models. We find that PyMT, Met-1 and D2.0R cell lines contain CSCs that display both short- and long-term metastatic dormancy in vivo, which is dependent on the host immune system. Natural killer cells were key for the metastatic dormancy phenotype observed for D2.0R and the role of NK cells in regulating CSCs was further investigated.Quiescent D2.0R CSC are resistant to NK cytotoxicity, while proliferative D2.0R CSC were sensitive to NK cytotoxicity both in vitro and in vivo. This resistance was mediated, in part, by the expression of Bach1 and Sox2 transcription factors. NK killing was enhanced by the STING agonist MSA-2. Collectively, our findings demonstrate the important role of immune regulation of breast tumor dormancy and highlight the importance of utilizing immunocompetent models to study this phenomenon.
Collapse
|
14
|
Yoshimura T, Li C, Wang Y, Matsukawa A. The chemokine monocyte chemoattractant protein-1/CCL2 is a promoter of breast cancer metastasis. Cell Mol Immunol 2023:10.1038/s41423-023-01013-0. [PMID: 37208442 DOI: 10.1038/s41423-023-01013-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 03/17/2023] [Indexed: 05/21/2023] Open
Abstract
Breast cancer is the most prevalent cancer worldwide, and metastasis is the leading cause of death in cancer patients. Human monocyte chemoattractant protein-1 (MCP-1/CCL2) was isolated from the culture supernatants of not only mitogen-activated peripheral blood mononuclear leukocytes but also malignant glioma cells based on its in vitro chemotactic activity toward human monocytes. MCP-1 was subsequently found to be identical to a previously described tumor cell-derived chemotactic factor thought to be responsible for the accumulation of tumor-associated macrophages (TAMs), and it became a candidate target of clinical intervention; however, the role of TAMs in cancer development was still controversial at the time of the discovery of MCP-1. The in vivo role of MCP-1 in cancer progression was first evaluated by examining human cancer tissues, including breast cancers. Positive correlations between the level of MCP-1 production in tumors and the degree of TAM infiltration and cancer progression were established. The contribution of MCP-1 to the growth of primary tumors and metastasis to the lung, bone, and brain was examined in mouse breast cancer models. The results of these studies strongly suggested that MCP-1 is a promoter of breast cancer metastasis to the lung and brain but not bone. Potential mechanisms of MCP-1 production in the breast cancer microenvironment have also been reported. In the present manuscript, we review studies in which the role of MCP-1 in breast cancer development and progression and the mechanisms of its production were examined and attempt to draw a consensus and discuss the potential use of MCP-1 as a biomarker for diagnosis.
Collapse
Affiliation(s)
- Teizo Yoshimura
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan.
| | - Chunning Li
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| | - Yuze Wang
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| | - Akihiro Matsukawa
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama, 700-8558, Japan
| |
Collapse
|
15
|
VanderVorst K, Dreyer CA, Hatakeyama J, Bell GRR, Learn JA, Berg AL, Hernandez M, Lee H, Collins SR, Carraway KL. Vangl-dependent Wnt/planar cell polarity signaling mediates collective breast carcinoma motility and distant metastasis. Breast Cancer Res 2023; 25:52. [PMID: 37147680 PMCID: PMC10163820 DOI: 10.1186/s13058-023-01651-2] [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: 12/18/2022] [Accepted: 04/23/2023] [Indexed: 05/07/2023] Open
Abstract
BACKGROUND In light of the growing appreciation for the role of collective cell motility in metastasis, a deeper understanding of the underlying signaling pathways will be critical to translating these observations to the treatment of advanced cancers. Here, we examine the contribution of Wnt/planar cell polarity (Wnt/PCP), one of the non-canonical Wnt signaling pathways and defined by the involvement of the tetraspanin-like proteins Vangl1 and Vangl2, to breast tumor cell motility, collective cell invasiveness and mammary tumor metastasis. METHODS Vangl1 and Vangl2 knockdown and overexpression and Wnt5a stimulation were employed to manipulate Wnt/PCP signaling in a battery of breast cancer cell lines representing all breast cancer subtypes, and in tumor organoids from MMTV-PyMT mice. Cell migration was assessed by scratch and organoid invasion assays, Vangl protein subcellular localization was assessed by confocal fluorescence microscopy, and RhoA activation was assessed in real time by fluorescence imaging with an advanced FRET biosensor. The impact of Wnt/PCP suppression on mammary tumor growth and metastasis was assessed by determining the effect of conditional Vangl2 knockout on the MMTV-NDL mouse mammary tumor model. RESULTS We observed that Vangl2 knockdown suppresses the motility of all breast cancer cell lines examined, and overexpression drives the invasiveness of collectively migrating MMTV-PyMT organoids. Vangl2-dependent RhoA activity is localized in real time to a subpopulation of motile leader cells displaying a hyper-protrusive leading edge, Vangl protein is localized to leader cell protrusions within leader cells, and actin cytoskeletal regulator RhoA is preferentially activated in the leader cells of a migrating collective. Mammary gland-specific knockout of Vangl2 results in a striking decrease in lung metastases in MMTV-NDL mice, but does not impact primary tumor growth characteristics. CONCLUSIONS We conclude that Vangl-dependent Wnt/PCP signaling promotes breast cancer collective cell migration independent of breast tumor subtype and facilitates distant metastasis in a genetically engineered mouse model of breast cancer. Our observations are consistent with a model whereby Vangl proteins localized at the leading edge of leader cells in a migrating collective act through RhoA to mediate the cytoskeletal rearrangements required for pro-migratory protrusion formation.
Collapse
Affiliation(s)
- Kacey VanderVorst
- Department of Biochemistry and Molecular Medicine and University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Courtney A Dreyer
- Department of Biochemistry and Molecular Medicine and University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Jason Hatakeyama
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - George R R Bell
- Department of Microbiology and Molecular Genetics, University of California Davis, Davis, CA, USA
| | - Julie A Learn
- Department of Biochemistry and Molecular Medicine and University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Anastasia L Berg
- Department of Biochemistry and Molecular Medicine and University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Maria Hernandez
- Department of Biochemistry and Molecular Medicine and University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Hyun Lee
- Department of Biochemistry and Molecular Medicine and University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Sean R Collins
- Department of Microbiology and Molecular Genetics, University of California Davis, Davis, CA, USA
| | - Kermit L Carraway
- Department of Biochemistry and Molecular Medicine and University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
16
|
Ganguly D, Schmidt MO, Coleman M, Ngo TVC, Sorrelle N, Dominguez AT, Murimwa GZ, Toombs JE, Lewis C, Fang YV, Valdes-Mora F, Gallego-Ortega D, Wellstein A, Brekken RA. Pleiotrophin drives a prometastatic immune niche in breast cancer. J Exp Med 2023; 220:e20220610. [PMID: 36828390 PMCID: PMC9998964 DOI: 10.1084/jem.20220610] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 11/21/2022] [Accepted: 01/09/2023] [Indexed: 11/04/2022] Open
Abstract
Metastatic cancer cells adapt to thrive in secondary organs. To investigate metastatic adaptation, we performed transcriptomic analysis of metastatic and non-metastatic murine breast cancer cells. We found that pleiotrophin (PTN), a neurotrophic cytokine, is a metastasis-associated factor that is expressed highly by aggressive breast cancers. Moreover, elevated PTN in plasma correlated significantly with metastasis and reduced survival of breast cancer patients. Mechanistically, we find that PTN activates NF-κB in cancer cells leading to altered cytokine production, subsequent neutrophil recruitment, and an immune suppressive microenvironment. Consequently, inhibition of PTN, pharmacologically or genetically, reduces the accumulation of tumor-associated neutrophils and reverts local immune suppression, resulting in increased T cell activation and attenuated metastasis. Furthermore, inhibition of PTN significantly enhanced the efficacy of immune checkpoint blockade and chemotherapy in reducing metastatic burden in mice. These findings establish PTN as a previously unrecognized driver of a prometastatic immune niche and thus represents a promising therapeutic target for the treatment of metastatic breast cancer.
Collapse
Affiliation(s)
- Debolina Ganguly
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cancer Biology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marcel O. Schmidt
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Morgan Coleman
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tuong-Vi Cindy Ngo
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Noah Sorrelle
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Adrian T.A. Dominguez
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gilbert Z. Murimwa
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jason E. Toombs
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Cheryl Lewis
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yisheng V. Fang
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Fatima Valdes-Mora
- Cancer Epigenetic Biology and Therapeutics group, Precision Medicine Theme, Children’s Cancer Institute, Sydney, Australia
- School of Clinical Medicine, University of NSW Sydney, Sydney, Australia
| | - David Gallego-Ortega
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, Australia
- Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, Australia
- School of Clinical Medicine, St Vincent’s Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
| | - Anton Wellstein
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Rolf A. Brekken
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cancer Biology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
17
|
Webb ER, Dodd GL, Noskova M, Bullock E, Muir M, Frame MC, Serrels A, Brunton VG. Kindlin-1 regulates IL-6 secretion and modulates the immune environment in breast cancer models. eLife 2023; 12:e85739. [PMID: 36883731 PMCID: PMC10023156 DOI: 10.7554/elife.85739] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/08/2023] [Indexed: 03/09/2023] Open
Abstract
The adhesion protein Kindlin-1 is over-expressed in breast cancer where it is associated with metastasis-free survival; however, the mechanisms involved are poorly understood. Here, we report that Kindlin-1 promotes anti-tumor immune evasion in mouse models of breast cancer. Deletion of Kindlin-1 in Met-1 mammary tumor cells led to tumor regression following injection into immunocompetent hosts. This was associated with a reduction in tumor infiltrating Tregs. Similar changes in T cell populations were seen following depletion of Kindlin-1 in the polyomavirus middle T antigen (PyV MT)-driven mouse model of spontaneous mammary tumorigenesis. There was a significant increase in IL-6 secretion from Met-1 cells when Kindlin-1 was depleted and conditioned media from Kindlin-1-depleted cells led to a decrease in the ability of Tregs to suppress the proliferation of CD8+ T cells, which was dependent on IL-6. In addition, deletion of tumor-derived IL-6 in the Kindlin-1-depleted tumors reversed the reduction of tumor-infiltrating Tregs. Overall, these data identify a novel function for Kindlin-1 in regulation of anti-tumor immunity, and that Kindlin-1 dependent cytokine secretion can impact the tumor immune environment.
Collapse
Affiliation(s)
- Emily R Webb
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Georgia L Dodd
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Michaela Noskova
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Esme Bullock
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Morwenna Muir
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Margaret C Frame
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Alan Serrels
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Valerie G Brunton
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
18
|
Bouchalova P, Bouchal P. Current methods for studying metastatic potential of tumor cells. Cancer Cell Int 2022; 22:394. [PMID: 36494720 PMCID: PMC9733110 DOI: 10.1186/s12935-022-02801-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
Cell migration and invasiveness significantly contribute to desirable physiological processes, such as wound healing or embryogenesis, as well as to serious pathological processes such as the spread of cancer cells to form tumor metastasis. The availability of appropriate methods for studying these processes is essential for understanding the molecular basis of cancer metastasis and for identifying suitable therapeutic targets for anti-metastatic treatment. This review summarizes the current status of these methods: In vitro methods for studying cell migration involve two-dimensional (2D) assays (wound-healing/scratch assay), and methods based on chemotaxis (the Dunn chamber). The analysis of both cell migration and invasiveness in vitro require more complex systems based on the Boyden chamber principle (Transwell migration/invasive test, xCELLigence system), or microfluidic devices with three-dimensional (3D) microscopy visualization. 3D culture techniques are rapidly becoming routine and involve multicellular spheroid invasion assays or array chip-based, spherical approaches, multi-layer/multi-zone culture, or organoid non-spherical models, including multi-organ microfluidic chips. The in vivo methods are mostly based on mice, allowing genetically engineered mice models and transplant models (syngeneic mice, cell line-derived xenografts and patient-derived xenografts including humanized mice models). These methods currently represent a solid basis for the state-of-the art research that is focused on understanding metastatic fundamentals as well as the development of targeted anti-metastatic therapies, and stratified treatment in oncology.
Collapse
Affiliation(s)
- Pavla Bouchalova
- grid.10267.320000 0001 2194 0956Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| | - Pavel Bouchal
- grid.10267.320000 0001 2194 0956Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| |
Collapse
|
19
|
Wang W, Hapach LA, Griggs L, Smart K, Wu Y, Taufalele PV, Rowe MM, Young KM, Bates ME, Johnson AC, Ferrell NJ, Pozzi A, Reinhart-King CA. Diabetic hyperglycemia promotes primary tumor progression through glycation-induced tumor extracellular matrix stiffening. SCIENCE ADVANCES 2022; 8:eabo1673. [PMID: 36399580 PMCID: PMC9674287 DOI: 10.1126/sciadv.abo1673] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 09/29/2022] [Indexed: 05/31/2023]
Abstract
Diabetes mellitus is a complex metabolic disorder that is associated with an increased risk of breast cancer. Despite this correlation, the interplay between tumor progression and diabetes, particularly with regard to stiffening of the extracellular matrix, is still mechanistically unclear. Here, we established a murine model where hyperglycemia was induced before breast tumor development. Using the murine model, in vitro systems, and patient samples, we show that hyperglycemia increases tumor growth, extracellular matrix stiffness, glycation, and epithelial-mesenchymal transition of tumor cells. Upon inhibition of glycation or mechanotransduction in diabetic mice, these same metrics are reduced to levels comparable with nondiabetic tumors. Together, our study describes a novel biomechanical mechanism by which diabetic hyperglycemia promotes breast tumor progression via glycating the extracellular matrix. In addition, our work provides evidence that glycation inhibition is a potential adjuvant therapy for diabetic cancer patients due to the key role of matrix stiffening in both diseases.
Collapse
Affiliation(s)
- Wenjun Wang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Lauren A. Hapach
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Lauren Griggs
- College of Engineering, Pennsylvania State University, State College, PA 16802, USA
| | - Kyra Smart
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Yusheng Wu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Paul V. Taufalele
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Matthew M. Rowe
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Katherine M. Young
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Madison E. Bates
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Andrew C. Johnson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Nicholas J. Ferrell
- Department of Internal Medicine, Division of Nephrology, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Ambra Pozzi
- Division of Nephrology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Veterans Affairs Hospitals, Nashville, TN 37684, USA
| | - Cynthia A. Reinhart-King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
20
|
Mulens-Arias V, Nicolás-Boluda A, Carn F, Gazeau F. Cationic Polyethyleneimine (PEI)–Gold Nanocomposites Modulate Macrophage Activation and Reprogram Mouse Breast Triple-Negative MET-1 Tumor Immunological Microenvironment. Pharmaceutics 2022; 14:pharmaceutics14102234. [PMID: 36297669 PMCID: PMC9607133 DOI: 10.3390/pharmaceutics14102234] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Nanomedicines based on inorganic nanoparticles have grown in the last decades due to the nanosystems’ versatility in the coating, tuneability, and physical and chemical properties. Nonetheless, concerns have been raised regarding the immunotropic profile of nanoparticles and how metallic nanoparticles affect the immune system. Cationic polymer nanoparticles are widely used for cell transfection and proved to exert an adjuvant immunomodulatory effect that improves the efficiency of conventional vaccines against infection or cancer. Likewise, gold nanoparticles (AuNPs) also exhibit diverse effects on immune response depending on size or coatings. Photothermal or photodynamic therapy, radiosensitization, and drug or gene delivery systems take advantage of the unique properties of AuNPs to deeply modify the tumoral ecosystem. However, the collective effects that AuNPs combined with cationic polymers might exert on their own in the tumor immunological microenvironment remain elusive. The purpose of this study was to analyze the triple-negative breast tumor immunological microenvironment upon intratumoral injection of polyethyleneimine (PEI)–AuNP nanocomposites (named AuPEI) and elucidate how it might affect future immunotherapeutic approaches based on this nanosystem. AuPEI nanocomposites were synthesized through a one-pot synthesis method with PEI as both a reducing and capping agent, resulting in fractal assemblies of about 10 nm AuNPs. AuPEI induced an inflammatory profile in vitro in the mouse macrophage-like cells RAW264.7 as determined by the secretion of TNF-α and CCL5 while the immunosuppressor IL-10 was not increased. However, in vivo in the mouse breast MET-1 tumor model, AuPEI nanocomposites shifted the immunological tumor microenvironment toward an M2 phenotype with an immunosuppressive profile as determined by the infiltration of PD-1-positive lymphocytes. This dichotomy in AuPEI nanocomposites in vitro and in vivo might be attributed to the highly complex tumor microenvironment and highlights the importance of testing the immunogenicity of nanomaterials in vitro and more importantly in vivo in relevant immunocompetent mouse tumor models to better elucidate any adverse or unexpected effect.
Collapse
Affiliation(s)
- Vladimir Mulens-Arias
- Matière et Systèmes Complexes (MSC), Université Paris Cité, CNRS, 45 rue des Saints Pères, 75006 Paris, France
- Integrative Biomedical Materials and Nanomedicine Lab, Department of Medicine and Life Sciences (MELIS), Pompeu Fabra University, PRBB, Carrer Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Alba Nicolás-Boluda
- Matière et Systèmes Complexes (MSC), Université Paris Cité, CNRS, 45 rue des Saints Pères, 75006 Paris, France
| | - Florent Carn
- Matière et Systèmes Complexes (MSC), Université Paris Cité, CNRS, 45 rue des Saints Pères, 75006 Paris, France
| | - Florence Gazeau
- Matière et Systèmes Complexes (MSC), Université Paris Cité, CNRS, 45 rue des Saints Pères, 75006 Paris, France
- Correspondence:
| |
Collapse
|
21
|
Roweth HG, Malloy MW, Goreczny GJ, Becker IC, Guo Q, Mittendorf EA, Italiano JE, McAllister SS, Battinelli EM. Pro-inflammatory megakaryocyte gene expression in murine models of breast cancer. SCIENCE ADVANCES 2022; 8:eabo5224. [PMID: 36223471 PMCID: PMC9555784 DOI: 10.1126/sciadv.abo5224] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 08/24/2022] [Indexed: 06/16/2023]
Abstract
Despite abundant research demonstrating that platelets can promote tumor cell metastasis, whether primary tumors affect platelet-producing megakaryocytes remains understudied. In this study, we used a spontaneous murine model of breast cancer to show that tumor burden reduced megakaryocyte number and size and disrupted polyploidization. Single-cell RNA sequencing demonstrated that megakaryocytes from tumor-bearing mice exhibit a pro-inflammatory phenotype, epitomized by increased Ctsg, Lcn2, S100a8, and S100a9 transcripts. Protein S100A8/A9 and lipocalin-2 levels were also increased in platelets, suggesting that tumor-induced alterations to megakaryocytes are passed on to their platelet progeny, which promoted in vitro tumor cell invasion and tumor cell lung colonization to a greater extent than platelets from wild-type animals. Our study is the first to demonstrate breast cancer-induced alterations in megakaryocytes, leading to qualitative changes in platelet content that may feedback to promote tumor metastasis.
Collapse
Affiliation(s)
- Harvey G. Roweth
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Michael W. Malloy
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Gregory J. Goreczny
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Isabelle C. Becker
- Harvard Medical School, Boston, MA 02115, USA
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Qiuchen Guo
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Elizabeth A. Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Breast Oncology Program, Dana-Farber/Brigham and Women’s Cancer Center, Boston, MA 02215, USA
- Ludwig Centre for Cancer Research at Harvard, Harvard Medical School, Boston, MA 02215, USA
| | - Joseph E. Italiano
- Harvard Medical School, Boston, MA 02115, USA
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Sandra S. McAllister
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Elisabeth M. Battinelli
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
22
|
Bismuth M, Katz S, Mano T, Aronovich R, Hershkovitz D, Exner AA, Ilovitsh T. Low frequency nanobubble-enhanced ultrasound mechanotherapy for noninvasive cancer surgery. NANOSCALE 2022; 14:13614-13627. [PMID: 36070492 DOI: 10.1039/d2nr01367c] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Scaling down the size of microbubble contrast agents to the nanometer level holds the promise for noninvasive cancer therapy. However, the small size of nanobubbles limits the obtained bioeffects as a result of ultrasound cavitation, when operating near the nanobubble resonance frequency. Here we show that coupled with low energy insonation at a frequency of 80 kHz, well below the resonance frequency of these agents, nanobubbles serve as noninvasive therapeutic warheads that trigger potent mechanical effects in tumors following a systemic injection. We demonstrate these capabilities in tissue mimicking phantoms, where a comparison of the acoustic response of micro- and nano-bubbles after insonation at a frequency of 250 or 80 kHz revealed that higher pressures were needed to implode the nanobubbles compared to microbubbles. Complete nanobubble destruction was achieved at a mechanical index of 2.6 for the 250 kHz insonation vs. 1.2 for the 80 kHz frequency. Thus, the 80 kHz insonation complies with safety regulations that recommend operation below a mechanical index of 1.9. In vitro in breast cancer tumor cells, the cell viability was reduced to 17.3 ± 1.7% of live cells. In vivo, in a breast cancer tumor mouse model, nanobubble tumor distribution and accumulation were evaluated by high frequency ultrasound imaging. Finally, nanobubble-mediated low frequency insonation of breast cancer tumors resulted in effective mechanical tumor ablation and tumor tissue fractionation. This approach provides a unique theranostic platform for safe, noninvasive and low energy tumor mechanotherapy.
Collapse
Affiliation(s)
- Mike Bismuth
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Sharon Katz
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel.
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tamar Mano
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Ramona Aronovich
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Dov Hershkovitz
- Department of Pathology, Tel-Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 6997800, Israel
| | - Agata A Exner
- Departments of Radiology and Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Tali Ilovitsh
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel.
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
23
|
Ieguchi K, Funakoshi M, Mishima T, Takizawa K, Omori T, Nakamura F, Watanabe M, Tsuji M, Kiuchi Y, Kobayashi S, Tsunoda T, Maru Y, Wada S. The Sympathetic Nervous System Contributes to the Establishment of Pre-Metastatic Pulmonary Microenvironments. Int J Mol Sci 2022; 23:ijms231810652. [PMID: 36142564 PMCID: PMC9501257 DOI: 10.3390/ijms231810652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 12/02/2022] Open
Abstract
Emerging evidence suggests that neural activity contributes to tumor initiation and its acquisition of metastatic properties. More specifically, it has been reported that the sympathetic nervous system regulates tumor angiogenesis, tumor growth, and metastasis. The function of the sympathetic nervous system in primary tumors has been gradually elucidated. However, its functions in pre-metastatic environments and/or the preparation of metastatic environments far from the primary sites are still unknown. To investigate the role of the sympathetic nervous system in pre-metastatic environments, we performed chemical sympathectomy using 6-OHDA in mice and observed a decrease in lung metastasis by attenuating the recruitment of myeloid-derived suppressor cells. Furthermore, we note that neuro-immune cell interactions could be observed in tumor-bearing mouse lungs in conjunction with the decreased expression of Sema3A. These data indicate that the sympathetic nervous system contributes to the preparation of pre-metastatic microenvironments in the lungs, which are mediated by neuro-immune cell interactions.
Collapse
Affiliation(s)
- Katsuaki Ieguchi
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya, Tokyo 157-8577, Japan
- Department of Pharmacology, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya, Tokyo 157-8577, Japan
| | - Masabumi Funakoshi
- Department of Pharmacology, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
- Department of Peripheral Nervous System Research, National Center of Neurology and Psychiatry, National Institute of Neuroscience, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8551, Japan
| | - Taishi Mishima
- Department of Pharmacology, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Kohtaro Takizawa
- Department of Biochemistry, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Tsutomu Omori
- Department of Pharmacology, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Fumio Nakamura
- Department of Biochemistry, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Makoto Watanabe
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya, Tokyo 157-8577, Japan
- Department of Pharmacology, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
- Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
| | - Mayumi Tsuji
- Department of Pharmacology, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
- Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
| | - Yuji Kiuchi
- Department of Pharmacology, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
- Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
| | - Shinichi Kobayashi
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya, Tokyo 157-8577, Japan
| | - Takuya Tsunoda
- Department of Medicine, Division of Medical Oncology, School of Medicine, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
| | - Yoshiro Maru
- Department of Pharmacology, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
- Correspondence: (Y.M.); (S.W.); Tel.: +81-3-5269-7417 (Y.M.); +81-3-3300-5257 (S.W.)
| | - Satoshi Wada
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya, Tokyo 157-8577, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya, Tokyo 157-8577, Japan
- Department of Medicine, Division of Medical Oncology, School of Medicine, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
- Correspondence: (Y.M.); (S.W.); Tel.: +81-3-5269-7417 (Y.M.); +81-3-3300-5257 (S.W.)
| |
Collapse
|
24
|
Rodriguez-Tirado C, Entenberg D, Li J, Qian BZ, Condeelis JS, Pollard JW. Interleukin 4 Controls the Pro-Tumoral Role of Macrophages in Mammary Cancer Pulmonary Metastasis in Mice. Cancers (Basel) 2022; 14:4336. [PMID: 36077870 PMCID: PMC9454655 DOI: 10.3390/cancers14174336] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Metastasis is the systemic manifestation of cancer and the main cause of death from breast cancer. In mouse models of lung metastases, recruitment of classical monocytes from blood to the lung and their differentiation to metastasis-associated macrophages (MAMs) facilitate cancer cell extravasation, survival and growth. Ablation of MAMs or their monocytic progenitors inhibits metastasis. We hypothesized that factors controlling macrophage polarization modulate tumor cell extravasation in the lung. We evaluated whether signaling by Th1 or Th2 cytokines in macrophages affected transendothelial migration of tumor cells in vitro. Interferon gamma and LPS inhibited macrophage-dependent tumor cell extravasation while the Th2 cytokine interleukin-4 (IL4) enhanced this process. We demonstrated that IL4 receptor (IL4rα)-null mice developed fewer and smaller lung metastasis in E0771-LG mammary cancer models of this disease. Adoptive transfer of wild-type monocytes to IL4rα-deficient mice partially rescued this phenotype. IL4 signaling in macrophages controlled the expression of the chemokine receptor CXCR2, necessary for IL4-mediated tumor cell extravasation in vitro. Furthermore, IL4 signaling in macrophages regulated the transcript abundance of several other genes already causally associated with mammary cancer lung metastasis including Ccl2, Csf1, Ccr1, Hgf and Flt1. The central role of IL4 signaling in MAMs was confirmed by high-resolution intravital imaging of the lung in mice at the time of metastatic seeding, which showed reduced physical interaction between tumor cells and IL4rα-deficient macrophages. This interaction with wild-type MAMs enhanced tumor cell survival and seeding, which was lost in the IL4rα mice. These data indicate that IL4 signaling in monocytes and macrophages is key during seeding and growth of breast metastasis in the lung, as it regulates pro-tumoral paracrine signaling between cancer cells and macrophages.
Collapse
Affiliation(s)
- Carolina Rodriguez-Tirado
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - David Entenberg
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
| | - Jiufeng Li
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
| | - Bin-Zhi Qian
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
- Medical Research Council Centre for Reproductive Health, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - John S. Condeelis
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
- Department of Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
| | - Jeffrey W. Pollard
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10461, USA
| |
Collapse
|
25
|
Bell ES, Shah P, Zuela-Sopilniak N, Kim D, Varlet AA, Morival JL, McGregor AL, Isermann P, Davidson PM, Elacqua JJ, Lakins JN, Vahdat L, Weaver VM, Smolka MB, Span PN, Lammerding J. Low lamin A levels enhance confined cell migration and metastatic capacity in breast cancer. Oncogene 2022; 41:4211-4230. [PMID: 35896617 PMCID: PMC9925375 DOI: 10.1038/s41388-022-02420-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 02/07/2023]
Abstract
Aberrations in nuclear size and shape are commonly used to identify cancerous tissue. However, it remains unclear whether the disturbed nuclear structure directly contributes to the cancer pathology or is merely a consequence of other events occurring during tumorigenesis. Here, we show that highly invasive and proliferative breast cancer cells frequently exhibit Akt-driven lower expression of the nuclear envelope proteins lamin A/C, leading to increased nuclear deformability that permits enhanced cell migration through confined environments that mimic interstitial spaces encountered during metastasis. Importantly, increasing lamin A/C expression in highly invasive breast cancer cells reflected gene expression changes characteristic of human breast tumors with higher LMNA expression, and specifically affected pathways related to cell-ECM interactions, cell metabolism, and PI3K/Akt signaling. Further supporting an important role of lamins in breast cancer metastasis, analysis of lamin levels in human breast tumors revealed a significant association between lower lamin A levels, Akt signaling, and decreased disease-free survival. These findings suggest that downregulation of lamin A/C in breast cancer cells may influence both cellular physical properties and biochemical signaling to promote metastatic progression.
Collapse
Affiliation(s)
- Emily S. Bell
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY,Current address: Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA
| | - Pragya Shah
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | | | - Dongsung Kim
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Alice-Anais Varlet
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Julien L.P. Morival
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Alexandra L. McGregor
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY,Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY
| | - Philipp Isermann
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | | | - Joshua J. Elacqua
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Jonathan N. Lakins
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - Linda Vahdat
- Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Valerie M. Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA,Helen Diller Cancer Center, Department of Bioengineering and Therapeutic Sciences, and Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA
| | - Marcus B. Smolka
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
| | - Paul N. Span
- Department of Radiation Oncology, Radiotherapy & OncoImmunology laboratory, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands
| | - Jan Lammerding
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA. .,Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
26
|
Guo Q, Spasic M, Maynard AG, Goreczny GJ, Bizuayehu A, Olive JF, van Galen P, McAllister SS. Clonal barcoding with qPCR detection enables live cell functional analyses for cancer research. Nat Commun 2022; 13:3837. [PMID: 35788590 PMCID: PMC9252988 DOI: 10.1038/s41467-022-31536-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 06/21/2022] [Indexed: 11/27/2022] Open
Abstract
Single-cell analysis methods are valuable tools; however, current approaches do not easily enable live cell retrieval. That is a particular issue when further study of cells that were eliminated during experimentation could provide critical information. We report a clonal molecular barcoding method, called SunCatcher, that enables longitudinal tracking and live cell functional analysis. From complex cell populations, we generate single cell-derived clonal populations, infect each with a unique molecular barcode, and retain stocks of individual barcoded clones (BCs). We develop quantitative PCR-based and next-generation sequencing methods that we employ to identify and quantify BCs in vitro and in vivo. We apply SunCatcher to various breast cancer cell lines and combine respective BCs to create versions of the original cell lines. While the heterogeneous BC pools reproduce their original parental cell line proliferation and tumor progression rates, individual BCs are phenotypically and functionally diverse. Early spontaneous metastases can also be identified and quantified. SunCatcher thus provides a rapid and sensitive approach for studying live single-cell clones and clonal evolution, and performing functional analyses.
Collapse
Affiliation(s)
- Qiuchen Guo
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Milos Spasic
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Adam G Maynard
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Gregory J Goreczny
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Amanuel Bizuayehu
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Jessica F Olive
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Peter van Galen
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Sandra S McAllister
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA.
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA.
| |
Collapse
|
27
|
Berg AL, Rowson-Hodel A, Hu M, Keeling M, Wu H, VanderVorst K, Chen JJ, Hatakeyama J, Jilek J, Dreyer CA, Wheeler MR, Yu AM, Li Y, Carraway KL. The Cationic Amphiphilic Drug Hexamethylene Amiloride Eradicates Bulk Breast Cancer Cells and Therapy-Resistant Subpopulations with Similar Efficiencies. Cancers (Basel) 2022; 14:cancers14040949. [PMID: 35205696 PMCID: PMC8869814 DOI: 10.3390/cancers14040949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/02/2022] [Accepted: 02/02/2022] [Indexed: 12/07/2022] Open
Abstract
The resistance of cancer cell subpopulations, including cancer stem cell (CSC) populations, to apoptosis-inducing chemotherapeutic agents is a key barrier to improved outcomes for cancer patients. The cationic amphiphilic drug hexamethylene amiloride (HMA) has been previously demonstrated to efficiently kill bulk breast cancer cells independent of tumor subtype or species but acts poorly toward non-transformed cells derived from multiple tissues. Here, we demonstrate that HMA is similarly cytotoxic toward breast CSC-related subpopulations that are resistant to conventional chemotherapeutic agents, but poorly cytotoxic toward normal mammary stem cells. HMA inhibits the sphere-forming capacity of FACS-sorted human and mouse mammary CSC-related cells in vitro, specifically kills tumor but not normal mammary organoids ex vivo, and inhibits metastatic outgrowth in vivo, consistent with CSC suppression. Moreover, HMA inhibits viability and sphere formation by lung, colon, pancreatic, brain, liver, prostate, and bladder tumor cell lines, suggesting that its effects may be applicable to multiple malignancies. Our observations expose a key vulnerability intrinsic to cancer stem cells and point to novel strategies for the exploitation of cationic amphiphilic drugs in cancer treatment.
Collapse
Affiliation(s)
- Anastasia L. Berg
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Ashley Rowson-Hodel
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Michelle Hu
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Michael Keeling
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Hao Wu
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Kacey VanderVorst
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Jenny J. Chen
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Jason Hatakeyama
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Joseph Jilek
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Courtney A. Dreyer
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Madelyn R. Wheeler
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Kermit L. Carraway
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Correspondence:
| |
Collapse
|
28
|
Omokehinde T, Jotte A, Johnson RW. gp130 Cytokines Activate Novel Signaling Pathways and Alter Bone Dissemination in ER+ Breast Cancer Cells. J Bone Miner Res 2022; 37:185-201. [PMID: 34477239 PMCID: PMC8828687 DOI: 10.1002/jbmr.4430] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 08/09/2021] [Accepted: 08/29/2021] [Indexed: 02/03/2023]
Abstract
Breast cancer cells frequently home to the bone marrow, where they encounter signals that promote survival and quiescence or stimulate their proliferation. The interleukin-6 (IL-6) cytokines signal through the co-receptor glycoprotein130 (gp130) and are abundantly secreted within the bone microenvironment. Breast cancer cell expression of leukemia inhibitory factor (LIF) receptor (LIFR)/STAT3 signaling promotes tumor dormancy in the bone, but it is unclear which, if any of the cytokines that signal through LIFR, including LIF, oncostatin M (OSM), and ciliary neurotrophic factor (CNTF), promote tumor dormancy and which signaling pathways are induced. We first confirmed that LIF, OSM, and CNTF and their receptor components were expressed across a panel of breast cancer cell lines, although expression was lower in estrogen receptor-negative (ER- ) bone metastatic clones compared with parental cell lines. In estrogen receptor-positive (ER+ ) cells, OSM robustly stimulated phosphorylation of known gp130 signaling targets STAT3, ERK, and AKT, while CNTF activated STAT3 signaling. In ER- breast cancer cells, OSM alone stimulated AKT and ERK signaling. Overexpression of OSM, but not CNTF, reduced dormancy gene expression and increased ER+ breast cancer bone dissemination. Reverse-phase protein array revealed distinct and overlapping pathways stimulated by OSM, LIF, and CNTF with known roles in breast cancer progression and metastasis. In breast cancer patients, downregulation of the cytokines or receptors was associated with reduced relapse-free survival, but OSM was significantly elevated in patients with invasive disease and distant metastasis. Together these data indicate that the gp130 cytokines induce multiple signaling cascades in breast cancer cells, with a potential pro-tumorigenic role for OSM and pro-dormancy role for CNTF. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Tolu Omokehinde
- Graduate Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alec Jotte
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Biochemistry, Vanderbilt University, Nashville, TN, USA
| | - Rachelle W Johnson
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
29
|
Kheirolomoom A, Kare AJ, Ingham ES, Paulmurugan R, Robinson ER, Baikoghli M, Inayathullah M, Seo JW, Wang J, Fite BZ, Wu B, Tumbale SK, Raie MN, Cheng RH, Nichols L, Borowsky AD, Ferrara KW. In situ T-cell transfection by anti-CD3-conjugated lipid nanoparticles leads to T-cell activation, migration, and phenotypic shift. Biomaterials 2022; 281:121339. [PMID: 35078042 PMCID: PMC8892572 DOI: 10.1016/j.biomaterials.2021.121339] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 12/24/2021] [Indexed: 02/03/2023]
Abstract
Ex vivo programming of T cells can be efficacious but is complex and expensive; therefore, the development of methods to transfect T cells in situ is important. We developed and optimized anti-CD3-targeted lipid nanoparticles (aCD3-LNPs) to deliver tightly packed, reporter gene mRNA specifically to T cells. In vitro, targeted LNPs efficiently delivered mCherry mRNA to Jurkat T cells, and T-cell activation and depletion were associated with aCD3 antibody coating on the surface of LNPs. aCD3-LNPs, but not non-targeted LNPs, accumulated within the spleen following systemic injection, with mCherry and Fluc signals visible within 30 min after injection. At 24 h after aCD3-LNP injection, 2-4% of all splenic T cells and 2-7% of all circulating T cells expressed mCherry, and this was dependent on aCD3 coating density. Targeting and transfection were accompanied by systemic CD25+, OX40+, and CD69+ T-cell activation with temporary CD3e ligand loss and depletion of splenic and circulating subsets. Migration of splenic CD8a+ T cells from the white-pulp to red-pulp, and differentiation from naïve to memory and effector phenotypes, followed upon aCD3-LNP delivery. Additionally, aCD3-LNP injection stimulated the secretion of myeloid-derived chemokines and T-helper cytokines into plasma. Lastly, we administered aCD3-LNPs to tumor bearing mice and found that transfected T cells localized within tumors and tumor-draining lymph nodes following immunotherapy treatment. In summary, we show that CD3-targeted transfection is feasible, yet associated with complex immunological consequences that must be further studied for potential therapeutic applications.
Collapse
Affiliation(s)
| | - Aris J. Kare
- Stanford University, Department of Bioengineering,
Stanford, CA, USA
| | - Elizabeth S. Ingham
- University of California, Davis, Department of Biomedical
Engineering, Davis, CA 95616, USA
| | | | | | - Mo Baikoghli
- University of California, Davis, Department of Molecular
and Cellular Biology, Davis, CA, USA
| | | | - Jai W. Seo
- Stanford University, Department of Radiology, Palo Alto,
CA, USA
| | - James Wang
- Stanford University, Department of Radiology, Palo Alto,
CA, USA
| | - Brett Z. Fite
- Stanford University, Department of Radiology, Palo Alto,
CA, USA
| | - Bo Wu
- Stanford University, Department of Radiology, Palo Alto,
CA, USA
| | | | - Marina N. Raie
- Stanford University, Department of Radiology, Palo Alto,
CA, USA
| | - R. Holland Cheng
- University of California, Davis, Department of Molecular
and Cellular Biology, Davis, CA, USA
| | - Lisa Nichols
- Stanford Shared FACS Facility, Stanford University,
Stanford, CA, USA
| | | | - Katherine W. Ferrara
- Stanford University, Department of Radiology, Palo Alto,
CA, USA,Corresponding author: Katherine W. Ferrara, PhD,
Professor and Division Chief, Molecular Imaging Program at Stanford, Department
of Radiology, 3165 Porter Drive, Stanford University, Palo Alto, CA 94304,
Phone: (650)723-8906,
| |
Collapse
|
30
|
TMEM176B Regulates AKT/mTOR Signaling and Tumor Growth in Triple-Negative Breast Cancer. Cells 2021; 10:cells10123430. [PMID: 34943938 PMCID: PMC8700203 DOI: 10.3390/cells10123430] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
Abstract
TMEM176B is a member of the membrane spanning 4-domains (MS4) family of transmembrane proteins, and a putative ion channel that is expressed in immune cells and certain cancers. We aimed to understand the role of TMEM176B in cancer cell signaling, gene expression, cell proliferation, and migration in vitro, as well as tumor growth in vivo. We generated breast cancer cell lines with overexpressed and silenced TMEM176B, and a therapeutic antibody targeting TMEM176B. Proliferation and migration assays were performed in vitro, and tumor growth was evaluated in vivo. We performed gene expression and Western blot analyses to identify the most differentially regulated genes and signaling pathways in cells with TMEM176B overexpression and silencing. Silencing TMEM176B or inhibiting it with a therapeutic antibody impaired cell proliferation, while overexpression increased proliferation in vitro. Syngeneic and xenograft tumor studies revealed the attenuated growth of tumors with TMEM176B gene silencing compared with controls. We found that the AKT/mTOR signaling pathway was activated or repressed in cells overexpressing or silenced for TMEM176B, respectively. Overall, our results suggest that TMEM176B expression in breast cancer cells regulates key signaling pathways and genes that contribute to cancer cell growth and progression, and is a potential target for therapeutic antibodies.
Collapse
|
31
|
Williams MM, Hafeez SA, Christenson JL, O’Neill KI, Hammond NG, Richer JK. Reversing an Oncogenic Epithelial-to-Mesenchymal Transition Program in Breast Cancer Reveals Actionable Immune Suppressive Pathways. Pharmaceuticals (Basel) 2021; 14:ph14111122. [PMID: 34832904 PMCID: PMC8622696 DOI: 10.3390/ph14111122] [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: 09/30/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/16/2022] Open
Abstract
Approval of checkpoint inhibitors for treatment of metastatic triple negative breast cancer (mTNBC) has opened the door for the use of immunotherapies against this disease. However, not all patients with mTNBC respond to current immunotherapy approaches such as checkpoint inhibitors. Recent evidence demonstrates that TNBC metastases are more immune suppressed than primary tumors, suggesting that combination or additional immunotherapy strategies may be required to activate an anti-tumor immune attack at metastatic sites. To identify other immune suppressive mechanisms utilized by mTNBC, our group and others manipulated oncogenic epithelial-to-mesenchymal transition (EMT) programs in TNBC models to reveal differences between this breast cancer subtype and its more epithelial counterpart. This review will discuss how EMT modulation revealed several mechanisms, including tumor cell metabolism, cytokine milieu and secretion of additional immune modulators, by which mTNBC cells may suppress both the innate and adaptive anti-tumor immune responses. Many of these pathways/proteins are under preclinical or clinical investigation as therapeutic targets in mTNBC and other advanced cancers to enhance their response to chemotherapy and/or checkpoint inhibitors.
Collapse
|
32
|
Honkala A, Malhotra SV, Kummar S, Junttila MR. Harnessing the predictive power of preclinical models for oncology drug development. Nat Rev Drug Discov 2021; 21:99-114. [PMID: 34702990 DOI: 10.1038/s41573-021-00301-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2021] [Indexed: 12/21/2022]
Abstract
Recent progress in understanding the molecular basis of cellular processes, identification of promising therapeutic targets and evolution of the regulatory landscape makes this an exciting and unprecedented time to be in the field of oncology drug development. However, high costs, long development timelines and steep rates of attrition continue to afflict the drug development process. Lack of predictive preclinical models is considered one of the key reasons for the high rate of attrition in oncology. Generating meaningful and predictive results preclinically requires a firm grasp of the relevant biological questions and alignment of the model systems that mirror the patient context. In doing so, the ability to conduct both forward translation, the process of implementing basic research discoveries into practice, as well as reverse translation, the process of elucidating the mechanistic basis of clinical observations, greatly enhances our ability to develop effective anticancer treatments. In this Review, we outline issues in preclinical-to-clinical translatability of molecularly targeted cancer therapies, present concepts and examples of successful reverse translation, and highlight the need to better align tumour biology in patients with preclinical model systems including tracking of strengths and weaknesses of preclinical models throughout programme development.
Collapse
Affiliation(s)
- Alexander Honkala
- Department of Cell Development & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Sanjay V Malhotra
- Department of Cell Development & Cancer Biology, Oregon Health & Science University, Portland, OR, USA.,Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Shivaani Kummar
- Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA. .,Division of Hematology & Medical Oncology, Oregon Health & Science University, Portland, OR, USA.
| | | |
Collapse
|
33
|
Huang W, Liu Y, Luz A, Berrong M, Meyer JN, Zou Y, Swann E, Sundaramoorthy P, Kang Y, Jauhari S, Lento W, Chao N, Racioppi L. Calcium/Calmodulin Dependent Protein Kinase Kinase 2 Regulates the Expansion of Tumor-Induced Myeloid-Derived Suppressor Cells. Front Immunol 2021; 12:754083. [PMID: 34712241 PMCID: PMC8546266 DOI: 10.3389/fimmu.2021.754083] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/27/2021] [Indexed: 12/24/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a hetero geneous group of cells, which can suppress the immune response, promote tumor progression and impair the efficacy of immunotherapies. Consequently, the pharmacological targeting of MDSC is emerging as a new immunotherapeutic strategy to stimulate the natural anti-tumor immune response and potentiate the efficacy of immunotherapies. Herein, we leveraged genetically modified models and a small molecule inhibitor to validate Calcium-Calmodulin Kinase Kinase 2 (CaMKK2) as a druggable target to control MDSC accumulation in tumor-bearing mice. The results indicated that deletion of CaMKK2 in the host attenuated the growth of engrafted tumor cells, and this phenomenon was associated with increased antitumor T cell response and decreased accumulation of MDSC. The adoptive transfer of MDSC was sufficient to restore the ability of the tumor to grow in Camkk2-/- mice, confirming the key role of MDSC in the mechanism of tumor rejection. In vitro studies indicated that blocking of CaMKK2 is sufficient to impair the yield of MDSC. Surprisingly, MDSC generated from Camkk2-/- bone marrow cells also showed a higher ability to terminally differentiate toward more immunogenic cell types (e.g inflammatory macrophages and dendritic cells) compared to wild type (WT). Higher intracellular levels of reactive oxygen species (ROS) accumulated in Camkk2-/- MDSC, increasing their susceptibility to apoptosis and promoting their terminal differentiation toward more mature myeloid cells. Mechanistic studies indicated that AMP-activated protein kinase (AMPK), which is a known CaMKK2 proximal target controlling the oxidative stress response, fine-tunes ROS accumulation in MDSC. Accordingly, failure to activate the CaMKK2-AMPK axis can account for the elevated ROS levels in Camkk2-/- MDSC. These results highlight CaMKK2 as an important regulator of the MDSC lifecycle, identifying this kinase as a new druggable target to restrain MDSC expansion and enhance the efficacy of anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Wei Huang
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Yaping Liu
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Anthony Luz
- Duke University Nicholas School of the Environment, Durham, NC, United States
| | - Mark Berrong
- Duke Human Vaccine Institute, Durham, NC, United States
| | - Joel N. Meyer
- Duke University Nicholas School of the Environment, Durham, NC, United States
| | - Yujing Zou
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Excel Swann
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Pasupathi Sundaramoorthy
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Yubin Kang
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Shekeab Jauhari
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - William Lento
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Nelson Chao
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Luigi Racioppi
- Division of Hematological Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
34
|
Giacobbe A, Abate-Shen C. Modeling metastasis in mice: a closer look. Trends Cancer 2021; 7:916-929. [PMID: 34303648 DOI: 10.1016/j.trecan.2021.06.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/27/2021] [Accepted: 06/29/2021] [Indexed: 02/07/2023]
Abstract
Unraveling the multifaceted cellular and physiological processes associated with metastasis is best achieved by using in vivo models that recapitulate the requisite tumor cell-intrinsic and -extrinsic mechanisms at the organismal level. We discuss the current status of mouse models of metastasis. We consider how mouse models can refine our understanding of the underlying biological and molecular processes that promote metastasis, and we envisage how the application of new technologies will further enhance investigations of metastasis at single-cell resolution in the context of the whole organism. Our view is that investigations based on state-of-the-art mouse models can propel a holistic understanding of the biology of metastasis, which will ultimately lead to the discovery of new therapeutic opportunities.
Collapse
Affiliation(s)
- Arianna Giacobbe
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Cory Abate-Shen
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Urology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY 10032, USA; Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Avenue, New York, NY10032, USA; Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Avenue, New York, NY 10032, USA.
| |
Collapse
|
35
|
Bismuth M, Katz S, Rosenblatt H, Twito M, Aronovich R, Ilovitsh T. Acoustically Detonated Microbubbles Coupled with Low Frequency Insonation: Multiparameter Evaluation of Low Energy Mechanical Ablation. Bioconjug Chem 2021; 33:1069-1079. [PMID: 34280311 PMCID: PMC9204695 DOI: 10.1021/acs.bioconjchem.1c00203] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
![]()
Noninvasive
ultrasound surgery can be achieved using focused ultrasound
to locally affect the targeted site without damaging intervening tissues.
Mechanical ablation and histotripsy use short and intense acoustic
pulses to destroy the tissue via a purely mechanical effect. Here,
we show that coupled with low-frequency excitation, targeted microbubbles
can serve as mechanical therapeutic warheads that trigger potent mechanical
effects in tumors using focused ultrasound. Upon low frequency excitation
(250 kHz and below), high amplitude microbubble oscillations occur
at substantially lower pressures as compared to higher MHz ultrasonic
frequencies. For example, inertial cavitation was initiated at a pressure
of 75 kPa for a center frequency of 80 kHz. Low frequency insonation
of targeted microbubbles was then used to achieve low energy tumor
cell fractionation at pressures below a mechanical index of 1.9, and
in accordance with the Food and Drug Administration guidelines. We
demonstrate these capabilities in vitro and in vivo. In cell cultures,
cell viability was reduced to 16% at a peak negative pressure of 800
kPa at the 250 kHz frequency (mechanical index of 1.6) and to 10%
at a peak negative pressure of 250 kPa at a frequency of 80 kHz (mechanical
index of 0.9). Following an intratumoral injection of targeted microbubbles
into tumor-bearing mice, and coupled with low frequency ultrasound
application, significant tumor debulking and cancer cell death was
observed. Our findings suggest that reducing the center frequency
enhances microbubble-mediated mechanical ablation; thus, this technology
provides a unique theranostic platform for safe low energy tumor fractionation,
while reducing off-target effects.
Collapse
Affiliation(s)
- Mike Bismuth
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Sharon Katz
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel.,The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Hagar Rosenblatt
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Maayan Twito
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ramona Aronovich
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tali Ilovitsh
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel.,The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
36
|
Shani O, Raz Y, Monteran L, Scharff Y, Levi-Galibov O, Megides O, Shacham H, Cohen N, Silverbush D, Avivi C, Sharan R, Madi A, Scherz-Shouval R, Barshack I, Tsarfaty I, Erez N. Evolution of fibroblasts in the lung metastatic microenvironment is driven by stage-specific transcriptional plasticity. eLife 2021; 10:e60745. [PMID: 34169837 PMCID: PMC8257251 DOI: 10.7554/elife.60745] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 06/24/2021] [Indexed: 12/21/2022] Open
Abstract
Mortality from breast cancer is almost exclusively a result of tumor metastasis, and lungs are one of the main metastatic sites. Cancer-associated fibroblasts are prominent players in the microenvironment of breast cancer. However, their role in the metastatic niche is largely unknown. In this study, we profiled the transcriptional co-evolution of lung fibroblasts isolated from transgenic mice at defined stage-specific time points of metastases formation. Employing multiple knowledge-based platforms of data analysis provided powerful insights on functional and temporal regulation of the transcriptome of fibroblasts. We demonstrate that fibroblasts in lung metastases are transcriptionally dynamic and plastic, and reveal stage-specific gene signatures that imply functional tasks, including extracellular matrix remodeling, stress response, and shaping the inflammatory microenvironment. Furthermore, we identified Myc as a central regulator of fibroblast rewiring and found that stromal upregulation of Myc transcriptional networks is associated with disease progression in human breast cancer.
Collapse
Affiliation(s)
- Ophir Shani
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Yael Raz
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
- Department of Obstetrics and Gynecology, Tel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Lea Monteran
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Ye'ela Scharff
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Oshrat Levi-Galibov
- Department of Biomolecular Sciences, The Weizmann Institute of ScienceRehovotIsrael
| | - Or Megides
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Hila Shacham
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Noam Cohen
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Dana Silverbush
- Blavatnik School of Computer Sciences, Faculty of Exact Sciences, Tel Aviv UniversityTel AvivIsrael
| | - Camilla Avivi
- Department of Pathology, Sheba Medical Center, Tel Hashomer, affiliated with Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Roded Sharan
- Blavatnik School of Computer Sciences, Faculty of Exact Sciences, Tel Aviv UniversityTel AvivIsrael
| | - Asaf Madi
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Ruth Scherz-Shouval
- Department of Biomolecular Sciences, The Weizmann Institute of ScienceRehovotIsrael
| | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Tel Hashomer, affiliated with Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Ilan Tsarfaty
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Neta Erez
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| |
Collapse
|
37
|
Nicolas-Boluda A, Vaquero J, Vimeux L, Guilbert T, Barrin S, Kantari-Mimoun C, Ponzo M, Renault G, Deptula P, Pogoda K, Bucki R, Cascone I, Courty J, Fouassier L, Gazeau F, Donnadieu E. Tumor stiffening reversion through collagen crosslinking inhibition improves T cell migration and anti-PD-1 treatment. eLife 2021; 10:58688. [PMID: 34106045 PMCID: PMC8203293 DOI: 10.7554/elife.58688] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/05/2021] [Indexed: 12/17/2022] Open
Abstract
Only a fraction of cancer patients benefits from immune checkpoint inhibitors. This may be partly due to the dense extracellular matrix (ECM) that forms a barrier for T cells. Comparing five preclinical mouse tumor models with heterogeneous tumor microenvironments, we aimed to relate the rate of tumor stiffening with the remodeling of ECM architecture and to determine how these features affect intratumoral T cell migration. An ECM-targeted strategy, based on the inhibition of lysyl oxidase, was used. In vivo stiffness measurements were found to be strongly correlated with tumor growth and ECM crosslinking but negatively correlated with T cell migration. Interfering with collagen stabilization reduces ECM content and tumor stiffness leading to improved T cell migration and increased efficacy of anti-PD-1 blockade. This study highlights the rationale of mechanical characterizations in solid tumors to understand resistance to immunotherapy and of combining treatment strategies targeting the ECM with anti-PD-1 therapy.
Collapse
Affiliation(s)
- Alba Nicolas-Boluda
- Institut Cochin, INSERM U1016/CNRS UMR 8104, Université de Paris, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Laboratoire Matière et Systèmes Complexes (MSC), CNRS, Université de Paris, Paris, France
| | - Javier Vaquero
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, Paris, France.,TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain.,LPP (Laboratoire de physique des plasmas, UMR 7648), Sorbonne Université, Centre national de la recherche scientifique (CNRS), Ecole Polytechnique, Paris, France.,Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Barcelona, Spain
| | - Lene Vimeux
- Institut Cochin, INSERM U1016/CNRS UMR 8104, Université de Paris, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Thomas Guilbert
- Institut Cochin, INSERM U1016/CNRS UMR 8104, Université de Paris, Paris, France
| | - Sarah Barrin
- Institut Cochin, INSERM U1016/CNRS UMR 8104, Université de Paris, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Chahrazade Kantari-Mimoun
- Institut Cochin, INSERM U1016/CNRS UMR 8104, Université de Paris, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Matteo Ponzo
- CNRS ERL 9215, CRRET laboratory, University of Paris-Est Créteil (UPEC), Paris, France
| | - Gilles Renault
- Institut Cochin, INSERM U1016/CNRS UMR 8104, Université de Paris, Paris, France
| | - Piotr Deptula
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Katarzyna Pogoda
- Institute of Nuclear Physics, Polish Academy of Sciences, Kraków, Poland
| | - Robert Bucki
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Ilaria Cascone
- CNRS ERL 9215, CRRET laboratory, University of Paris-Est Créteil (UPEC), Paris, France
| | - José Courty
- CNRS ERL 9215, CRRET laboratory, University of Paris-Est Créteil (UPEC), Paris, France
| | - Laura Fouassier
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, Paris, France
| | - Florence Gazeau
- Laboratoire Matière et Systèmes Complexes (MSC), CNRS, Université de Paris, Paris, France
| | - Emmanuel Donnadieu
- Institut Cochin, INSERM U1016/CNRS UMR 8104, Université de Paris, Paris, France.,Equipe Labellisée Ligue Contre le Cancer, Paris, France
| |
Collapse
|
38
|
Bushnell GG, Deshmukh AP, den Hollander P, Luo M, Soundararajan R, Jia D, Levine H, Mani SA, Wicha MS. Breast cancer dormancy: need for clinically relevant models to address current gaps in knowledge. NPJ Breast Cancer 2021; 7:66. [PMID: 34050189 PMCID: PMC8163741 DOI: 10.1038/s41523-021-00269-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/08/2021] [Indexed: 02/04/2023] Open
Abstract
Breast cancer is the most commonly diagnosed cancer in the USA. Although advances in treatment over the past several decades have significantly improved the outlook for this disease, most women who are diagnosed with estrogen receptor positive disease remain at risk of metastatic relapse for the remainder of their life. The cellular source of late relapse in these patients is thought to be disseminated tumor cells that reactivate after a long period of dormancy. The biology of these dormant cells and their natural history over a patient's lifetime is largely unclear. We posit that research on tumor dormancy has been significantly limited by the lack of clinically relevant models. This review will discuss existing dormancy models, gaps in biological understanding, and propose criteria for future models to enhance their clinical relevance.
Collapse
Affiliation(s)
- Grace G Bushnell
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Abhijeet P Deshmukh
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Petra den Hollander
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ming Luo
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Rama Soundararajan
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dongya Jia
- Center for Theoretical Biological Physics, Rice University, Houston, TX, USA
| | - Herbert Levine
- Center for Theoretical Biological Physics and Departments of Physics and Bioengineering, Northeastern University, Boston, MA, USA.
| | - Sendurai A Mani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Max S Wicha
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
39
|
Williams MM, Christenson JL, O'Neill KI, Hafeez SA, Ihle CL, Spoelstra NS, Slansky JE, Richer JK. MicroRNA-200c restoration reveals a cytokine profile to enhance M1 macrophage polarization in breast cancer. NPJ Breast Cancer 2021; 7:64. [PMID: 34045467 PMCID: PMC8160264 DOI: 10.1038/s41523-021-00273-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 05/03/2021] [Indexed: 11/21/2022] Open
Abstract
Many immune suppressive mechanisms utilized by triple negative breast cancer (TNBC) are regulated by oncogenic epithelial-to-mesenchymal transition (EMT). How TNBC EMT impacts innate immune cells is not fully understood. To determine how TNBC suppresses antitumor macrophages, we used microRNA-200c (miR-200c), a powerful repressor of EMT, to drive mesenchymal-like mouse mammary carcinoma and human TNBC cells toward a more epithelial state. MiR-200c restoration significantly decreased growth of mouse mammary carcinoma Met-1 cells in culture and in vivo. Cytokine profiling of Met-1 and human BT549 cells revealed that miR-200c upregulated cytokines, such as granulocyte-macrophage colony-stimulating factor (GM-CSF), promoted M1 antitumor macrophage polarization. Cytokines upregulated by miR-200c correlated with an epithelial gene signature and M1 macrophage polarization in BC patients and predicted a more favorable overall survival for TNBC patients. Our findings demonstrate that immunogenic cytokines (e.g., GM-CSF) are suppressed in aggressive TNBC, warranting further investigation of cytokine-based therapies to limit disease recurrence.
Collapse
Affiliation(s)
- Michelle M Williams
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jessica L Christenson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kathleen I O'Neill
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sabrina A Hafeez
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Claire L Ihle
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Nicole S Spoelstra
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jill E Slansky
- Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
40
|
Vogel CFA, Lazennec G, Kado SY, Dahlem C, He Y, Castaneda A, Ishihara Y, Vogeley C, Rossi A, Haarmann-Stemmann T, Jugan J, Mori H, Borowsky AD, La Merrill MA, Sweeney C. Targeting the Aryl Hydrocarbon Receptor Signaling Pathway in Breast Cancer Development. Front Immunol 2021; 12:625346. [PMID: 33763068 PMCID: PMC7982668 DOI: 10.3389/fimmu.2021.625346] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/11/2021] [Indexed: 01/09/2023] Open
Abstract
Activation of the aryl hydrocarbon receptor (AhR) through environmental exposure to known human carcinogens including dioxins can lead to the promotion of breast cancer. While the repressor protein of the AhR (AhRR) blocks the canonical AhR pathway, the function of AhRR in the development of breast cancer is not well-known. In the current study we examined the impact of suppressing AhR activity using its dedicated repressor protein AhRR. AhRR is a putative tumor suppressor and is silenced in several cancer types, including breast, where its loss correlates with shorter patient survival. Using the AhRR transgenic mouse, we demonstrate that AhRR overexpression opposes AhR-driven and inflammation-induced growth of mammary tumors in two different murine models of breast cancer. These include a syngeneic model using E0771 mammary tumor cells as well as the Polyoma Middle T antigen (PyMT) transgenic model. Further AhRR overexpression or knockout of AhR in human breast cancer cells enhanced apoptosis induced by chemotherapeutics and inhibited the growth of mouse mammary tumor cells. This study provides the first in vivo evidence that AhRR suppresses mammary tumor development and suggests that strategies which lead to its functional restoration and expression may have therapeutic benefit.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Antigens, Polyomavirus Transforming/genetics
- Antineoplastic Agents/pharmacology
- Apoptosis
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Proliferation
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Doxorubicin/pharmacology
- Drug Resistance, Neoplasm
- Etoposide/pharmacology
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- MCF-7 Cells
- Mice, Inbred C57BL
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, Aryl Hydrocarbon/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Signal Transduction/drug effects
- Time Factors
- Tumor Burden
- Tumor Cells, Cultured
- Mice
Collapse
Affiliation(s)
- Christoph F. A. Vogel
- Department of Environmental Toxicology, University of California, Davis, Davis, CA, United States
- Center for Health and the Environment, University of California, Davis, Davis, CA, United States
| | | | - Sarah Y. Kado
- Center for Health and the Environment, University of California, Davis, Davis, CA, United States
| | - Carla Dahlem
- Center for Health and the Environment, University of California, Davis, Davis, CA, United States
| | - Yi He
- Center for Health and the Environment, University of California, Davis, Davis, CA, United States
| | - Alejandro Castaneda
- Center for Health and the Environment, University of California, Davis, Davis, CA, United States
| | - Yasuhiro Ishihara
- Center for Health and the Environment, University of California, Davis, Davis, CA, United States
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Christian Vogeley
- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Andrea Rossi
- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | | | - Juliann Jugan
- Department of Environmental Toxicology, University of California, Davis, Davis, CA, United States
| | - Hidetoshi Mori
- Center for Comparative Medicine, University of California, Davis, Davis, CA, United States
| | - Alexander D. Borowsky
- Center for Comparative Medicine, University of California, Davis, Davis, CA, United States
| | - Michele A. La Merrill
- Department of Environmental Toxicology, University of California, Davis, Davis, CA, United States
| | - Colleen Sweeney
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
41
|
Hillers-Ziemer LE, Williams AE, Janquart A, Grogan C, Thompson V, Sanchez A, Arendt LM. Obesity-Activated Lung Stromal Cells Promote Myeloid Lineage Cell Accumulation and Breast Cancer Metastasis. Cancers (Basel) 2021; 13:1005. [PMID: 33670906 PMCID: PMC7957630 DOI: 10.3390/cancers13051005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/13/2021] [Accepted: 02/23/2021] [Indexed: 12/25/2022] Open
Abstract
Obesity is correlated with increased incidence of breast cancer metastasis; however, the mechanisms underlying how obesity promotes metastasis are unclear. In a diet-induced obese mouse model, obesity enhanced lung metastasis in both the presence and absence of primary mammary tumors and increased recruitment of myeloid lineage cells into the lungs. In the absence of tumors, obese mice demonstrated increased numbers of myeloid lineage cells and elevated collagen fibers within the lung stroma, reminiscent of premetastatic niches formed by primary tumors. Lung stromal cells isolated from obese tumor-naïve mice showed increased proliferation, contractility, and expression of extracellular matrix, inflammatory markers and transforming growth factor beta-1 (TGFβ1). Conditioned media from lung stromal cells from obese mice promoted myeloid lineage cell migration in vitro in response to colony-stimulating factor 2 (CSF2) expression and enhanced invasion of tumor cells. Together, these results suggest that prior to tumor formation, obesity alters the lung microenvironment, creating niches conducive to metastatic growth.
Collapse
Affiliation(s)
- Lauren E. Hillers-Ziemer
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Abbey E. Williams
- Program in Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Amanda Janquart
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (A.J.); (C.G.); (V.T.); (A.S.)
| | - Caitlin Grogan
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (A.J.); (C.G.); (V.T.); (A.S.)
| | - Victoria Thompson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (A.J.); (C.G.); (V.T.); (A.S.)
| | - Adriana Sanchez
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (A.J.); (C.G.); (V.T.); (A.S.)
| | - Lisa M. Arendt
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA;
- Program in Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA;
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (A.J.); (C.G.); (V.T.); (A.S.)
| |
Collapse
|
42
|
Zhang H, Tang WL, Kheirolomoom A, Fite BZ, Wu B, Lau K, Baikoghli M, Raie MN, Tumbale SK, Foiret J, Ingham ES, Mahakian LM, Tam SM, Cheng RH, Borowsky AD, Ferrara KW. Development of thermosensitive resiquimod-loaded liposomes for enhanced cancer immunotherapy. J Control Release 2021; 330:1080-1094. [PMID: 33189786 PMCID: PMC7906914 DOI: 10.1016/j.jconrel.2020.11.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 11/01/2020] [Accepted: 11/08/2020] [Indexed: 12/12/2022]
Abstract
Resiquimod (R848) is a toll-like receptor 7 and 8 (TLR7/8) agonist with potent antitumor and immunostimulatory activity. However, systemic delivery of R848 is poorly tolerated because of its poor solubility in water and systemic immune activation. In order to address these limitations, we developed an intravenously-injectable formulation with R848 using thermosensitive liposomes (TSLs) as a delivery vehicle. R848 was remotely loaded into TSLs composed of DPPC: DSPC: DSPE-PEG2K (85:10:5, mol%) with 100 mM FeSO4 as the trapping agent inside. The final R848 to lipid ratio of the optimized R848-loaded TSLs (R848-TSLs) was 0.09 (w/w), 10-fold higher than the previously-reported values. R848-TSLs released 80% of R848 within 5 min at 42 °C. These TSLs were then combined with αPD-1, an immune checkpoint inhibitor, and ultrasound-mediated hyperthermia in a neu deletion (NDL) mouse mammary carcinoma model (Her2+, ER/PR negative). Combined with αPD-1, local injection of R848-TSLs showed superior efficacy with complete NDL tumor regression in both treated and abscopal sites achieved in 8 of 11 tumor bearing mice over 100 days. Immunohistochemistry confirmed enhanced CD8+ T cell infiltration and accumulation by R848-TSLs. Systemic delivery of R848-TSLs, combined with local hyperthermia and αPD-1, inhibited tumor growth and extended median survival from 28 days (non-treatment control) to 94 days. Upon re-challenge with reinjection of tumor cells, none of the previously cured mice developed tumors, as compared with 100% of age-matched control mice. The dose of R848 (10 μg for intra-tumoral injection or 6 mg/kg for intravenous injection delivered up to 4 times) was well-tolerated without weight loss or organ hypertrophy. In summary, we developed R848-TSLs that can be administered locally or systematically, resulting in tumor regression and enhanced survival when combined with αPD-1 in mouse models of breast cancer.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA; Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Wei-Lun Tang
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Azadeh Kheirolomoom
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA; Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Brett Z Fite
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Bo Wu
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Kenneth Lau
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Mo Baikoghli
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | - Marina Nura Raie
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Spencer K Tumbale
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Josquin Foiret
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Elizabeth S Ingham
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Lisa M Mahakian
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Sarah M Tam
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - R Holland Cheng
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | | | - Katherine W Ferrara
- Molecular Imaging Program, Department of Radiology, Stanford University, 3165 Porter Drive, Palo Alto, CA 94304, USA.
| |
Collapse
|
43
|
Kong D, Zhou H, Neelakantan D, Hughes CJ, Hsu JY, Srinivasan RR, Lewis MT, Ford HL. VEGF-C mediates tumor growth and metastasis through promoting EMT-epithelial breast cancer cell crosstalk. Oncogene 2021; 40:964-979. [PMID: 33299122 PMCID: PMC7867573 DOI: 10.1038/s41388-020-01539-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 10/07/2020] [Accepted: 10/22/2020] [Indexed: 12/19/2022]
Abstract
It is well established that a subset of cells within primary breast cancers can undergo an epithelial-to-mesenchymal transition (EMT), although the role of EMT in metastasis remains controversial. We previously demonstrated that breast cancer cells that had undergone an oncogenic EMT could increase metastasis of neighboring cancer cells via non-canonical paracrine-mediated activation of GLI activity that is dependent on SIX1 expression in the EMT cancer cells. However, the mechanism by which these SIX1-expressing EMT cells activate GLI signaling remained unclear. In this study, we demonstrate a novel mechanism for activation of GLI-mediated signaling in epithelial breast tumor cells via EMT cell-induced production and secretion of VEGF-C. We show that VEGF-C, secreted by breast cancer cells that have undergone an EMT, promotes paracrine-mediated increases in proliferation, migration, and invasion of epithelial breast cancer cells, via non-canonical activation of GLI-signaling. We further show that the aggressive phenotypes, including metastasis, imparted by EMT cells on adjacent epithelial cancer cells can be disrupted by either inhibiting VEGF-C in EMT cells or by knocking down NRP2, a receptor which interacts with VEGF-C, in neighboring epithelial cancer cells. Interrogation of TCGA and GEO public datasets supports the relevance of this pathway in human breast cancer, demonstrating that VEGF-C strongly correlates with activation of Hedgehog signaling and EMT in the human disease. Our study suggests that the VEGF-C/NRP2/GLI axis is a novel and conserved paracrine means by which EMT cells enhance metastasis, and provides potential targets for therapeutic intervention in this heterogeneous disease.
Collapse
Affiliation(s)
- Deguang Kong
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of General Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Hengbo Zhou
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Program in Cancer Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Deepika Neelakantan
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Program in Molecular Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Connor J Hughes
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Program in Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jessica Y Hsu
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Program in Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Michael T Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Heide L Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Program in Cancer Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Program in Molecular Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Program in Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
44
|
Utz B, Turpin R, Lampe J, Pouwels J, Klefström J. Assessment of the WAP-Myc mouse mammary tumor model for spontaneous metastasis. Sci Rep 2020; 10:18733. [PMID: 33127915 PMCID: PMC7599250 DOI: 10.1038/s41598-020-75411-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022] Open
Abstract
Breast cancer is the most common form of cancer in women. Despite significant therapeutic advances in recent years, breast cancer also still causes the greatest number of cancer-related deaths in women, the vast majority of which (> 90%) are caused by metastases. However, very few mouse mammary cancer models exist that faithfully recapitulate the multistep metastatic process in human patients. Here we assessed the suitability of a syngrafting protocol for a Myc-driven mammary tumor model (WAP-Myc) to study autochthonous metastasis. A moderate but robust spontaneous lung metastasis rate of around 25% was attained. In addition, increased T cell infiltration was observed in metastatic tumors compared to donor and syngrafted primary tumors. Thus, the WAP-Myc syngrafting protocol is a suitable tool to study the mechanisms of metastasis in MYC-driven breast cancer.
Collapse
Affiliation(s)
- Begüm Utz
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Rita Turpin
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Johanna Lampe
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jeroen Pouwels
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Juha Klefström
- Cancer Cell Circuitry Laboratory, Translational Cancer Medicine Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
45
|
Mei D, Zhao B, Zhang J, Xu H, Huang B. Nephronectin is a prognostic biomarker and promotes gastric cancer cell proliferation, migration and invasion. Histol Histopathol 2020; 35:1263-1274. [PMID: 32935851 DOI: 10.14670/hh-18-260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Gastric cancer (GC) is a malignant disease with high incidence and mortality rates worldwide. Nephronectin (NPNT) was found to be dysregulated in some kinds of cancer. The goal of our study was to explore the expression profile of NPNT based on large numbers of GC samples with detailed clinicopathological and prognostic data from our institution and the data from a public database. A total of 117 GC samples and 73 corresponding non-tumorous adjacent tissues (NATs) were obtained from GC patients and used to detect expression of NPNT through immunohistochemistry. Western blot and qRT-PCR were performed to examine expression of NPNT in GC cell lines. Our results found that the positive expression ratio of NPNT in GC tissues is significantly higher than that in NATs (p<0.001). Chi-squared analysis results showed positive expression ratio of NPNT was significantly associated with depth of tumor invasion (p=0.049) and TNM stage (p=0.017). Kaplan-Meier survival and cox analysis results showed that patients with positive NPNT protein expression tend to have poorer prognosis than those with negative NPNT expression (p=0.0032) and NPNT expression was independent prognostic factor. High expression level was seen in GC cell lines. Furthermore, through a series of cancer cell proliferation, invasion and migration associated experiments, we found that NPNT could evidently promote GC cell proliferation, invasion and migration, as well as epithelial-mesenthymal transition. In summary, NPNT was evidently overexpressed in GC and had an oncogenic role. In the future, NPNT could serve as a promising therapeutic target for treating GC patients.
Collapse
Affiliation(s)
- Di Mei
- Department of Surgical Oncology, the First Affiliated Hospital of China Medical University, Shenyang, PR China.,Department of General Surgery, Huludao Center Hospital, Huludao, PR China
| | - Bochao Zhao
- Department of Surgical Oncology, the First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Jiale Zhang
- Department of Surgical Oncology, the First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Huimian Xu
- Department of Surgical Oncology, the First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Baojun Huang
- Department of Surgical Oncology, the First Affiliated Hospital of China Medical University, Shenyang, PR China.
| |
Collapse
|
46
|
Thies KA, Hammer AM, Hildreth BE, Steck SA, Spehar JM, Kladney RD, Geisler JA, Das M, Russell LO, Bey JF, Bolyard CM, Pilarski R, Trimboli AJ, Cuitiño MC, Koivisto CS, Stover DG, Schoenfield L, Otero J, Godbout JP, Chakravarti A, Ringel MD, Ramaswamy B, Li Z, Kaur B, Leone G, Ostrowski MC, Sizemore ST, Sizemore GM. Stromal Platelet-Derived Growth Factor Receptor-β Signaling Promotes Breast Cancer Metastasis in the Brain. Cancer Res 2020; 81:606-618. [PMID: 32327406 DOI: 10.1158/0008-5472.can-19-3731] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/25/2020] [Accepted: 04/20/2020] [Indexed: 11/16/2022]
Abstract
Platelet-derived growth factor receptor-beta (PDGFRβ) is a receptor tyrosine kinase found in cells of mesenchymal origin such as fibroblasts and pericytes. Activation of this receptor is dependent on paracrine ligand induction, and its preferred ligand PDGFB is released by neighboring epithelial and endothelial cells. While expression of both PDGFRβ and PDGFB has been noted in patient breast tumors for decades, how PDGFB-to-PDGFRβ tumor-stroma signaling mediates breast cancer initiation, progression, and metastasis remains unclear. Here we demonstrate this paracrine signaling pathway that mediates both primary tumor growth and metastasis, specifically, metastasis to the brain. Elevated levels of PDGFB accelerated orthotopic tumor growth and intracranial growth of mammary tumor cells, while mesenchymal-specific expression of an activating mutant PDGFRβ (PDGFRβD849V) exerted proproliferative signals on adjacent mammary tumor cells. Stromal expression of PDGFRβD849V also promoted brain metastases of mammary tumor cells expressing high PDGFB when injected intravenously. In the brain, expression of PDGFRβD849V was observed within a subset of astrocytes, and aged mice expressing PDGFRβD849V exhibited reactive gliosis. Importantly, the PDGFR-specific inhibitor crenolanib significantly reduced intracranial growth of mammary tumor cells. In a tissue microarray comprised of 363 primary human breast tumors, high PDGFB protein expression was prognostic for brain metastases, but not metastases to other sites. Our results advocate the use of mice expressing PDGFRβD849V in their stromal cells as a preclinical model of breast cancer-associated brain metastases and support continued investigation into the clinical prognostic and therapeutic use of PDGFB-to-PDGFRβ signaling in women with breast cancer. SIGNIFICANCE: These studies reveal a previously unknown role for PDGFB-to-PDGFRβ paracrine signaling in the promotion of breast cancer brain metastases and support the prognostic and therapeutic clinical utility of this pathway for patients.See related article by Wyss and colleagues, p. 594.
Collapse
Affiliation(s)
- Katie A Thies
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Anisha M Hammer
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Blake E Hildreth
- O'Neal Comprehensive Cancer Center, University of Alabama-Birmingham, Birmingham, Alabama.,Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama-Birmingham, Birmingham, Alabama
| | - Sarah A Steck
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Jonathan M Spehar
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Raleigh D Kladney
- Department of Medicine, Molecular Oncology Division, Washington University School of Medicine, St. Louis, Missouri
| | - Jennifer A Geisler
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Manjusri Das
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Luke O Russell
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Neurological Surgery, The Ohio State University, Columbus, Ohio
| | - Jerome F Bey
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Chelsea M Bolyard
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Neurological Surgery, The Ohio State University, Columbus, Ohio
| | - Robert Pilarski
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Division of Human Genetics, The Ohio State University, Columbus, Ohio
| | - Anthony J Trimboli
- The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.,Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Maria C Cuitiño
- The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.,Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Christopher S Koivisto
- The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.,Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Daniel G Stover
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Lynn Schoenfield
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Jose Otero
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University, Columbus, Ohio.,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio
| | - Arnab Chakravarti
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Matthew D Ringel
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Bhuvaneswari Ramaswamy
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Zaibo Li
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Balveen Kaur
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Gustavo Leone
- The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.,Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Michael C Ostrowski
- The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.,Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Steven T Sizemore
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Gina M Sizemore
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio. .,Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
47
|
Huang Y, Hakamivala A, Li S, Nair A, Saxena R, Hsieh JT, Tang L. Chemokine releasing particle implants for trapping circulating prostate cancer cells. Sci Rep 2020; 10:4433. [PMID: 32157115 PMCID: PMC7064596 DOI: 10.1038/s41598-020-60696-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 02/04/2020] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer (PCa) is the most prevalent cancer in U.S. men and many other countries. Although primary PCa can be controlled with surgery or radiation, treatment options of preventing metastatic PCa are still limited. To develop a new treatment of eradicating metastatic PCa, we have created an injectable cancer trap that can actively recruit cancer cells in bloodstream. The cancer trap is composed of hyaluronic acid microparticles that have good cell and tissue compatibility and can extend the release of chemokines to 4 days in vitro. We find that erythropoietin (EPO) and stromal derived factor-1α can attract PCa in vitro. Animal results show that EPO-releasing cancer trap attracted large number of circulating PCa and significantly reduced cancer spreading to other organs compared with controls. These results support that cancer trap may serve as a unique device to sequester circulating PCa cells and subsequently reduce distant metastasis.
Collapse
Affiliation(s)
- YiHui Huang
- Department of Bioengineering, the University of Texas at Arlington, Arlington, Texas, 76019, USA
| | - Amirhossein Hakamivala
- Department of Bioengineering, the University of Texas at Arlington, Arlington, Texas, 76019, USA
| | - Shuxin Li
- Department of Bioengineering, the University of Texas at Arlington, Arlington, Texas, 76019, USA
| | - Ashwin Nair
- Department of Bioengineering, the University of Texas at Arlington, Arlington, Texas, 76019, USA
| | - Ramesh Saxena
- Division of Nephrology, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Liping Tang
- Department of Bioengineering, the University of Texas at Arlington, Arlington, Texas, 76019, USA.
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| |
Collapse
|
48
|
Patel H, Li J, Herrero A, Kroboth J, Byron A, Kriegsheim AV, Brunton V, Carragher N, Hurd T, Frame M. Novel roles of PRK1 and PRK2 in cilia and cancer biology. Sci Rep 2020; 10:3902. [PMID: 32127582 PMCID: PMC7054267 DOI: 10.1038/s41598-020-60604-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 02/10/2020] [Indexed: 12/24/2022] Open
Abstract
PRK1 and PRK2 are two closely related AGC-family serine/threonine protein kinases. Here we demonstrate novel roles for them at cilia and in cancer biology. In both instances serum withdrawal leads to increased activating PRK1 and PRK2 phosphorylation (pPRK1/pPRK2) and their depletion results in reduced spheroid growth. pPRK1/pPRK2 localise to the transition zone of cilia and their co-depletion results in reduced cilia size, impaired planer polarity and impaired cilia associated signalling. High PRK2 (but not PRK1) expression correlates with poor outcome in patients with basal-like/Triple Negative (TN) Breast Cancer (BC) where there is also higher expression relative to other BC tumour subtypes. In agreement, depletion of PRK1 and PRK2 in mouse TNBC cells, or CRISPR/Cas9 mediated deletion of PRK2 alone, significantly reduces cell proliferation and spheroid growth. Finally proteomic analysis to identify PRK2 binding partners in mouse TNBC cells revealed proteins that are important for both cilia and BC biology. Taken together these data demonstrate novel roles for PRK1 and PRK2 at cilia and in BC biology and in the case of PRK2 in particular, identifies it as a novel TNBC therapeutic target.
Collapse
Affiliation(s)
- Hitesh Patel
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom.
- University of Sussex, Sussex Drug Discovery Centre, School of Life Sciences, Brighton, BN1 9QJ, United Kingdom.
| | - Jun Li
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom
| | - Ana Herrero
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom
| | - Jakob Kroboth
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom
| | - Adam Byron
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom
| | - Alex Von Kriegsheim
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom
| | - Valerie Brunton
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom
| | - Neil Carragher
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom
| | - Toby Hurd
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom.
| | - Margaret Frame
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom
| |
Collapse
|
49
|
Obesity Promotes Cooperation of Cancer Stem-Like Cells and Macrophages to Enhance Mammary Tumor Angiogenesis. Cancers (Basel) 2020; 12:cancers12020502. [PMID: 32098183 PMCID: PMC7072330 DOI: 10.3390/cancers12020502] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/13/2020] [Accepted: 02/19/2020] [Indexed: 02/06/2023] Open
Abstract
Obesity is correlated with worsened prognosis and treatment resistance in breast cancer. Macrophage-targeted therapies are currently in clinical trials, however, little is known about how obesity may impact treatment efficacy. Within breast adipose tissue, obesity leads to chronic, macrophage-driven inflammation, suggesting that obese breast cancer patients may benefit from these therapies. Using a high fat diet model of obesity, we orthotopically transplanted cancer cell lines into the mammary glands of obese and lean mice. We quantified changes in tumor invasiveness, angiogenesis and metastasis, and examined the efficacy of macrophage depletion to diminish tumor progression in obese and lean mice. Mammary tumors from obese mice grew significantly faster, were enriched for cancer stem-like cells (CSCs) and were more locally invasive and metastatic. Tumor cells isolated from obese mice demonstrated enhanced expression of stem cell-related pathways including Sox2 and Notch2. Despite more rapid growth, mammary tumors from obese mice had reduced necrosis, higher blood vessel density, and greater macrophage recruitment. Depletion of macrophages in obese tumor-bearing mice resulted in increased tumor necrosis, reduced endothelial cells, and enhanced recruitment of CD8+ T cells compared to IgG-treated controls. Macrophages may be an important clinical target to improve treatment options for obese breast cancer patients.
Collapse
|
50
|
Cai D, Wang J, Gao B, Li J, Wu F, Zou JX, Xu J, Jiang Y, Zou H, Huang Z, Borowsky AD, Bold RJ, Lara PN, Li JJ, Chen X, Lam KS, To KF, Kung HJ, Fiehn O, Zhao R, Evans RM, Chen HW. RORγ is a targetable master regulator of cholesterol biosynthesis in a cancer subtype. Nat Commun 2019; 10:4621. [PMID: 31604910 PMCID: PMC6789042 DOI: 10.1038/s41467-019-12529-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 08/12/2019] [Indexed: 02/04/2023] Open
Abstract
Tumor subtype-specific metabolic reprogrammers could serve as targets of therapeutic intervention. Here we show that triple-negative breast cancer (TNBC) exhibits a hyper-activated cholesterol-biosynthesis program that is strongly linked to nuclear receptor RORγ, compared to estrogen receptor-positive breast cancer. Genetic and pharmacological inhibition of RORγ reduces tumor cholesterol content and synthesis rate while preserving host cholesterol homeostasis. We demonstrate that RORγ functions as an essential activator of the entire cholesterol-biosynthesis program, dominating SREBP2 via its binding to cholesterol-biosynthesis genes and its facilitation of the recruitment of SREBP2. RORγ inhibition disrupts its association with SREBP2 and reduces chromatin acetylation at cholesterol-biosynthesis gene loci. RORγ antagonists cause tumor regression in patient-derived xenografts and immune-intact models. Their combination with cholesterol-lowering statins elicits superior anti-tumor synergy selectively in TNBC. Together, our study uncovers a master regulator of the cholesterol-biosynthesis program and an attractive target for TNBC.
Collapse
Affiliation(s)
- Demin Cai
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Junjian Wang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Bei Gao
- West Coast Metabolomics Center, University of California Davis, Davis, CA, USA
| | - Jin Li
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Feng Wu
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - June X Zou
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Jianzhen Xu
- Shantou University Medical College, Shantou, China
| | - Yuqian Jiang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Hongye Zou
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Zenghong Huang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
| | - Alexander D Borowsky
- Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA, USA
| | - Richard J Bold
- Department of Surgery, University of California Davis, Sacramento, CA, USA
- Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
| | - Primo N Lara
- Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
| | - Jian Jian Li
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, USA
| | - Xinbin Chen
- Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
- Comparative Oncology Laboratory, University of California Davis, Davis, CA, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
- Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hsing-Jien Kung
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA
- Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California Davis, Davis, CA, USA
| | - Ruqian Zhao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing, China
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute, Howard Hughes Medical Institute, Salk Institute, La Jolla, CA, USA
| | - Hong-Wu Chen
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, USA.
- Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA.
| |
Collapse
|