1
|
Das K, Schulte M, Gerhart M, Bayoumi H, Paulson D, Fink DM, Parrish C, Willand-Charnley R. Early in vitro results indicate that de-O-acetylated sialic acids increase Selectin binding in cancers. Front Oncol 2024; 14:1443303. [PMID: 39717751 PMCID: PMC11663943 DOI: 10.3389/fonc.2024.1443303] [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: 06/03/2024] [Accepted: 11/11/2024] [Indexed: 12/25/2024] Open
Abstract
Cancers utilize a simple glycan, Sialic Acid, to engage in metastatic processes via the Sialic acid (Sia) -Selectin pathway. Selectins recognize and bind to sialylated substrates, resulting in adhesion, migration, and extravasation, however, how deviations from the canonical form of Sia regulate binding to Selectin receptors (E, L, and P) on hemopoietic cells resulting in these metastatic processes, remained a gap in knowledge. De-O-acetylated Sias has been recently shown to be an integral substrate to the binding of sialic acid binding proteins. The two proteins responsible for regulating the acetyl functional group on Sia's C6 tail, are Sialic acid acetylesterase (SIAE) and Sialic acid O acetyltransferase (CASD1). To elucidate the contribution of functional group alterations on Sia, 9-O and 7,9-O-acetylation of Sia was modulated via the use of CRISRP-Cas9 gene editing and with Sialyl Glycan Recognition Probes, to produce either O-acetylated-Sia or de-O-acetylated- Sia, respectively. In vitro experiments revealed that increased cell surface expression of de-O-acetylated- Sia resulted in an increase in Selectin binding, enhanced cell proliferation, and increased migration capabilities both in lung and colon cancer. These results delineate for the first time the mechanistic contribution of de-O-acetylated-Sia to Selectin binding, reinforcing the importance of elucidating functional group alterations on Sia and their contribution. Without accurate identification of which functionalized form of Sia is being utilized to bind to sialic acid binding proteins, we cannot accurately produce glycan therapeutics with the correct specificity and reactivity, thus this work contributes an integral component in the development of promising therapeutic avenues, for example in the realm of enzyme antibody drug conjugates.
Collapse
Affiliation(s)
- Kakali Das
- Department of Chemistry, Biochemistry and Physics, South Dakota State University, Brookings, SD, United States
| | - Megan Schulte
- Department of Chemistry, Biochemistry and Physics, South Dakota State University, Brookings, SD, United States
| | - Megan Gerhart
- Department of Chemistry, Biochemistry and Physics, South Dakota State University, Brookings, SD, United States
| | - Hala Bayoumi
- Department of Chemistry, Biochemistry and Physics, South Dakota State University, Brookings, SD, United States
| | - Delayna Paulson
- Department of Chemistry, Biochemistry and Physics, South Dakota State University, Brookings, SD, United States
| | - Darci M. Fink
- Department of Chemistry, Biochemistry and Physics, South Dakota State University, Brookings, SD, United States
| | - Colin Parrish
- Department of Microbiology and Immunology, Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Rachel Willand-Charnley
- Department of Chemistry, Biochemistry and Physics, South Dakota State University, Brookings, SD, United States
| |
Collapse
|
2
|
Alnaqbi H, Becker LM, Mousa M, Alshamsi F, Azzam SK, Emini Veseli B, Hymel LA, Alhosani K, Alhusain M, Mazzone M, Alsafar H, Carmeliet P. Immunomodulation by endothelial cells: prospects for cancer therapy. Trends Cancer 2024; 10:1072-1091. [PMID: 39289084 DOI: 10.1016/j.trecan.2024.08.002] [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: 04/22/2024] [Revised: 07/26/2024] [Accepted: 08/09/2024] [Indexed: 09/19/2024]
Abstract
Growing evidence highlights the importance of tumor endothelial cells (TECs) in the tumor microenvironment (TME) for promoting tumor growth and evading immune responses. Immunomodulatory endothelial cells (IMECs) represent a distinct plastic phenotype of ECs that exerts the ability to modulate immunity in health and disease. This review discusses our current understanding of IMECs in cancer biology, scrutinizing insights from single-cell reports to compare their characteristics and function dynamics across diverse tumor types, conditions, and species. We investigate possible implications of exploiting IMECs in the context of cancer treatment, particularly examining their influence on the efficacy of existing therapies and the potential to leverage them as targets in optimizing immunotherapeutic strategies.
Collapse
Affiliation(s)
- Halima Alnaqbi
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates; Department of Biomedical Engineering and Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Lisa M Becker
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Mira Mousa
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates; Department of Public Health and Epidemiology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Fatima Alshamsi
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates; Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Sarah K Azzam
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Besa Emini Veseli
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Lauren A Hymel
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Khalood Alhosani
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Marwa Alhusain
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium.
| | - Habiba Alsafar
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates; Department of Biomedical Engineering and Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Peter Carmeliet
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates; Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, University of Leuven, Leuven, Belgium.
| |
Collapse
|
3
|
Lu K, Xia Y, Cheng P, Li Y, He L, Tao L, Wei Z, Lu Y. Synergistic potentiation of the anti-metastatic effect of a Ginseng-Salvia miltiorrhiza herbal pair and its biological ingredients via the suppression of CD62E-dependent neutrophil infiltration and NETformation. J Adv Res 2024:S2090-1232(24)00490-9. [PMID: 39481643 DOI: 10.1016/j.jare.2024.10.036] [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: 05/06/2024] [Revised: 09/19/2024] [Accepted: 10/29/2024] [Indexed: 11/02/2024] Open
Abstract
INTRODUCTION The combination of the roots of ginseng and Salvia miltiorrhiza is an effective approach for treating metastatic cancer in patients with Qi stagnation and blood stasis patterns. However, the molecular mechanism underlying the combined use of ginseng and Salvia miltiorrhiza is unknown. OBJECTIVES This study unveils the pharmacological foundation of ginseng and Salvia miltiorrhiza by examining the involvement of neutrophils in the critical process of tumor hematogenous metastasis. Additionally, by employing a reverse pharmacology research model (effect-target-constituent), potential potent components were screened, and the dominant component formulations were determined. METHODS An experimental lung metastatic model was constructed to compare the antitumor effects of ginseng and Salvia miltiorrhiza. RNA sequencing was employed to identify pivotal biological events and key targets, while the detection of CD62E expression and neutrophil extracellular traps (NETs) release was used to screen for effective substances in ginseng and Salvia miltiorrhiza. In addition, a comprehensive array of in vitro and in vivo experiments was conducted to explore the underlying mechanisms and therapeutic significance. RESULTS Compared with single-herb use, the use of ginseng or Salvia miltiorrhiza significantly reduced tumor metastasis, which was accompanied by reduced neutrophil infiltration into the lungs. Cryptotanshinone (CPT), an active constituent of Salvia miltiorrhiza, can inhibit neutrophil adhesion and recruitment to lung tissue by downregulating the expression of E-selectin (CD62E) in endothelial cells. Moreover, the ginseng-derived ginsenoside Rg1 mitigated the formation of NETs in lung tissues and reversed the protumor effects of NETs. We further explored the efficacy of combination therapy with Rg1 and CPT, which also reduced tumor metastasis in vivo. CONCLUSION Ginseng and Salvia miltiorrhiza exhibited a mutual potentiation of the anti-metastatic effect by suppressing both early and late stages of neutrophil-initiated metastasis cascade. Rg1 and CPT represent the synergistic ingredients from ginseng and Salvia miltiorrhiza, respectively.
Collapse
Affiliation(s)
- Keqin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yawen Xia
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Peng Cheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yanan Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Liang He
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Li Tao
- Department of Pharmacy, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China.
| | - Zhonghong Wei
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
4
|
Bromberger S, Zadorozhna Y, Ressler JM, Holzner S, Nawrocki A, Zila N, Springer A, Røssel Larsen M, Schossleitner K. Off-targets of BRAF inhibitors disrupt endothelial signaling and vascular barrier function. Life Sci Alliance 2024; 7:e202402671. [PMID: 38839106 PMCID: PMC11153892 DOI: 10.26508/lsa.202402671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/07/2024] Open
Abstract
Targeted therapies against mutant BRAF are effectively used in combination with MEK inhibitors (MEKi) to treat advanced melanoma. However, treatment success is affected by resistance and adverse events (AEs). Approved BRAF inhibitors (BRAFi) show high levels of target promiscuity, which can contribute to these effects. The blood vessel lining is in direct contact with high plasma concentrations of BRAFi, but effects of the inhibitors in this cell type are unknown. Hence, we aimed to characterize responses to approved BRAFi for melanoma in the vascular endothelium. We showed that clinically approved BRAFi induced a paradoxical activation of endothelial MAPK signaling. Moreover, phosphoproteomics revealed distinct sets of off-targets per inhibitor. Endothelial barrier function and junction integrity were impaired upon treatment with vemurafenib and the next-generation dimerization inhibitor PLX8394, but not with dabrafenib or encorafenib. Together, these findings provide insights into the surprisingly distinct side effects of BRAFi on endothelial signaling and functionality. Better understanding of off-target effects could help to identify molecular mechanisms behind AEs and guide the continued development of therapies for BRAF-mutant melanoma.
Collapse
Affiliation(s)
- Sophie Bromberger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Yuliia Zadorozhna
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | | - Silvio Holzner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Arkadiusz Nawrocki
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Nina Zila
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- University of Applied Sciences FH Campus Wien, Division of Biomedical Science, Vienna, Austria
| | - Alexander Springer
- Department of Pediatric Surgery, Medical University of Vienna, Vienna, Austria
| | - Martin Røssel Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | | |
Collapse
|
5
|
Ryan AT, Kim M, Lim K. Immune Cell Migration to Cancer. Cells 2024; 13:844. [PMID: 38786066 PMCID: PMC11120175 DOI: 10.3390/cells13100844] [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: 03/23/2024] [Revised: 04/27/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Immune cell migration is required for the development of an effective and robust immune response. This elegant process is regulated by both cellular and environmental factors, with variables such as immune cell state, anatomical location, and disease state that govern differences in migration patterns. In all cases, a major factor is the expression of cell surface receptors and their cognate ligands. Rapid adaptation to environmental conditions partly depends on intrinsic cellular immune factors that affect a cell's ability to adjust to new environment. In this review, we discuss both myeloid and lymphoid cells and outline key determinants that govern immune cell migration, including molecules required for immune cell adhesion, modes of migration, chemotaxis, and specific chemokine signaling. Furthermore, we summarize tumor-specific elements that contribute to immune cell trafficking to cancer, while also exploring microenvironment factors that can alter these cellular dynamics within the tumor in both a pro and antitumor fashion. Specifically, we highlight the importance of the secretome in these later aspects. This review considers a myriad of factors that impact immune cell trajectory in cancer. We aim to highlight the immunotherapeutic targets that can be harnessed to achieve controlled immune trafficking to and within tumors.
Collapse
Affiliation(s)
- Allison T. Ryan
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (A.T.R.); (M.K.)
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (A.T.R.); (M.K.)
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Kihong Lim
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (A.T.R.); (M.K.)
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| |
Collapse
|
6
|
Kuznetsova AB, Kolesova EP, Parodi A, Zamyatnin AA, Egorova VS. Reprogramming Tumor-Associated Macrophage Using Nanocarriers: New Perspectives to Halt Cancer Progression. Pharmaceutics 2024; 16:636. [PMID: 38794298 PMCID: PMC11124960 DOI: 10.3390/pharmaceutics16050636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Cancer remains a significant challenge for public healthcare systems worldwide. Within the realm of cancer treatment, considerable attention is focused on understanding the tumor microenvironment (TME)-the complex network of non-cancerous elements surrounding the tumor. Among the cells in TME, tumor-associated macrophages (TAMs) play a central role, traditionally categorized as pro-inflammatory M1 macrophages or anti-inflammatory M2 macrophages. Within the TME, M2-like TAMs can create a protective environment conducive to tumor growth and progression. These TAMs secrete a range of factors and molecules that facilitate tumor angiogenesis, increased vascular permeability, chemoresistance, and metastasis. In response to this challenge, efforts are underway to develop adjuvant therapy options aimed at reprogramming TAMs from the M2 to the anti-tumor M1 phenotype. Such reprogramming holds promise for suppressing tumor growth, alleviating chemoresistance, and impeding metastasis. Nanotechnology has enabled the development of nanoformulations that may soon offer healthcare providers the tools to achieve targeted drug delivery, controlled drug release within the TME for TAM reprogramming and reduce drug-related adverse events. In this review, we have synthesized the latest data on TAM polarization in response to TME factors, highlighted the pathological effects of TAMs, and provided insights into existing nanotechnologies aimed at TAM reprogramming and depletion.
Collapse
Affiliation(s)
- Alyona B. Kuznetsova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
| | - Ekaterina P. Kolesova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
| | - Alessandro Parodi
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
| | - Andrey A. Zamyatnin
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Department of Biological Chemistry, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Vera S. Egorova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia; (A.B.K.); (E.P.K.); (A.P.)
| |
Collapse
|
7
|
Wang M, Chen S, He X, Yuan Y, Wei X. Targeting inflammation as cancer therapy. J Hematol Oncol 2024; 17:13. [PMID: 38520006 PMCID: PMC10960486 DOI: 10.1186/s13045-024-01528-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/07/2024] [Indexed: 03/25/2024] Open
Abstract
Inflammation has accompanied human beings since the emergence of wounds and infections. In the past decades, numerous efforts have been undertaken to explore the potential role of inflammation in cancer, from tumor development, invasion, and metastasis to the resistance of tumors to treatment. Inflammation-targeted agents not only demonstrate the potential to suppress cancer development, but also to improve the efficacy of other therapeutic modalities. In this review, we describe the highly dynamic and complex inflammatory tumor microenvironment, with discussion on key inflammation mediators in cancer including inflammatory cells, inflammatory cytokines, and their downstream intracellular pathways. In addition, we especially address the role of inflammation in cancer development and highlight the action mechanisms of inflammation-targeted therapies in antitumor response. Finally, we summarize the results from both preclinical and clinical studies up to date to illustrate the translation potential of inflammation-targeted therapies.
Collapse
Affiliation(s)
- Manni Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Siyuan Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xuemei He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yong Yuan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
8
|
Kikuchi H, Maishi N, Yu L, Jia Z, Li C, Sato M, Takeda R, Ishizuka K, Hida Y, Shinohara N, Hida K. Low-dose metronomic cisplatin as an antiangiogenic and anti-inflammatory strategy for cancer. Br J Cancer 2024; 130:336-345. [PMID: 38036665 PMCID: PMC10803316 DOI: 10.1038/s41416-023-02498-2] [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: 03/16/2023] [Revised: 10/28/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Conventional chemotherapy is based on the maximum tolerated dose (MTD) and requires treatment-free intervals to restore normal host cells. MTD chemotherapy may induce angiogenesis or immunosuppressive cell infiltration during treatment-free intervals. Low-dose metronomic (LDM) chemotherapy is defined as frequent administration at lower doses and causes less inflammatory change, whereas MTD chemotherapy induces an inflammatory change. Although several LDM regimens have been applied, LDM cisplatin (CDDP) has been rarely reported. This study addressed the efficacy of LDM CDDP on tumour endothelial cell phenotypic alteration compared to MTD CDDP. METHODS Tumour growth and metastasis were assessed in bladder cancer-bearing mice treated with LDM or MTD gemcitabine (GEM) and CDDP. To elucidate the therapeutic effects of LDM CDDP, the change of tumour vasculature, tumour-infiltrating immune cells and inflammatory changes were evaluated by histological analysis and mRNA expression in tumour tissues. RESULTS Tumour growth and bone metastasis were more suppressed by LDM CDDP + MTD GEM treatment than MTD CDDP + MTD GEM. Myeloid-derived suppressor cell accumulation was reduced by LDM CDDP, whereas inflammatory change was induced in the tumour microenvironment by MTD CDDP. CONCLUSION LDM CDDP does not cause inflammatory change unlike MTD CDDP, suggesting that it is a promising strategy in chemotherapy.
Collapse
Affiliation(s)
- Hiroshi Kikuchi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Nako Maishi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, Japan
| | - Li Yu
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, Japan
| | - Zi Jia
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, Japan
| | - Cong Li
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, Japan
| | - Masumi Sato
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Ryo Takeda
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, Japan
| | - Keita Ishizuka
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Yasuhiro Hida
- Department of Cardiovascular and Thoracic Surgery, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Advanced Robotic and Endoscopic Surgery, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Nobuo Shinohara
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kyoko Hida
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, Japan.
| |
Collapse
|
9
|
Rodrigues DB, Reis RL, Pirraco RP. Modelling the complex nature of the tumor microenvironment: 3D tumor spheroids as an evolving tool. J Biomed Sci 2024; 31:13. [PMID: 38254117 PMCID: PMC10804490 DOI: 10.1186/s12929-024-00997-9] [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: 05/30/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Cancer remains a serious burden in society and while the pace in the development of novel and more effective therapeutics is increasing, testing platforms that faithfully mimic the tumor microenvironment are lacking. With a clear shift from animal models to more complex in vitro 3D systems, spheroids emerge as strong options in this regard. Years of development have allowed spheroid-based models to better reproduce the biomechanical cues that are observed in the tumor-associated extracellular matrix (ECM) and cellular interactions that occur in both a cell-cell and cell-ECM manner. Here, we summarize some of the key cellular interactions that drive tumor development, progression and invasion, and how successfully are these interactions recapitulated in 3D spheroid models currently in use in the field. We finish by speculating on future advancements in the field and on how these can shape the relevance of spherical 3D models for tumor modelling.
Collapse
Affiliation(s)
- Daniel B Rodrigues
- 3B's Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017, Guimarães, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, 4805-017, Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017, Guimarães, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, 4805-017, Guimarães, Portugal
| | - Rogério P Pirraco
- 3B's Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017, Guimarães, Portugal.
- ICVS/3B's, PT Government Associate Laboratory, Braga, 4805-017, Guimarães, Portugal.
| |
Collapse
|
10
|
van Gogh M, Glaus Garzon JF, Sahin D, Knopfova L, Benes P, Boyman O, Jurisica I, Borsig L. Tumor Cell-Intrinsic c-Myb Upregulation Stimulates Antitumor Immunity in a Murine Colorectal Cancer Model. Cancer Immunol Res 2023; 11:1432-1444. [PMID: 37478172 PMCID: PMC10548106 DOI: 10.1158/2326-6066.cir-22-0912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/08/2023] [Accepted: 07/20/2023] [Indexed: 07/23/2023]
Abstract
The transcription factor c-Myb is overexpressed in many different types of solid tumors, including colorectal cancer. However, its exact role in tumorigenesis is unclear. In this study, we show that tumor-intrinsic c-Myb expression in mouse models of colon cancer and melanoma suppresses tumor growth. Although no differences in proliferation, apoptosis, and angiogenesis of tumors were evident in tumors with distinct levels of c-Myb expression, we observed changes in intratumoral immune cell infiltrates. MC38 tumors with upregulated c-Myb expression showed increased numbers of CD103+ dendritic cells and eosinophils, but decreased tumor-associated macrophages (TAM). Concomitantly, an increase in the number of activated cytotoxic CD8+ T cells upon c-Myb upregulation was observed, which correlated with a pro-inflammatory tumor microenvironment and increased numbers of M1 polarized TAMs. Mechanistically, c-Myb upregulation in immunogenic MC38 colon cancer cells resulted in enhanced expression of immunomodulatory genes, including those encoding β2-microglobulin and IFNβ, and decreased expression of the gene encoding the chemokine receptor CCR2. The increased numbers of activated cytotoxic CD8+ T cells contributed to tumor growth attenuation. In poorly immunogenic CT26, LLC, and B16-BL6 tumor cells, c-Myb upregulation did not affect the immunomodulatory gene expression. Despite this, c-Myb upregulation led to reduced B16-BL6 tumor growth but it did not affect tumor growth of CT26 and LLC tumors. Altogether, we postulate that c-Myb functions as a tumor suppressor in a tumor cell-type specific manner and modulates antitumor immunity.
Collapse
Affiliation(s)
- Merel van Gogh
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | - Dilara Sahin
- Department of Immunology, University Hospital Zurich, Zurich, Switzerland
| | - Lucia Knopfova
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Petr Benes
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Igor Jurisica
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute and, Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, Toronto Western Hospital (UHN), Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Department of Computer Science, University of Toronto, Toronto, Canada
- Faculty of Dentistry, University of Toronto, Toronto, Canada
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lubor Borsig
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Comprehensive Cancer Center Zurich, University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
11
|
Leimbacher AC, Villiger P, Desboeufs N, Aboouf MA, Nanni M, Armbruster J, Ademi H, Flüchter P, Ruetten M, Gantenbein F, Haider TJ, Gassmann M, Thiersch M. Voluntary exercise does not always suppress lung cancer progression. iScience 2023; 26:107298. [PMID: 37520731 PMCID: PMC10374464 DOI: 10.1016/j.isci.2023.107298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/11/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023] Open
Abstract
Physical exercise can lower lung cancer incidence. However, its effect on lung cancer progression is less understood. Studies on exercising mice have shown decreased ectopic lung cancer growth through the secretion of interleukin-6 from muscles and the recruitment of natural killer (NK) cells to tumors. We asked if exercise suppresses lung cancer in an orthotopic model also. Single-housed C57Bl/6 male mice in cages with running wheels were tail vein-injected with LLC1.1 lung cancer cells, and lung tumor nodules were analyzed. Exercise did not affect lung cancer. Therefore, we also tested the effect of exercise on a subcutaneous LLC1 tumor and a tail vein-injected B16F10 melanoma model. Except for one case of excessive exercise, tumor progression was not influenced. Moderately exercising mice did not increase IL-6 or recruit NK cells to the tumor. Our data suggest that the exercise dose may dictate how efficiently the immune system is stimulated and controls tumor progression.
Collapse
Affiliation(s)
- Aurelia C. Leimbacher
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Philipp Villiger
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Nina Desboeufs
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Mostafa A. Aboouf
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
- Center for Clinical Studies, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Monica Nanni
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Julia Armbruster
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Hyrije Ademi
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Pascal Flüchter
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Maja Ruetten
- PathoVet AG, Pathology Diagnostic Laboratory, 8317 Tagelswangen ZH, Switzerland
| | - Felix Gantenbein
- Zurich Integrative Rodent Physiology (ZIRP), University of Zurich, 8057 Zurich, Switzerland
| | - Thomas J. Haider
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| | - Markus Thiersch
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
- Center for Clinical Studies, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
12
|
Jang H, Ojha U, Jeong JH, Park KG, Lee SY, Lee YM. Myriocin suppresses tumor growth by modulating macrophage polarization and function through the PI3K/Akt/mTOR pathway. Arch Pharm Res 2023; 46:629-645. [PMID: 37468765 DOI: 10.1007/s12272-023-01454-1] [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: 02/21/2023] [Accepted: 07/02/2023] [Indexed: 07/21/2023]
Abstract
Macrophages within the tumor microenvironment (TME), referred to as tumor-associated macrophages (TAMs), are involved in various aspects of tumor progression including initiation, angiogenesis, metastasis, immunosuppression, and resistance to therapy. Myriocin, a natural compound isolated from Mycelia sterilia, is an immunosuppressant that inhibits tumor growth and angiogenesis. However, the mechanisms underlying the effects of myriocin on TAMs and TAM-mediated tumor growth have not yet been elucidated. In this study, we determined the effects of myriocin on TAMs and the underlying mechanism in vitro and in vivo. Myriocin significantly suppressed monocyte-macrophage differentiation and M2 polarization of macrophages but not M1 polarization. In addition, myriocin inhibited the expression of anti-inflammatory cytokines and secretion of proangiogenic factors, such as vascular endothelial growth factor, in M2 macrophages as well as M2-induced endothelial cell permeability. Myriocin also inhibited the PI3K/Akt/mTOR signaling pathway in M2 macrophages. Myriocin reduced the population of M2-like TAMs within the tumor tissue of a mouse allograft tumor model but not that of M1-like TAMs. Moreover, combined treatment with myriocin and cisplatin synergistically suppressed tumor growth and enhanced survival rate in mice by reducing the population of M2-like TAMs. Overall, these results suggest that myriocin inhibits tumor growth by remodeling the TME through suppression of differentiation and polarization of M2-like TAMs via the PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Hyeonha Jang
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Uttam Ojha
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Ji-Hak Jeong
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Keun-Gyu Park
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
| | - Shin Yup Lee
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
| | - You Mie Lee
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea.
- Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea.
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
13
|
Karreman MA, Bauer AT, Solecki G, Berghoff AS, Mayer CD, Frey K, Hebach N, Feinauer MJ, Schieber NL, Tehranian C, Mercier L, Singhal M, Venkataramani V, Schubert MC, Hinze D, Hölzel M, Helfrich I, Schadendorf D, Schneider SW, Westphal D, Augustin HG, Goetz JG, Schwab Y, Wick W, Winkler F. Active Remodeling of Capillary Endothelium via Cancer Cell-Derived MMP9 Promotes Metastatic Brain Colonization. Cancer Res 2023; 83:1299-1314. [PMID: 36652557 PMCID: PMC7614438 DOI: 10.1158/0008-5472.can-22-3964] [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: 12/19/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
Crossing the blood-brain barrier is a crucial, rate-limiting step of brain metastasis. Understanding of the mechanisms of cancer cell extravasation from brain microcapillaries is limited as the underlying cellular and molecular processes cannot be adequately investigated using in vitro models and endpoint in vivo experiments. Using ultrastructural and functional imaging, we demonstrate that dynamic changes of activated brain microcapillaries promote the mandatory first steps of brain colonization. Successful extravasation of arrested cancer cells occurred when adjacent capillary endothelial cells (EC) entered into a distinct remodeling process. After extravasation, capillary loops were formed, which was characteristic of aggressive metastatic growth. Upon cancer cell arrest in brain microcapillaries, matrix-metalloprotease 9 (MMP9) was expressed. Inhibition of MMP2/9 and genetic perturbation of MMP9 in cancer cells, but not the host, reduced EC projections, extravasation, and brain metastasis outgrowth. These findings establish an active role of ECs in the process of cancer cell extravasation, facilitated by cross-talk between the two cell types. This extends our understanding of how host cells can contribute to brain metastasis formation and how to prevent it. SIGNIFICANCE Tracking single extravasating cancer cells using multimodal correlative microscopy uncovers a brain seeding mechanism involving endothelial remodeling driven by cancer cell-derived MMP9, which might enable the development of approaches to prevent brain metastasis. See related commentary by McCarty, p. 1167.
Collapse
Affiliation(s)
- Matthia A. Karreman
- Neurology Clinic and National Center for Tumor Diseases, University
Hospital Heidelberg, INF 400, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium
(DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Cell Biology and Biophysics Unit, European Molecular Biology
Laboratory, Heidelberg, Germany
| | - Alexander T. Bauer
- Department of Dermatology and Venereology, University Medical Center
Hamburg-Eppendorf, Hamburg, Germany
| | - Gergely Solecki
- Neurology Clinic and National Center for Tumor Diseases, University
Hospital Heidelberg, INF 400, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium
(DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Business Unit Service and Customer Care, Carl Zeiss Microscopy GmbH,
Jena, Germany
| | - Anna S. Berghoff
- Neurology Clinic and National Center for Tumor Diseases, University
Hospital Heidelberg, INF 400, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium
(DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine I, Division of Oncology, Medical University
of Vienna, Comprehensive Cancer Center Vienna, Vienna, Austria
| | - Chanté D. Mayer
- Neurology Clinic and National Center for Tumor Diseases, University
Hospital Heidelberg, INF 400, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium
(DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katharina Frey
- Neurology Clinic and National Center for Tumor Diseases, University
Hospital Heidelberg, INF 400, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium
(DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nils Hebach
- Neurology Clinic and National Center for Tumor Diseases, University
Hospital Heidelberg, INF 400, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium
(DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manuel J. Feinauer
- Neurology Clinic and National Center for Tumor Diseases, University
Hospital Heidelberg, INF 400, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium
(DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nicole L. Schieber
- Cell Biology and Biophysics Unit, European Molecular Biology
Laboratory, Heidelberg, Germany
- Centre for Microscopy and Microanalyses, The University of
Queensland, Brisbane, Australia
| | - Cedric Tehranian
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium
(DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Luc Mercier
- National Institute of Health and Medical Research (INSERM)
UMR_S1109, Tumor Biomechanics, Université de Strasbourg,
Fédération de Médecine Translationnelle de Strasbourg (FMTS),
Strasbourg, France
| | - Mahak Singhal
- European Center for Angioscience (ECAS), Medical Faculty Mannheim,
Heidelberg University, Germany
- Division of Vascular Oncology and Metastasis, German Cancer
Research Center Heidelberg (DKFZ-ZMBH Alliance), Germany
| | - Varun Venkataramani
- Neurology Clinic and National Center for Tumor Diseases, University
Hospital Heidelberg, INF 400, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium
(DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Functional Neuroanatomy, Institute for Anatomy and
Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Marc C. Schubert
- Department of Functional Neuroanatomy, Institute for Anatomy and
Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Daniel Hinze
- LAMPseq Diagnostics GmbH, Bonn, Germany
- Institute of Experimental Oncology, University Hospital Bonn,
University of Bonn, Bonn, Germany
| | - Michael Hölzel
- Institute of Experimental Oncology, University Hospital Bonn,
University of Bonn, Bonn, Germany
| | - Iris Helfrich
- Skin Cancer Unit of the Dermatology Department, Medical Faculty,
West German Cancer Center, University Duisburg-Essen, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Dermatology and Allergology, Medical Faculty of the
Ludwig Maximilian University of Munich, Munich, Germany
| | - Dirk Schadendorf
- Skin Cancer Unit of the Dermatology Department, Medical Faculty,
West German Cancer Center, University Duisburg-Essen, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Stefan W. Schneider
- Department of Dermatology and Venereology, University Medical Center
Hamburg-Eppendorf, Hamburg, Germany
| | - Dana Westphal
- Department of Dermatology, Medical Faculty and University Hospital
Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Hellmut G. Augustin
- European Center for Angioscience (ECAS), Medical Faculty Mannheim,
Heidelberg University, Germany
- Division of Vascular Oncology and Metastasis, German Cancer
Research Center Heidelberg (DKFZ-ZMBH Alliance), Germany
| | - Jacky G. Goetz
- National Institute of Health and Medical Research (INSERM)
UMR_S1109, Tumor Biomechanics, Université de Strasbourg,
Fédération de Médecine Translationnelle de Strasbourg (FMTS),
Strasbourg, France
| | - Yannick Schwab
- Cell Biology and Biophysics Unit, European Molecular Biology
Laboratory, Heidelberg, Germany
- Electron Microscopy Core Facility, European Molecular Biology
Laboratory, Heidelberg, Germany
| | - Wolfgang Wick
- Neurology Clinic and National Center for Tumor Diseases, University
Hospital Heidelberg, INF 400, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium
(DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Winkler
- Neurology Clinic and National Center for Tumor Diseases, University
Hospital Heidelberg, INF 400, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium
(DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
14
|
Jing H, Wu X, Xiang M, Wang C, Novakovic VA, Shi J. Microparticle Phosphatidylserine Mediates Coagulation: Involvement in Tumor Progression and Metastasis. Cancers (Basel) 2023; 15:cancers15071957. [PMID: 37046617 PMCID: PMC10093313 DOI: 10.3390/cancers15071957] [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: 01/31/2023] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023] Open
Abstract
Tumor progression and cancer metastasis has been linked to the release of microparticles (MPs), which are shed upon cell activation or apoptosis and display parental cell antigens, phospholipids such as phosphatidylserine (PS), and nucleic acids on their external surfaces. In this review, we highlight the biogenesis of MPs as well as the pathophysiological processes of PS externalization and its involvement in coagulation activation. We review the available evidence, suggesting that coagulation factors (mainly tissue factor, thrombin, and fibrin) assist in multiple steps of tumor dissemination, including epithelial-mesenchymal transition, extracellular matrix remodeling, immune escape, and tumor angiogenesis to support the formation of the pre-metastatic niche. Platelets are not just bystander cells in circulation but are functional players in primary tumor growth and metastasis. Tumor-induced platelet aggregation protects circulating tumor cells (CTCs) from the blood flow shear forces and immune cell attack while also promoting the binding of CTCs to endothelial cells and extravasation, which activates tumor invasion and sustains metastasis. Finally, in terms of therapy, lactadherin can inhibit coagulation by competing effectively with coagulation factors for PS binding sites and may similarly delay tumor progression. Furthermore, we also investigate the therapeutic potential of coagulation factor inhibitors within the context of cancer treatment. The development of multiple therapies targeting platelet activation and platelet-tumor cell interactions may not only reduce the lethal consequences of thrombosis but also impede tumor growth and spread.
Collapse
Affiliation(s)
- Haijiao Jing
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
| | - Xiaoming Wu
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
| | - Mengqi Xiang
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
| | - Chengyue Wang
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
| | - Valerie A Novakovic
- Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA 02132, USA
| | - Jialan Shi
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
- Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA 02132, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02132, USA
| |
Collapse
|
15
|
Long Non-Coding RNAs as Novel Targets for Phytochemicals to Cease Cancer Metastasis. Molecules 2023; 28:molecules28030987. [PMID: 36770654 PMCID: PMC9921150 DOI: 10.3390/molecules28030987] [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: 11/23/2022] [Revised: 12/31/2022] [Accepted: 01/11/2023] [Indexed: 01/21/2023] Open
Abstract
Metastasis is a multi-step phenomenon during cancer development leading to the propagation of cancer cells to distant organ(s). According to estimations, metastasis results in over 90% of cancer-associated death around the globe. Long non-coding RNAs (LncRNAs) are a group of regulatory RNA molecules more than 200 base pairs in length. The main regulatory activity of these molecules is the modulation of gene expression. They have been reported to affect different stages of cancer development including proliferation, apoptosis, migration, invasion, and metastasis. An increasing number of medical data reports indicate the probable function of LncRNAs in the metastatic spread of different cancers. Phytochemical compounds, as the bioactive agents of plants, show several health benefits with a variety of biological activities. Several phytochemicals have been demonstrated to target LncRNAs to defeat cancer. This review article briefly describes the metastasis steps, summarizes data on some well-established LncRNAs with a role in metastasis, and identifies the phytochemicals with an ability to suppress cancer metastasis by targeting LncRNAs.
Collapse
|
16
|
Zhang X, Bai W, Hu L, Ha H, Du Y, Xiong W, Wang H, Shang P. The pleiotropic mode and molecular mechanism of macrophages in promoting tumor progression and metastasis. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:91-104. [PMID: 36071369 DOI: 10.1007/s12094-022-02932-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 08/22/2022] [Indexed: 01/07/2023]
Abstract
Macrophages are the most abundant immune cells in primary and metastatic tumor tissues. Studies have shown that macrophages mainly exhibit a tumor-promoting phenotype and play a key role in tumor progression and metastasis. Therefore, many macrophage-targeted drugs have entered clinical trials. However, compared to preclinical studies, some clinical trial results showed that macrophage-targeted therapy did not achieve the desired effect. This may be because most of what we know about macrophages comes from in vitro experiments and animal models, while macrophages in the more complex human microenvironment are still poorly understood. With the development of technologies such as single-cell RNA sequencing, we have gained a new understanding of the origin, classification and functional mechanism of tumor-associated macrophages. Therefore, this study reviewed the recent progress of macrophages in promoting tumor progression and metastasis, aiming to provide some help for the formulation of optimal strategies for macrophage-targeted therapy.
Collapse
Affiliation(s)
- Xingxing Zhang
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Wenxiu Bai
- Ultrasonic Special Examination Department, Tai An TSCM Hospital, Taian, 271000, Shandong, China
| | - Lisha Hu
- Ultrasonic Special Examination Department, Tai An TSCM Hospital, Taian, 271000, Shandong, China
| | - Hualan Ha
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Yuelin Du
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Wei Xiong
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Hongbo Wang
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Panfeng Shang
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
17
|
Nakayama A, Roquid KA, Iring A, Strilic B, Günther S, Chen M, Weinstein LS, Offermanns S. Suppression of CCL2 angiocrine function by adrenomedullin promotes tumor growth. J Exp Med 2022; 220:213682. [PMID: 36374225 PMCID: PMC9665902 DOI: 10.1084/jem.20211628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/19/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Within the tumor microenvironment, tumor cells and endothelial cells regulate each other. While tumor cells induce angiogenic responses in endothelial cells, endothelial cells release angiocrine factors, which act on tumor cells and other stromal cells. We report that tumor cell-derived adrenomedullin has a pro-angiogenic as well as a direct tumor-promoting effect, and that endothelium-derived CC chemokine ligand 2 (CCL2) suppresses adrenomedullin-induced tumor cell proliferation. Loss of the endothelial adrenomedullin receptor CALCRL or of the G-protein Gs reduced endothelial proliferation. Surprisingly, tumor cell proliferation was also reduced after endothelial deletion of CALCRL or Gs. We identified CCL2 as a critical angiocrine factor whose formation is inhibited by adrenomedullin. Furthermore, CCL2 inhibited adrenomedullin formation in tumor cells through its receptor CCR2. Consistently, loss of endothelial CCL2 or tumor cell CCR2 normalized the reduced tumor growth seen in mice lacking endothelial CALCRL or Gs. Our findings show tumor-promoting roles of adrenomedullin and identify CCL2 as an angiocrine factor controlling adrenomedullin formation by tumor cells.
Collapse
Affiliation(s)
- Akiko Nakayama
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany,Correspondence to Akiko Nakayama:
| | - Kenneth Anthony Roquid
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - András Iring
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Boris Strilic
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Günther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Min Chen
- Metabolic Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MA
| | - Lee S. Weinstein
- Metabolic Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MA
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany,Center for Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany,Cardiopulmonary Institute, Bad Nauheim, Germany,German Center for Cardiovascular Research, Bad Nauheim, Germany,Stefan Offermanns:
| |
Collapse
|
18
|
Singh AK, Malviya R. Coagulation and inflammation in cancer: Limitations and prospects for treatment. Biochim Biophys Acta Rev Cancer 2022; 1877:188727. [PMID: 35378243 DOI: 10.1016/j.bbcan.2022.188727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 02/08/2023]
Abstract
The development of so-called immune checkpoint inhibitors (ICIs), which target specific molecular processes of tumour growth, has had a transformative effect on cancer treatment. Widespread use of antibody-based medicines to inhibit tumour cell immune evasion by modulating T cell responses is becoming more common. Despite this, response rates are still low, and secondary resistance is an issue that arises often. In addition, a wide range of serious adverse effects is triggered by enhancing the immunological response. As a result of an increased mortality rate, a higher prevalence of thrombotic complications is connected with an increased incidence of immunological reactions, complement activation, and skin toxicity. This suggests that the tumour microenvironment's interaction between coagulation and inflammation is important at every stage of the tumour's life cycle. The coagulation system's function in tumour formation is the topic of this review. By better understanding the molecular mechanisms in which tumour cells circulate, plasmatic coagulation and immune system cells are engaged, new therapy options for cancer sufferers may be discovered.
Collapse
Affiliation(s)
- Arun Kumar Singh
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India.
| |
Collapse
|
19
|
Smeda M, Stojak M, Przyborowski K, Sternak M, Suraj-Prazmowska J, Kus K, Derszniak K, Jasztal A, Kij A, Kurpinska A, Kieronska-Rudek A, Wojnar-Lason K, Buczek E, Mohaissen T, Chlopicki S. Direct Thrombin Inhibitor Dabigatran Compromises Pulmonary Endothelial Integrity in a Murine Model of Breast Cancer Metastasis to the Lungs; the Role of Platelets and Inflammation-Associated Haemostasis. Front Pharmacol 2022; 13:834472. [PMID: 35295330 PMCID: PMC8918823 DOI: 10.3389/fphar.2022.834472] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Activation of the coagulation cascade favours metastatic spread, but antithrombotic therapy might also have detrimental effects on cancer progression. In this study, we characterized the effects of dabigatran, a direct reversible thrombin inhibitor, on the pulmonary endothelial barrier and metastatic spread in a murine model of breast cancer metastasis. Dabigatran etexilate (100 mg kg−1) was administered to mice twice daily by oral gavage. Pulmonary metastasis, pulmonary endothelium permeability in vivo, and platelet reactivity were evaluated after intravenous injection of 4T1 breast cancer cells into BALB/c mice. The effect of dabigatran on platelet-dependent protection of pulmonary endothelial barrier in the presence of an inflammatory stimulus was also verified in vitro using human lung microvascular endothelial cell (HLMVEC) cultures. Dabigatran-treated mice harbored more metastases in their lungs and displayed increased pulmonary endothelium permeability after cancer cell injection. It was not associated with altered lung fibrin deposition, changes in INFγ, or complement activation. In the in vitro model of the pulmonary endothelial barrier, dabigatran inhibited platelet-mediated protection of pulmonary endothelium. In a murine model of breast cancer metastasis, dabigatran treatment promoted pulmonary metastasis by the inhibition of platelet-dependent protection of pulmonary endothelial barrier integrity.
Collapse
Affiliation(s)
- Marta Smeda
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- *Correspondence: Marta Smeda, ; Stefan Chlopicki,
| | - Marta Stojak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Kamil Przyborowski
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Magdalena Sternak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Joanna Suraj-Prazmowska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Kamil Kus
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Katarzyna Derszniak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- Faculty of Chemistry, Jagiellonian University, Krakow, Poland
| | - Agnieszka Jasztal
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Agnieszka Kij
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Anna Kurpinska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Anna Kieronska-Rudek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- Department of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
| | - Kamila Wojnar-Lason
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- Department of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
| | - Elzbieta Buczek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Tasnim Mohaissen
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- Department of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
- *Correspondence: Marta Smeda, ; Stefan Chlopicki,
| |
Collapse
|
20
|
Bauer AT, Gorzelanny C, Gebhardt C, Pantel K, Schneider SW. Interplay between coagulation and inflammation in cancer: Limitations and therapeutic opportunities. Cancer Treat Rev 2022; 102:102322. [DOI: 10.1016/j.ctrv.2021.102322] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 12/12/2022]
|
21
|
Stefanescu C, Van Gogh M, Roblek M, Heikenwalder M, Borsig L. TGFβ Signaling in Myeloid Cells Promotes Lung and Liver Metastasis Through Different Mechanisms. Front Oncol 2021; 11:765151. [PMID: 34868988 PMCID: PMC8637420 DOI: 10.3389/fonc.2021.765151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/01/2021] [Indexed: 11/13/2022] Open
Abstract
TGFβ overexpression is commonly detected in cancer patients and correlates with poor prognosis and metastasis. Cancer progression is often associated with an enhanced recruitment of myeloid-derived cells to the tumor microenvironment. Here we show that functional TGFβ-signaling in myeloid cells is required for metastasis to the lungs and the liver. Myeloid-specific deletion of Tgfbr2 resulted in reduced spontaneous lung metastasis, which was associated with a reduction of proinflammatory cytokines in the metastatic microenvironment. Notably, CD8+ T cell depletion in myeloid-specific Tgfbr2-deficient mice rescued lung metastasis. Myeloid-specific Tgfbr2-deficiency resulted in reduced liver metastasis with an almost complete absence of myeloid cells within metastatic foci. On contrary, an accumulation of Tgfβ-responsive myeloid cells was associated with an increased recruitment of monocytes and granulocytes and higher proinflammatory cytokine levels in control mice. Monocytic cells isolated from metastatic livers of Tgfbr2-deficient mice showed increased polarization towards the M1 phenotype, Tnfα and Il-1β expression, reduced levels of M2 markers and reduced production of chemokines responsible for myeloid-cell recruitment. No significant differences in Tgfβ levels were observed at metastatic sites of any model. These data demonstrate that Tgfβ signaling in monocytic myeloid cells suppresses CD8+ T cell activity during lung metastasis, while these cells actively contribute to tumor growth during liver metastasis. Thus, myeloid cells modulate metastasis through different mechanisms in a tissue-specific manner.
Collapse
Affiliation(s)
| | - Merel Van Gogh
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Marko Roblek
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lubor Borsig
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,Comprehensive Cancer Center Zurich, Zurich, Switzerland
| |
Collapse
|
22
|
D’Arcy C, Kiel C. Cell Adhesion Molecules in Normal Skin and Melanoma. Biomolecules 2021; 11:biom11081213. [PMID: 34439879 PMCID: PMC8391223 DOI: 10.3390/biom11081213] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/11/2022] Open
Abstract
Cell adhesion molecules (CAMs) of the cadherin, integrin, immunoglobulin, and selectin protein families are indispensable for the formation and maintenance of multicellular tissues, especially epithelia. In the epidermis, they are involved in cell–cell contacts and in cellular interactions with the extracellular matrix (ECM), thereby contributing to the structural integrity and barrier formation of the skin. Bulk and single cell RNA sequencing data show that >170 CAMs are expressed in the healthy human skin, with high expression levels in melanocytes, keratinocytes, endothelial, and smooth muscle cells. Alterations in expression levels of CAMs are involved in melanoma propagation, interaction with the microenvironment, and metastasis. Recent mechanistic analyses together with protein and gene expression data provide a better picture of the role of CAMs in the context of skin physiology and melanoma. Here, we review progress in the field and discuss molecular mechanisms in light of gene expression profiles, including recent single cell RNA expression information. We highlight key adhesion molecules in melanoma, which can guide the identification of pathways and strategies for novel anti-melanoma therapies.
Collapse
|
23
|
Malhab LJB, Saber-Ayad MM, Al-Hakm R, Nair VA, Paliogiannis P, Pintus G, Abdel-Rahman WM. Chronic Inflammation and Cancer: The Role of Endothelial Dysfunction and Vascular Inflammation. Curr Pharm Des 2021; 27:2156-2169. [PMID: 33655853 DOI: 10.2174/1381612827666210303143442] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 12/17/2020] [Indexed: 01/17/2023]
Abstract
Long-lasting subclinical inflammation is associated with a wide range of human diseases, particularly at a middle and older age. Recent reports showed that there is a direct causal link between inflammation and cancer development, as several cancers were found to be associated with chronic inflammatory conditions. In patients with cancer, healthy endothelial cells regulate vascular homeostasis, and it is believed that they can limit tumor growth, invasiveness, and metastasis. Conversely, dysfunctional endothelial cells that have been exposed to the inflammatory tumor microenvironment can support cancer progression and metastasis. Dysfunctional endothelial cells can exert these effects via diverse mechanisms, including dysregulated adhesion, permeability, and activation of NF-κB and STAT3 signaling. In this review, we highlight the role of vascular inflammation in predisposition to cancer within the context of two common disease risk factors: obesity and smoking. In addition, we discuss the molecular triggers, pathophysiological mechanisms, and the biological consequences of vascular inflammation during cancer development and metastasis. Finally, we summarize the current therapies and pharmacological agents that target vascular inflammation and endothelial dysfunction.
Collapse
Affiliation(s)
- Lara J Bou Malhab
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Maha M Saber-Ayad
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Ranyah Al-Hakm
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Vidhya A Nair
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Panagiotis Paliogiannis
- Department of Medical, Surgical, and Experimental Surgery, University of Sassari, Viale San Pietro 43,07100 Sassari, Italy
| | - Gianfranco Pintus
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Wael M Abdel-Rahman
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
24
|
Delprat V, Michiels C. A bi-directional dialog between vascular cells and monocytes/macrophages regulates tumor progression. Cancer Metastasis Rev 2021; 40:477-500. [PMID: 33783686 PMCID: PMC8213675 DOI: 10.1007/s10555-021-09958-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/23/2021] [Indexed: 02/06/2023]
Abstract
Cancer progression largely depends on tumor blood vessels as well on immune cell infiltration. In various tumors, vascular cells, namely endothelial cells (ECs) and pericytes, strongly regulate leukocyte infiltration into tumors and immune cell activation, hence the immune response to cancers. Recently, a lot of compelling studies unraveled the molecular mechanisms by which tumor vascular cells regulate monocyte and tumor-associated macrophage (TAM) recruitment and phenotype, and consequently tumor progression. Reciprocally, TAMs and monocytes strongly modulate tumor blood vessel and tumor lymphatic vessel formation by exerting pro-angiogenic and lymphangiogenic effects, respectively. Finally, the interaction between monocytes/TAMs and vascular cells is also impacting several steps of the spread of cancer cells throughout the body, a process called metastasis. In this review, the impact of the bi-directional dialog between blood vascular cells and monocytes/TAMs in the regulation of tumor progression is discussed. All together, these data led to the design of combinations of anti-angiogenic and immunotherapy targeting TAMs/monocyte whose effects are briefly discussed in the last part of this review.
Collapse
Affiliation(s)
- Victor Delprat
- Biochemistry and Cellular Biology Research Unit (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 61 Rue de Bruxelles, B-5000, Namur, Belgium
| | - Carine Michiels
- Biochemistry and Cellular Biology Research Unit (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 61 Rue de Bruxelles, B-5000, Namur, Belgium.
| |
Collapse
|
25
|
Control of Tumor Progression by Angiocrine Factors. Cancers (Basel) 2021; 13:cancers13112610. [PMID: 34073394 PMCID: PMC8198241 DOI: 10.3390/cancers13112610] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 12/24/2022] Open
Abstract
Tumor progression, therapy resistance and metastasis are profoundly controlled by the tumor microenvironment. The contribution of endothelial cells to tumor progression was initially only attributed to the formation of new blood vessels (angiogenesis). Research in the last decade has revealed however that endothelial cells control their microenvironment through the expression of membrane-bound and secreted factors. Such angiocrine functions are frequently hijacked by cancer cells, which deregulate the signaling pathways controlling the expression of angiocrine factors. Here, we review the crosstalk between cancer cells and endothelial cells and how this contributes to the cancer stem cell phenotype, epithelial to mesenchymal transition, immunosuppression, remodeling of the extracellular matrix and intravasation of cancer cells into the bloodstream. We also address the long-distance crosstalk of a primary tumor with endothelial cells at the pre-metastatic niche and how this contributes to metastasis.
Collapse
|
26
|
Song Y, Sun H, Wu K, Lyu J, Zhang J, Gu F, Ma Y, Shen B, Wang C, Chen X, Xu J, Li W, Liu F, Fu L. sLe x expression in invasive micropapillary breast carcinoma is associated with poor prognosis and can be combined with MUC1/EMA as a supplementary diagnostic indicator. Cancer Biol Med 2021; 18:j.issn.2095-3941.2020.0422. [PMID: 33893728 PMCID: PMC8185870 DOI: 10.20892/j.issn.2095-3941.2020.0422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 12/08/2020] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE Mucin 1 (MUC1/EMA) and sialyl Lewis X (sLex) indicate polarity reversal in invasive micropapillary carcinoma (IMPC). The purpose of this study was to evaluate the expression of MUC1/EMA and sLex and to assess their diagnostic and prognostic value in patients with IMPC. METHODS The expression of sLex and MUC1/EMA in 100 patients with IMPC and a control group of 89 patients with invasive ductal carcinoma not otherwise specified (IDC-NOS) were analyzed with IHC. Fresh tumor tissues were collected from patients with IMPC or IDC-NOS for primary culture and immunofluorescence analysis. RESULTS The rate of nodal metastasis was higher in patients with IMPC than those with IDC-NOS, and IMPC cells tended to express more sLex and MUC1/EMA in the cytomembranes (the stroma-facing surfaces of the micropapillary clusters) than IDC-NOS cells. In IMPC, high cytomembrane expression of sLex, but not MUC1/EMA, indicated poor prognosis. In addition, among the 100 patients with IMPC, 10 patients had sLex+/EMA- expression patterns, and 8 patients had sLex-/EMA+ expression patterns. The primary IMPC cells were suspended, non-adherent tumor cell clusters, whereas the primary IDC cells were adherent tumor cells. Immunofluorescence analysis showed that MUC1/EMA and sLex were co-expressed on the cytomembranes in IMPC cell clusters and in the cytoplasm in IDC-NOS cells. CONCLUSIONS sLex can be used as a prognostic indicator and can be combined with MUC1/EMA as a complementary diagnostic indicator to avoid missed IMPC diagnosis.
Collapse
Affiliation(s)
- Yawen Song
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Breast Cancer Innovation Team of the Ministry of Education; State Key Laboratory of Breast Cancer Research, Tianjin 300060, China
| | - Hui Sun
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Breast Cancer Innovation Team of the Ministry of Education; State Key Laboratory of Breast Cancer Research, Tianjin 300060, China
| | - Kailiang Wu
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Breast Cancer Innovation Team of the Ministry of Education; State Key Laboratory of Breast Cancer Research, Tianjin 300060, China
| | - Jianke Lyu
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Breast Cancer Innovation Team of the Ministry of Education; State Key Laboratory of Breast Cancer Research, Tianjin 300060, China
| | - Jingyue Zhang
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Breast Cancer Innovation Team of the Ministry of Education; State Key Laboratory of Breast Cancer Research, Tianjin 300060, China
| | - Feng Gu
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Breast Cancer Innovation Team of the Ministry of Education; State Key Laboratory of Breast Cancer Research, Tianjin 300060, China
| | - Yongjie Ma
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Breast Cancer Innovation Team of the Ministry of Education; State Key Laboratory of Breast Cancer Research, Tianjin 300060, China
| | - Beibei Shen
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Breast Cancer Innovation Team of the Ministry of Education; State Key Laboratory of Breast Cancer Research, Tianjin 300060, China
| | - Chijuan Wang
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Breast Cancer Innovation Team of the Ministry of Education; State Key Laboratory of Breast Cancer Research, Tianjin 300060, China
| | - Xiaojiao Chen
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Breast Cancer Innovation Team of the Ministry of Education; State Key Laboratory of Breast Cancer Research, Tianjin 300060, China
| | - Jing Xu
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Breast Cancer Innovation Team of the Ministry of Education; State Key Laboratory of Breast Cancer Research, Tianjin 300060, China
| | - Weidong Li
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Breast Cancer Innovation Team of the Ministry of Education; State Key Laboratory of Breast Cancer Research, Tianjin 300060, China
| | - Fangfang Liu
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Breast Cancer Innovation Team of the Ministry of Education; State Key Laboratory of Breast Cancer Research, Tianjin 300060, China
| | - Li Fu
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Breast Cancer Innovation Team of the Ministry of Education; State Key Laboratory of Breast Cancer Research, Tianjin 300060, China
| |
Collapse
|
27
|
Aberrant protein glycosylation in cancer: implications in targeted therapy. Biochem Soc Trans 2021; 49:843-854. [PMID: 33704376 DOI: 10.1042/bst20200763] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/12/2021] [Accepted: 02/15/2021] [Indexed: 12/28/2022]
Abstract
Aberrant cell surface glycosylation signatures are currently known to actively drive the neoplastic transformation of healthy cells. By disrupting the homeostatic functions of their protein carriers, cancer-associated glycans mechanistically underpin several molecular hallmarks of human malignancy. Furthermore, such aberrant glycan structures play key roles in the acquisition of molecular resistance to targeted therapeutic agents, which compromises their clinical efficacy, by modulating tumour cell aggressiveness and supporting the establishment of an immunosuppressive microenvironment. Recent advances in the study of the tumour cell glycoproteome have unravelled previously elusive molecular mechanisms of therapeutic resistance, guided the rational design of novel personalized therapeutic strategies, and may further improve the clinical performance of currently approved anti-cancer targeted agents. In this review, we highlight the impact of glycosylation in cancer targeted therapy, with particular focus on receptor tyrosine kinase-targeted therapy, immune checkpoints blockade therapy, and current developments on therapeutic strategies directed to glycan-binding proteins and other innovative glycan therapeutic strategies.
Collapse
|
28
|
Dúcka M, Kučeríková M, Trčka F, Červinka J, Biglieri E, Šmarda J, Borsig L, Beneš P, Knopfová L. c-Myb interferes with inflammatory IL1α-NF-κB pathway in breast cancer cells. Neoplasia 2021; 23:326-336. [PMID: 33621853 PMCID: PMC7905261 DOI: 10.1016/j.neo.2021.01.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/12/2021] [Accepted: 01/20/2021] [Indexed: 12/14/2022] Open
Abstract
The transcription factor c-Myb can be involved in the activation of many genes with protumorigenic function; however, its role in breast cancer (BC) development is still under discussion. c-Myb is considered as a tumor-promoting factor in the early phases of BC, on the other hand, its expression in BC patients relates to a good prognosis. Previously, we have shown that c-Myb controls the capacity of BC cells to form spontaneous lung metastasis. Reduced seeding of BC cells to the lungs is linked to high expression of c-Myb and a decline in expression of a specific set of inflammatory genes. Here, we unraveled a c-Myb-IL1α-NF-κB signaling axis that takes place in tumor cells. We report that an overexpression of c-Myb interfered with the activity of NF-κB in several BC cell lines. We identified IL1α to be essential for this interference since it was abrogated in the IL1α-deficient cells. Overexpression of IL1α, as well as addition of recombinant IL1α protein, activated NF-κB signaling and restored expression of the inflammatory signature genes suppressed by c-Myb. The endogenous levels of c-Myb negatively correlated with IL1α on both transcriptional and protein levels across BC cell lines. We concluded that inhibition of IL1α expression by c-Myb reduces NF-κB activity and disconnects the inflammatory circuit, a potentially targetable mechanism to mimic the antimetastatic effect of c-Myb with therapeutic perspective.
Collapse
Affiliation(s)
- Monika Dúcka
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, Center for Biological and Cellular Engineering, St. Anne's University Hospital, Brno, Czech Republic
| | - Martina Kučeríková
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Filip Trčka
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, Center for Biological and Cellular Engineering, St. Anne's University Hospital, Brno, Czech Republic
| | - Jakub Červinka
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, Center for Biological and Cellular Engineering, St. Anne's University Hospital, Brno, Czech Republic
| | - Elisabetta Biglieri
- Institute of Physiology, University of Zurich and Comprehensive Cancer Center, Zurich, Switzerland
| | - Jan Šmarda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Lubor Borsig
- Institute of Physiology, University of Zurich and Comprehensive Cancer Center, Zurich, Switzerland
| | - Petr Beneš
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, Center for Biological and Cellular Engineering, St. Anne's University Hospital, Brno, Czech Republic
| | - Lucia Knopfová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, Center for Biological and Cellular Engineering, St. Anne's University Hospital, Brno, Czech Republic.
| |
Collapse
|
29
|
Stock C. Circulating Tumor Cells: Does Ion Transport Contribute to Intravascular Survival, Adhesion, Extravasation, and Metastatic Organotropism? Rev Physiol Biochem Pharmacol 2021; 182:139-175. [DOI: 10.1007/112_2021_68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
30
|
Hussain S, Peng B, Cherian M, Song JW, Ahirwar DK, Ganju RK. The Roles of Stroma-Derived Chemokine in Different Stages of Cancer Metastases. Front Immunol 2020; 11:598532. [PMID: 33414786 PMCID: PMC7783453 DOI: 10.3389/fimmu.2020.598532] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
The intricate interplay between malignant cells and host cellular and non-cellular components play crucial role in different stages of tumor development, progression, and metastases. Tumor and stromal cells communicate to each other through receptors such as integrins and secretion of signaling molecules like growth factors, cytokines, chemokines and inflammatory mediators. Chemokines mediated signaling pathways have emerged as major mechanisms underlying multifaceted roles played by host cells during tumor progression. In response to tumor stimuli, host cells-derived chemokines further activates signaling cascades that support the ability of tumor cells to invade surrounding basement membrane and extra-cellular matrix. The host-derived chemokines act on endothelial cells to increase their permeability and facilitate tumor cells intravasation and extravasation. The tumor cells-host neutrophils interaction within the vasculature initiates chemokines driven recruitment of inflammatory cells that protects circulatory tumor cells from immune attack. Chemokines secreted by tumor cells and stromal immune and non-immune cells within the tumor microenvironment enter the circulation and are responsible for formation of a "pre-metastatic niche" like a "soil" in distant organs whereby circulating tumor cells "seed' and colonize, leading to formation of metastatic foci. Given the importance of host derived chemokines in cancer progression and metastases several drugs like Mogamulizumab, Plerixafor, Repertaxin among others are part of ongoing clinical trial which target chemokines and their receptors against cancer pathogenesis. In this review, we focus on recent advances in understanding the complexity of chemokines network in tumor microenvironment, with an emphasis on chemokines secreted from host cells. We especially summarize the role of host-derived chemokines in different stages of metastases, including invasion, dissemination, migration into the vasculature, and seeding into the pre-metastatic niche. We finally provide a brief description of prospective drugs that target chemokines in different clinical trials against cancer.
Collapse
Affiliation(s)
- Shahid Hussain
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Bo Peng
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Mathew Cherian
- Division of Medical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Jonathan W Song
- Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Department of Mechanical and Aerospace Engineering, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Dinesh K Ahirwar
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Ramesh K Ganju
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
31
|
Duru G, van Egmond M, Heemskerk N. A Window of Opportunity: Targeting Cancer Endothelium to Enhance Immunotherapy. Front Immunol 2020; 11:584723. [PMID: 33262763 PMCID: PMC7686513 DOI: 10.3389/fimmu.2020.584723] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/19/2020] [Indexed: 12/19/2022] Open
Abstract
Vascular abnormalities in tumors have a major impact on the immune microenvironment in tumors. The consequences of abnormal vasculature include increased hypoxia, acidosis, high intra-tumoral fluid pressure, and angiogenesis. This introduces an immunosuppressive microenvironment that alters immune cell maturation, activation, and trafficking, which supports tumor immune evasion and dissemination of tumor cells. Increasing data suggests that cancer endothelium is a major barrier for traveling leukocytes, ranging from a partial blockade resulting in a selective endothelial barrier, to a complete immune infiltration blockade associated with immune exclusion and immune desert cancer phenotypes. Failed immune cell trafficking as well as immunosuppression within the tumor microenvironment limits the efficacy of immunotherapeutic approaches. As such, targeting proteins with key roles in angiogenesis may potentially reduce immunosuppression and might restore infiltration of anti-tumor immune cells, creating a therapeutic window for successful immunotherapy. In this review, we provide a comprehensive overview of established as well as more controversial endothelial pathways that govern selective immune cell trafficking across cancer endothelium. Additionally, we discuss recent insights and strategies that target tumor vasculature in order to increase infiltration of cytotoxic immune cells during the therapeutic window of vascular normalization hereby improving the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Gizem Duru
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection & Immunity, Amsterdam, Netherlands
| | - Marjolein van Egmond
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection & Immunity, Amsterdam, Netherlands
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Surgery, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Niels Heemskerk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection & Immunity, Amsterdam, Netherlands
| |
Collapse
|
32
|
Birmingham KG, O'Melia MJ, Bordy S, Reyes Aguilar D, El-Reyas B, Lesinski G, Thomas SN. Lymph Node Subcapsular Sinus Microenvironment-On-A-Chip Modeling Shear Flow Relevant to Lymphatic Metastasis and Immune Cell Homing. iScience 2020; 23:101751. [PMID: 33241198 PMCID: PMC7672279 DOI: 10.1016/j.isci.2020.101751] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 10/11/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022] Open
Abstract
A lymph node sinus-on-a-chip adhesion microfluidic platform that recapitulates the hydrodynamic microenvironment of the lymph node subcapsular sinus was engineered. This device was used to interrogate the effects of lymph node remodeling on cellular adhesion in fluid flow relevant to lymphatic metastasis. Wall shear stress levels analytically estimated and modeled after quiescent and diseased/inflamed lymph nodes were experimentally recapitulated using a flow-based microfluidic perfusion system to assess the effects of physiological flow fields on human metastatic cancer cell adhesion. Results suggest that both altered fluid flow profiles and presentation of adhesive ligands, which are predicted to manifest within the lymph node subcapsular sinus as a result of inflammation-induced remodeling, and the presence of lymph-borne monocytic cells may synergistically contribute to the dynamic extent of cell adhesion in flow relevant to lymph node invasion by cancer and monocytic immune cells during lymphatic metastasis.
Collapse
Affiliation(s)
- Katherine G. Birmingham
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, IBB 2310 315 Ferst Drive NW, Atlanta, GA 30332, USA
| | - Meghan J. O'Melia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Samantha Bordy
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - David Reyes Aguilar
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, IBB 2310 315 Ferst Drive NW, Atlanta, GA 30332, USA
| | - Bassel El-Reyas
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Gregory Lesinski
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Susan N. Thomas
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, IBB 2310 315 Ferst Drive NW, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Corresponding author
| |
Collapse
|
33
|
Nasrollahzadeh E, Razi S, Keshavarz-Fathi M, Mazzone M, Rezaei N. Pro-tumorigenic functions of macrophages at the primary, invasive and metastatic tumor site. Cancer Immunol Immunother 2020; 69:1673-1697. [PMID: 32500231 DOI: 10.1007/s00262-020-02616-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 05/16/2020] [Indexed: 12/19/2022]
Abstract
The tumor microenvironment (TME) not only facilitates cancer progression from the early formation to distant metastasis, but also it differs itself from time to time alongside the tumor evolution. Tumor-associated macrophages (TAMs), whether as pre-existing tissue-resident macrophages or recruited monocytes, are an inseparable part of this microenvironment. As their parents are broadly classified into a dichotomic, simplistic M1 and M2 subtypes, TAMs also exert paradoxical and diverse phenotypes as they are settled in different regions of TME and receive different microenvironmental signals. Briefly, M1 macrophages induce an inflammatory precancerous niche and flame the early oncogenic mutations, whereas their M2 counterparts are reprogrammed to release various growth factors and providing an immunosuppressive state in TME as long as abetting hypoxic cancer cells to set up a new vasculature. Further, they mediate stromal micro-invasion and co-migrate with invasive cancer cells to invade the vascular wall and neural sheath, while another subtype of TAMs prepares suitable niches much earlier than metastatic cells arrive at the target tissues. Accordingly, at the neoplastic transformation, during the benign-to-malignant transition and through the metastatic cascade, macrophages are involved in shaping the primary, micro-invasive and pre-metastatic TMEs. Whether their behavioral plasticity is derived from distinct genotypes or is fueled by microenvironmental cues, it could define these cells as remarkably interesting therapeutic targets.
Collapse
Affiliation(s)
- Elaheh Nasrollahzadeh
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer Biology, VIB, KU Leuven, Louvain, B3000, Belgium
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, 14194, Tehran, Iran. .,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran. .,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden.
| |
Collapse
|
34
|
Abstract
Metastatic disease is the leading cause of death in patients with solid cancers. The progression to metastasis is a multistep process that involves detachment of tumor cells from their constraining basement membrane at the primary site, migration and intravasation into the circulation, survival in the circulation, extravasation into the secondary organ, and survival and growth at the secondary site. During these steps, tumor and immune cells interact and influence each other both within the tumor microenvironment and systemically. In particular, myeloid cells such as monocytes, macrophages, neutrophils, and myeloid-derived suppressor cells (myeloid regulatory cells) have been shown to play important roles in the metastatic process. These interactions open new avenues for targeting cancer metastasis, especially given the increasing interest in development of cancer immunotherapies. In this review, we describe the currently reported pathways and mechanisms involved in myeloid cell enhancement of the metastatic cascade.
Collapse
Affiliation(s)
- Agnieszka Swierczak
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Jeffrey W Pollard
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| |
Collapse
|
35
|
Heparanase in Cancer Metastasis – Heparin as a Potential Inhibitor of Cell Adhesion Molecules. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:309-329. [DOI: 10.1007/978-3-030-34521-1_11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
36
|
Abstract
The dynamic interplay between neoplastic cells and the immune microenvironment regulates every step of the metastatic process. Immune cells contribute to invasion by secreting a cornucopia of inflammatory factors that promote epithelial-to-mesenchymal transition and remodeling of the stroma. Cancer cells then intravasate to the circulatory system assisted by macrophages and use several pathways to avoid recognition by cytotoxtic lymphocytes and phagocytes. Circulating tumor cells that manage to adhere to the vasculature and encounter premetastic niches are able to use the associated myeloid cells to extravasate into ectopic organs and establish a dormant microscopic colony. If successful at avoiding repetitive immune attack, dormant cells can subsequently grow into overt, clinically detectable metastatic lesions, which ultimately account to most cancer-related deaths. Understanding how disseminated tumor cells evade and corrupt the immune system during the final stages of metastasis will be pivotal in developing new therapeutic modalities that combat metastasis.
Collapse
Affiliation(s)
- Asmaa El-Kenawi
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| | - Kay Hänggi
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| | - Brian Ruffell
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| |
Collapse
|
37
|
The endothelial barrier and cancer metastasis: Does the protective facet of platelet function matter? Biochem Pharmacol 2020; 176:113886. [PMID: 32113813 DOI: 10.1016/j.bcp.2020.113886] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/24/2020] [Indexed: 12/16/2022]
Abstract
Overwhelming evidence suggests that platelets have a detrimental role in promoting cancer spread via platelet-cancer cell interactions linked to thrombotic mechanisms. On the other hand, a beneficial role of platelets in the preservation of the endothelial barrier in inflammatory conditions has been recently described, a phenomenon that could also operate in cancer-related inflammation. It is tempting to speculate that some antiplatelet strategies to combat cancer metastasis may impair the endogenous platelet-dependent mechanisms preserving endothelial barrier function. If the protective function of platelets is impaired, it may lead to increased endothelial permeability and more efficient cancer cell intravasation in the primary tumor and cancer cell extravasation at metastatic sites. In this commentary, we discuss current evidence that could support this hypothesis.
Collapse
|
38
|
The contribution of platelets to intravascular arrest, extravasation, and outgrowth of disseminated tumor cells. Clin Exp Metastasis 2020; 37:47-67. [PMID: 31758288 DOI: 10.1007/s10585-019-10009-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/15/2019] [Indexed: 12/16/2022]
Abstract
Platelets are primarily known for their contribution to hemostasis and subsequent wound healing. In addition to these functions, platelets play a role in the process of metastasis. Since the first study that suggested a metastasis-promoting function for platelets was published in 1968, various mechanisms have been proposed to explain how platelets contribute to the metastatic process. These include roles in the intravascular arrest of tumor cells, in tumor cell transendothelial migration, in the degradation of basement membrane barriers, in migration and invasion at the metastatic site, and in the proliferation of disseminated tumor cells. Nevertheless, conflicting observations about the role of platelets in these processes have also been reported. Here, we review the in vivo evidence that supports a role for platelets in metastasis formation, propose several scenarios for the contribution of platelets to tumor cell arrest and transendothelial migration, and discuss the evidence that platelets contribute to metastatic invasion and outgrowth.
Collapse
|
39
|
Zhou X, Yang G, Guan F. Biological Functions and Analytical Strategies of Sialic Acids in Tumor. Cells 2020; 9:E273. [PMID: 31979120 PMCID: PMC7072699 DOI: 10.3390/cells9020273] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/21/2022] Open
Abstract
Sialic acids, a subset of nine carbon acidic sugars, often exist as the terminal sugars of glycans on either glycoproteins or glycolipids on the cell surface. Sialic acids play important roles in many physiological and pathological processes via carbohydrate-protein interactions, including cell-cell communication, bacterial and viral infections. In particular, hypersialylation in tumors, as well as their roles in tumor growth and metastasis, have been widely described. Recent studies have indicated that the aberrant sialylation is a vital way for tumor cells to escape immune surveillance and keep malignance. In this article, we outline the present state of knowledge on the metabolic pathway of human sialic acids, the function of hypersialylation in tumors, as well as the recent labeling and analytical techniques for sialic acids. It is expected to offer a brief introduction of sialic acid metabolism and provide advanced analytical strategies in sialic acid studies.
Collapse
Affiliation(s)
- Xiaoman Zhou
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Ganglong Yang
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Feng Guan
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Joint International Research Laboratory of Glycobiology and Medicinal Chemistry, College of Life Science, Northwest University, Xi’an 710069, China
| |
Collapse
|
40
|
Wettschureck N, Strilic B, Offermanns S. Passing the Vascular Barrier: Endothelial Signaling Processes Controlling Extravasation. Physiol Rev 2019; 99:1467-1525. [PMID: 31140373 DOI: 10.1152/physrev.00037.2018] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A central function of the vascular endothelium is to serve as a barrier between the blood and the surrounding tissue of the body. At the same time, solutes and cells have to pass the endothelium to leave or to enter the bloodstream to maintain homeostasis. Under pathological conditions, for example, inflammation, permeability for fluid and cells is largely increased in the affected area, thereby facilitating host defense. To appropriately function as a regulated permeability filter, the endothelium uses various mechanisms to allow solutes and cells to pass the endothelial layer. These include transcellular and paracellular pathways of which the latter requires remodeling of intercellular junctions for its regulation. This review provides an overview on endothelial barrier regulation and focuses on the endothelial signaling mechanisms controlling the opening and closing of paracellular pathways for solutes and cells such as leukocytes and metastasizing tumor cells.
Collapse
Affiliation(s)
- Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Boris Strilic
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| |
Collapse
|
41
|
Läubli H, Borsig L. Altered Cell Adhesion and Glycosylation Promote Cancer Immune Suppression and Metastasis. Front Immunol 2019; 10:2120. [PMID: 31552050 PMCID: PMC6743365 DOI: 10.3389/fimmu.2019.02120] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 08/23/2019] [Indexed: 12/14/2022] Open
Abstract
Cell-cell interactions and cell adhesion are key mediators of cancer progression and facilitate hallmarks of cancer including immune evasion and metastatic dissemination. Many cell adhesion molecules within the tumor microenvironment are changed and significant alterations of glycosylation are observed. These changes in cell adhesion molecules alter the ability of tumor cells to interact with other cells and extracellular matrix proteins. Three families of cell-cell interaction molecules selectins, Siglecs, and integrins have been associated with cancer progression in many pre-clinical studies, yet inhibition of cell adhesion as a therapeutic target is just beginning to be explored. We review how cell-cell interactions mediated by integrins and the glycan-binding receptors selectins and Siglec receptors support cancer progression. The discussion focuses on mechanisms during immune evasion and metastasis that can be therapeutically targeted by blocking these cell-cell interactions.
Collapse
Affiliation(s)
- Heinz Läubli
- Laboratory for Cancer Immunotherapy, Department of Biomedicine and Medical Oncology, Department of Internal Medicine, University Hospital, Basel, Switzerland
| | - Lubor Borsig
- Department of Physiology, University of Zurich, Zurich, Switzerland.,Comprehensive Cancer Center, Zurich, Switzerland
| |
Collapse
|
42
|
Pape J, Magdeldin T, Ali M, Walsh C, Lythgoe M, Emberton M, Cheema U. Cancer invasion regulates vascular complexity in a three-dimensional biomimetic model. Eur J Cancer 2019; 119:179-193. [PMID: 31470251 DOI: 10.1016/j.ejca.2019.07.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/03/2019] [Accepted: 07/05/2019] [Indexed: 11/20/2022]
Abstract
INTRODUCTION There is a growing appreciation for including a complex, vascularised stroma in three-dimensional (3D) tumour models to recapitulate the native tumour microenvironment in situ. METHODS Using a compartmentalised, biomimetic, 3D cancer model, comprising a central cancer mass surrounded by a vascularised stroma, we have tested the invasive capability of colorectal cancer cells. RESULTS We show histological analysis of dense collagen I/laminin scaffolds, forming necrotic cores with cellular debris. Furthermore, cancer cells within this 3D matrix form spheroids, which is corroborated with high EpCAM expression. We validate the invasive growth of cancer cells into the stroma through quantitative image analysis and upregulation of known invasive gene markers, including metastasis associated in colon cancer 1, matrix metalloproteinase 7 and heparinase. Tumouroids containing highly invasive HCT116 cancer masses form less complex and less branched vascular networks, recapitulating 'leaky' vasculature associated with highly metastatic cancers. Angiogenic factors regulating this were vascular endothelial growth factor A and hepatocyte growth factor active protein. Where vascular networks were formed with less invasive cancer masses (HT29), higher expression of vascular endothelial cadherin active protein resulted in more complex and branched networks. To eliminate the cell-cell interaction between the cancer mass and stroma, we developed a three-compartment model containing an acellular ring to test the chemoattractant pull from the cancer mass. This resulted in migration of endothelial networks through the acellular ring accompanied by alignment of vascular networks at the cancer/stroma boundary. DISCUSSION This work interrogates to the gene and protein level how cancer cells influence the development of a complex stroma, which shows to be directly influenced by the invasive capability of the cancer.
Collapse
Affiliation(s)
- Judith Pape
- Institute of Orthopaedics and Musculoskeletal Sciences, Division of Surgery and Interventional Science, University College London, Stanmore Campus, Brockley Hill, HA7 4LP, London, United Kingdom
| | - Tarig Magdeldin
- Institute of Orthopaedics and Musculoskeletal Sciences, Division of Surgery and Interventional Science, University College London, Stanmore Campus, Brockley Hill, HA7 4LP, London, United Kingdom
| | - Morium Ali
- Center for Advanced Biomedical Imaging, Paul O'Gorman Building, 72 Huntley Street, University College London, WC1E 6DD, London, United Kingdom
| | - Claire Walsh
- Center for Advanced Biomedical Imaging, Paul O'Gorman Building, 72 Huntley Street, University College London, WC1E 6DD, London, United Kingdom
| | - Mark Lythgoe
- Center for Advanced Biomedical Imaging, Paul O'Gorman Building, 72 Huntley Street, University College London, WC1E 6DD, London, United Kingdom
| | - Mark Emberton
- Faculty of Medical Sciences, University College London, Bloomsbury Campus Maple House, 149 Tottenham Court Road, W1T 7NF, London, United Kingdom
| | - Umber Cheema
- Institute of Orthopaedics and Musculoskeletal Sciences, Division of Surgery and Interventional Science, University College London, Stanmore Campus, Brockley Hill, HA7 4LP, London, United Kingdom.
| |
Collapse
|
43
|
McDowell SAC, Quail DF. Immunological Regulation of Vascular Inflammation During Cancer Metastasis. Front Immunol 2019; 10:1984. [PMID: 31497019 PMCID: PMC6712555 DOI: 10.3389/fimmu.2019.01984] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 08/06/2019] [Indexed: 12/30/2022] Open
Abstract
Metastasis is the predominant cause of cancer-related mortality, despite being a highly inefficient process overall. The vasculature is the gatekeeper for tumor cell seeding within the secondary tissue microenvironment—the rate limiting step of the metastatic cascade. Therefore, factors that regulate vascular physiology dramatically influence cancer outcomes. There are a myriad of physiologic circumstances that not only influence the intrinsic capacity of tumor cells to cross the endothelial barrier, but also that regulate vascular inflammation and barrier integrity to enable extravasation into the metastatic niche. These processes are highly dependent on inflammatory cues largely initiated by the innate immune compartment, that are meant to help re-establish tissue homeostasis, but instead become hijacked by cancer cells. Here, we discuss the scientific advances in understanding the interactions between innate immune cells and the endothelium, describe their influence on cancer metastasis, and evaluate potential therapeutic interventions for the alleviation of metastatic disease. By triangulating the relationship between immune cells, endothelial cells, and tumor cells, we will gain greater insight into how to impede the metastatic process by focusing on its most vulnerable phases, thereby reducing metastatic spread and cancer-related mortality.
Collapse
Affiliation(s)
- Sheri A C McDowell
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada.,Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - Daniela F Quail
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada.,Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.,Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
44
|
Liu L, Liu L, Li Y, Huang X, Gu D, Wei B, Su D, Jin G. Ultrasmall superparamagnetic nanoparticles targeting E-selectin: synthesis and effects in mice in vitro and in vivo. Int J Nanomedicine 2019; 14:4517-4528. [PMID: 31354271 PMCID: PMC6590629 DOI: 10.2147/ijn.s199571] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 05/01/2019] [Indexed: 11/23/2022] Open
Abstract
Purpose: We developed a contrast agent for targeting E-selectin expression. We detected the agent using magnetic resonance imaging (MRI) in vivo in nude mice that had undergone nasopharyngeal carcinoma (NPC) metastasis. Methods: Sialyl Lewis X (sLeX) was conjugated with ultrasmall superparamagnetic iron oxide (USPIO) nanoparticles. Hydrodynamic size, polydispersity index, and ζ-potential of USPIO–polyethylene glycol (PEG) nanoparticles and USPIO-PEG-sLeX nanoparticles were measured. Component changes in nanoparticles of USPIO, USPIO-PEG, and USPIO-PEG-sLeX were analyzed by thermogravimetric analysis and Fourier-transform infrared spectroscopy. A model of NPC metastasis to inguinal lymph nodes in nude mice was used to investigate characteristics of the USPIO-PEG-sLeX nanoparticles in vivo. We investigated the ability of the T2* value, change in T2* value (ΔT2* value), and enhancement rate (ER) to assess accumulation of USPIO-PEG-sLeX nanoparticles quantitatively in mice of a metastasis group and control group. Four MRI scans were undertaken for each mouse. The first scan (t0) was done before administration of USPIO-PEG-sLeX nanoparticles (0.1 mL) via the tail vein. The other scans were carried out at 0 (t1), 1 (t2), and 2 hours (t3) postinjection. The mean optical density was used to reflect E-selectin expression. Results: sLeX was labeled onto USPIO successfully. In vivo, there were significant interactions between the groups and time for T2* values after administration of USPIO-PEG-sLeX nanoparticles. Six parameters (T2* at t2, ΔT2* at t1, ΔT2* at t2, ER at t1, ER at t2, and ER at t3) were correlated with the mean optical density. Conclusion: USPIO-PEG-sLeX nanoparticles can be used to assess E-selectin expression quantitatively. Use of such molecular probes could enable detection of early metastasis of NPC, more accurate staging, and treatment monitoring.
Collapse
Affiliation(s)
- Lijuan Liu
- Centre of Imaging Diagnosis, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Lu Liu
- Centre of Imaging Diagnosis, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Yin Li
- Centre of Imaging Diagnosis, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Xiaoxin Huang
- Centre of Imaging Diagnosis, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Donglian Gu
- Centre of Imaging Diagnosis, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Bo Wei
- Centre of Imaging Diagnosis, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Danke Su
- Centre of Imaging Diagnosis, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Guanqiao Jin
- Centre of Imaging Diagnosis, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| |
Collapse
|
45
|
Martin EW, Malgor R, Resto VA, Goetz DJ, Burdick MM. Dynamic biochemical tissue analysis detects functional selectin ligands on human cancer tissues. Sci Rep 2019; 9:8511. [PMID: 31186472 PMCID: PMC6560120 DOI: 10.1038/s41598-019-44838-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/15/2019] [Indexed: 12/12/2022] Open
Abstract
Cell adhesion mediated by selectins (expressed by activated endothelium, activated platelets, and leukocytes) binding to their resepective selectin ligands (expressed by cancer cells) may be involved in metastasis. Therefore, methods of characterizing selectin ligands expressed on human tissue may serve as valuable assays. Presented herein is an innovative method for detecting functional selectin ligands expressed on human tissue that uses a dynamic approach, which allows for control over the force applied to the bonds between the probe and target molecules. This new method of tissue interrogation, known as dynamic biochemical tissue analysis (DBTA), involves the perfusion of molecular probe-coated microspheres over tissues. DBTA using selectin-coated probes is able to detect functional selectin ligands expressed on tissue from multiple cancer types at both primary and metastatic sites.
Collapse
Affiliation(s)
- Eric W Martin
- Biomedical Engineering Program, Russ College of Engineering and Technology, Athens, USA
- Department of Chemical and Biomolecular Engineering, Russ College of Engineering and Technology, Athens, USA
| | - Ramiro Malgor
- Biomedical Engineering Program, Russ College of Engineering and Technology, Athens, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Athens, USA
| | - Vicente A Resto
- Department of Otolaryngology, University of Texas-Medical Branch, Galveston, TX, 77555, USA
| | - Douglas J Goetz
- Biomedical Engineering Program, Russ College of Engineering and Technology, Athens, USA
- Department of Chemical and Biomolecular Engineering, Russ College of Engineering and Technology, Athens, USA
| | - Monica M Burdick
- Biomedical Engineering Program, Russ College of Engineering and Technology, Athens, USA.
- Department of Chemical and Biomolecular Engineering, Russ College of Engineering and Technology, Athens, USA.
- Edison Biotechnology Institute, Ohio University, Athens, OH, 45701, USA.
| |
Collapse
|
46
|
Kim H, Chung H, Kim J, Choi D, Shin Y, Kang YG, Kim B, Seo S, Chung S, Seok SH. Macrophages-Triggered Sequential Remodeling of Endothelium-Interstitial Matrix to Form Pre-Metastatic Niche in Microfluidic Tumor Microenvironment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900195. [PMID: 31179226 PMCID: PMC6548952 DOI: 10.1002/advs.201900195] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/01/2019] [Indexed: 05/07/2023]
Abstract
The primed microenvironment of future metastatic sites, called the pre-metastatic niche, is a prerequisite for overt metastasis. However, a mechanistic understanding of the contributions of recruited cells to the niche is hindered by complex in vivo systems. Herein, a microfluidic platform that incorporates endothelial cells and extracellular matrix (ECM) scaffolds is developed, and the distinct role of recruited monocytes and macrophages in establishing pre-metastatic niches is delineated. It is observed that monocyte-derived matrix metalloproteinase 9 facilitates cancer cell extravasation through destruction of endothelial tight junctions. Furthermore, subsequent cancer cell invasiveness is significantly enhanced. Close examination of ECM structures reveals that cancer cells move within characteristic "microtracks" generated by macrophages, suggesting that macrophages could serve as a compensatory mechanism for the reduced migratory capacity of cancer cells. Thus, the first evidence of monocyte/macrophage-induced remodeling is shown, and these findings will open up new horizons for improving characterization of the pre-metastatic niche and corresponding immunotherapies.
Collapse
Affiliation(s)
- Hyunho Kim
- School of Mechanical EngineeringCollege of EngineeringKorea UniversitySeoul02841Republic of Korea
| | - Hyewon Chung
- Department of Microbiology and ImmunologyInstitute of Endemic DiseaseCollege of MedicineSeoul National UniversitySeoul03080Republic of Korea
| | - Jaehoon Kim
- School of Mechanical EngineeringCollege of EngineeringKorea UniversitySeoul02841Republic of Korea
| | - Dong‐Hee Choi
- School of Mechanical EngineeringCollege of EngineeringKorea UniversitySeoul02841Republic of Korea
| | - Yoojin Shin
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Yong Guk Kang
- Department of Bio‐Convergence EngineeringCollege of Health ScienceKorea UniversitySeoul02841Republic of Korea
| | - Beop‐Min Kim
- Department of Bio‐Convergence EngineeringCollege of Health ScienceKorea UniversitySeoul02841Republic of Korea
| | - Sang‐Uk Seo
- Department of Biomedical SciencesCollege of MedicineSeoul National UniversitySeoul03080Republic of Korea
| | - Seok Chung
- School of Mechanical EngineeringCollege of EngineeringKorea UniversitySeoul02841Republic of Korea
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
| | - Seung Hyeok Seok
- Department of Microbiology and ImmunologyInstitute of Endemic DiseaseCollege of MedicineSeoul National UniversitySeoul03080Republic of Korea
| |
Collapse
|
47
|
S-nitrosylation and its role in breast cancer angiogenesis and metastasis. Nitric Oxide 2019; 87:52-59. [PMID: 30862477 DOI: 10.1016/j.niox.2019.03.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 01/23/2019] [Accepted: 03/06/2019] [Indexed: 12/24/2022]
Abstract
S-nitrosylation, the modification by nitric oxide of free sulfhydryl groups in cysteines, has become an important regulatory mechanism in carcinogenesis and metastasis. S-nitrosylation of targets in tumor cells contributes to metastasis regulating epithelial to mesenchymal transition, migration and invasion. In the tumor environment, the role of S-nitrosylation in endothelium has not been addressed; however, the evidence points out that S-nitrosylation of endothelial proteins may regulate angiogenesis, adhesion of tumor cells to the endothelium, intra and extravasation of tumor cells and contribute to metastasis.
Collapse
|
48
|
Windisch R, Pirschtat N, Kellner C, Chen-Wichmann L, Lausen J, Humpe A, Krause DS, Wichmann C. Oncogenic Deregulation of Cell Adhesion Molecules in Leukemia. Cancers (Basel) 2019; 11:E311. [PMID: 30841639 PMCID: PMC6468598 DOI: 10.3390/cancers11030311] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/26/2019] [Accepted: 02/28/2019] [Indexed: 01/01/2023] Open
Abstract
Numerous cell⁻cell and cell⁻matrix interactions within the bone marrow microenvironment enable the controlled lifelong self-renewal and progeny of hematopoietic stem and progenitor cells (HSPCs). On the cellular level, this highly mutual interaction is granted by cell adhesion molecules (CAMs) integrating differentiation, proliferation, and pro-survival signals from the surrounding microenvironment to the inner cell. However, cell⁻cell and cell⁻matrix interactions are also critically involved during malignant transformation of hematopoietic stem/progenitor cells. It has become increasingly apparent that leukemia-associated gene products, such as activated tyrosine kinases and fusion proteins resulting from chromosomal translocations, directly regulate the activation status of adhesion molecules, thereby directing the leukemic phenotype. These observations imply that interference with adhesion molecule function represents a promising treatment strategy to target pre-leukemic and leukemic lesions within the bone marrow niche. Focusing on myeloid leukemia, we provide a current overview of the mechanisms by which leukemogenic gene products hijack control of cellular adhesion to subsequently disturb normal hematopoiesis and promote leukemia development.
Collapse
Affiliation(s)
- Roland Windisch
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, LMU Munich, 81377 Munich, Germany.
| | - Nina Pirschtat
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, LMU Munich, 81377 Munich, Germany.
| | - Christian Kellner
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, LMU Munich, 81377 Munich, Germany.
| | - Linping Chen-Wichmann
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, LMU Munich, 81377 Munich, Germany.
| | - Jörn Lausen
- Institute for Transfusion Medicine and Immunohematology, Johann-Wolfgang-Goethe University and German Red Cross Blood Service, 60528 Frankfurt am Main, Germany.
| | - Andreas Humpe
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, LMU Munich, 81377 Munich, Germany.
| | - Daniela S Krause
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt am Main, Germany.
| | - Christian Wichmann
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, LMU Munich, 81377 Munich, Germany.
| |
Collapse
|
49
|
Roblek M, Protsyuk D, Becker PF, Stefanescu C, Gorzelanny C, Glaus Garzon JF, Knopfova L, Heikenwalder M, Luckow B, Schneider SW, Borsig L. CCL2 Is a Vascular Permeability Factor Inducing CCR2-Dependent Endothelial Retraction during Lung Metastasis. Mol Cancer Res 2018; 17:783-793. [PMID: 30552233 DOI: 10.1158/1541-7786.mcr-18-0530] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/30/2018] [Accepted: 12/04/2018] [Indexed: 12/24/2022]
Abstract
Increased levels of the chemokine CCL2 in cancer patients are associated with poor prognosis. Experimental evidence suggests that CCL2 correlates with inflammatory monocyte recruitment and induction of vascular activation, but the functionality remains open. Here, we show that endothelial Ccr2 facilitates pulmonary metastasis using an endothelial-specific Ccr2-deficient mouse model (Ccr2ecKO). Similar levels of circulating monocytes and equal leukocyte recruitment to metastatic lesions of Ccr2ecKO and Ccr2fl/fl littermates were observed. The absence of endothelial Ccr2 strongly reduced pulmonary metastasis, while the primary tumor growth was unaffected. Despite a comparable cytokine milieu in Ccr2ecKO and Ccr2fl/fl littermates the absence of vascular permeability induction was observed only in Ccr2ecKO mice. CCL2 stimulation of pulmonary endothelial cells resulted in increased phosphorylation of MLC2, endothelial cell retraction, and vascular leakiness that was blocked by an addition of a CCR2 inhibitor. These data demonstrate that endothelial CCR2 expression is required for tumor cell extravasation and pulmonary metastasis. IMPLICATIONS: The findings provide mechanistic insight into how CCL2-CCR2 signaling in endothelial cells promotes their activation through myosin light chain phosphorylation, resulting in endothelial retraction and enhanced tumor cell migration and metastasis.
Collapse
Affiliation(s)
- Marko Roblek
- Institute of Physiology, University of Zurich and Zurich Center for Integrative Human Physiology, Zurich, Switzerland
| | - Darya Protsyuk
- Institute of Physiology, University of Zurich and Zurich Center for Integrative Human Physiology, Zurich, Switzerland
| | - Paul F Becker
- Institute of Virology, Technische Universität München/Helmholtz Zentrum Munich, Munich, Germany
| | - Cristina Stefanescu
- Institute of Physiology, University of Zurich and Zurich Center for Integrative Human Physiology, Zurich, Switzerland
| | - Christian Gorzelanny
- Department of Dermatology and Venerology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Jesus F Glaus Garzon
- Institute of Physiology, University of Zurich and Zurich Center for Integrative Human Physiology, Zurich, Switzerland
| | - Lucia Knopfova
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.,International Clinical Research Center, Center for Biological and Cellular Engineering, St. Anne's University Hospital, Brno, Czech Republic
| | - Mathias Heikenwalder
- Institute of Virology, Technische Universität München/Helmholtz Zentrum Munich, Munich, Germany.,German Cancer Research Centre (DKFZ), Division of Chronic Inflammation and Cancer, Heidelberg, Germany
| | - Bruno Luckow
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU München, Munich, Germany
| | - Stefan W Schneider
- Department of Dermatology and Venerology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Lubor Borsig
- Institute of Physiology, University of Zurich and Zurich Center for Integrative Human Physiology, Zurich, Switzerland.
| |
Collapse
|
50
|
Borsig L. Selectins in cancer immunity. Glycobiology 2018; 28:648-655. [PMID: 29272415 DOI: 10.1093/glycob/cwx105] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/18/2017] [Indexed: 12/12/2022] Open
Abstract
Selectins are vascular adhesion molecules that mediate physiological responses such as inflammation, immunity and hemostasis. During cancer progression, selectins promote various steps enabling the interactions between tumor cells and the blood constituents, including platelets, endothelial cells and leukocytes. Selectins are carbohydrate-binding molecules that bind to sialylated, fucosylated glycan structures. The increased selectin ligand expression on tumor cells correlates with enhanced metastasis and poor prognosis for cancer patients. While, many studies focused on the role of selectin as a mediator of tumor cell adhesion and extravasation during metastasis, there is evidence for selectins to activate signaling cascade that regulates immune responses within a tumor microenvironment. L-Selectin binding induces activation of leukocytes, which can be further modulated by selectin-mediated interactions with platelets and endothelial cells. Selectin ligand on leukocytes, PSGL-1, triggers intracellular signaling in leukocytes that are induced through platelet's P-selectin or endothelial E-selectin binding. In this review, I summarize the evidence for selectin-induced immune modulation in cancer progression that represents a possible target for controlling tumor immunity.
Collapse
Affiliation(s)
- Lubor Borsig
- Institute of Physiology, University of Zurich and Zurich Center for Integrative Human Physiology, Winterthurerstrasse 190, Zurich, Switzerland
| |
Collapse
|