51
|
Surve CR, Duran CL, Ye X, Chen X, Lin Y, Harney AS, Wang Y, Sharma VP, Stanley ER, Cox D, McAuliffe JC, Entenberg D, Oktay MH, Condeelis JS. Signaling events at TMEM doorways provide potential targets for inhibiting breast cancer dissemination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.08.574676. [PMID: 38260319 PMCID: PMC10802469 DOI: 10.1101/2024.01.08.574676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Tumor cell intravasation is essential for metastatic dissemination, but its exact mechanism is incompletely understood. We have previously shown that in breast cancer, the direct and stable association of a tumor cell expressing Mena, a Tie2hi/VEGFhi macrophage, and a vascular endothelial cell, creates an intravasation portal, called a "tumor microenvironment of metastasis" (TMEM) doorway, for tumor cell intravasation, leading to dissemination to distant sites. The density of TMEM doorways, also called TMEM doorway score, is a clinically validated prognostic marker of distant metastasis in breast cancer patients. Although we know that tumor cells utilize TMEM doorway-associated transient vascular openings to intravasate, the precise signaling mechanisms involved in TMEM doorway function are only partially understood. Using two mouse models of breast cancer and an in vitro assay of intravasation, we report that CSF-1 secreted by the TMEM doorway tumor cell stimulates local secretion of VEGF-A from the Tie2hi TMEM doorway macrophage, leading to the dissociation of endothelial junctions between TMEM doorway associated endothelial cells, supporting tumor cell intravasation. Acute blockade of CSF-1R signaling decreases macrophage VEGF-A secretion as well as TMEM doorway-associated vascular opening, tumor cell trans-endothelial migration, and dissemination. These new insights into signaling events regulating TMEM doorway function should be explored further as treatment strategies for metastatic disease.
Collapse
Affiliation(s)
- Chinmay R. Surve
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, New York
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York
| | - Camille L. Duran
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, New York
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York
- Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine, Bronx, New York
| | - Xianjun Ye
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, New York
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York
- Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine, Bronx, New York
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York
| | - Xiaoming Chen
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| | - Yu Lin
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| | - Allison S. Harney
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, New York
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York
| | - Yarong Wang
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, New York
| | - Ved P. Sharma
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York
| | - E. Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Dianne Cox
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York
- Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine, Bronx, New York
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - John C. McAuliffe
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, New York
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York
- Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine, Bronx, New York
- Department of Surgery, Albert Einstein College of Medicine, Bronx, New York
| | - David Entenberg
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, New York
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York
- Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine, Bronx, New York
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York
| | - Maja H. Oktay
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, New York
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York
- Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine, Bronx, New York
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York
- Department of Surgery, Albert Einstein College of Medicine, Bronx, New York
| | - John S. Condeelis
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, New York
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, New York
- Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine, Bronx, New York
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York
- Department of Surgery, Albert Einstein College of Medicine, Bronx, New York
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
52
|
Wijler LA, Viergever BJ, Strating E, van Schelven SJ, Poghosyan S, Frenkel NC, Te Rietmole H, Verheem A, Raats DAE, Borel Rinkes IHM, Hagendoorn J, Kranenburg O. Onward Spread from Liver Metastases Is a Major Cause of Multi-Organ Metastasis in a Mouse Model of Metastatic Colon Cancer. Cancers (Basel) 2024; 16:1073. [PMID: 38473429 DOI: 10.3390/cancers16051073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/15/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Colorectal cancer metastasizes predominantly to the liver but also to the lungs and the peritoneum. The presence of extra-hepatic metastases limits curative (surgical) treatment options and is associated with very poor survival. The mechanisms governing multi-organ metastasis formation are incompletely understood. Here, we tested the hypothesis that the site of tumor growth influences extra-hepatic metastasis formation. To this end, we implanted murine colon cancer organoids into the primary tumor site (i.e., the caecum) and into the primary metastasis site (i.e., the liver) in immunocompetent mice. The organoid-initiated liver tumors were significantly more efficient in seeding distant metastases compared to tumors of the same origin growing in the caecum (intra-hepatic: 51 vs. 40%, p = 0.001; peritoneal cavity: 51% vs. 33%, p = 0.001; lungs: 30% vs. 7%, p = 0.017). The enhanced metastatic capacity of the liver tumors was associated with the formation of 'hotspots' of vitronectin-positive blood vessels surrounded by macrophages. RNA sequencing analysis of clinical samples showed a high expression of vitronectin in liver metastases, along with signatures reflecting hypoxia, angiogenesis, coagulation, and macrophages. We conclude that 'onward spread' from liver metastases is facilitated by liver-specific microenvironmental signals that cause the formation of macrophage-associated vascular hotspots. The therapeutic targeting of these signals may help to contain the disease within the liver and prevent onward spread.
Collapse
Affiliation(s)
- Liza A Wijler
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Bastiaan J Viergever
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Esther Strating
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Susanne J van Schelven
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Susanna Poghosyan
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Nicola C Frenkel
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Hedy Te Rietmole
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Andre Verheem
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Danielle A E Raats
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Inne H M Borel Rinkes
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Jeroen Hagendoorn
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Onno Kranenburg
- Laboratory of Translational Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- Department of Surgical Oncology, Division of Imaging and Cancer, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- Utrecht Platform for Organoid Technology, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, The Netherlands
| |
Collapse
|
53
|
Albain KS, Yau C, Petricoin EF, Wolf DM, Lang JE, Chien AJ, Haddad T, Forero-Torres A, Wallace AM, Kaplan H, Pusztai L, Euhus D, Nanda R, Elias AD, Clark AS, Godellas C, Boughey JC, Isaacs C, Tripathy D, Lu J, Yung RL, Gallagher RI, Wulfkuhle JD, Brown-Swigart L, Krings G, Chen YY, Potter DA, Stringer-Reasor E, Blair S, Asare SM, Wilson A, Hirst GL, Singhrao R, Buxton M, Clennell JL, Sanil A, Berry S, Asare AL, Matthews JB, DeMichele AM, Hylton NM, Melisko M, Perlmutter J, Rugo HS, Symmans WF, van’t Veer LJ, Yee D, Berry DA, Esserman LJ. Neoadjuvant Trebananib plus Paclitaxel-based Chemotherapy for Stage II/III Breast Cancer in the Adaptively Randomized I-SPY2 Trial-Efficacy and Biomarker Discovery. Clin Cancer Res 2024; 30:729-740. [PMID: 38109213 PMCID: PMC10956403 DOI: 10.1158/1078-0432.ccr-22-2256] [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: 10/28/2022] [Revised: 10/11/2023] [Accepted: 12/13/2023] [Indexed: 12/20/2023]
Abstract
PURPOSE The neutralizing peptibody trebananib prevents angiopoietin-1 and angiopoietin-2 from binding with Tie2 receptors, inhibiting angiogenesis and proliferation. Trebananib was combined with paclitaxel±trastuzumab in the I-SPY2 breast cancer trial. PATIENTS AND METHODS I-SPY2, a phase II neoadjuvant trial, adaptively randomizes patients with high-risk, early-stage breast cancer to one of several experimental therapies or control based on receptor subtypes as defined by hormone receptor (HR) and HER2 status and MammaPrint risk (MP1, MP2). The primary endpoint is pathologic complete response (pCR). A therapy "graduates" if/when it achieves 85% Bayesian probability of success in a phase III trial within a given subtype. Patients received weekly paclitaxel (plus trastuzumab if HER2-positive) without (control) or with weekly intravenous trebananib, followed by doxorubicin/cyclophosphamide and surgery. Pathway-specific biomarkers were assessed for response prediction. RESULTS There were 134 participants randomized to trebananib and 133 to control. Although trebananib did not graduate in any signature [phase III probabilities: Hazard ratio (HR)-negative (78%), HR-negative/HER2-positive (74%), HR-negative/HER2-negative (77%), and MP2 (79%)], it demonstrated high probability of superior pCR rates over control (92%-99%) among these subtypes. Trebananib improved 3-year event-free survival (HR 0.67), with no significant increase in adverse events. Activation levels of the Tie2 receptor and downstream signaling partners predicted trebananib response in HER2-positive disease; high expression of a CD8 T-cell gene signature predicted response in HR-negative/HER2-negative disease. CONCLUSIONS The angiopoietin (Ang)/Tie2 axis inhibitor trebananib combined with standard neoadjuvant therapy increased estimated pCR rates across HR-negative and MP2 subtypes, with probabilities of superiority >90%. Further study of Ang/Tie2 receptor axis inhibitors in validated, biomarker-predicted sensitive subtypes is warranted.
Collapse
Affiliation(s)
- Kathy S. Albain
- Loyola University Chicago Stritch School of Medicine, Chicago, IL
| | - Christina Yau
- University of California San Francisco, San Francisco, CA
| | | | - Denise M. Wolf
- University of California San Francisco, San Francisco, CA
| | | | - A. Jo Chien
- University of California San Francisco, San Francisco, CA
| | | | | | | | | | | | | | | | | | | | | | | | | | - Debu Tripathy
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Janice Lu
- University of Southern California, Los Angeles, CA
| | | | | | | | | | - Gregor Krings
- University of California San Francisco, San Francisco, CA
| | - Yunn Yi Chen
- University of California San Francisco, San Francisco, CA
| | | | | | - Sarah Blair
- University of California San Diego, La Jolla, CA
| | - Smita M. Asare
- Quantum Leap Healthcare Collaborative, San Francisco, CA
| | - Amy Wilson
- Quantum Leap Healthcare Collaborative, San Francisco, CA
| | | | - Ruby Singhrao
- University of California San Francisco, San Francisco, CA
| | | | | | | | | | - Adam L. Asare
- Quantum Leap Healthcare Collaborative, San Francisco, CA
| | | | | | - Nola M. Hylton
- University of California San Francisco, San Francisco, CA
| | | | | | - Hope S. Rugo
- University of California San Francisco, San Francisco, CA
| | | | | | | | | | | |
Collapse
|
54
|
Bull JA, Mulholland EJ, Leedham SJ, Byrne HM. Extended correlation functions for spatial analysis of multiplex imaging data. BIOLOGICAL IMAGING 2024; 4:e2. [PMID: 38516631 PMCID: PMC10951806 DOI: 10.1017/s2633903x24000011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 01/11/2024] [Accepted: 01/28/2024] [Indexed: 03/23/2024]
Abstract
Imaging platforms for generating highly multiplexed histological images are being continually developed and improved. Significant improvements have also been made in the accuracy of methods for automated cell segmentation and classification. However, less attention has focused on the quantification and analysis of the resulting point clouds, which describe the spatial coordinates of individual cells. We focus here on a particular spatial statistical method, the cross-pair correlation function (cross-PCF), which can identify positive and negative spatial correlation between cells across a range of length scales. However, limitations of the cross-PCF hinder its widespread application to multiplexed histology. For example, it can only consider relations between pairs of cells, and cells must be classified using discrete categorical labels (rather than labeling continuous labels such as stain intensity). In this paper, we present three extensions to the cross-PCF which address these limitations and permit more detailed analysis of multiplex images: topographical correlation maps can visualize local clustering and exclusion between cells; neighbourhood correlation functions can identify colocalization of two or more cell types; and weighted-PCFs describe spatial correlation between points with continuous (rather than discrete) labels. We apply the extended PCFs to synthetic and biological datasets in order to demonstrate the insight that they can generate.
Collapse
Affiliation(s)
- Joshua A. Bull
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, OxfordOX2 6GG, UK
| | - Eoghan J. Mulholland
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, OxfordOX3 7BN, UK
| | - Simon J. Leedham
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, OxfordOX3 7BN, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, OxfordOX3 9DU, UK
- Oxford NIHR Biomedical Research Centre, John Radcliffe Hospital, University of Oxford, OxfordOX3 9DU, UK
| | - Helen M. Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, OxfordOX2 6GG, UK
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, OxfordOX3 7DQ, UK
| |
Collapse
|
55
|
Kim DE, Roh HS, Kim GH, Bhang DH, Um SH, Singh R, Baek KH. S6K1 deficiency in tumor stroma impairs lung metastasis of melanoma in mice. Biochem Biophys Res Commun 2024; 696:149469. [PMID: 38194806 DOI: 10.1016/j.bbrc.2024.149469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/26/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024]
Abstract
Accumulating data suggest that ribosomal protein S6 kinase 1 (S6K1), an effector in the mammalian target of rapamycin (mTOR) pathway, plays pleiotropic roles in tumor progression. However, to date, while the tumorigenic function of S6K1 in tumor cells has been well elucidated, its role in the tumor stroma remains poorly understood. We recently showed that S6K1 mediates vascular endothelial growth factor A (VEGF-A) production in macrophages, thereby supporting tumor angiogenesis and growth. As macrophage-derived VEGF-A is crucial for both tumor cell intravasation and extravasation across the vascular endothelium, our previous findings suggest that stromal S6K1 signaling is required for tumor metastatic spread. Therefore, we aimed to determine the impact of host S6K1 depletion on tumor metastasis using a murine model of pulmonary metastasis (S6k1-/- mice implanted with B16F10 melanoma). The ablation of S6K1 in the host microenvironment significantly reduced the metastasized B16F10 melanoma cells on the lung surface in both spontaneous and intravenous lung metastasis mouse models without affecting the incidence of metastasis to distant lymph nodes. In addition, stromal S6K1 loss decreased the number of tumor cells circulating in the peripheral blood of mice bearing B16F10 xenografts without affecting the vascular leakage induced by VEGF-A in vivo. These observations demonstrate that S6K1 signaling in host cells other than endothelial cells is required to modulate the host microenvironment to facilitate the metastatic spread of tumors via blood circulation, thus revealing its novel role in the tumor stroma during tumor progression.
Collapse
Affiliation(s)
- Da-Eun Kim
- Department of Molecular and Cellular Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Hyun-Soo Roh
- Department of Molecular and Cellular Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Ga-Hee Kim
- Department of Molecular and Cellular Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Dong Ha Bhang
- Department of Molecular and Cellular Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Sung Hee Um
- Department of Molecular and Cellular Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Rohit Singh
- Immuno-oncology Branch, Division of Rare and Refractory Cancer, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
| | - Kwan-Hyuck Baek
- Department of Molecular and Cellular Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi, 16419, Republic of Korea.
| |
Collapse
|
56
|
Mierke CT. Phenotypic Heterogeneity, Bidirectionality, Universal Cues, Plasticity, Mechanics, and the Tumor Microenvironment Drive Cancer Metastasis. Biomolecules 2024; 14:184. [PMID: 38397421 PMCID: PMC10887446 DOI: 10.3390/biom14020184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Tumor diseases become a huge problem when they embark on a path that advances to malignancy, such as the process of metastasis. Cancer metastasis has been thoroughly investigated from a biological perspective in the past, whereas it has still been less explored from a physical perspective. Until now, the intraluminal pathway of cancer metastasis has received the most attention, while the interaction of cancer cells with macrophages has received little attention. Apart from the biochemical characteristics, tumor treatments also rely on the tumor microenvironment, which is recognized to be immunosuppressive and, as has recently been found, mechanically stimulates cancer cells and thus alters their functions. The review article highlights the interaction of cancer cells with other cells in the vascular metastatic route and discusses the impact of this intercellular interplay on the mechanical characteristics and subsequently on the functionality of cancer cells. For instance, macrophages can guide cancer cells on their intravascular route of cancer metastasis, whereby they can help to circumvent the adverse conditions within blood or lymphatic vessels. Macrophages induce microchannel tunneling that can possibly avoid mechanical forces during extra- and intravasation and reduce the forces within the vascular lumen due to vascular flow. The review article highlights the vascular route of cancer metastasis and discusses the key players in this traditional route. Moreover, the effects of flows during the process of metastasis are presented, and the effects of the microenvironment, such as mechanical influences, are characterized. Finally, the increased knowledge of cancer metastasis opens up new perspectives for cancer treatment.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth System Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
57
|
Kienzler JC, Becher B. Immunity in malignant brain tumors: Tumor entities, role of immunotherapy, and specific contribution of myeloid cells to the brain tumor microenvironment. Eur J Immunol 2024; 54:e2250257. [PMID: 37940552 DOI: 10.1002/eji.202250257] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 10/30/2023] [Accepted: 11/07/2023] [Indexed: 11/10/2023]
Abstract
Malignant brain tumors lack effective treatment, that can improve their poor overall survival achieved with standard of care. Advancement in different cancer treatments has shifted the focus in brain tumor research and clinical trials toward immunotherapy-based approaches. The investigation of the immune cell landscape revealed a dominance of myeloid cells in the tumor microenvironment. Their exact roles and functions are the subject of ongoing research. Current evidence suggests a complex interplay of tumor cells and myeloid cells with competing functions toward support vs. control of tumor growth. Here, we provide a brief overview of the three most abundant brain tumor entities: meningioma, glioma, and brain metastases. We also describe the field of ongoing immunotherapy trials and their results, including immune checkpoint inhibitors, vaccination studies, oncolytic viral therapy, and CAR-T cells. Finally, we summarize the phenotypes of microglia, monocyte-derived macrophages, border-associated macrophages, neutrophils, and potential novel therapy targets.
Collapse
Affiliation(s)
- Jenny C Kienzler
- Institute of Experimental Immunology, Inflammation Research Lab, University of Zurich, Zurich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, Inflammation Research Lab, University of Zurich, Zurich, Switzerland
| |
Collapse
|
58
|
Chung H, Gyu-mi P, Na YR, Lee YS, Choi H, Seok SH. Comprehensive characterization of early-programmed tumor microenvironment by tumor-associated macrophages reveals galectin-1 as an immune modulatory target in breast cancer. Theranostics 2024; 14:843-860. [PMID: 38169569 PMCID: PMC10758049 DOI: 10.7150/thno.88917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
Background: In recent years, there has been considerable interest in the therapeutic targeting of tumor-associated macrophages (TAMs) to modulate the tumor microenvironment (TME), resulting in antitumoral phenotypes. However, key mediators suitable for TAM-mediated remodeling of the TME remain poorly understood. Methods: In this study, we used single-cell RNA sequencing analyses to analyze the landscape of the TME modulated by TAMs in terms of a protumor microenvironment during early tumor development. Results: Our data revealed that the depletion of TAMs leads to a decreased epithelial-to-mesenchymal transition (EMT) signature in cancer cells and a distinct transcriptional state characterized by CD8+ T cell activation. Moreover, notable alterations in gene expression were observed upon the depletion of TAMs, identifying Galectin-1 (Gal-1) as a crucial molecular factor responsible for the observed effect. Gal-1 inhibition reversed immune suppression via the reinvigoration of CD8+ T cells, impairing tumor growth and potentiating immune checkpoint inhibitors in breast tumor models. Conclusion: These results provide comprehensive insights into TAM-mediated early tumor microenvironments and reveal immune evasion mechanisms that can be targeted by Gal-1 to induce antitumor immune responses.
Collapse
Affiliation(s)
- Hyewon Chung
- Macrophage Lab, Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul 110-799, South Korea
- Institute of Endemic Diseases, Seoul National University Medical Research Center (SNUMRC), Seoul, Republic of Korea
| | - Park Gyu-mi
- Macrophage Lab, Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul 110-799, South Korea
- Department of Biomedical Sciences and Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yi Rang Na
- Transdisciplinary Department of Medicine and Advanced Technology, Seoul National University Hospital, Seoul, South Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hongyoon Choi
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung Hyeok Seok
- Macrophage Lab, Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul 110-799, South Korea
- Department of Biomedical Sciences and Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, South Korea
| |
Collapse
|
59
|
Ding B, Chen Q, Wu Z, Li X, Ding Y, Wu Q, Han L, Wu H. In Vitro and In Vivo Analyses Reveal Tumor-Derived Exosome miR-558 Promotes Angiogenesis in Tongue Squamous Cell Carcinoma by Targeting Heparinase. Technol Cancer Res Treat 2024; 23:15330338241261615. [PMID: 38887096 PMCID: PMC11185026 DOI: 10.1177/15330338241261615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
This study aimed to investigate the role of miR-558 in tumor angiogenesis by targeting heparinase (HPSE) in tongue squamous cell carcinoma (TSCC)-derived exosomes. In the present study, the role of exosome miR-558 in angiogenesis in vitro and in vivo was investigated by cell proliferation, migration, tube formation, subcutaneous tumor formation in mice, and in vivo Matrigel plug assay. The target genes of miR-558 were detected by means of dual luciferase assay. It was found that TSCC cells secrete miR-558 into the extracellular environment, with exosome as the carrier. Human umbilical vein endothelial cells (HUVEC) ingested exosomes, which not only increased the expression level of miR-558, but also enhanced their proliferation, migration, and tube formation functions. In vivo Matrigel plug assay demonstrated that TSCC cell-derived exosome miR-558 promoted neovascularization in vivo. Compared with negative control cells, TSCC cells overexpressing miR-558 formed subcutaneous tumors in nude mice, with larger volume, heavier mass, and more vascularization. Dual luciferase assay confirmed that HPSE was the direct target gene regulated by miR-558. HPSE promoted the proliferation, migration, and tube formation of HUVECs, and the knockout of HPSE could downregulate the pro-angiogenic effect of miR-558. In summary, miR-558 in TSCC exosomes promotes the proliferation, migration, and tube formation of HUVECs by targeting HPSE, and enhancing tumor angiogenesis.
Collapse
Affiliation(s)
- Bixiao Ding
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Nantong University, Nantong, China
| | - Qingwen Chen
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Zhen Wu
- Department of Otolaryngology-Head and Neck Surgery, Changshu Second People's Hospital, Suzhou, China
| | - Xiaoguang Li
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine; Shanghai Key Lab, Shanghai, China
| | - Yuancheng Ding
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Nantong University, Nantong, China
| | - Qiong Wu
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Nantong University, Nantong, China
| | - Liang Han
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Hao Wu
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
60
|
Kerboeuf M, Haugeberg DA, Olsen T, Sørling LK, Koppang EO, Moe L, Haaland AH. Tumor-associated macrophages in canine visceral hemangiosarcoma. Vet Pathol 2024; 61:32-45. [PMID: 37341055 PMCID: PMC10687809 DOI: 10.1177/03009858231179947] [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] [Indexed: 06/22/2023]
Abstract
Canine hemangiosarcoma (HSA) is a highly malignant tumor derived from hematopoietic stem cells and commonly occurs in visceral organs or skin. Visceral HSAs are particularly aggressive and progress rapidly despite multimodal treatment. Tumor-associated macrophages (TAMs) play a central role in carcinogenesis, tumor progression, and metastasis in humans and murine models. In this retrospective study, we investigated the prevalence and phenotype of TAMs in privately owned, treatment-naïve dogs with naturally occurring HSA. We used CD204 as a general macrophage marker and CD206 as a marker for M2-polarized macrophages. Formalin-fixed paraffin-embedded tissues from HSAs in the spleen (n = 9), heart (n = 6), and other locations (n = 12) from 17 dogs were sectioned and immunohistochemically labeled with CD204 and CD206 antibodies. The mean number of log(CD204)- and log(CD206)-positive cells and the ratio of log(CD206/CD204)-positive cells were compared with normal surrounding tissues and between tumor locations. There were significantly more macrophages and M2 macrophages, and a higher ratio of M2 macrophages to total macrophages in tumor hot spots (P = .0002, P < .0001, and P = .0002, respectively) and in tumor tissues outside of hot spots (P = .009, P = .002, and P = .007, respectively) than in normal surrounding tissues. There were no significant differences between tumor locations, but there was a trend toward higher numbers of CD204-positive macrophages within the splenic tumors. There was no association between histological parameters or clinical stage and TAM numbers or phenotype. As in humans, TAMs in dogs with HSA have a predominantly M2-skewed phenotype. Dogs with HSA could serve as excellent models to evaluate new TAM-reprogramming therapies.
Collapse
Affiliation(s)
| | | | - Tobias Olsen
- Norwegian University of Life Sciences, Ås, Norway
| | | | | | - Lars Moe
- Norwegian University of Life Sciences, Ås, Norway
| | | |
Collapse
|
61
|
Kang X, Huang Y, Wang H, Jadhav S, Yue Z, Tiwari AK, Babu RJ. Tumor-Associated Macrophage Targeting of Nanomedicines in Cancer Therapy. Pharmaceutics 2023; 16:61. [PMID: 38258072 PMCID: PMC10819517 DOI: 10.3390/pharmaceutics16010061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/24/2023] [Accepted: 12/25/2023] [Indexed: 01/24/2024] Open
Abstract
The tumor microenvironment (TME) is pivotal in tumor growth and metastasis, aligning with the "Seed and Soil" theory. Within the TME, tumor-associated macrophages (TAMs) play a central role, profoundly influencing tumor progression. Strategies targeting TAMs have surfaced as potential therapeutic avenues, encompassing interventions to block TAM recruitment, eliminate TAMs, reprogram M2 TAMs, or bolster their phagocytic capabilities via specific pathways. Nanomaterials including inorganic materials, organic materials for small molecules and large molecules stand at the forefront, presenting significant opportunities for precise targeting and modulation of TAMs to enhance therapeutic efficacy in cancer treatment. This review provides an overview of the progress in designing nanoparticles for interacting with and influencing the TAMs as a significant strategy in cancer therapy. This comprehensive review presents the role of TAMs in the TME and various targeting strategies as a promising frontier in the ever-evolving field of cancer therapy. The current trends and challenges associated with TAM-based therapy in cancer are presented.
Collapse
Affiliation(s)
- Xuejia Kang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA;
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Yongzhuo Huang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Guangzhou 528400, China;
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China;
| | - Huiyuan Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China;
| | - Sanika Jadhav
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA;
| | - Zongliang Yue
- Department of Health Outcome and Research Policy, Harrison School of Pharmacy, Auburn University, Auburn, AL 36849, USA;
| | - Amit K. Tiwari
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas of Medical Sciences, Little Rock, AR 72205, USA;
| | - R. Jayachandra Babu
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA;
| |
Collapse
|
62
|
Banerjee K, Kerzel T, Bekkhus T, de Souza Ferreira S, Wallmann T, Wallerius M, Landwehr LS, Agardy DA, Schauer N, Malmerfeldt A, Bergh J, Bartish M, Hartman J, Östman A, Squadrito ML, Rolny C. VEGF-C-expressing TAMs rewire the metastatic fate of breast cancer cells. Cell Rep 2023; 42:113507. [PMID: 38041815 DOI: 10.1016/j.celrep.2023.113507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 10/11/2023] [Accepted: 11/13/2023] [Indexed: 12/04/2023] Open
Abstract
The expression of pro-lymphangiogenic VEGF-C in primary tumors is associated with sentinel lymph node metastasis in most solid cancer types. However, the impact of VEGF-C on distant organ metastasis remains unclear. Perivascular tumor-associated macrophages (TAMs) play a crucial role in guiding hematogenous spread of cancer cells by establishing metastatic pathways within the tumor microenvironment. This process supports breast cancer cell intravasation and metastatic dissemination. We show here that VEGF-C-expressing TAMs reduce the dissemination of mammary cancer cells to the lungs while concurrently increasing lymph node metastasis. These TAMs express podoplanin and interact with normalized tumor blood vessels expressing VEGFR3. Moreover, clinical data suggest inverse association between VEGF-C-expressing TAMs and breast cancer malignancy. Thus, our study elucidates the paradoxical role of VEGF-C-expressing TAMs in redirecting cancer cells to preferentially disseminate to lymph nodes rather than to lungs, partially achieved by normalizing tumor blood vessels and promoting lymphangiogenesis.
Collapse
Affiliation(s)
- Kaveri Banerjee
- Department of Oncology-Pathology, Karolinska Institutet, 17164 Stockholm, Sweden
| | - Thomas Kerzel
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Vita Salute San Raffaele University, 20132 Milan, Italy
| | - Tove Bekkhus
- Department of Oncology-Pathology, Karolinska Institutet, 17164 Stockholm, Sweden
| | | | - Tatjana Wallmann
- Department of Oncology-Pathology, Karolinska Institutet, 17164 Stockholm, Sweden
| | - Majken Wallerius
- Department of Oncology-Pathology, Karolinska Institutet, 17164 Stockholm, Sweden
| | | | | | - Nele Schauer
- Department of Oncology-Pathology, Karolinska Institutet, 17164 Stockholm, Sweden
| | - Anna Malmerfeldt
- Department of Oncology-Pathology, Karolinska Institutet, 17164 Stockholm, Sweden
| | - Jonas Bergh
- Department of Oncology-Pathology, Karolinska Institutet, 17164 Stockholm, Sweden; Breast Center, Karolinska Comprehensive Cancer Center and Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Margarita Bartish
- Department of Oncology-Pathology, Karolinska Institutet, 17164 Stockholm, Sweden; Gerald Bronfman Department of Oncology, Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - Johan Hartman
- Department of Oncology-Pathology, Karolinska Institutet, 17164 Stockholm, Sweden; Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Arne Östman
- Department of Oncology-Pathology, Karolinska Institutet, 17164 Stockholm, Sweden
| | - Mario Leonardo Squadrito
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Vita Salute San Raffaele University, 20132 Milan, Italy.
| | - Charlotte Rolny
- Department of Oncology-Pathology, Karolinska Institutet, 17164 Stockholm, Sweden.
| |
Collapse
|
63
|
Peng X, Wang Y, Zhang J, Zhang Z, Qi S. Intravital imaging of the functions of immune cells in the tumor microenvironment during immunotherapy. Front Immunol 2023; 14:1288273. [PMID: 38124754 PMCID: PMC10730658 DOI: 10.3389/fimmu.2023.1288273] [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: 09/04/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023] Open
Abstract
Cancer immunotherapy has developed rapidly in recent years and stands as one of the most promising techniques for combating cancer. To develop and optimize cancer immunotherapy, it is crucial to comprehend the interactions between immune cells and tumor cells in the tumor microenvironment (TME). The TME is complex, with the distribution and function of immune cells undergoing dynamic changes. There are several research techniques to study the TME, and intravital imaging emerges as a powerful tool for capturing the spatiotemporal dynamics, especially the movement behavior and the immune function of various immune cells in real physiological state. Intravital imaging has several advantages, such as high spatio-temporal resolution, multicolor, dynamic and 4D detection, making it an invaluable tool for visualizing the dynamic processes in the TME. This review summarizes the workflow for intravital imaging technology, multi-color labeling methods, optical imaging windows, methods of imaging data analysis and the latest research in visualizing the spatio-temporal dynamics and function of immune cells in the TME. It is essential to investigate the role played by immune cells in the tumor immune response through intravital imaging. The review deepens our understanding of the unique contribution of intravital imaging to improve the efficiency of cancer immunotherapy.
Collapse
Affiliation(s)
- Xuwen Peng
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, China
- MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuke Wang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, China
- MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jie Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, China
- MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhihong Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, China
- MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuhong Qi
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei, China
- MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
64
|
Nasir I, McGuinness C, Poh AR, Ernst M, Darcy PK, Britt KL. Tumor macrophage functional heterogeneity can inform the development of novel cancer therapies. Trends Immunol 2023; 44:971-985. [PMID: 37995659 DOI: 10.1016/j.it.2023.10.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/25/2023]
Abstract
Macrophages represent a key component of the tumor microenvironment (TME) and are largely associated with poor prognosis. Therapeutic targeting of macrophages has historically focused on inhibiting their recruitment or reprogramming their phenotype from a protumor (M2-like) to an antitumor (M1-like) one. Unfortunately, this approach has not provided clinical breakthroughs that have changed practice. Emerging studies utilizing single-cell RNA-sequencing (scRNA-seq) and spatial transcriptomics have improved our understanding of the ontogeny, phenotype, and functional plasticity of macrophages. Overlaying the wealth of current information regarding macrophage molecular subtypes and functions has also identified novel therapeutic vulnerabilities that might drive better control of tumor-associated macrophages (TAMs). Here, we discuss the functional profiling of macrophages and provide an update of novel macrophage-targeted therapies in development.
Collapse
Affiliation(s)
- Ibraheem Nasir
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Conor McGuinness
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Melbourne, VIC 3000, Australia
| | - Ashleigh R Poh
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria 3084, Australia; La Trobe University School of Cancer Medicine, Heidelberg, Victoria 3084, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria 3084, Australia; La Trobe University School of Cancer Medicine, Heidelberg, Victoria 3084, Australia
| | - Phillip K Darcy
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Melbourne, VIC 3000, Australia; Cancer Immunology Research Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Kara L Britt
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Melbourne, VIC 3000, Australia.
| |
Collapse
|
65
|
Chai K, Wang C, Zhou J, Mu W, Gao M, Fan Z, Lv G. Quenching thirst with poison? Paradoxical effect of anticancer drugs. Pharmacol Res 2023; 198:106987. [PMID: 37949332 DOI: 10.1016/j.phrs.2023.106987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
Anticancer drugs have been developed with expectations to provide long-term or at least short-term survival benefits for patients with cancer. Unfortunately, drug therapy tends to provoke malignant biological and clinical behaviours of cancer cells relating not only to the evolution of resistance to specific drugs but also to the enhancement of their proliferation and metastasis abilities. Thus, drug therapy is suspected to impair long-term survival in treated patients under certain circumstances. The paradoxical therapeutic effects could be described as 'quenching thirst with poison', where temporary relief is sought regardless of the consequences. Understanding the underlying mechanisms by which tumours react on drug-induced stress to maintain viability is crucial to develop rational targeting approaches which may optimize survival in patients with cancer. In this review, we describe the paradoxical adverse effects of anticancer drugs, in particular how cancer cells complete resistance evolution, enhance proliferation, escape from immune surveillance and metastasize efficiently when encountered with drug therapy. We also describe an integrative therapeutic framework that may diminish such paradoxical effects, consisting of four main strategies: (1) targeting endogenous stress response pathways, (2) targeting new identities of cancer cells, (3) adaptive therapy- exploiting subclonal competition of cancer cells, and (4) targeting tumour microenvironment.
Collapse
Affiliation(s)
- Kaiyuan Chai
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Chuanlei Wang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Jianpeng Zhou
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Wentao Mu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Menghan Gao
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhongqi Fan
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China.
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
66
|
Chen JG, Chávez-Fuentes JC, O'Brien M, Xu J, Ruiz E, Wang W, Amin I, Sarfraz I, Guckhool P, Sistig A, Yuan GC, Dries R. Giotto Suite: a multi-scale and technology-agnostic spatial multi-omics analysis ecosystem. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.26.568752. [PMID: 38077085 PMCID: PMC10705291 DOI: 10.1101/2023.11.26.568752] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2023]
Abstract
Emerging spatial omics technologies continue to advance the molecular mapping of tissue architecture and the investigation of gene regulation and cellular crosstalk, which in turn provide new mechanistic insights into a wide range of biological processes and diseases. Such technologies provide an increasingly large amount of information content at multiple spatial scales. However, representing and harmonizing diverse spatial datasets efficiently, including combining multiple modalities or spatial scales in a scalable and flexible manner, remains a substantial challenge. Here, we present Giotto Suite, a suite of open-source software packages that underlies a fully modular and integrated spatial data analysis toolbox. At its core, Giotto Suite is centered around an innovative and technology-agnostic data framework embedded in the R software environment, which allows the representation and integration of virtually any type of spatial omics data at any spatial resolution. In addition, Giotto Suite provides both scalable and extensible end-to-end solutions for data analysis, integration, and visualization. Giotto Suite integrates molecular, morphology, spatial, and annotated feature information to create a responsive and flexible workflow for multi-scale, multi-omic data analyses, as demonstrated here by applications to several state-of-the-art spatial technologies. Furthermore, Giotto Suite builds upon interoperable interfaces and data structures that bridge the established fields of genomics and spatial data science, thereby enabling independent developers to create custom-engineered pipelines. As such, Giotto Suite creates an immersive ecosystem for spatial multi-omic data analysis.
Collapse
Affiliation(s)
- Jiaji George Chen
- Section of Hematology and Medical Oncology, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
| | | | - Matthew O'Brien
- Section of Hematology and Medical Oncology, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
| | - Junxiang Xu
- Section of Hematology and Medical Oncology, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
| | - Edward Ruiz
- Section of Hematology and Medical Oncology, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
| | - Wen Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Iqra Amin
- Section of Hematology and Medical Oncology, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
| | - Irzam Sarfraz
- Section of Hematology and Medical Oncology, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
| | - Pratishtha Guckhool
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Adriana Sistig
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ruben Dries
- Section of Hematology and Medical Oncology, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
- Division of Computational Biomedicine, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
67
|
Aizaz M, Khan A, Khan F, Khan M, Musad Saleh EA, Nisar M, Baran N. The cross-talk between macrophages and tumor cells as a target for cancer treatment. Front Oncol 2023; 13:1259034. [PMID: 38033495 PMCID: PMC10682792 DOI: 10.3389/fonc.2023.1259034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/17/2023] [Indexed: 12/02/2023] Open
Abstract
Macrophages represent an important component of the innate immune system. Under physiological conditions, macrophages, which are essential phagocytes, maintain a proinflammatory response and repair damaged tissue. However, these processes are often impaired upon tumorigenesis, in which tumor-associated macrophages (TAMs) protect and support the growth, proliferation, and invasion of tumor cells and promote suppression of antitumor immunity. TAM abundance is closely associated with poor outcome of cancer, with impediment of chemotherapy effectiveness and ultimately a dismal therapy response and inferior overall survival. Thus, cross-talk between cancer cells and TAMs is an important target for immune checkpoint therapies and metabolic interventions, spurring interest in it as a therapeutic vulnerability for both hematological cancers and solid tumors. Furthermore, targeting of this cross-talk has emerged as a promising strategy for cancer treatment with the antibody against CD47 protein, a critical macrophage checkpoint recognized as the "don't eat me" signal, as well as other metabolism-focused strategies. Therapies targeting CD47 constitute an important milestone in the advancement of anticancer research and have had promising effects on not only phagocytosis activation but also innate and adaptive immune system activation, effectively counteracting tumor cells' evasion of therapy as shown in the context of myeloid cancers. Targeting of CD47 signaling is only one of several possibilities to reverse the immunosuppressive and tumor-protective tumor environment with the aim of enhancing the antitumor response. Several preclinical studies identified signaling pathways that regulate the recruitment, polarization, or metabolism of TAMs. In this review, we summarize the current understanding of the role of macrophages in cancer progression and the mechanisms by which they communicate with tumor cells. Additionally, we dissect various therapeutic strategies developed to target macrophage-tumor cell cross-talk, including modulation of macrophage polarization, blockade of signaling pathways, and disruption of physical interactions between leukemia cells and macrophages. Finally, we highlight the challenges associated with tumor hypoxia and acidosis as barriers to effective cancer therapy and discuss opportunities for future research in this field.
Collapse
Affiliation(s)
- Muhammad Aizaz
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Aakif Khan
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Faisal Khan
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Maria Khan
- Center of Biotechnology and Microbiology, University of Peshawar, Peshawar, Pakistan
| | - Ebraheem Abdu Musad Saleh
- Department of Chemistry, College of Arts & Science, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Maryum Nisar
- School of Interdisciplinary Engineering & Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Natalia Baran
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
68
|
Snyder CM, Gill SI. Good CARMA: Turning bad tumor-resident myeloid cells good with chimeric antigen receptor macrophages. Immunol Rev 2023; 320:236-249. [PMID: 37295964 DOI: 10.1111/imr.13231] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023]
Abstract
In religious philosophy, the concept of karma represents the effect of one's past and present actions on one's future. Macrophages are highly plastic cells with myriad roles in health and disease. In the setting of cancer, macrophages are among the most plentiful members of the immune microenvironment where they generally support tumor growth and restrain antitumor immunity. However, macrophages are not necessarily born bad. Macrophages or their immediate progenitors, monocytes, are induced to traffic to the tumor microenvironment (TME) and during this process they are polarized toward a tumor-promoting phenotype. Efforts to deplete or repolarize tumor-associated macrophages (TAM) for therapeutic benefit in cancer have to date disappointed. By contrast, genetic engineering of macrophages followed by their transit into the TME may allow these impressionable cells to mend their ways. In this review, we summarize and discuss recent advances in the genetic engineering of macrophages for the treatment of cancer.
Collapse
Affiliation(s)
- Christopher M Snyder
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Saar I Gill
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
69
|
Wu J, Chan YT, Lu Y, Wang N, Feng Y. The tumor microenvironment in the postsurgical liver: Mechanisms and potential targets of postoperative recurrence in human hepatocellular carcinoma. Med Res Rev 2023; 43:1946-1973. [PMID: 37102365 DOI: 10.1002/med.21967] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/23/2023] [Accepted: 04/13/2023] [Indexed: 04/28/2023]
Abstract
Surgery remains to be the mainstay of treatment for hepatocellular carcinoma (HCC). Nonetheless, its therapeutic efficacy is significantly impaired by postoperative recurrence, which occurs in more than half of cases as a result of intrahepatic metastasis or de novo tumorigenesis. For decades, most therapeutic strategies on inhibiting postoperative HCC recurrence have been focused on the residual tumor cells but satisfying therapeutic outcomes are barely observed in the clinic. In recent years, a better understanding of tumor biology allows us to shift our focus from tumor cells toward the postoperative tumor microenvironment (TME), which is gradually identified to play a pivotal role in tumor recurrence. In this review, we describe various surgical stress and surgical perturbation on postoperative TME. Besides, we discuss how such alternations in TME give rise to postoperative recurrence of HCC. Based on its clinical significance, we additionally highlight the potential of the postoperative TME as a target for postoperative adjuvant therapeutics.
Collapse
Affiliation(s)
- Junyu Wu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yau-Tuen Chan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yuanjun Lu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
70
|
Nowosad A, Marine JC, Karras P. Perivascular niches: critical hubs in cancer evolution. Trends Cancer 2023; 9:897-910. [PMID: 37453870 DOI: 10.1016/j.trecan.2023.06.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023]
Abstract
Tumors are heterogeneous ecosystems in which cancer cells coexist within a complex tumor immune microenvironment (TIME). The malignant, stromal, and immune cell compartments establish a plethora of bidirectional cell-cell communication crosstalks that influence tumor growth and metastatic dissemination, which we are only beginning to understand. Cancer cells either co-opt or promote the formation of new blood and lymphatic vessels to cope with their need for nutrients and oxygen. Recent studies have highlighted additional key roles for the tumor vasculature and have identified the perivascular niche as a cellular hub, where intricate and dynamic cellular interactions promote cancer stemness, immune evasion, dormancy, and metastatic spreading. Here, we review these findings, and discuss how they may be exploited therapeutically.
Collapse
Affiliation(s)
- Ada Nowosad
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium.
| | - Panagiotis Karras
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
71
|
Genna A, Alter J, Poletti M, Meirson T, Sneh T, Gendler M, Saleev N, Karagiannis GS, Wang Y, Cox D, Entenberg D, Oktay MH, Korcsmaros T, Condeelis JS, Gil-Henn H. FAK family proteins regulate in vivo breast cancer metastasis via distinct mechanisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.27.564212. [PMID: 37961438 PMCID: PMC10634866 DOI: 10.1101/2023.10.27.564212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Breast cancer is the most commonly diagnosed malignancy and the major leading cause of tumor-related deaths in women. It is estimated that the majority of breast tumor-related deaths are a consequence of metastasis, to which no cure exists at present. The FAK family proteins Proline-rich tyrosine kinase (PYK2) and focal adhesion kinase (FAK) are highly expressed in breast cancer, but the exact cellular and signaling mechanisms by which they regulate in vivo tumor cell invasiveness and consequent metastatic dissemination are mostly unknown. Using a PYK2 and FAK knockdown xenograft model we show here, for the first time, that ablation of either PYK2 or FAK decreases primary tumor size and significantly reduces Tumor MicroEnvironment of Metastasis (TMEM) doorway activation, leading to decreased intravasation and reduced spontaneous lung metastasis. Intravital imaging analysis further demonstrates that PYK2, but not FAK, regulates a motility phenotype switch between focal adhesion-mediated fast motility and invadopodia-dependent, ECM-degradation associated slow motility within the primary tumor. Furthermore, we validate our in vivo and intravital imaging results with integrated transcriptomic and proteomic data analysis from xenograft knockdown tumors and reveal new and distinct pathways by which these two homologous kinases regulate breast tumor cell invasiveness and consequent metastatic dissemination. Our findings identify PYK2 and FAK as novel mediators of mammary tumor progression and metastasis and as candidate therapeutic targets for breast cancer metastasis.
Collapse
|
72
|
Shen K, Song W, Wang H, Wang L, Yang Y, Hu Q, Ren M, Gao Z, Wang Q, Zheng S, Zhu M, Yang Y, Zhang Y, Wei C, Gu J. Decoding the metastatic potential and optimal postoperative adjuvant therapy of melanoma based on metastasis score. Cell Death Discov 2023; 9:397. [PMID: 37880239 PMCID: PMC10600209 DOI: 10.1038/s41420-023-01678-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/26/2023] [Accepted: 10/11/2023] [Indexed: 10/27/2023] Open
Abstract
Metastasis is a formidable challenge in the prognosis of melanoma. Accurately predicting the metastatic potential of non-metastatic melanoma (NMM) and determining effective postoperative adjuvant treatments for inhibiting metastasis remain uncertain. In this study, we conducted comprehensive analyses of melanoma metastases using bulk and single-cell RNA sequencing data, enabling the construction of a metastasis score (MET score) through diverse machine-learning algorithms. The reliability and robustness of the MET score were validated using various in vitro assays and in vivo models. Our findings revealed a distinct molecular landscape in metastatic melanoma characterized by the enrichment of metastasis-related pathways, intricate cell-cell communication, and heightened infiltration of pro-angiogenic tumor-associated macrophages compared to NMM. Importantly, patients in the high MET score group exhibited poorer prognoses and an immunosuppressive microenvironment, featuring increased infiltration of regulatory T cells and decreased infiltration of CD8+ T cells, compared to the low MET score patient group. Expression of PD-1 was markedly higher in patients with low MET scores. Anti-PD-1 (aPD-1) therapy profoundly affected antitumor immunity activation and metastasis inhibition in these patients. In summary, our study demonstrates the effectiveness of the MET score in predicting melanoma metastatic potential. For patients with low MET scores, aPD-1 therapy may be a potential treatment strategy to inhibit metastasis. Patients with high MET scores may benefit from combination therapies.
Collapse
Affiliation(s)
- Kangjie Shen
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenyu Song
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hongye Wang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Lu Wang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yang Yang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qianrong Hu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Min Ren
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zixu Gao
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiangcheng Wang
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Shaoluan Zheng
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, China
| | - Ming Zhu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanwen Yang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yong Zhang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chuanyuan Wei
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jianying Gu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China.
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, China.
| |
Collapse
|
73
|
Gadde M, Mehrabi-Dehdezi M, Debeb BG, Woodward WA, Rylander MN. Influence of Macrophages on Vascular Invasion of Inflammatory Breast Cancer Emboli Measured Using an In Vitro Microfluidic Multi-Cellular Platform. Cancers (Basel) 2023; 15:4883. [PMID: 37835577 PMCID: PMC10571588 DOI: 10.3390/cancers15194883] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Inflammatory breast cancer (IBC) is an aggressive disease with a poor prognosis and a lack of effective treatments. It is widely established that understanding the interactions between tumor-associated macrophages (TAMs) and the tumor microenvironment is essential for identifying distinct targeting markers that help with prognosis and subsequent development of effective treatments. In this study, we present a 3D in vitro microfluidic IBC platform consisting of THP1 M0, M1, or M2 macrophages, IBC cells, and endothelial cells. The platform comprises a collagen matrix that includes an endothelialized vessel, creating a physiologically relevant environment for cellular interactions. Through the utilization of this platform, it was discovered that the inclusion of tumor-associated macrophages (TAMs) led to an increase in the formation of new blood vessel sprouts and enhanced permeability of the endothelium, regardless of the macrophage phenotype. Interestingly, the platforms containing THP-1 M1 or M2 macrophages exhibited significantly greater porosity in the collagen extracellular matrix (ECM) compared to the platforms containing THP-1 M0 and the MDA-IBC3 cells alone. Cytokine analysis revealed that IL-8 and MMP9 showed selective increases when macrophages were cultured in the platforms. Notably, intravasation of tumor cells into the vessels was observed exclusively in the platform containing MDA-IBC3 and M0 macrophages.
Collapse
Affiliation(s)
- Manasa Gadde
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (M.G.); (M.M.-D.)
| | - Melika Mehrabi-Dehdezi
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (M.G.); (M.M.-D.)
| | - Bisrat G. Debeb
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Wendy A. Woodward
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marissa Nichole Rylander
- Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (M.G.); (M.M.-D.)
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
- Oden Institute for Computational and Engineering Sciences, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
74
|
Petersen J, Du W, Adkisson C, Gravekamp C, Oktay MH, Condeelis J, Panarelli NC, McAuliffe JC, Entenberg D. Stabilized Window for Intravital Imaging of the Murine Pancreas. J Vis Exp 2023:10.3791/65498. [PMID: 37870314 PMCID: PMC10731889 DOI: 10.3791/65498] [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] [Indexed: 10/24/2023] Open
Abstract
The physiology and pathophysiology of the pancreas are complex. Diseases of the pancreas, such as pancreatitis and pancreatic adenocarcinoma (PDAC) have high morbidity and mortality. Intravital imaging (IVI) is a powerful technique enabling the high-resolution imaging of tissues in both healthy and diseased states, allowing for real-time observation of cell dynamics. IVI of the murine pancreas presents significant challenges due to the deep visceral and compliant nature of the organ, which make it highly prone to damage and motion artifacts. Described here is the process of implantation of the Stabilized Window for Intravital imaging of the murine Pancreas (SWIP). The SWIP allows IVI of the murine pancreas in normal healthy states, during the transformation from the healthy pancreas to acute pancreatitis induced by cerulein, and in malignant states such as pancreatic tumors. In conjunction with genetically labeled cells or the administration of fluorescent dyes, the SWIP enables the measurement of single-cell and subcellular dynamics (including single-cell and collective migration) as well as serial imaging of the same region of interest over multiple days. The ability to capture tumor cell migration is of particular importance as the primary cause of cancer-related mortality in PDAC is the overwhelming metastatic burden. Understanding the physiological dynamics of metastasis in PDAC is a critical unmet need and crucial for improving patient prognosis. Overall, the SWIP provides improved imaging stability and expands the application of IVI in the healthy pancreas and malignant pancreas diseases.
Collapse
Affiliation(s)
- Jakeb Petersen
- Department of Surgery, Einstein College of Medicine / Montefiore Medical Center; Department of Pathology, Einstein College of Medicine / Montefiore Medical Center; Integrated Imaging Program for Cancer Research, Einstein College of Medicine / Montefiore Medical Center
| | - Wei Du
- Department of Anatomy and Structural Biology, Einstein College of Medicine / Montefiore Medical Center; Breast Center, Peking University People's Hospital
| | - Christian Adkisson
- Department of Surgery, Einstein College of Medicine / Montefiore Medical Center; Department of Anatomy and Structural Biology, Einstein College of Medicine / Montefiore Medical Center
| | - Claudia Gravekamp
- Department of Microbiology & Immunology, Einstein College of Medicine / Montefiore Medical Center; Montefiore Einstein Cancer Center, Einstein College of Medicine / Montefiore Medical Center
| | - Maja H Oktay
- Department of Pathology, Einstein College of Medicine / Montefiore Medical Center; Integrated Imaging Program for Cancer Research, Einstein College of Medicine / Montefiore Medical Center; Department of Anatomy and Structural Biology, Einstein College of Medicine / Montefiore Medical Center; Montefiore Einstein Cancer Center, Einstein College of Medicine / Montefiore Medical Center; Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine / Montefiore Medical Center; Gruss-Lipper Biophotonics Center, Einstein College of Medicine / Montefiore Medical Center
| | - John Condeelis
- Department of Surgery, Einstein College of Medicine / Montefiore Medical Center; Integrated Imaging Program for Cancer Research, Einstein College of Medicine / Montefiore Medical Center; Department of Anatomy and Structural Biology, Einstein College of Medicine / Montefiore Medical Center; Montefiore Einstein Cancer Center, Einstein College of Medicine / Montefiore Medical Center; Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine / Montefiore Medical Center; Gruss-Lipper Biophotonics Center, Einstein College of Medicine / Montefiore Medical Center; Department of Cell Biology, Einstein College of Medicine / Montefiore Medical Center
| | - Nicole C Panarelli
- Department of Pathology, Einstein College of Medicine / Montefiore Medical Center; Integrated Imaging Program for Cancer Research, Einstein College of Medicine / Montefiore Medical Center; Montefiore Einstein Cancer Center, Einstein College of Medicine / Montefiore Medical Center
| | - John C McAuliffe
- Department of Surgery, Einstein College of Medicine / Montefiore Medical Center; Department of Pathology, Einstein College of Medicine / Montefiore Medical Center; Integrated Imaging Program for Cancer Research, Einstein College of Medicine / Montefiore Medical Center; Montefiore Einstein Cancer Center, Einstein College of Medicine / Montefiore Medical Center; Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine / Montefiore Medical Center
| | - David Entenberg
- Department of Pathology, Einstein College of Medicine / Montefiore Medical Center; Integrated Imaging Program for Cancer Research, Einstein College of Medicine / Montefiore Medical Center; Department of Anatomy and Structural Biology, Einstein College of Medicine / Montefiore Medical Center; Montefiore Einstein Cancer Center, Einstein College of Medicine / Montefiore Medical Center; Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine / Montefiore Medical Center; Gruss-Lipper Biophotonics Center, Einstein College of Medicine / Montefiore Medical Center;
| |
Collapse
|
75
|
Ildiz ES, Gvozdenovic A, Kovacs WJ, Aceto N. Travelling under pressure - hypoxia and shear stress in the metastatic journey. Clin Exp Metastasis 2023; 40:375-394. [PMID: 37490147 PMCID: PMC10495280 DOI: 10.1007/s10585-023-10224-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/05/2023] [Indexed: 07/26/2023]
Abstract
Cancer cell invasion, intravasation and survival in the bloodstream are early steps of the metastatic process, pivotal to enabling the spread of cancer to distant tissues. Circulating tumor cells (CTCs) represent a highly selected subpopulation of cancer cells that tamed these critical steps, and a better understanding of their biology and driving molecular principles may facilitate the development of novel tools to prevent metastasis. Here, we describe key research advances in this field, aiming at describing early metastasis-related processes such as collective invasion, shedding, and survival of CTCs in the bloodstream, paying particular attention to microenvironmental factors like hypoxia and mechanical stress, considered as important influencers of the metastatic journey.
Collapse
Affiliation(s)
- Ece Su Ildiz
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Ana Gvozdenovic
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Werner J Kovacs
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Nicola Aceto
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland.
| |
Collapse
|
76
|
Yang Y, Chen Y, Pei P, Fan Y, Wang S, Zhang H, Zhao D, Qian BZ, Zhang F. Fluorescence-amplified nanocrystals in the second near-infrared window for in vivo real-time dynamic multiplexed imaging. NATURE NANOTECHNOLOGY 2023; 18:1195-1204. [PMID: 37349506 DOI: 10.1038/s41565-023-01422-2] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 05/16/2023] [Indexed: 06/24/2023]
Abstract
Optical imaging in the second near-infrared window (NIR-II, 1,000-1,700 nm) holds great promise for non-invasive in vivo detection. However, real-time dynamic multiplexed imaging remains challenging due to the lack of available fluorescence probes and multiplexing techniques in the ideal NIR-IIb (1,500-1,700 nm) 'deep-tissue-transparent' sub-window. Here we report on thulium-based cubic-phase downshifting nanoparticles (α-TmNPs) with 1,632 nm fluorescence amplification. This strategy was also validated for the fluorescence enhancement of nanoparticles doped with NIR-II Er3+ (α-ErNPs) or Ho3+ (α-HoNPs). In parallel, we developed a simultaneous dual-channel imaging system with high spatiotemporal synchronization and accuracy. The NIR-IIb α-TmNPs and α-ErNPs facilitated the non-invasive real-time dynamic multiplexed imaging of cerebrovascular vasomotion activity and the single-cell-level neutrophil behaviour in mouse subcutaneous tissue and ischaemic stroke model.
Collapse
Affiliation(s)
- Yiwei Yang
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers and iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, China
| | - Ying Chen
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers and iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, China
| | - Peng Pei
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers and iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, China
| | - Yong Fan
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers and iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, China.
| | - Shangfeng Wang
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers and iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, China
| | - Hongxin Zhang
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers and iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, China
| | - Dongyuan Zhao
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers and iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, China
| | - Bin-Zhi Qian
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, China
- Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Fan Zhang
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers and iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, China.
| |
Collapse
|
77
|
Nguyen LNM, Lin ZP, Sindhwani S, MacMillan P, Mladjenovic SM, Stordy B, Ngo W, Chan WCW. The exit of nanoparticles from solid tumours. NATURE MATERIALS 2023; 22:1261-1272. [PMID: 37592029 DOI: 10.1038/s41563-023-01630-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/07/2023] [Indexed: 08/19/2023]
Abstract
Nanoparticles enter tumours through endothelial cells, gaps or other mechanisms, but how they exit is unclear. The current paradigm states that collapsed tumour lymphatic vessels impair the exit of nanoparticles and lead to enhanced retention. Here we show that nanoparticles exit the tumour through the lymphatic vessels within or surrounding the tumour. The dominant lymphatic exit mechanism depends on the nanoparticle size. Nanoparticles that exit the tumour through the lymphatics are returned to the blood system, allowing them to recirculate and interact with the tumour in another pass. Our results enable us to define a mechanism of nanoparticle delivery to solid tumours alternative to the enhanced permeability and retention effect. We call this mechanism the active transport and retention principle. This delivery principle provides a new framework to engineer nanomedicines for cancer treatment and detection.
Collapse
Affiliation(s)
- Luan N M Nguyen
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Zachary P Lin
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Shrey Sindhwani
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Division of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Presley MacMillan
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Stefan M Mladjenovic
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin Stordy
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Wayne Ngo
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Warren C W Chan
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
78
|
Sigdel I, Ofori-Kwafo A, Heizelman RJ, Nestor-Kalinoski A, Prabhakarpandian B, Tiwari AK, Tang Y. Biomimetic on-chip assay reveals the anti-metastatic potential of a novel thienopyrimidine compound in triple-negative breast cancer cell lines. Front Bioeng Biotechnol 2023; 11:1227119. [PMID: 37840664 PMCID: PMC10569307 DOI: 10.3389/fbioe.2023.1227119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/19/2023] [Indexed: 10/17/2023] Open
Abstract
Introduction: This study presents a microfluidic tumor microenvironment (TME) model for evaluating the anti-metastatic efficacy of a novel thienopyrimidines analog with anti-cancer properties utilizing an existing commercial platform. The microfluidic device consists of a tissue compartment flanked by vascular channels, allowing for the co-culture of multiple cell types and providing a wide range of culturing conditions in one device. Methods: Human metastatic, drug-resistant triple-negative breast cancer (TNBC) cells (SUM159PTX) and primary human umbilical vein endothelial cells (HUVEC) were used to model the TME. A dynamic perfusion scheme was employed to facilitate EC physiological function and lumen formation. Results: The measured permeability of the EC barrier was comparable to observed microvessels permeability in vivo. The TNBC cells formed a 3D tumor, and co-culture with HUVEC negatively impacted EC barrier integrity. The microfluidic TME was then used to model the intravenous route of drug delivery. Paclitaxel (PTX) and a novel non-apoptotic agent TPH104c were introduced via the vascular channels and successfully reached the TNBC tumor, resulting in both time and concentration-dependent tumor growth inhibition. PTX treatment significantly reduced EC barrier integrity, highlighting the adverse effects of PTX on vascular ECs. TPH104c preserved EC barrier integrity and prevented TNBC intravasation. Discussion: In conclusion, this study demonstrates the potential of microfluidics for studying complex biological processes in a controlled environment and evaluating the efficacy and toxicity of chemotherapeutic agents in more physiologically relevant conditions. This model can be a valuable tool for screening potential anticancer drugs and developing personalized cancer treatment strategies.
Collapse
Affiliation(s)
- Indira Sigdel
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| | - Awurama Ofori-Kwafo
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| | - Robert J. Heizelman
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Andrea Nestor-Kalinoski
- Department of Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
| | | | - Amit K. Tiwari
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Yuan Tang
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| |
Collapse
|
79
|
Bahri M, Anstee JE, Opzoomer JW, Arnold JN. Perivascular tumor-associated macrophages and their role in cancer progression. Essays Biochem 2023; 67:919-928. [PMID: 37199172 PMCID: PMC10539944 DOI: 10.1042/ebc20220242] [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: 01/27/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/19/2023]
Abstract
Perivascular (Pv) tumor-associated macrophages (TAMs) are a highly specialized stromal subset within the tumor microenvironment (TME) that are defined by their spatial proximity, within one cell thickness, to blood vasculature. PvTAMs have been demonstrated to support a variety of pro-tumoral functions including angiogenesis, metastasis, and modulating the immune and stromal landscape. Furthermore, PvTAMs can also limit the response of anti-cancer and anti-angiogenic therapies and support tumor recurrence post-treatment. However, their role may not exclusively be pro-tumoral as PvTAMs can also have immune-stimulatory capabilities. PvTAMs are derived from a monocyte progenitor that develop and localize to the Pv niche as part of a multistep process which relies on a series of signals from tumor, endothelial and Pv mesenchymal cell populations. These cellular communications and signals create a highly specialized TAM subset that can also form CCR5-dependent multicellular 'nest' structures in the Pv niche. This review considers our current understanding of the role of PvTAMs, their markers for identification, development, and function in cancer. The role of PvTAMs in supporting disease progression and modulating the outcome from anti-cancer therapies highlight these cells as a therapeutic target. However, their resistance to pan-TAM targeting therapies, such as those targeting the colony stimulating factor-1 (CSF1)-CSF1 receptor axis, prompts the need for more targeted therapeutic approaches to be considered for this subset. This review highlights potential therapeutic strategies to target and modulate PvTAM development and function in the TME.
Collapse
Affiliation(s)
- Meriem Bahri
- School of Cancer and Pharmaceutical Sciences, King's College London, Faculty of Life Sciences and Medicine, Guy's Hospital, London SE1 1UL, United Kingdom
| | - Joanne E Anstee
- School of Cancer and Pharmaceutical Sciences, King's College London, Faculty of Life Sciences and Medicine, Guy's Hospital, London SE1 1UL, United Kingdom
| | - James W Opzoomer
- School of Cancer and Pharmaceutical Sciences, King's College London, Faculty of Life Sciences and Medicine, Guy's Hospital, London SE1 1UL, United Kingdom
| | - James N Arnold
- School of Cancer and Pharmaceutical Sciences, King's College London, Faculty of Life Sciences and Medicine, Guy's Hospital, London SE1 1UL, United Kingdom
| |
Collapse
|
80
|
Lin Q, Choyke PL, Sato N. Visualizing vasculature and its response to therapy in the tumor microenvironment. Theranostics 2023; 13:5223-5246. [PMID: 37908739 PMCID: PMC10614675 DOI: 10.7150/thno.84947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/30/2023] [Indexed: 11/02/2023] Open
Abstract
Tumor vasculature plays a critical role in the progression and metastasis of tumors, antitumor immunity, drug delivery, and resistance to therapies. The morphological and functional changes of tumor vasculature in response to therapy take place in a spatiotemporal-dependent manner, which can be predictive of treatment outcomes. Dynamic monitoring of intratumor vasculature contributes to an improved understanding of the mechanisms of action of specific therapies or reasons for treatment failure, leading to therapy optimization. There is a rich history of methods used to image the vasculature. This review describes recent advances in imaging technologies to visualize the tumor vasculature, with a focus on enhanced intravital imaging techniques and tumor window models. We summarize new insights on spatial-temporal vascular responses to various therapies, including changes in vascular perfusion and permeability and immune-vascular crosstalk, obtained from intravital imaging. Finally, we briefly discuss the clinical applications of intravital imaging techniques.
Collapse
Affiliation(s)
| | | | - Noriko Sato
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
81
|
Singh S, Barik D, Arukha AP, Prasad S, Mohapatra I, Singh A, Singh G. Small Molecule Targeting Immune Cells: A Novel Approach for Cancer Treatment. Biomedicines 2023; 11:2621. [PMID: 37892995 PMCID: PMC10604364 DOI: 10.3390/biomedicines11102621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/05/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Conventional and cancer immunotherapies encompass diverse strategies to address various cancer types and stages. However, combining these approaches often encounters limitations such as non-specific targeting, resistance development, and high toxicity, leading to suboptimal outcomes in many cancers. The tumor microenvironment (TME) is orchestrated by intricate interactions between immune and non-immune cells dictating tumor progression. An innovative avenue in cancer therapy involves leveraging small molecules to influence a spectrum of resistant cell populations within the TME. Recent discoveries have unveiled a phenotypically diverse cohort of innate-like T (ILT) cells and tumor hybrid cells (HCs) exhibiting novel characteristics, including augmented proliferation, migration, resistance to exhaustion, evasion of immunosurveillance, reduced apoptosis, drug resistance, and heightened metastasis frequency. Leveraging small-molecule immunomodulators to target these immune players presents an exciting frontier in developing novel tumor immunotherapies. Moreover, combining small molecule modulators with immunotherapy can synergistically enhance the inhibitory impact on tumor progression by empowering the immune system to meticulously fine-tune responses within the TME, bolstering its capacity to recognize and eliminate cancer cells. This review outlines strategies involving small molecules that modify immune cells within the TME, potentially revolutionizing therapeutic interventions and enhancing the anti-tumor response.
Collapse
Affiliation(s)
- Shilpi Singh
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Debashis Barik
- Center for Computational Natural Science and Bioinformatics, International Institute of Information Technology, Hyderabad 500032, Telangana, India
| | | | | | - Iteeshree Mohapatra
- Department of Veterinary and Biomedical Sciences, University of Minnesota—Twin Cities, Saint Paul, MN 55108, USA
| | - Amar Singh
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Gatikrushna Singh
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
82
|
Li H, Ma T, Hao M, Guo W, Gu J, Zhang X, Wei L. Decoding functional cell-cell communication events by multi-view graph learning on spatial transcriptomics. Brief Bioinform 2023; 24:bbad359. [PMID: 37824741 DOI: 10.1093/bib/bbad359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/25/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023] Open
Abstract
Cell-cell communication events (CEs) are mediated by multiple ligand-receptor (LR) pairs. Usually only a particular subset of CEs directly works for a specific downstream response in a particular microenvironment. We name them as functional communication events (FCEs) of the target responses. Decoding FCE-target gene relations is: important for understanding the mechanisms of many biological processes, but has been intractable due to the mixing of multiple factors and the lack of direct observations. We developed a method HoloNet for decoding FCEs using spatial transcriptomic data by integrating LR pairs, cell-type spatial distribution and downstream gene expression into a deep learning model. We modeled CEs as a multi-view network, developed an attention-based graph learning method to train the model for generating target gene expression with the CE networks, and decoded the FCEs for specific downstream genes by interpreting trained models. We applied HoloNet on three Visium datasets of breast cancer and liver cancer. The results detangled the multiple factors of FCEs by revealing how LR signals and cell types affect specific biological processes, and specified FCE-induced effects in each single cell. We conducted simulation experiments and showed that HoloNet is more reliable on LR prioritization in comparison with existing methods. HoloNet is a powerful tool to illustrate cell-cell communication landscapes and reveal vital FCEs that shape cellular phenotypes. HoloNet is available as a Python package at https://github.com/lhc17/HoloNet.
Collapse
Affiliation(s)
- Haochen Li
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Tianxing Ma
- MOE Key Lab of Bioinformatics, Bioinformatics Division of BNRIST and Department of Automation, Tsinghua University, Beijing 100084, China
| | - Minsheng Hao
- MOE Key Lab of Bioinformatics, Bioinformatics Division of BNRIST and Department of Automation, Tsinghua University, Beijing 100084, China
| | - Wenbo Guo
- MOE Key Lab of Bioinformatics, Bioinformatics Division of BNRIST and Department of Automation, Tsinghua University, Beijing 100084, China
| | - Jin Gu
- MOE Key Lab of Bioinformatics, Bioinformatics Division of BNRIST and Department of Automation, Tsinghua University, Beijing 100084, China
| | - Xuegong Zhang
- School of Medicine, Tsinghua University, Beijing 100084, China
- MOE Key Lab of Bioinformatics, Bioinformatics Division of BNRIST and Department of Automation, Tsinghua University, Beijing 100084, China
| | - Lei Wei
- MOE Key Lab of Bioinformatics, Bioinformatics Division of BNRIST and Department of Automation, Tsinghua University, Beijing 100084, China
| |
Collapse
|
83
|
Anstee JE, Feehan KT, Opzoomer JW, Dean I, Muller HP, Bahri M, Cheung TS, Liakath-Ali K, Liu Z, Choy D, Caron J, Sosnowska D, Beatson R, Muliaditan T, An Z, Gillett CE, Lan G, Zou X, Watt FM, Ng T, Burchell JM, Kordasti S, Withers DR, Lawrence T, Arnold JN. LYVE-1 + macrophages form a collaborative CCR5-dependent perivascular niche that influences chemotherapy responses in murine breast cancer. Dev Cell 2023; 58:1548-1561.e10. [PMID: 37442140 DOI: 10.1016/j.devcel.2023.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 04/05/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023]
Abstract
Tumor-associated macrophages (TAMs) are a heterogeneous population of cells that facilitate cancer progression. However, our knowledge of the niches of individual TAM subsets and their development and function remain incomplete. Here, we describe a population of lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1)-expressing TAMs, which form coordinated multi-cellular "nest" structures that are heterogeneously distributed proximal to vasculature in tumors of a spontaneous murine model of breast cancer. We demonstrate that LYVE-1+ TAMs develop in response to IL-6, which induces their expression of the immune-suppressive enzyme heme oxygenase-1 and promotes a CCR5-dependent signaling axis, which guides their nest formation. Blocking the development of LYVE-1+ TAMs or their nest structures, using gene-targeted mice, results in an increase in CD8+ T cell recruitment to the tumor and enhanced response to chemotherapy. This study highlights an unappreciated collaboration of a TAM subset to form a coordinated niche linked to immune exclusion and resistance to anti-cancer therapy.
Collapse
Affiliation(s)
- Joanne E Anstee
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK
| | - Karen T Feehan
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK
| | - James W Opzoomer
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK
| | - Isaac Dean
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Henrike P Muller
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK
| | - Meriem Bahri
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK
| | - Tik Shing Cheung
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK
| | | | - Ziyan Liu
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK
| | - Desmond Choy
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK
| | - Jonathan Caron
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK
| | - Dominika Sosnowska
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK
| | - Richard Beatson
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK
| | - Tamara Muliaditan
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK
| | - Zhengwen An
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK
| | - Cheryl E Gillett
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK
| | - Guocheng Lan
- Cancer Research UK Cambridge Research Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 ORE, UK
| | - Xiangang Zou
- Cancer Research UK Cambridge Research Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 ORE, UK
| | - Fiona M Watt
- Centre for Stem Cells and Regenerative Medicine, King's College London, London SE1 9RT, UK
| | - Tony Ng
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK; UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Joy M Burchell
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK
| | - Shahram Kordasti
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK; Haematology Department, Guy's Hospital, London SE1 9RT, UK
| | - David R Withers
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Toby Lawrence
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK; Aix Marseille University, CNRS, INSERM, CIML, Marseille, France; Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - James N Arnold
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK.
| |
Collapse
|
84
|
Diamantopoulou Z, Gvozdenovic A, Aceto N. A new time dimension in the fight against metastasis. Trends Cell Biol 2023; 33:736-748. [PMID: 36967300 DOI: 10.1016/j.tcb.2023.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/26/2023] [Accepted: 02/10/2023] [Indexed: 06/18/2023]
Abstract
Despite advances in uncovering vulnerabilities, identifying biomarkers, and developing more efficient treatments, cancer remains a threat because of its ability to progress while acquiring resistance to therapy. The circadian rhythm governs most of the cellular functions implicated in cancer progression, and its exploitation therefore opens new promising directions in the fight against metastasis. In this review we summarize the role of the circadian rhythm in tumor development and progression, with emphasis on the circadian rhythm-regulated elements that control the generation of circulating tumor cells (CTCs) and metastasis. We then present data on chronotherapy and discuss how circadian rhythm investigations may open new paths to more effective anticancer treatments.
Collapse
Affiliation(s)
- Zoi Diamantopoulou
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Ana Gvozdenovic
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Nicola Aceto
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.
| |
Collapse
|
85
|
Bied M, Ho WW, Ginhoux F, Blériot C. Roles of macrophages in tumor development: a spatiotemporal perspective. Cell Mol Immunol 2023; 20:983-992. [PMID: 37429944 PMCID: PMC10468537 DOI: 10.1038/s41423-023-01061-6] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/16/2023] [Indexed: 07/12/2023] Open
Abstract
Macrophages are critical regulators of tissue homeostasis but are also abundant in the tumor microenvironment (TME). In both primary tumors and metastases, such tumor-associated macrophages (TAMs) seem to support tumor development. While we know that TAMs are the dominant immune cells in the TME, their vast heterogeneity and associated functions are only just being unraveled. In this review, we outline the various known TAM populations found thus far and delineate their specialized roles associated with the main stages of cancer progression. We discuss how macrophages may prime the premetastatic niche to enable the growth of a metastasis and then how subsequent metastasis-associated macrophages can support secondary tumor growth. Finally, we speculate on the challenges that remain to be overcome in TAM research.
Collapse
Affiliation(s)
- Mathilde Bied
- Institut Gustave Roussy, INSERM U1015, Villejuif, France
| | - William W Ho
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Florent Ginhoux
- Institut Gustave Roussy, INSERM U1015, Villejuif, France.
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China.
- Translational Immunology Institute, SingHealth Duke-NUS, Singapore, Singapore.
| | - Camille Blériot
- Institut Gustave Roussy, INSERM U1015, Villejuif, France.
- Institut Necker des Enfants Malades, INSERM, CNRS, Université Paris Cité, Paris, France.
| |
Collapse
|
86
|
Sun L, Kienzler JC, Reynoso JG, Lee A, Shiuan E, Li S, Kim J, Ding L, Monteleone AJ, Owens GC, Phillips JJ, Everson RG, Nathanson D, Cloughesy TF, Li G, Liau LM, Hugo W, Kim W, Prins RM. Immune checkpoint blockade induces distinct alterations in the microenvironments of primary and metastatic brain tumors. J Clin Invest 2023; 133:e169314. [PMID: 37655659 PMCID: PMC10471177 DOI: 10.1172/jci169314] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 07/12/2023] [Indexed: 09/02/2023] Open
Abstract
In comparison with responses in recurrent glioblastoma (rGBM), the intracranial response of brain metastases (BrM) to immune checkpoint blockade (ICB) is less well studied. Here, we present an integrated single-cell RNA-Seq (scRNA-Seq) study of 19 ICB-naive and 9 ICB-treated BrM samples from our own and published data sets. We compared them with our previously published scRNA-Seq data from rGBM and found that ICB led to more prominent T cell infiltration into BrM than rGBM. These BrM-infiltrating T cells exhibited a tumor-specific phenotype and displayed greater activated/exhausted features. We also used multiplex immunofluorescence and spatial transcriptomics to reveal that ICB reduced a distinct CD206+ macrophage population in the perivascular space, which may modulate T cell entry into BrM. Furthermore, we identified a subset of progenitor exhausted T cells that correlated with longer overall survival in BrM patients. Our study provides a comprehensive immune cellular landscape of ICB's effect on metastatic brain tumors and offers insights into potential strategies for improving ICB efficacy for brain tumor patients.
Collapse
Affiliation(s)
- Lu Sun
- Department of Neurosurgery, UCLA, Los Angeles, California, USA
| | - Jenny C. Kienzler
- Department of Neurosurgery, UCLA, Los Angeles, California, USA
- Inflammation Research Group, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Alexander Lee
- Department of Neurosurgery, UCLA, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology
| | | | | | | | - Lizhong Ding
- UCLA Jonsson Comprehensive Cancer Center (JCCC), and
- Department of Medicine/Dermatology, UCLA, Los Angeles, California, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | | | | | - Joanna J. Phillips
- Helen Diller Family Comprehensive Cancer Center and
- Department of Neurological Surgery, UCSF, San Francisco, California, USA
| | - Richard G. Everson
- Department of Neurosurgery, UCLA, Los Angeles, California, USA
- UCLA Jonsson Comprehensive Cancer Center (JCCC), and
| | - David Nathanson
- Department of Molecular and Medical Pharmacology
- UCLA Jonsson Comprehensive Cancer Center (JCCC), and
| | - Timothy F. Cloughesy
- Department of Neurosurgery, UCLA, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology
- UCLA Jonsson Comprehensive Cancer Center (JCCC), and
- Department of Neurology/Neuro-Oncology, UCLA, Los Angeles, California, USA
| | | | - Linda M. Liau
- Department of Neurosurgery, UCLA, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology
- UCLA Jonsson Comprehensive Cancer Center (JCCC), and
| | - Willy Hugo
- UCLA Jonsson Comprehensive Cancer Center (JCCC), and
- Department of Medicine/Dermatology, UCLA, Los Angeles, California, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Won Kim
- Department of Neurosurgery, UCLA, Los Angeles, California, USA
- UCLA Jonsson Comprehensive Cancer Center (JCCC), and
| | - Robert M. Prins
- Department of Neurosurgery, UCLA, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology
- UCLA Jonsson Comprehensive Cancer Center (JCCC), and
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| |
Collapse
|
87
|
Ma H, Zhang Z, Hu Q, Chen H, Wu G, Zhou Y, Xue Q. Shedding light on macrophage immunotherapy in lung cancer. J Cancer Res Clin Oncol 2023; 149:8143-8152. [PMID: 37052632 DOI: 10.1007/s00432-023-04740-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/02/2023] [Indexed: 04/14/2023]
Abstract
The search for therapeutic options for lung cancer continues to advance, with rapid advances in the search for therapies to improve patient prognosis. At present, systemic chemotherapy, immune checkpoint inhibitor therapy, antiangiogenic therapy, and targeted therapy for driver gene positivity are available in the clinic. Common clinical treatments fail to achieve desired outcomes due to immunosuppression of the tumor microenvironment (TME). Tumor immune evasion is mediated by cytokines, chemokines, immune cells, and other cells such as vascular endothelial cells within the tumor immune microenvironment. Tumor-associated macrophages (TAMs) are important immune cells in the TME, inducing tumor angiogenesis, encouraging tumor cell proliferation and migration, and suppressing antitumor immune responses. Thus, TAM targeting becomes the key to lung cancer immunotherapy. This review focuses on macrophage phenotype, polarization mechanism, role in lung cancer, and advances in macrophage centric immunotherapies.
Collapse
Affiliation(s)
- Huiyun Ma
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Zhouwei Zhang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Qin Hu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Hongyu Chen
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Gujie Wu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Youlang Zhou
- Research Central of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China.
| | - Qun Xue
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China.
| |
Collapse
|
88
|
Liu X, Huangfu Y, Wang J, Kong P, Tian W, Liu P, Fang C, Li S, Nie Y, Feng Z, Huang P, Shi S, Zhang C, Dong A, Wang W. Supramolecular Polymer-Nanomedicine Hydrogel Loaded with Tumor Associated Macrophage-Reprogramming polyTLR7/8a Nanoregulator for Enhanced Anti-Angiogenesis Therapy of Orthotopic Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300637. [PMID: 37229748 PMCID: PMC10401096 DOI: 10.1002/advs.202300637] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/07/2023] [Indexed: 05/27/2023]
Abstract
Anti-angiogenic therapies targeting inhibition of vascular endothelial growth factor (VEGF) pathway show clinical benefit in hypervascular hepatocellular carcinoma (HCC) tumors. However, HCC expresses massive pro-angiogenic factors in the tumor microenvironment (TME) in response to anti-angiogenic therapy, recruiting tumor-associated macrophages (TAMs), leading to revascularization and tumor progression. To regulate cell types in TME and promote the therapeutic efficiency of anti-angiogenic therapy, a supramolecular hydrogel drug delivery system (PLDX-PMI) co-assembled by anti-angiogenic nanomedicines (PCN-Len nanoparticles (NPs)) and oxidized dextran (DX), and loaded with TAMs-reprogramming polyTLR7/8a nanoregulators (p(Man-IMDQ) NRs) is developed for orthotopic liver cancer therapy. PCN-Len NPs target tyrosine kinases of vascular endothelial cells and blocked VEGFR signaling pathway. p(Man-IMDQ) NRs repolarize pro-angiogenic M2-type TAMs into anti-angiogenic M1-type TAMs via mannose-binding receptors, reducing the secretion of VEGF, which further compromised the migration and proliferation of vascular endothelial cells. On highly malignant orthotopic liver cancer Hepa1-6 model, it is found that a single administration of the hydrogel formulation significantly decreases tumor microvessel density, promotes tumor vascular network maturation, and reduces M2-subtype TAMs, thereby effectively inhibiting tumor progression. Collectively, findings in this work highlight the great significance of TAMs reprogramming in enhancing anti-angiogenesis treatment for orthotopic HCC, and provides an advanced hydrogel delivery system-based synergistic approach for tumor therapy.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Polymer Science and EngineeringKey Laboratory of Systems Bioengineering (Ministry of Education)School of Chemical Engineering and TechnologyTianjin UniversityTianjin300072P. R. China
| | - Yini Huangfu
- Department of Polymer Science and EngineeringKey Laboratory of Systems Bioengineering (Ministry of Education)School of Chemical Engineering and TechnologyTianjin UniversityTianjin300072P. R. China
| | - Jingrong Wang
- Tianjin Key Laboratory of Biomaterial ResearchInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192P. R. China
| | - Pengxu Kong
- Department of Structural Heart DiseaseFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037P. R. China
| | - Weijun Tian
- Department of General SurgeryTianjin Medical University General HospitalTianjin300052P. R. China
| | - Peng Liu
- Department of General SurgeryTianjin Medical University General HospitalTianjin300052P. R. China
| | - Chuang Fang
- Department of General SurgeryTianjin Medical University General HospitalTianjin300052P. R. China
| | - Shuangyang Li
- Department of Polymer Science and EngineeringKey Laboratory of Systems Bioengineering (Ministry of Education)School of Chemical Engineering and TechnologyTianjin UniversityTianjin300072P. R. China
| | - Yu Nie
- Department of Gastrointestinal OncologyShandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanShandong250117P. R. China
| | - Zujian Feng
- Tianjin Key Laboratory of Biomaterial ResearchInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192P. R. China
| | - Pingsheng Huang
- Tianjin Key Laboratory of Biomaterial ResearchInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192P. R. China
| | - Shengbin Shi
- Department of Gastrointestinal OncologyShandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanShandong250117P. R. China
| | - Chuangnian Zhang
- Tianjin Key Laboratory of Biomaterial ResearchInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192P. R. China
| | - Anjie Dong
- Department of Polymer Science and EngineeringKey Laboratory of Systems Bioengineering (Ministry of Education)School of Chemical Engineering and TechnologyTianjin UniversityTianjin300072P. R. China
- Frontiers Science Center for Synthetic BiologyKey Laboratory of Systems Bioengineering (MOE)Tianjin UniversityTianjin300072P. R. China
| | - Weiwei Wang
- Tianjin Key Laboratory of Biomaterial ResearchInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192P. R. China
| |
Collapse
|
89
|
Geetha R, Iyer S, Keechilat P, N GI, Thankappan KK, N V S. Evaluation of premetastatic changes in lymph nodes(pN0) of oral tongue tumour: A prospective observational Study. F1000Res 2023; 12:889. [PMID: 37786649 PMCID: PMC10541534 DOI: 10.12688/f1000research.138951.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/17/2023] [Indexed: 10/04/2023] Open
Abstract
Background: Tongue tumors show intra and inter-tumoral heterogenicity with high incidence, relapse and mortality rates necessitating further research. Recurrence/metastasis that occurs after surgical resection of primary cancer is often the reason for poor survival in these patients. Lymph nodes are the most common site of metastasis in tongue tumors. Therefore, premetastatic molecular changes can be best evaluated in lymph nodes which may epitomize the earliest events in the metastasis cascades. The presence of circulating tumor cells(CTCs) in the absence of nodal disease (N0) may represent tumor aggressiveness, suggesting an immune escape which may have high metastatic potential. This trial was developed to investigate the earliest pre-metastatic changes which may regulate tumor dormancy and predict metastasis. A better understanding of organotropism or pre-metastatic changes can help in theragnostic, thereby preventing the outbreak of overt metastasis. Methods: A single-institutional prospective observational cohort study. This trial will be conducted at a tertiary care Centre (Amrita Institute of Medical Sciences Kochi). Eligible patients will be enrolled after obtaining informed consent. The dissected lymph nodes will be subjected to histopathological and immunohistochemical analyses for premetastatic niche (PMN) formation. In addition, circulating tumor cells will be evaluated before treatment and 6 months after treatment. The patients will be followed up for a period of two years to correlate the findings with the recurrence-free survival. Expected results: The pre-metastatic changes, if detected will be a predictive biomarker. It may help to define future drug targets for metastasis chemoprevention . CTCs may define the tumor aggressiveness ,there by prognostication and helps in better disease management. Ethics and dissemination: The study has received the following approval: Ethics Committee of Amrita School of Medicine (ECASM-AIMS-2022-048).Trial Registered Prospectively( CTRI/2022/03/041256 ) on 22/03/2022 under Clinical Trial Registry of India.
Collapse
Affiliation(s)
- Rajalakshmi Geetha
- Head and Neck Surgery/Oncology, Amrita Institute of Medical Sciences - Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Subramania Iyer
- Head and Neck Surgery/Oncology, Amrita Institute of Medical Sciences - Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Pavithran Keechilat
- Medical Oncology, Amrita Institute of Medical Sciences - Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | | | - Krishna Kumar Thankappan
- Head and Neck Surgery/Oncology, Amrita Institute of Medical Sciences - Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Smitha N V
- Department of Pathology, Amrita Institute of Medical Sciences -Amrita Vishwa Vidyapeetham, Kochi, India
| |
Collapse
|
90
|
Si H, Esquivel M, Mendoza Mendoza E, Roarty K. The covert symphony: cellular and molecular accomplices in breast cancer metastasis. Front Cell Dev Biol 2023; 11:1221784. [PMID: 37440925 PMCID: PMC10333702 DOI: 10.3389/fcell.2023.1221784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
Breast cancer has emerged as the most commonly diagnosed cancer and primary cause of cancer-related deaths among women worldwide. Although significant progress has been made in targeting the primary tumor, the effectiveness of systemic treatments to prevent metastasis remains limited. Metastatic disease continues to be the predominant factor leading to fatality in the majority of breast cancer patients. The existence of a prolonged latency period between initial treatment and eventual recurrence in certain patients indicates that tumors can both adapt to and interact with the systemic environment of the host, facilitating and sustaining the progression of the disease. In order to identify potential therapeutic interventions for metastasis, it will be crucial to gain a comprehensive framework surrounding the mechanisms driving the growth, survival, and spread of tumor cells, as well as their interaction with supporting cells of the microenvironment. This review aims to consolidate recent discoveries concerning critical aspects of breast cancer metastasis, encompassing the intricate network of cells, molecules, and physical factors that contribute to metastasis, as well as the molecular mechanisms governing cancer dormancy.
Collapse
Affiliation(s)
- Hongjiang Si
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Madelyn Esquivel
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Erika Mendoza Mendoza
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Kevin Roarty
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, United States
| |
Collapse
|
91
|
Healy S, Henderson ET, Ke S, Kelly J, Simons BW, Hu C, Ivkov R, Korangath P. Spatial analysis of nanoparticle distribution in human breast xenografts reveals nanoparticles targeted to cancer cells localized with tumor-associated stromal cells. Nanotheranostics 2023; 7:393-411. [PMID: 37426881 PMCID: PMC10327423 DOI: 10.7150/ntno.84255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/31/2023] [Indexed: 07/11/2023] Open
Abstract
The biological influence of physicochemical parameters of "targeted" nanoparticles on their delivery to cancer tumors remains poorly understood. A comparative analysis of nanoparticle distributions in tumors following systemic delivery across several models can provide valuable insights. Methods: Bionized nanoferrite nanoparticles (iron oxide core coated with starch), either conjugated with a targeted anti-HER2 antibody (BH), or unconjugated (BP), were intravenously injected into athymic nude or NOD-scid gamma (NSG) female mice bearing one of five human breast cancer tumor xenografts growing in a mammary fat pad. Tumors were harvested 24 hours after nanoparticle injection, fixed, mounted, and stained. We performed detailed histopathology analysis by comparing spatial distributions of nanoparticles (Prussian blue) with various stromal cells (CD31, SMA, F4/80, CD11c, etc.) and the target antigen-expressing (HER2) tumor cells. Results: Only BH nanoparticles were retained in tumors and generally concentrated in the tumor periphery, with nanoparticle content diminishing towards the tumor interior. Nanoparticle distribution correlated strongly with specific stromal cells within each tumor type, which varied among tumor types and between mouse strains. Weak or no correlation between nanoparticle distribution and HER2 positive cells, or CD31 cells was observed. Conclusion: Antibody-labeled nanoparticles were retained across all tumors, irrespective of presence of the "target" antigen. Though presence of antibody on nanoparticles correlated with retention, non-cancerous host stromal cells were responsible for their retention in the tumor microenvironment. This study highlights gaps in our understanding of the complex biological interplay between disease and host immune biology, and the need to account for the influence of underlying aberrant tumor biology as factors determining nanoparticle fate in vivo.
Collapse
Affiliation(s)
- Sean Healy
- Department of Biomedical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore 21218, USA
- Dept of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21231 USA
| | - Elizabeth T Henderson
- Dept of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21231 USA
| | - Suqi Ke
- Department of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Centre, School of Medicine, Johns Hopkins University, Baltimore, MD 21231 USA
| | - Jacqueline Kelly
- Dept of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21231 USA
| | - Brian W Simons
- Center for Comparative Medicine, Baylor College of Medicine, Houston, TX USA
| | - Chen Hu
- Department of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Centre, School of Medicine, Johns Hopkins University, Baltimore, MD 21231 USA
| | - Robert Ivkov
- Dept of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21231 USA
- Dept of Oncology, Sydney Kimmel Comprehensive Cancer Centre, School of Medicine, Johns Hopkins University, Baltimore, MD 21231 USA
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore 21218, USA
| | - Preethi Korangath
- Dept of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD 21231 USA
- Dept of Oncology, Sydney Kimmel Comprehensive Cancer Centre, School of Medicine, Johns Hopkins University, Baltimore, MD 21231 USA
| |
Collapse
|
92
|
Cencini E, Sicuranza A, Ciofini S, Fabbri A, Bocchia M, Gozzetti A. Tumor-Associated Macrophages in Multiple Myeloma: Key Role in Disease Biology and Potential Therapeutic Implications. Curr Oncol 2023; 30:6111-6133. [PMID: 37504315 PMCID: PMC10378698 DOI: 10.3390/curroncol30070455] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/14/2023] [Accepted: 06/23/2023] [Indexed: 07/29/2023] Open
Abstract
Multiple myeloma (MM) is characterized by multiple relapse and, despite the introduction of novel therapies, the disease becomes ultimately drug-resistant. The tumor microenvironment (TME) within the bone marrow niche includes dendritic cells, T-cytotoxic, T-helper, reactive B-lymphoid cells and macrophages, with a complex cross-talk between these cells and the MM tumor cells. Tumor-associated macrophages (TAM) have an important role in the MM pathogenesis, since they could promote plasma cells proliferation and angiogenesis, further supporting MM immune evasion and progression. TAM are polarized towards M1 (classically activated, antitumor activity) and M2 (alternatively activated, pro-tumor activity) subtypes. Many studies demonstrated a correlation between TAM, disease progression, drug-resistance and reduced survival in lymphoproliferative neoplasms, including MM. MM plasma cells in vitro could favor an M2 TAM polarization. Moreover, a possible correlation between the pro-tumor effect of M2 TAM and a reduced sensitivity to proteasome inhibitors and immunomodulatory drugs was hypothesized. Several clinical studies confirmed CD68/CD163 double-positive M2 TAM were associated with increased microvessel density, chemoresistance and reduced survival, independently of the MM stage. This review provided an overview of the biology and clinical relevance of TAM in MM, as well as a comprehensive evaluation of a potential TAM-targeted immunotherapy.
Collapse
Affiliation(s)
- Emanuele Cencini
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy
| | - Anna Sicuranza
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy
| | - Sara Ciofini
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy
| | - Alberto Fabbri
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy
| | - Monica Bocchia
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy
| | - Alessandro Gozzetti
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, University of Siena, 53100 Siena, Italy
| |
Collapse
|
93
|
Liu K, Dou R, Yang C, Di Z, Shi D, Zhang C, Song J, Fang Y, Huang S, Xiang Z, Zhang W, Wang S, Xiong B. Exosome-transmitted miR-29a induces colorectal cancer metastasis by destroying the vascular endothelial barrier. Carcinogenesis 2023; 44:356-367. [PMID: 36939367 DOI: 10.1093/carcin/bgad013] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 03/05/2023] [Accepted: 03/17/2023] [Indexed: 03/21/2023] Open
Abstract
Metastasis is the leading cause of colorectal cancer treatment failure and mortality. Communication between endothelium and tumor cells in the tumor microenvironment is required for cancer metastasis. Tumor-derived exosomes have been shown to increase vascular permeability by delivering microRNA (miRNA) to vascular endothelial cells, facilitating cancer metastasis. The mechanism by which Epithelial-mesenchymal transition (EMT) tumor cell-derived exosomes influence vascular permeability remains unknown. MicroRNA-29a (miR-29a) expression is up-regulated in colorectal cancer (CRC) tissues, which is clinically significant in metastasis. Exosomal miR-29a secreted by EMT-CRC cells has been found to decrease the expression of Zonula occlusion 1 (ZO-1), Claudin-5, and Occludin via targeting Kruppel-like factor 4 (KLF4). In vitro co-culture investigations further revealed that EMT-cancer cells release exosomal miR-29a, which alters vascular endothelial permeability. Furthermore, exosomal miR-29a promoted liver metastases in CRC mice. Our findings demonstrate that EMT-CRC cells may transport exosomal miR-29a to endothelial cells in the tumor microenvironment (TME). As a result, increased vascular permeability promotes the development and metastasis of CRC. Exosomal miR-29a has the potential to be a predictive marker for tumor metastasis as well as a viable therapeutic target for CRC.
Collapse
Affiliation(s)
- Keshu Liu
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Rongzhang Dou
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Chaogang Yang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Ziyang Di
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Dongdong Shi
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Chunxiao Zhang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Jialin Song
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Yan Fang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Sihao Huang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Zhenxian Xiang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Weisong Zhang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Shuyi Wang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Bin Xiong
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| |
Collapse
|
94
|
Kim G, Karadal-Ferrena B, Qin J, Sharma VP, Oktay IS, Lin Y, Ye X, Asiry S, Pastoriza JM, Cheng E, Ladak N, Condeelis JS, Adler E, Ginter PS, D'Alfonso T, Entenberg D, Xue X, Sparano JA, Oktay MH. Racial disparity in tumor microenvironment and distant recurrence in residual breast cancer after neoadjuvant chemotherapy. NPJ Breast Cancer 2023; 9:52. [PMID: 37311792 PMCID: PMC10264351 DOI: 10.1038/s41523-023-00547-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/28/2023] [Indexed: 06/15/2023] Open
Abstract
Black, compared to white, women with residual estrogen receptor-positive (ER+) breast cancer after neoadjuvant chemotherapy (NAC) have worse distant recurrence-free survival (DRFS). Such racial disparity may be due to difference in density of portals for systemic cancer cell dissemination, called TMEM doorways, and pro-metastatic tumor microenvironment (TME). Here, we evaluate residual cancer specimens after NAC from 96 Black and 87 white women. TMEM doorways are visualized by triple immunohistochemistry, and cancer stem cells by immunofluorescence for SOX9. The correlation between TMEM doorway score and pro-metastatic TME parameters with DRFS is examined using log-rank and multivariate Cox regression. Black, compared to white, patients are more likely to develop distant recurrence (49% vs 34.5%, p = 0.07), receive mastectomy (69.8% vs 54%, p = 0.04), and have higher grade tumors (p = 0.002). Tumors from Black patients have higher TMEM doorway and macrophages density overall (p = 0.002; p = 0.002, respectively) and in the ER+/HER2- (p = 0.02; p = 0.02, respectively), but not in the triple negative disease. Furthermore, high TMEM doorway score is associated with worse DRFS. TMEM doorway score is an independent prognostic factor in the entire study population (HR, 2.02; 95%CI, 1.18-3.46; p = 0.01), with a strong trend in ER+/HER2- disease (HR, 2.38; 95%CI, 0.96-5.95; p = 0.06). SOX9 expression is not associated with racial disparity in TME or outcome. In conclusion, higher TMEM doorway density in residual breast cancer after NAC is associated with higher distant recurrence risk, and Black patients are associated with higher TMEM doorway density, suggesting that TMEM doorway density may contribute to racial disparities in breast cancer.
Collapse
Affiliation(s)
- Gina Kim
- Department of Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Burcu Karadal-Ferrena
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Department of Basic Oncology, Hacettepe University, Ankara, Turkey
| | - Jiyue Qin
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Ved P Sharma
- Bio-Imaging Resource Center, The Rockefeller University, New York, NY, USA
| | - Isabelle S Oktay
- College of Art and Sciences, New York University, New York, NY, USA
| | - Yu Lin
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Xianjun Ye
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Saeed Asiry
- Department of Pathology, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Jessica M Pastoriza
- Department of Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | | | - Nurfiza Ladak
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - John S Condeelis
- Department of Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Esther Adler
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Paula S Ginter
- Department of Pathology, NYU Long Island School of Medicine, Mineola, NY, USA
| | - Timothy D'Alfonso
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David Entenberg
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Xiaonan Xue
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Joseph A Sparano
- Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, USA
| | - Maja H Oktay
- Department of Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
| |
Collapse
|
95
|
Hartkopf AD, Fehm TN, Welslau M, Müller V, Schütz F, Fasching PA, Janni W, Witzel I, Thomssen C, Beierlein M, Belleville E, Untch M, Thill M, Tesch H, Ditsch N, Lux MP, Aktas B, Banys-Paluchowski M, Kolberg-Liedtke C, Wöckel A, Kolberg HC, Harbeck N, Stickeler E, Bartsch R, Schneeweiss A, Ettl J, Würstlein R, Krug D, Taran FA, Lüftner D. Update Breast Cancer 2023 Part 1 - Early Stage Breast Cancer. Geburtshilfe Frauenheilkd 2023; 83:653-663. [PMID: 37916183 PMCID: PMC10617391 DOI: 10.1055/a-2074-0551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 11/03/2023] Open
Abstract
With abemaciclib (monarchE study) and olaparib (OlympiA study) gaining approval in the adjuvant treatment setting, a significant change in the standard of care for patients with early stage breast cancer has been established for some time now. Accordingly, some diverse developments are slowly being transferred from the metastatic to the adjuvant treatment setting. Recently, there have also been positive reports of the NATALEE study. Other clinical studies are currently investigating substances that are already established in the metastatic setting. These include, for example, the DESTINY Breast05 study with trastuzumab deruxtecan and the SASCIA study with sacituzumab govitecan. In this review paper, we summarize and place in context the latest developments over the past months.
Collapse
Affiliation(s)
- Andreas D. Hartkopf
- Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
| | - Tanja N. Fehm
- Department of Gynecology and Obstetrics, University Hospital Düsseldorf, Düsseldorf, Germany
| | | | - Volkmar Müller
- Department of Gynecology, Hamburg-Eppendorf University Medical Center, Hamburg, Germany
| | - Florian Schütz
- Gynäkologie und Geburtshilfe, Diakonissen-Stiftungs-Krankenhaus Speyer, Speyer, Germany
| | - Peter A. Fasching
- Erlangen University Hospital, Department of Gynecology and Obstetrics; Comprehensive Cancer Center Erlangen EMN, Friedrich-Alexander University Erlangen-Nuremberg,
Erlangen, Germany
| | - Wolfgang Janni
- Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
| | - Isabell Witzel
- Klinik für Gynäkologie, Universitätsspital Zürich, Zürich, Switzerland
| | - Christoph Thomssen
- Department of Gynaecology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Milena Beierlein
- Erlangen University Hospital, Department of Gynecology and Obstetrics; Comprehensive Cancer Center Erlangen EMN, Friedrich-Alexander University Erlangen-Nuremberg,
Erlangen, Germany
| | | | - Michael Untch
- Clinic for Gynecology and Obstetrics, Breast Cancer Center, Gynecologic Oncology Center, Helios Klinikum Berlin Buch, Berlin, Germany
| | - Marc Thill
- Department of Gynecology and Gynecological Oncology, Agaplesion Markus Krankenhaus, Frankfurt am Main, Germany
| | - Hans Tesch
- Oncology Practice at Bethanien Hospital, Frankfurt am Main, Germany
| | - Nina Ditsch
- Department of Gynecology and Obstetrics, University Hospital Augsburg, Augsburg, Germany
| | - Michael P. Lux
- Klinik für Gynäkologie und Geburtshilfe, Frauenklinik St. Louise, Paderborn, St. Josefs-Krankenhaus, Salzkotten, St. Vincenz Krankenhaus GmbH, Paderborn, Germany
| | - Bahriye Aktas
- Department of Gynecology, University of Leipzig Medical Center, Leipzig, Germany
| | - Maggie Banys-Paluchowski
- Department of Gynecology and Obstetrics, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | | | - Achim Wöckel
- Department of Gynecology and Obstetrics, University Hospital Würzburg, Würzburg, Germany
| | | | - Nadia Harbeck
- Breast Center, Department of Gynecology and Obstetrics and CCC Munich LMU, LMU University Hospital, München, Germany
| | - Elmar Stickeler
- Department of Obstetrics and Gynecology, Center for Integrated Oncology (CIO Aachen, Bonn, Cologne, Düsseldorf), University Hospital of RWTH Aachen, Aachen, Germany
| | - Rupert Bartsch
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Andreas Schneeweiss
- National Center for Tumor Diseases, University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - Johannes Ettl
- Klinik für Frauenheilkunde und Gynäkologie, Klinikum Kempten, Klinikverbund Allgäu, Kempten, Germany
| | - Rachel Würstlein
- Breast Center, Department of Gynecology and Obstetrics and CCC Munich LMU, LMU University Hospital, München, Germany
| | - David Krug
- Klinik für Strahlentherapie, Universitätsklinkum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Florin-Andrei Taran
- Department of Gynecology and Obstetrics, University Hospital Freiburg, Freiburg, Germany
| | - Diana Lüftner
- Medical University of Brandenburg Theodor-Fontane, Immanuel Hospital Märkische Schweiz, Buckow, Germany
| |
Collapse
|
96
|
Rios AC, van Rheenen J, Scheele CLGJ. Multidimensional Imaging of Breast Cancer. Cold Spring Harb Perspect Med 2023; 13:a041330. [PMID: 36167726 PMCID: PMC10153799 DOI: 10.1101/cshperspect.a041330] [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: 11/25/2022]
Abstract
Breast cancer is a pathological condition characterized by high morphological and molecular heterogeneity. Not only the breast cancer cells, but also their tumor micro-environment consists of a multitude of cell types and states, which continuously evolve throughout progression of the disease. To understand breast cancer evolution within this complex environment, in situ analysis of breast cancer and their co-evolving cells and structures in space and time are essential. In this review, recent technical advances in three-dimensional (3D) and intravital imaging of breast cancer are discussed. Moreover, we highlight the resulting new knowledge on breast cancer biology obtained through these innovative imaging technologies. Finally, we discuss how multidimensional imaging technologies can be integrated with molecular profiling to understand the full complexity of breast cancer and the tumor micro-environment during tumor progression and treatment response.
Collapse
Affiliation(s)
- Anne C Rios
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| | - Jacco van Rheenen
- Oncode Institute, 3521 AL Utrecht, The Netherlands
- Department of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Colinda L G J Scheele
- Laboratory for Intravital Imaging and Dynamics of Tumor Progression, VIB Center for Cancer Biology, KU Leuven, 3000 Leuven, Belgium
- Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
97
|
Sakibuzzaman M, Mahmud S, Afroze T, Fathma S, Zakia UB, Afroz S, Zafar F, Hossain M, Barua A, Akter S, Chowdhury HI, Ahsan E, Eshan SH, Fariza TT. Pathology of breast cancer metastasis and a view of metastasis to the brain. Int J Neurosci 2023; 133:544-554. [PMID: 34044732 DOI: 10.1080/00207454.2021.1935929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023]
Abstract
Despite the advances in diagnosis and management of breast cancer, metastasis has been responsible for the staggering percentage of breast cancer-related death. Mortality threat can be explained mostly by the lack of proper understanding of the diversity of pathological features and underlying mechanism of breast cancer metastasis and effective targeted therapy. Breast cancer stem cells (BCSCs) are the potential source of tumor cells spread to distant organs. BCSCs targeted therapy can suppress the breast cancer progression to metastasis. Spreading of tumor cells to the bone, lung, liver, and brain occurs through a distinct non-random process; called metastasis organotropism. Recently, brain metastasis in breast cancer patients has been detected more frequently, causing a significant clinical burden. BRCA1 and BRCA2 associated breast cancers carry a remarkably higher propensity of CNS metastasis. BRCA1 and BRCA2 associated breast cancers commonly have the propensity to be the triple-negative (TN) and hormone receptors (HR)-positive/human epidermal growth factor receptor 2 (HER2)-negative molecular subtypes, respectively. Regardless of molecular subtypes, metastasis is most commonly evident at the bone. Heterogeneity is a critical pathological feature, leads to therapeutic resistance. BCSCs, biomarkers expression patterns, and mutations contribute to heterogeneity. In this paper, we discuss crucial pathological features of breast cancer metastasis, emphasizing metastasis organotropism and heterogeneity; and mechanisms of breast cancer metastasis, highlighting the pathways of metastasis to the brain. We consider that this paper reinforces future research areas and benefits the general readers, physicians, and researchers to identify potential areas to develop targeted therapies.
Collapse
Affiliation(s)
- Md Sakibuzzaman
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Shahriar Mahmud
- Sher-E-Bangla Medical College and Hospital, Barisal, Bangladesh
| | | | - Sawsan Fathma
- Bangladesh Medical College and Hospital, Dhaka, Bangladesh
| | | | - Sabrina Afroz
- Faridpur Medical College and Hospital, Faridpur, Bangladesh
| | - Farzina Zafar
- Shaheed Suhrawardy Medical College and Hospital, Dhaka, Bangladesh
| | - Maksuda Hossain
- Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Amit Barua
- Institute of Applied Health Sciences, Chattogram, Bangladesh
| | - Sabiha Akter
- Sher-E-Bangla Medical College and Hospital, Barisal, Bangladesh
| | | | - Eram Ahsan
- Medical College for Women and Hospital, Dhaka, Bangladesh
| | - Shayet Hossain Eshan
- Department of Internal Medicine, Amita Health Saint Joseph Hospital Chicago, Chicago, IL, USA
| | | |
Collapse
|
98
|
Schuster E, Dashzeveg N, Jia Y, Golam K, Zhang T, Hoffman A, Zhang Y, Zheng C, Ramos E, Taftaf R, Shennawy LE, Scholten D, Kitata RB, Adorno-Cruz V, Reduzzi C, Spahija S, Xu R, Siziopikou KP, Platanias LC, Shah A, Gradishar WJ, Cristofanilli M, Tsai CF, Shi T, Liu H. Computational ranking-assisted identification of Plexin-B2 in homotypic and heterotypic clustering of circulating tumor cells in breast cancer metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.10.536233. [PMID: 37090580 PMCID: PMC10120645 DOI: 10.1101/2023.04.10.536233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Metastasis is the cause of over 90% of all deaths associated with breast cancer, yet the strategies to predict cancer spreading based on primary tumor profiles and therefore prevent metastasis are egregiously limited. As rare precursor cells to metastasis, circulating tumor cells (CTCs) in multicellular clusters in the blood are 20-50 times more likely to produce viable metastasis than single CTCs. However, the molecular mechanisms underlying various CTC clusters, such as homotypic tumor cell clusters and heterotypic tumor-immune cell clusters, are yet to be fully elucidated. Combining machine learning-assisted computational ranking with experimental demonstration to assess cell adhesion candidates, we identified a transmembrane protein Plexin- B2 (PB2) as a new therapeutic target that drives the formation of both homotypic and heterotypic CTC clusters. High PB2 expression in human primary tumors predicts an unfavorable distant metastasis-free survival and is enriched in CTC clusters compared to single CTCs in advanced breast cancers. Loss of PB2 reduces formation of homotypic tumor cell clusters as well as heterotypic tumor-myeloid cell clusters in triple-negative breast cancer. Interactions between PB2 and its ligand Sema4C on tumor cells promote homotypic cluster formation, and PB2 binding with Sema4A on myeloid cells (monocytes) drives heterotypic CTC cluster formation, suggesting that metastasizing tumor cells hijack the PB2/Sema family axis to promote lung metastasis in breast cancer. Additionally, using a global proteomic analysis, we identified novel downstream effectors of the PB2 pathway associated with cancer stemness, cell cycling, and tumor cell clustering in breast cancer. Thus, PB2 is a novel therapeutic target for preventing new metastasis.
Collapse
|
99
|
Iovan L, Liliac IM, Istrate-Ofiteru AM, Rosu CG, Mogoanta L. Clinical and Morphological Study of Cervical Squamous Intraepithelial Lesions. CURRENT HEALTH SCIENCES JOURNAL 2023; 49:220-229. [PMID: 37779826 PMCID: PMC10541069 DOI: 10.12865/chsj.49.02.220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 03/18/2023] [Indexed: 10/03/2023]
Abstract
Squamous intraepithelial lesions (SILs) are cancer precursors targeted by secondary prevention of cervical cancer programs that are sometimes difficult to grade accurately. Mena is an actin regulatory protein involved in membrane protrusion, cell motility, in tumor invasion and metastasis. We studied retrospectively 68 cases of patients diagnosed with squamous intraepithelial lesions that received expedited treatment (treatment without colposcopic biopsy). We analyzed demographic, behavioral data, obstetrical and medical history, from the patients' medical charts and we studied the cervical fragments or cones harvested after the excisional procedure. Our study failed to identify a correlation between SILs and risk factors such as low socioeconomic status, combined oral contraceptive use, intrauterine device use, parity, gravity, except for the tobacco smoking habit that proved to be related to the cervical lesions' development. Mena was expressed in most of the analyzed SILs and its expression was correlated with lesions' grade in terms of both area and intensity, suggesting that Mena stains especially abnormal cells and that its expression intensity correlates with the risk of malignant transformation. Further studies are needed to validate Mena as an early stage of cervical carcinogenesis marker.
Collapse
Affiliation(s)
- Larisa Iovan
- Department of Histology, University of Medicine and Pharmacy of Craiova, Romania
| | - Ilona Mihaela Liliac
- Department of Histology, University of Medicine and Pharmacy of Craiova, Romania
| | | | | | - Laurentiu Mogoanta
- Department of Histology, University of Medicine and Pharmacy of Craiova, Romania
| |
Collapse
|
100
|
Li X, Pan J, Liu T, Yin W, Miao Q, Zhao Z, Gao Y, Zheng W, Li H, Deng R, Huang D, Qiu S, Zhang Y, Qi Q, Deng L, Huang M, Tang PMK, Cao Y, Chen M, Ye W, Zhang D. Novel TCF21 high pericyte subpopulation promotes colorectal cancer metastasis by remodelling perivascular matrix. Gut 2023; 72:710-721. [PMID: 36805487 PMCID: PMC10086488 DOI: 10.1136/gutjnl-2022-327913] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 08/20/2022] [Indexed: 02/19/2023]
Abstract
OBJECTIVE Haematogenous dissemination is a prevalent route of colorectal cancer (CRC) metastasis. However, as the gatekeeper of vessels, the role of tumour pericytes (TPCs) in haematogenous metastasis remains largely unknown. Here, we aimed to investigate the heterogeneity of TPCs and their effects on CRC metastasis. DESIGN TPCs were isolated from patients with CRC with or without liver metastases and analysed by single-cell RNA sequencing (scRNA-seq). Clinical CRC specimens were collected to analyse the association between the molecular profiling of TPCs and CRC metastasis. RNA-sequencing, chromatin immunoprecipitation-sequencing and bisulfite-sequencing were performed to investigate the TCF21-regulated genes and mechanisms underlying integrin α5 on TCF21 DNA hypermethylation. Pericyte-conditional Tcf21-knockout mice were constructed to investigate the effects of TCF21 in TPCs on CRC metastasis. Masson staining, atomic force microscopy, second-harmonic generation and two-photon fluorescence microscopy were employed to observe perivascular extracellular matrix (ECM) remodelling. RESULTS Thirteen TPC subpopulations were identified by scRNA-seq. A novel subset of TCF21high TPCs, termed 'matrix-pericytes', was associated with liver metastasis in patients with CRC. TCF21 in TPCs increased perivascular ECM stiffness, collagen rearrangement and basement membrane degradation, establishing a perivascular metastatic microenvironment to instigate colorectal cancer liver metastasis (CRCLM). Tcf21 depletion in TPCs mitigated perivascular ECM remodelling and CRCLM, whereas the coinjection of TCF21high TPCs and CRC cells markedly promoted CRCLM. Mechanistically, loss of integrin α5 inhibited the FAK/PI3K/AKT/DNMT1 axis to impair TCF21 DNA hypermethylation in TCF21high TPCs. CONCLUSION This study uncovers a previously unidentified role of TPCs in haematogenous metastasis and provides a potential diagnostic marker and therapeutic target for CRC metastasis.
Collapse
Affiliation(s)
- Xiaobo Li
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Jinghua Pan
- Department of General Surgery, Jinan University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Tongzheng Liu
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Wenqian Yin
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Qun Miao
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Zhan Zhao
- Department of General Surgery, Jinan University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Yufeng Gao
- Chinese Academy of Sciences Shenzhen Institutes of Advanced Technology, Shenzhen, Guangdong, China
| | - Wei Zheng
- Chinese Academy of Sciences Shenzhen Institutes of Advanced Technology, Shenzhen, Guangdong, China
| | - Hang Li
- College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Rong Deng
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Dandan Huang
- Department of Coloproctology & Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, Guangdong, China
| | - Shenghui Qiu
- Department of General Surgery, Jinan University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Yiran Zhang
- Department of General Surgery, Jinan University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Qi Qi
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Lijuan Deng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Maohua Huang
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Stockholm, Sweden
| | - Minfeng Chen
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Wencai Ye
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Dongmei Zhang
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|