1
|
Delle Cave D, Mangini M, Tramontano C, De Stefano L, Corona M, Rea I, De Luca AC, Lonardo E. Hybrid Biosilica Nanoparticles for in-vivo Targeted Inhibition of Colorectal Cancer Growth and Label-Free Imaging. Int J Nanomedicine 2024; 19:12079-12098. [PMID: 39583322 PMCID: PMC11585298 DOI: 10.2147/ijn.s480168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/17/2024] [Indexed: 11/26/2024] Open
Abstract
Background Metastasis-initiating cells are key players in progression, resistance, and relapse of colorectal cancer (CRC), by leveraging the regulatory relationship between Transforming Growth Factor-beta (TGF-β) signaling and anti-L1 cell adhesion molecule (L1CAM). Methods This study introduces a novel strategy for CRC targeted therapy and imaging based on the use of a hybrid nanosystem made of gold nanoparticles-covered porous biosilica further modified with the (L1CAM) antibody. Results The nanosystem intracellularly delivers galunisertib (LY), a TGF-β inhibitor, aiming to inhibit epithelial-mesenchymal transition (EMT), a process pivotal for metastasis. Anti-L1CAM antibody-functionalized nanoparticles (NPs) target tumor-initiating cells expressing L1CAM, inhibiting cancer growth. The number of antibody molecules conjugated to the single NP is precisely quantified, revealing a high surface coverage that facilitates the tumor targeting. The therapeutic efficacy of the nanosystem is investigated in organoid-like cultures of CRC cells and in vivo mouse models, showing a significant reduction in tumor growth. The spatial distribution of NPs within CRC tumors from mice is investigated using a label-free optical approach based on Raman micro-spectroscopy. Conclusion This research highlights the multifunctional capabilities of engineered biosilica NPs, which offer new insights in targeted CRC therapy and imaging, improving patient outcomes and paving the way for personalized therapies.
Collapse
Affiliation(s)
- Donatella Delle Cave
- National Research Council, Institute of Genetics and Biophysics, Naples, 80131, Italy
| | - Maria Mangini
- National Research Council, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, Second Unit, Naples, 80131, Italy
| | - Chiara Tramontano
- National Research Council, Institute of Applied Sciences and Intelligent Systems, Unit of Naples, Naples, 80131, Italy
| | - Luca De Stefano
- National Research Council, Institute of Applied Sciences and Intelligent Systems, Unit of Naples, Naples, 80131, Italy
| | - Marco Corona
- National Research Council, Institute of Genetics and Biophysics, Naples, 80131, Italy
| | - Ilaria Rea
- National Research Council, Institute of Applied Sciences and Intelligent Systems, Unit of Naples, Naples, 80131, Italy
| | - Anna Chiara De Luca
- National Research Council, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, Second Unit, Naples, 80131, Italy
| | - Enza Lonardo
- National Research Council, Institute of Genetics and Biophysics, Naples, 80131, Italy
| |
Collapse
|
2
|
Shahhosseini R, Pakmehr S, Elhami A, Shakir MN, Alzahrani AA, Al-Hamdani MM, Abosoda M, Alsalamy A, Mohammadi-Dehcheshmeh M, Maleki TE, Saffarfar H, Ali-Khiavi P. Current biological implications and clinical relevance of metastatic circulating tumor cells. Clin Exp Med 2024; 25:7. [PMID: 39546080 PMCID: PMC11567993 DOI: 10.1007/s10238-024-01518-6] [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: 10/09/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
Metastatic disease and cancer recurrence are the primary causes of cancer-related deaths. Circulating tumor cells (CTCs) and disseminated tumor cells (DTCs) are the driving forces behind the spread of cancer cells. The emergence and development of liquid biopsy using rare CTCs as a minimally invasive strategy for early-stage tumor detection and improved tumor management is a promising advancement in recent years. However, before blood sample analysis and clinical translation, precise isolation of CTCs from patients' blood based on their biophysical properties, followed by molecular identification of CTCs using single-cell multi-omics technologies is necessary to understand tumor heterogeneity and provide effective diagnosis and monitoring of cancer progression. Additionally, understanding the origin, morphological variation, and interaction between CTCs and the primary and metastatic tumor niche, as well as and regulatory immune cells, will offer new insights into the development of CTC-based advanced tumor targeting in the future clinical trials.
Collapse
Affiliation(s)
| | - SeyedAbbas Pakmehr
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Ahvaz Jundishapur University of Medical Sciences Ahvaz, Ahvaz, Iran
| | - Anis Elhami
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maha Noori Shakir
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | | | | | - Munther Abosoda
- College of Pharmacy, The Islamic University, Najaf, Iraq
- College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, The Islamic University of Babylon, Babylon, Iraq
| | - Ali Alsalamy
- College of Pharmacy, Imam Ja'afar Al-Sadiq University, Al-Samawa, Al-Muthanna, 66002, Iraq
| | | | | | - Hossein Saffarfar
- Cardiovascular Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Payam Ali-Khiavi
- Medical Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Chen G, Zhang M, Lin X, Shi Q, Xu C, Sun B, Xiao X, Feng H. Single-cell RNA transcriptomic analyses of tumor microenvironment of ovarian metastasis in gastric cancer. Cell Oncol (Dordr) 2024; 47:1911-1925. [PMID: 39162990 DOI: 10.1007/s13402-024-00974-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2024] [Indexed: 08/21/2024] Open
Abstract
PURPOSE Ovarian metastasis of gastric cancer (GC), commonly referred to as Krukenberg tumors, leads to a poor prognosis. However, the cause of metastasis remains unknown. Here, we present an integrated single-cell RNA sequencing (scRNA-Seq) analysis of the immunological microenvironment of two paired clinical specimens with ovarian metastasis of GC. METHODS scRNA-Seq was performed to determine the immunological microenvironment in ovarian metastasis of gastric cancer. CellChat was employed to analyze cell-cell communications across different cell types. Functional enrichment analysis was done by enrichKEGG in clusterProfiler. GEPIA2 was used to assess the influence of certain genes and gene signatures on prognosis. RESULTS The ovarian metastasis tissues exhibit a heterogenous immunological microenvironment compared to the primary tumors. Exhaustion of T and B cells is observed in the ovarian metastasis tissues. Compared to the paired adjacent non-tumoral and primary tumors, the ratio of endothelial cells and fibroblasts is high in the ovarian metastasis tissues. Compared to primary ovarian cancers, we identify a specific group of tumor-associated fibroblasts with MFAP4 and CAPNS1 expression in the ovarian metastatic tissues of GC. We further define metastasis-related-endothelial and metastasis-related-fibroblast signatures and indicate that patients with these high signature scores have a poor prognosis. In addition, the ovarian metastasis tissue has a lower level of intercellular communications compared to the primary tumor. CONCLUSION Our findings reveal the immunological microenvironment of ovarian metastasis of gastric cancer and will promote the discovery of new therapeutic strategies for ovarian metastasis in gastric cancer.
Collapse
Affiliation(s)
- Guoyu Chen
- State Key Laboratory of Systems Medicine for Cancer, Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Mingda Zhang
- State Key Laboratory of Systems Medicine for Cancer, Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xiaolin Lin
- Department of Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Qiqi Shi
- State Key Laboratory of Systems Medicine for Cancer, Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Chenxin Xu
- State Key Laboratory of Systems Medicine for Cancer, Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Bowen Sun
- State Key Laboratory of Systems Medicine for Cancer, Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xiuying Xiao
- Department of Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Haizhong Feng
- State Key Laboratory of Systems Medicine for Cancer, Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
4
|
Hu Y, Xu Z, Zhou D, Hou H, Liu B, Long H, Hu W, Tang Y, Wang J, Wei D, Zhao Q. CXCR4 promotes migration, invasion, and epithelial-mesenchymal transition of papillary thyroid carcinoma by activating STAT3 signaling pathway. J Cancer Res Ther 2024; 20:1241-1250. [PMID: 39206986 DOI: 10.4103/jcrt.jcrt_2395_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 05/02/2024] [Indexed: 09/04/2024]
Abstract
AIMS Papillary thyroid cancer (PTC) is a serious threat to human health worldwide, while metastasis in the early phase limits therapeutic success and leads to poor survival outcomes. The CXC chemokine receptor type 4 (CXCR4) plays an important role in many cellular movements such as transcriptional modulation, cell skeleton rearrangement, and cell migration, and the change in CXCR4 levels are crucial in various diseases including cancer. In this study, we explored the role of CXCR4 in the migration and invasion of PTC and investigated the potential mechanisms underlying its effects. SUBJECTS AND METHODS We analyzed the expression levels of CXCR4 in PTC tissues and cell lines. Would healing migration, Transwell invasion assay in vitro, and tail-vein lung metastasis assay In vivo were performed to evaluated the migration and invasion abilities of PTC cells with stable CXCR4 knockdown or overexpression. Signal transducers and activators of transcription (STAT3) signaling pathway-related protein expressions were examined by Western blotting assays. RESULTS The results showed that CXCR4 was highly expressed in PTC cell lines and PTC tissues. CXCR4 knockdown in PTC cells dampened the migration, invasion, and epithelial-mesenchymal transition (EMT), whereas CXCR4 overexpression enhanced these properties. In vivo, we also found that CXCR4 promoted the metastasis of PTC. Mechanistic studies showed that CXCR4 played these vital roles through the STAT3 signaling pathway. Furthermore, PTC patients with high CXCR4 or p-STAT3 expression correlated with aggressive clinical characteristics such as extrathyroidal extension (ETE), and lymph node metastasis (LNM). CONCLUSIONS We provided evidence that CXCR4 might activate the STAT3 signaling pathway and further promote PTC development. Thus, CXCR4 might be a novel therapeutic target for PTC.
Collapse
Affiliation(s)
- Yajie Hu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zhipeng Xu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Department of Urology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Organ Transplantation and Nephrosis, Shandong Institute of Nephrology, Jinan, Shandong, China
| | - Dongsheng Zhou
- Department of Thyroid Surgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Haitao Hou
- Department of Breast and Thyroid Surgery, Tengzhou Central People's Hospital, Zaozhuang, Shandong, China
| | - Bin Liu
- Department of Breast and Thyroid Surgery, Tengzhou Central People's Hospital, Zaozhuang, Shandong, China
| | - Houlong Long
- Department of Breast and Thyroid Surgery, Tengzhou Central People's Hospital, Zaozhuang, Shandong, China
| | - Wenxin Hu
- Department of Urology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Organ Transplantation and Nephrosis, Shandong Institute of Nephrology, Jinan, Shandong, China
| | - Yuanqi Tang
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, Shandong, China
| | - Jianning Wang
- Department of Urology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Organ Transplantation and Nephrosis, Shandong Institute of Nephrology, Jinan, Shandong, China
| | - Dan Wei
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, Shandong, China
| | - Quanlin Zhao
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
5
|
Fan C, Xiong F, Zhang S, Gong Z, Liao Q, Li G, Guo C, Xiong W, Huang H, Zeng Z. Role of adhesion molecules in cancer and targeted therapy. SCIENCE CHINA. LIFE SCIENCES 2024; 67:940-957. [PMID: 38212458 DOI: 10.1007/s11427-023-2417-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/17/2023] [Indexed: 01/13/2024]
Abstract
Adhesion molecules mediate cell-to-cell and cell-to-extracellular matrix interactions and transmit mechanical and chemical signals among them. Various mechanisms deregulate adhesion molecules in cancer, enabling tumor cells to proliferate without restraint, invade through tissue boundaries, escape from immune surveillance, and survive in the tumor microenvironment. Recent studies have revealed that adhesion molecules also drive angiogenesis, reshape metabolism, and are involved in stem cell self-renewal. In this review, we summarize the functions and mechanisms of adhesion molecules in cancer and the tumor microenvironment, as well as the therapeutic strategies targeting adhesion molecules. These studies have implications for furthering our understanding of adhesion molecules in cancer and providing a paradigm for exploring novel therapeutic approaches.
Collapse
Affiliation(s)
- Chunmei Fan
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410000, China
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Fang Xiong
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Shanshan Zhang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Zhaojian Gong
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Qianjin Liao
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410000, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410000, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, China
| | - Can Guo
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410000, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410000, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, China
| | - He Huang
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, 410013, China.
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410000, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, China.
| |
Collapse
|
6
|
Papavassiliou AG, Delle Cave D. Novel Therapeutic Approaches for Colorectal Cancer Treatment. Int J Mol Sci 2024; 25:2228. [PMID: 38396903 PMCID: PMC10889277 DOI: 10.3390/ijms25042228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
According to GLOBOCAN 2020 data, colorectal cancer (CRC) represents the third most common malignancy and the second most deadly cancer worldwide [...].
Collapse
Affiliation(s)
- Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Donatella Delle Cave
- Institute of Genetics and Biophysics ‘Adriano Buzzati-Traverso’, CNR, 80131 Naples, Italy
| |
Collapse
|
7
|
Sivaccumar JP, Iaccarino E, Oliver A, Cantile M, Olimpieri P, Leonardi A, Ruvo M, Sandomenico A. Production in Bacteria and Characterization of Engineered Humanized Fab Fragment against the Nodal Protein. Pharmaceuticals (Basel) 2023; 16:1130. [PMID: 37631045 PMCID: PMC10459755 DOI: 10.3390/ph16081130] [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: 06/29/2023] [Revised: 07/29/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Drug development in recent years is increasingly focused on developing personalized treatments based on blocking molecules selective for therapeutic targets specifically present in individual patients. In this perspective, the specificity of therapeutic targets and blocking agents plays a crucial role. Monoclonal antibodies (mAbs) and their surrogates are increasingly used in this context thanks to their ability to bind therapeutic targets and to inhibit their activity or to transport bioactive molecules into the compartments in which the targets are expressed. Small antibody-like molecules, such as Fabs, are often used in certain clinical settings where small size and better tissue penetration are required. In the wake of this research trend, we developed a murine mAb (3D1) neutralizing the activity of Nodal, an oncofetal protein that is attracting an ever-increasing interest as a selective therapeutic target for several cancer types. Here, we report the preparation of a recombinant Fab of 3D1 that has been humanized through a computational approach starting from the sequence of the murine antibody. The Fab has been expressed in bacterial cells (1 mg/L bacterial culture), biochemically characterized in terms of stability and binding properties by circular dichroism and bio-layer interferometry techniques and tested in vitro on Nodal-positive cancer cells.
Collapse
Affiliation(s)
- Jwala P. Sivaccumar
- Institute of Biostructures and Bioimaging, CNR, Via P. Castellino, 111, 80131 Naples, Italy (E.I.)
| | - Emanuela Iaccarino
- Institute of Biostructures and Bioimaging, CNR, Via P. Castellino, 111, 80131 Naples, Italy (E.I.)
| | - Angela Oliver
- Institute of Biostructures and Bioimaging, CNR, Via P. Castellino, 111, 80131 Naples, Italy (E.I.)
- Università degli Studi della Campania Luigi Vanvitelli, Via Vivaldi 43, 81100 Caserta, Italy
| | | | | | - Antonio Leonardi
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, via Pansini 5, 80131 Naples, Italy
| | - Menotti Ruvo
- Institute of Biostructures and Bioimaging, CNR, Via P. Castellino, 111, 80131 Naples, Italy (E.I.)
| | - Annamaria Sandomenico
- Institute of Biostructures and Bioimaging, CNR, Via P. Castellino, 111, 80131 Naples, Italy (E.I.)
| |
Collapse
|
8
|
Hjazi A, Nasir F, Noor R, Alsalamy A, Zabibah RS, Romero-Parra RM, Ullah MI, Mustafa YF, Qasim MT, Akram SV. The pathological role of C-X-C chemokine receptor type 4 (CXCR4) in colorectal cancer (CRC) progression; special focus on molecular mechanisms and possible therapeutics. Pathol Res Pract 2023; 248:154616. [PMID: 37379710 DOI: 10.1016/j.prp.2023.154616] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/07/2023] [Accepted: 06/10/2023] [Indexed: 06/30/2023]
Abstract
Colorectal cancer (CRC) is comprised of transformed cells and non-malignant cells including cancer-associated fibroblasts (CAF), endothelial vasculature cells, and tumor-infiltrating cells. These nonmalignant cells, as well as soluble factors (e.g., cytokines), and the extracellular matrix (ECM), form the tumor microenvironment (TME). In general, the cancer cells and their surrounding TME can crosstalk by direct cell-to-cell contact and via soluble factors, such as cytokines (e.g., chemokines). TME not only promotes cancer progression through growth-promoting cytokines but also provides resistance to chemotherapy. Understanding the mechanisms of tumor growth and progression and the roles of chemokines in CRC will likely suggest new therapeutic targets. In this line, a plethora of reports has evidenced the critical role of chemokine receptor type 4 (CXCR4)/C-X-C motif chemokine ligand 12 (CXCL12 or SDF-1) axis in CRC pathogenesis. In the current review, we take a glimpse into the role of the CXCR4/CXCL12 axis in CRC growth, metastasis, angiogenesis, drug resistance, and immune escape. Also, a summary of recent reports concerning targeting CXCR4/CXCL12 axis for CRC management and therapy has been delivered.
Collapse
Affiliation(s)
- Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | | | - Rabia Noor
- Amna Inayat Medical College, Lahore, Pakistan
| | - Ali Alsalamy
- College of Medical Technique, Imam Ja'afar Al-Sadiq University, Al-Muthanna 66002, Iraq
| | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | | | - Muhammad Ikram Ullah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 75471, Aljouf, Saudi Arabia
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| | - Maytham T Qasim
- Department of Anesthesia, College of Health and Medical Technololgy, Al-Ayen University, Thi-Qar, Iraq
| | - Shaik Vaseem Akram
- Uttaranchal Institute of Technology, Division of Research & Innovation, Uttaranchal University, Dehradun 248007, India
| |
Collapse
|
9
|
Neto Í, Rocha J, Gaspar MM, Reis CP. Experimental Murine Models for Colorectal Cancer Research. Cancers (Basel) 2023; 15:2570. [PMID: 37174036 PMCID: PMC10177088 DOI: 10.3390/cancers15092570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Colorectal cancer (CRC) is the third most prevalent malignancy worldwide and in both sexes. Numerous animal models for CRC have been established to study its biology, namely carcinogen-induced models (CIMs) and genetically engineered mouse models (GEMMs). CIMs are valuable for assessing colitis-related carcinogenesis and studying chemoprevention. On the other hand, CRC GEMMs have proven to be useful for evaluating the tumor microenvironment and systemic immune responses, which have contributed to the discovery of novel therapeutic approaches. Although metastatic disease can be induced by orthotopic injection of CRC cell lines, the resulting models are not representative of the full genetic diversity of the disease due to the limited number of cell lines suitable for this purpose. On the other hand, patient-derived xenografts (PDX) are the most reliable for preclinical drug development due to their ability to retain pathological and molecular characteristics. In this review, the authors discuss the various murine CRC models with a focus on their clinical relevance, benefits, and drawbacks. From all models discussed, murine CRC models will continue to be an important tool in advancing our understanding and treatment of this disease, but additional research is required to find a model that can correctly reflect the pathophysiology of CRC.
Collapse
Affiliation(s)
- Íris Neto
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (Í.N.); (J.R.)
| | - João Rocha
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (Í.N.); (J.R.)
| | - Maria Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (Í.N.); (J.R.)
| | - Catarina P. Reis
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (Í.N.); (J.R.)
- Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|
10
|
Wu Z, Wu Y, Liu Z, Song Y, Ge L, Du T, Liu Y, Liu L, Liu C, Ma L. L1CAM deployed perivascular tumor niche promotes vessel wall invasion of tumor thrombus and metastasis of renal cell carcinoma. Cell Death Discov 2023; 9:112. [PMID: 37015905 PMCID: PMC10073121 DOI: 10.1038/s41420-023-01410-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 04/06/2023] Open
Abstract
The survival of tumor cells in the bloodstream, and vasculature adhesion at metastatic sites are crucial for tumor metastasis. Perivascular invasion aids tumor cell self-renewal, survival, and formation of metastases by facilitating readily available oxygen, nutrients, and endothelial-derived paracrine factors. Renal cell carcinoma (RCC) is among the most prevalent tumors of the urinary system, and the formation of venous tumor thrombus (VTT) is a characteristic feature of RCC. We observed high expression of L1CAM in the VTT with vessel wall invasion. L1CAM promotes the adhesion, migration, and invasion ability of RCC and enhances metastasis by interacting with ITGA5, which elicits activation of signaling downstream of integrin α5β1. L1CAM promotes ADAM17 transcription to facilitate transmembrane ectodomain cleavage and release of soluble L1CAM. In response to soluble L1CAM, vascular endothelial cells release several cytokines and chemokines. Endothelial-derived CXCL5 and its receptor CXCR2 promote the migration and intravasation of RCC toward endothelial cells suggesting that crosstalk between endothelial cells and tumor cells has a direct guiding role in driving the metastatic spread of RCC. LICAM plays a crucial role in the invasive ability of RCC, and regulation of L1CAM expression may contribute therapeutically to preventing RCC progression.
Collapse
Affiliation(s)
- Zonglong Wu
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Yaqian Wu
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Zhuo Liu
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Yimeng Song
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Liyuan Ge
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Tan Du
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Yunchong Liu
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Li Liu
- School of Nursing, Beijing University of Chinese Medicine, Beijing, 100191, P.R. China
| | - Cheng Liu
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P.R. China.
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P.R. China.
| | - Lulin Ma
- Department of Urology, Peking University Third Hospital, Beijing, 100191, P.R. China.
| |
Collapse
|
11
|
Wu T, Wan J, Qu X, Xia K, Wang F, Zhang Z, Yang M, Wu X, Gao R, Yuan X, Fang L, Chen C, Yin L. Nodal promotes colorectal cancer survival and metastasis through regulating SCD1-mediated ferroptosis resistance. Cell Death Dis 2023; 14:229. [PMID: 37002201 PMCID: PMC10066180 DOI: 10.1038/s41419-023-05756-6] [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: 09/17/2022] [Revised: 03/11/2023] [Accepted: 03/17/2023] [Indexed: 04/03/2023]
Abstract
Re-expression of an embryonic morphogen, Nodal, has been seen in several types of malignant tumours. By far, studies about Nodal's role in colorectal cancer (CRC) remain limited. Ferroptosis is essential for CRC progression, which is caused by cellular redox imbalance and characterized by lipid peroxidation. Herein, we observed that Nodal enhanced CRC cell's proliferative rate, motility, invasiveness, and epithelial-mesenchymal transition (EMT) in vivo and in vitro. Notably, Nodal overexpression induced monounsaturated fatty acids synthesis and increased the lipid unsaturation level. Nodal knockdown resulted in increased CRC cell lipid peroxidation. Stearoyl-coenzyme A desaturase 1 (SCD1) inhibition at least partially abolished the resistance of Nodal-overexpressing cells to RSL3-induced ferroptosis. Mechanistically, SCD1 was transcriptionally up-regulated by Smad2/3 pathway activation in response to Nodal overexpression. Significant Nodal and SCD1 up-regulation were observed in CRC tissues and were associated with CRC metastasis and poor clinical outcomes. Furthermore, bovine serum albumin nanoparticles/si-Nodal nanocomplexes targeting Nodal had anti-tumour effects on CRC progression and metastasis. This research elucidated the role of Nodal in CRC development and revealed a potential gene-based therapeutic strategy targeting Nodal for improving CRC treatment.
Collapse
Affiliation(s)
- Tianqi Wu
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Jian Wan
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226000, China
| | - Xiao Qu
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Kai Xia
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Fangtao Wang
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Zichao Zhang
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Muqing Yang
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Xiaocai Wu
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Renyuan Gao
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Xiaoqi Yuan
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Lin Fang
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Chunqiu Chen
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Lu Yin
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| |
Collapse
|
12
|
Tramontano C, Martins JP, De Stefano L, Kemell M, Correia A, Terracciano M, Borbone N, Rea I, Santos HA. Microfluidic-Assisted Production of Gastro-Resistant Active-Targeted Diatomite Nanoparticles for the Local Release of Galunisertib in Metastatic Colorectal Cancer Cells. Adv Healthc Mater 2023; 12:e2202672. [PMID: 36459471 PMCID: PMC11468342 DOI: 10.1002/adhm.202202672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/17/2022] [Indexed: 12/03/2022]
Abstract
The oral route is highly desirable for colorectal cancer (CRC) treatment because it allows concentrating the drug in the colon and achieving a localized effect. However, orally administered drugs are often metabolized in the liver, resulting in reduced efficacy and the need for higher doses. Nanoparticle-based drug delivery systems can be engineered to prevent the diffusion of the drug in the stomach, addressing the release at the target site, and enhancing the efficacy of the delivered drug. Here, an orally administrable galunisertib delivery system is developed with gelatin-covered diatomite nanoparticles targeting the ligand 1-cell adhesion molecule (L1-CAM) on metastatic cells, and further encapsulated in an enteric matrix by microfluidics. The gastro-resistant polymer protects the nanoparticles from the action of the digestive enzymes and allows for a sustained release of galunisertib at the intestinal pH. The efficacy of antibody-antigen interactions to drive the internalization of nanoparticles in the targeted cells is investigated in CRC cells expressing abnormal (SW620) or basal levels (Caco-2, HT29-MTX) of L1-CAM. The combination of local drug release and active targeting enhances the effect of the delivered galunisertib, which inhibits the migration of the SW620 cells with greater efficiency compared to the free drug.
Collapse
Affiliation(s)
- Chiara Tramontano
- Institute of Applied Sciences and Intelligent SystemsUnit of NaplesNational Research CouncilNaples80131Italy
- Department of PharmacyUniversity of Naples Federico IINaples80131Italy
| | - João Pedro Martins
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
| | - Luca De Stefano
- Institute of Applied Sciences and Intelligent SystemsUnit of NaplesNational Research CouncilNaples80131Italy
| | - Marianna Kemell
- Department of ChemistryUniversity of HelsinkiHelsinkiFI‐00014Finland
| | - Alexandra Correia
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
| | | | - Nicola Borbone
- Department of PharmacyUniversity of Naples Federico IINaples80131Italy
| | - Ilaria Rea
- Institute of Applied Sciences and Intelligent SystemsUnit of NaplesNational Research CouncilNaples80131Italy
| | - Hélder A. Santos
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
- Department of Biomedical EngineeringUniversity Medical Center GroningenUniversity of GroningenGroningen9713 AVThe Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials ScienceUniversity Medical Center GroningenUniversity of GroningenGroningen9713 AVThe Netherlands
| |
Collapse
|
13
|
Dana P, Thumrongsiri N, Tanyapanyachon P, Chonniyom W, Punnakitikashem P, Saengkrit N. Resveratrol Loaded Liposomes Disrupt Cancer Associated Fibroblast Communications within the Tumor Microenvironment to Inhibit Colorectal Cancer Aggressiveness. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 13:107. [PMID: 36616017 PMCID: PMC9824711 DOI: 10.3390/nano13010107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 06/17/2023]
Abstract
Colorectal cancer (CRC) is a cancer-associated fibroblast, CAF-rich tumor. CAF promotes cancer cell proliferation, metastasis, drug resistance via secretes soluble factors, and extracellular matrices which leads to dense stroma, a major barrier for drug delivery. Resveratrol (RES) is a polyphenolic compound, has several pharmacologic functions including anti-inflammation and anticancer effects. Considering tumor microenvironment of CRC, resveratrol-loaded liposome (L-RES) was synthesized and employed to inhibit CAF functions. The L-RES was synthesized by thin-film hydration method. The cytotoxicity of L-RES was evaluated using MTT assay. Effect of L-RES treated CAF on tumor spheroid growth was performed. Cell invasion was determined using spheroid invasion assay. The effect of L-RES on 5-fluorouracil (5-FU) sensitivity of CRC cells was determined in co-cultured tumor spheroids. Subtoxic dose of L-RES was selected to study possible inhibiting CAF functions. Decreased CAF markers, α-SMA and IL-6 levels, were observed in L-RES treated activated fibroblast. Interestingly, the activated fibroblast promoted invasive ability and drug resistance of CRC cells in co-culture condition of both 2D and 3D cultures and was attenuated by L-RES treatment in the activated fibroblast. Therefore, L-RES provides a promising drug delivery strategy for CRC treatment by disrupting the crosstalk between CRC cells and CAF.
Collapse
Affiliation(s)
- Paweena Dana
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Nutthanit Thumrongsiri
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Prattana Tanyapanyachon
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Walailuk Chonniyom
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| | - Primana Punnakitikashem
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok 10700, Thailand
- Research Network NANOTEC-Mahidol University in Theranostic Nanomedicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok 10700, Thailand
| | - Nattika Saengkrit
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand
| |
Collapse
|
14
|
Wang X, Wu C, Zhang S, Yu P, Li L, Guo C, Li R. A novel deep learning segmentation model for organoid-based drug screening. Front Pharmacol 2022; 13:1080273. [PMID: 36588731 PMCID: PMC9794595 DOI: 10.3389/fphar.2022.1080273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
Organoids are self-organized three-dimensional in vitro cell cultures derived from stem cells. They can recapitulate organ development, tissue regeneration, and disease progression and, hence, have broad applications in drug discovery. However, the lack of effective graphic algorithms for organoid growth analysis has slowed the development of organoid-based drug screening. In this study, we take advantage of a bladder cancer organoid system and develop a deep learning model, the res-double dynamic conv attention U-Net (RDAU-Net) model, to improve the efficiency and accuracy of organoid-based drug screenings. In this RDAU-Net model, the dynamic convolution and attention modules are integrated. The feature-extracting capability of the encoder and the utilization of multi-scale information are substantially enhanced, and the semantic gap caused by skip connections has been filled, which substantially improved its anti-interference ability. A total of 200 images of bladder cancer organoids on culture days 1, 3, 5, and 7, with or without drug treatment, were employed for training and testing. Compared with the other variations of the U-Net model, the segmentation indicators, such as Intersection over Union and dice similarity coefficient, in the RDAU-Net model have been improved. In addition, this algorithm effectively prevented false identification and missing identification, while maintaining a smooth edge contour of segmentation results. In summary, we proposed a novel method based on a deep learning model which could significantly improve the efficiency and accuracy of high-throughput drug screening and evaluation using organoids.
Collapse
Affiliation(s)
- Xiaowen Wang
- School of Information, Yunnan University, Kunming, China
| | - Chunyue Wu
- School of Life Science, Yunnan University, Kunming, China
| | - Shudi Zhang
- School of Information, Yunnan University, Kunming, China
| | - Pengfei Yu
- School of Information, Yunnan University, Kunming, China
| | - Lu Li
- School of Life Science, Yunnan University, Kunming, China
| | - Chunming Guo
- School of Life Science, Yunnan University, Kunming, China
| | - Rui Li
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
15
|
Cave DD, Buonaiuto S, Sainz B, Fantuz M, Mangini M, Carrer A, Di Domenico A, Iavazzo TT, Andolfi G, Cortina C, Sevillano M, Heeschen C, Colonna V, Corona M, Cucciardi A, Di Guida M, Batlle E, De Luca A, Lonardo E. LAMC2 marks a tumor-initiating cell population with an aggressive signature in pancreatic cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:315. [PMID: 36289544 PMCID: PMC9609288 DOI: 10.1186/s13046-022-02516-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 10/09/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Tumor-initiating cells (TIC), also known as cancer stem cells, are considered a specific subpopulation of cells necessary for cancer initiation and metastasis; however, the mechanisms by which they acquire metastatic traits are not well understood. METHODS LAMC2 transcriptional levels were evaluated using publicly available transcriptome data sets, and LAMC2 immunohistochemistry was performed using a tissue microarray composed of PDAC and normal pancreas tissues. Silencing and tracing of LAMC2 was performed using lentiviral shRNA constructs and CRISPR/Cas9-mediated homologous recombination, respectively. The contribution of LAMC2 to PDAC tumorigenicity was explored in vitro by tumor cell invasion, migration, sphere-forming and organoids assays, and in vivo by tumor growth and metastatic assays. mRNA sequencing was performed to identify key cellular pathways upregulated in LAMC2 expressing cells. Metastatic spreading induced by LAMC2- expressing cells was blocked by pharmacological inhibition of transforming growth factor beta (TGF-β) signaling. RESULTS We report a LAMC2-expressing cell population, which is endowed with enhanced self-renewal capacity, and is sufficient for tumor initiation and differentiation, and drives metastasis. mRNA profiling of these cells indicates a prominent squamous signature, and differentially activated pathways critical for tumor growth and metastasis, including deregulation of the TGF-β signaling pathway. Treatment with Vactosertib, a new small molecule inhibitor of the TGF-β type I receptor (activin receptor-like kinase-5, ALK5), completely abrogated lung metastasis, primarily originating from LAMC2-expressing cells. CONCLUSIONS We have identified a highly metastatic subpopulation of TICs marked by LAMC2. Strategies aimed at targeting the LAMC2 population may be effective in reducing tumor aggressiveness in PDAC patients. Our results prompt further study of this TIC population in pancreatic cancer and exploration as a potential therapeutic target and/or biomarker.
Collapse
Affiliation(s)
- Donatella Delle Cave
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| | - Silvia Buonaiuto
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| | - Bruno Sainz
- grid.466793.90000 0004 1803 1972Department of Cancer Biology, Instituto de Investigaciones Biomedicas “Alberto Sols” (IIBM), CSIC-UAM, 28029 Madrid, Spain ,grid.420232.50000 0004 7643 3507Chronic Diseases and Cancer, Area 3-Instituto Ramon Y Cajal de Investigacion Sanitaria (IRYCIS), 28034 Madrid, Spain ,grid.510933.d0000 0004 8339 0058Centro de Investigación Biomédica en Red, Área Cáncer, CIBERONC, ISCIII, 28029 Madrid, Spain
| | - Marco Fantuz
- grid.5608.b0000 0004 1757 3470Department of Biology, University of Padova, 35129 Padova, Italy ,grid.428736.cVeneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Maria Mangini
- grid.5326.20000 0001 1940 4177Institute for Experimental Endocrinology and Oncology, “G. Salvatore” (IEOS), Second Unit, Consiglio Nazionale Delle Ricerche (CNR), 801310 Naples, Italy
| | - Alessandro Carrer
- grid.5608.b0000 0004 1757 3470Department of Biology, University of Padova, 35129 Padova, Italy ,grid.428736.cVeneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Annalisa Di Domenico
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| | - Tea Teresa Iavazzo
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| | - Gennaro Andolfi
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| | - Carme Cortina
- grid.7722.00000 0001 1811 6966Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain ,grid.510933.d0000 0004 8339 0058Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 08028 Barcelona, Spain
| | - Marta Sevillano
- grid.7722.00000 0001 1811 6966Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain ,grid.510933.d0000 0004 8339 0058Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 08028 Barcelona, Spain
| | - Christopher Heeschen
- grid.16821.3c0000 0004 0368 8293State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Vincenza Colonna
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| | - Marco Corona
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| | - Antonio Cucciardi
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| | - Martina Di Guida
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| | - Eduard Batlle
- grid.7722.00000 0001 1811 6966Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain ,grid.510933.d0000 0004 8339 0058Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 08028 Barcelona, Spain
| | - Annachiara De Luca
- grid.5326.20000 0001 1940 4177Institute for Experimental Endocrinology and Oncology, “G. Salvatore” (IEOS), Second Unit, Consiglio Nazionale Delle Ricerche (CNR), 801310 Naples, Italy
| | - Enza Lonardo
- grid.5326.20000 0001 1940 4177Institute of Genetics and Biophysics “A. Buzzati-Traverso”, National Research Council (CNR-IGB), 80131 Naples, Italy
| |
Collapse
|
16
|
Jiao L, Yang Q, Miao G, Wang Y, Yang Z, Liu X. Bone Marrow Mesenchymal Stem Cell (BMSC) Restrains the Angiogenesis in Melanoma Through Stromal-Derived-Factor-1/C-X-C Chemokine Receptor Type 4 (SDF-1/CXCR4). J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study analyzes the effect of BMSC on restraining the angiogenesis in melanoma through inducing SDF-1/CXCR4 channel. 50 female naked rates were equally assigned into NC group, model group, BMSC group, agonist group and positive NC group randomly followed by analysis of pathological
changes, and the level of HIF-1, VEG, MVD, SDF-1 and CXCR4. Agonist group showed the highest level of HIF-1α and VEGF and MVD followed by, model group BMSC group, positive NC group and NC group with no different between BMSC group and positive NC group. SDF-1 and CXCR4 expression
was highest in agonist group, followed by that in model group, positive NC group, BMSC group and NC group without difference between model group and positive NC group. In conclusion, SDF-1/CXCR4 activity could be restrained by BMSC partly along with reduced level of HIF-1α and
VEGF. This is mainly related with restraining the SDF-1/CXCR4 channel, indicating that it could be adopted as a brand-new therapeutic target for treating melanoma.
Collapse
Affiliation(s)
- Liyan Jiao
- Department of Dermatological, Affiliated Hospital of Hebei University of Engineering, Handan, Hebei, 056000, China
| | - Qingyan Yang
- Department of Neurology, Affiliated Hospital of Hebei University of Engineering, Handan, Hebei, 056000, China
| | - Guoying Miao
- Department of Dermatological, Affiliated Hospital of Hebei University of Engineering, Handan, Hebei, 056000, China
| | - Youming Wang
- Department of Neurology, Affiliated Hospital of Hebei University of Engineering, Handan, Hebei, 056000, China
| | - Zhitang Yang
- Department of Neurology, Affiliated Hospital of Hebei University of Engineering, Handan, Hebei, 056000, China
| | - Xiaojuan Liu
- Department of Dermatological, Affiliated Hospital of Hebei University of Engineering, Handan, Hebei, 056000, China
| |
Collapse
|
17
|
Montalbán-Hernández K, Cantero-Cid R, Casalvilla-Dueñas JC, Avendaño-Ortiz J, Marín E, Lozano-Rodríguez R, Terrón-Arcos V, Vicario-Bravo M, Marcano C, Saavedra-Ambrosy J, Prado-Montero J, Valentín J, Pérez de Diego R, Córdoba L, Pulido E, del Fresno C, Dueñas M, López-Collazo E. Colorectal Cancer Stem Cells Fuse with Monocytes to Form Tumour Hybrid Cells with the Ability to Migrate and Evade the Immune System. Cancers (Basel) 2022; 14:3445. [PMID: 35884505 PMCID: PMC9324286 DOI: 10.3390/cancers14143445] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/09/2022] [Accepted: 07/13/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The cancer cell fusion theory could be one of the best explanations for the metastasis from primary tumours. METHODS Herein, we co-cultured colorectal cancer (CRC) stem cells with human monocytes and analysed the properties of the generated tumour hybrid cells (THCs). The presence of THCs in the bloodstream together with samples from primary and metastatic lesions and their clinical correlations were evaluated in CRC patients and were detected by both FACS and immunofluorescence methods. Additionally, the role of SIGLEC5 as an immune evasion molecule in colorectal cancer was evaluated. RESULTS Our data demonstrated the generation of THCs after the in vitro co-culture of CRC stem cells and monocytes. These cells, defined as CD45+CD14+EpCAM+, showed enhanced migratory and proliferative abilities. The THC-specific cell surface signature allows identification in matched primary tumour tissues and metastases as well as in the bloodstream from patients with CRC, thus functioning as a biomarker. Moreover, SIG-LEC5 expression on in vitro generated THCs has shown to be involved in the mechanism for immune evasion. Additionally, sSIGLEC5 levels correlated with THC numbers in the prospective cohort of patients. CONCLUSIONS Our results indicate the generation of a hybrid entity after the in vitro co-culture between CRC stem cells and human monocytes. Moreover, THC numbers present in patients are related to both prognosis and the later spread of metastases in CRC patients.
Collapse
Affiliation(s)
- Karla Montalbán-Hernández
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain; (K.M.-H.); (R.C.-C.); (J.C.C.-D.); (J.A.-O.); (E.M.); (R.L.-R.); (V.T.-A.); (J.P.-M.); (J.V.); (R.P.d.D.); (L.C.); (E.P.); (C.d.F.)
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
| | - Ramón Cantero-Cid
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain; (K.M.-H.); (R.C.-C.); (J.C.C.-D.); (J.A.-O.); (E.M.); (R.L.-R.); (V.T.-A.); (J.P.-M.); (J.V.); (R.P.d.D.); (L.C.); (E.P.); (C.d.F.)
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
- Digestive Surgery Service, La Paz Univeristy Hospital, 28046 Madrid, Spain; (M.V.-B.); (C.M.); (J.S.-A.)
- Translational Research and Innovation in Surgery Group, La Paz Univeristy Hospital, 28046 Madrid, Spain
| | - José Carlos Casalvilla-Dueñas
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain; (K.M.-H.); (R.C.-C.); (J.C.C.-D.); (J.A.-O.); (E.M.); (R.L.-R.); (V.T.-A.); (J.P.-M.); (J.V.); (R.P.d.D.); (L.C.); (E.P.); (C.d.F.)
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
| | - José Avendaño-Ortiz
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain; (K.M.-H.); (R.C.-C.); (J.C.C.-D.); (J.A.-O.); (E.M.); (R.L.-R.); (V.T.-A.); (J.P.-M.); (J.V.); (R.P.d.D.); (L.C.); (E.P.); (C.d.F.)
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
- Biobank Platform, IdiPAZ, La Paz Universitary Hospital, 28046 Madrid, Spain
| | - Elvira Marín
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain; (K.M.-H.); (R.C.-C.); (J.C.C.-D.); (J.A.-O.); (E.M.); (R.L.-R.); (V.T.-A.); (J.P.-M.); (J.V.); (R.P.d.D.); (L.C.); (E.P.); (C.d.F.)
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
| | - Roberto Lozano-Rodríguez
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain; (K.M.-H.); (R.C.-C.); (J.C.C.-D.); (J.A.-O.); (E.M.); (R.L.-R.); (V.T.-A.); (J.P.-M.); (J.V.); (R.P.d.D.); (L.C.); (E.P.); (C.d.F.)
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
| | - Verónica Terrón-Arcos
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain; (K.M.-H.); (R.C.-C.); (J.C.C.-D.); (J.A.-O.); (E.M.); (R.L.-R.); (V.T.-A.); (J.P.-M.); (J.V.); (R.P.d.D.); (L.C.); (E.P.); (C.d.F.)
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
| | - Marina Vicario-Bravo
- Digestive Surgery Service, La Paz Univeristy Hospital, 28046 Madrid, Spain; (M.V.-B.); (C.M.); (J.S.-A.)
| | - Cristóbal Marcano
- Digestive Surgery Service, La Paz Univeristy Hospital, 28046 Madrid, Spain; (M.V.-B.); (C.M.); (J.S.-A.)
| | - Jorge Saavedra-Ambrosy
- Digestive Surgery Service, La Paz Univeristy Hospital, 28046 Madrid, Spain; (M.V.-B.); (C.M.); (J.S.-A.)
| | - Julia Prado-Montero
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain; (K.M.-H.); (R.C.-C.); (J.C.C.-D.); (J.A.-O.); (E.M.); (R.L.-R.); (V.T.-A.); (J.P.-M.); (J.V.); (R.P.d.D.); (L.C.); (E.P.); (C.d.F.)
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
| | - Jaime Valentín
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain; (K.M.-H.); (R.C.-C.); (J.C.C.-D.); (J.A.-O.); (E.M.); (R.L.-R.); (V.T.-A.); (J.P.-M.); (J.V.); (R.P.d.D.); (L.C.); (E.P.); (C.d.F.)
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
| | - Rebeca Pérez de Diego
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain; (K.M.-H.); (R.C.-C.); (J.C.C.-D.); (J.A.-O.); (E.M.); (R.L.-R.); (V.T.-A.); (J.P.-M.); (J.V.); (R.P.d.D.); (L.C.); (E.P.); (C.d.F.)
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
| | - Laura Córdoba
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain; (K.M.-H.); (R.C.-C.); (J.C.C.-D.); (J.A.-O.); (E.M.); (R.L.-R.); (V.T.-A.); (J.P.-M.); (J.V.); (R.P.d.D.); (L.C.); (E.P.); (C.d.F.)
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
- Biobank Platform, IdiPAZ, La Paz Universitary Hospital, 28046 Madrid, Spain
| | - Elisa Pulido
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain; (K.M.-H.); (R.C.-C.); (J.C.C.-D.); (J.A.-O.); (E.M.); (R.L.-R.); (V.T.-A.); (J.P.-M.); (J.V.); (R.P.d.D.); (L.C.); (E.P.); (C.d.F.)
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
| | - Carlos del Fresno
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain; (K.M.-H.); (R.C.-C.); (J.C.C.-D.); (J.A.-O.); (E.M.); (R.L.-R.); (V.T.-A.); (J.P.-M.); (J.V.); (R.P.d.D.); (L.C.); (E.P.); (C.d.F.)
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
| | - Marta Dueñas
- Molecular Oncology Unit, Biomedical Innovation Department, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain;
- Centre for Biomedical Research Network of Oncological Diseases (CIBERONC), 29029 Madrid, Spain
| | - Eduardo López-Collazo
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain; (K.M.-H.); (R.C.-C.); (J.C.C.-D.); (J.A.-O.); (E.M.); (R.L.-R.); (V.T.-A.); (J.P.-M.); (J.V.); (R.P.d.D.); (L.C.); (E.P.); (C.d.F.)
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
- Centre for Biomedical Research Network of Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| |
Collapse
|
18
|
Fan T, Kuang G, Long R, Han Y, Wang J. The overall process of metastasis: From initiation to a new tumor. Biochim Biophys Acta Rev Cancer 2022; 1877:188750. [PMID: 35728735 DOI: 10.1016/j.bbcan.2022.188750] [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: 02/06/2022] [Revised: 06/09/2022] [Accepted: 06/09/2022] [Indexed: 11/26/2022]
Abstract
Metastasis-a process that involves the migration of cells from the primary site to distant organs-is the leading cause of cancer-associated death. Improved technology and in-depth research on tumors have furthered our understanding of the various mechanisms involved in tumor metastasis. Metastasis is initiated by cancer cells of a specific phenotype, which migrate with the assistance of extracellular components and metastatic traits conferred via epigenetic regulation while modifying their behavior in response to the complex and dynamic human internal environment. In this review, we have summarized the general steps involved in tumor metastasis and their characteristics, incorporating recent studies and topical issues, including epithelial-mesenchymal transition, cancer stem cells, neutrophil extracellular traps, pre-metastatic niche, extracellular vesicles, and dormancy. Several feasible treatment directions have also been summarized. In addition, the correlation between cancer metastasis and lifestyle factors, such as obesity and circadian rhythm, has been illustrated.
Collapse
Affiliation(s)
- Tianyue Fan
- Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Guicheng Kuang
- Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Runmin Long
- Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Yunwei Han
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Jing Wang
- Department of Blood Transfusion, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China.
| |
Collapse
|
19
|
Alsayed RKME, Khan AQ, Ahmad F, Ansari AW, Alam MA, Buddenkotte J, Steinhoff M, Uddin S, Ahmad A. Epigenetic Regulation of CXCR4 Signaling in Cancer Pathogenesis and Progression. Semin Cancer Biol 2022; 86:697-708. [PMID: 35346802 DOI: 10.1016/j.semcancer.2022.03.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 02/08/2023]
Abstract
Signaling involving chemokine receptor CXCR4 and its ligand SDF-1/CXL12 has been investigated for many years for its possible role in cancer progression and pathogenesis. Evidence emerging from clinical studies in recent years has further established diagnostic as well as prognostic importance of CXCR4 signaling. CXCR4 and SDF-1 are routinely reported to be elevated in tumors, distant metastases, which correlates with poor survival of patients. These findings have kindled interest in the mechanisms that regulate CXCR4/SDF-1 expression. Of note, there is a particular interest in the epigenetic regulation of CXCR4 signaling that may be responsible for upregulated CXCR4 in primary as well as metastatic cancers. This review first lists the clinical evidence supporting CXCR4 signaling as putative cancer diagnostic and/or prognostic biomarker, followed by a discussion on reported epigenetic mechanisms that affect CXCR4 expression. These mechanisms include regulation by non-coding RNAs, such as, microRNAs, long non-coding RNAs and circular RNAs. Additionally, we also discuss the regulation of CXCR4 expression through methylation and acetylation. Better understanding and appreciation of epigenetic regulation of CXCR4 signaling can invariably lead to identification of novel therapeutic targets as well as therapies to regulate this oncogenic signaling.
Collapse
Affiliation(s)
- Reem Khaled M E Alsayed
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar
| | - Abdul Q Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar
| | - Fareed Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha, 3050, Qatar
| | - Abdul Wahid Ansari
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha, 3050, Qatar
| | - Majid Ali Alam
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha, 3050, Qatar
| | - Jorg Buddenkotte
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha, 3050, Qatar
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha, 3050, Qatar; Weill Cornell Medicine-Qatar, Medical School, Doha, 24144, Qatar; Dept. of Dermatology, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Laboratory Animal Research Center, Qatar University, Doha, 2713, Qatar
| | - Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha, 3050, Qatar.
| |
Collapse
|
20
|
The Small-Molecule Wnt Inhibitor ICG-001 Efficiently Inhibits Colorectal Cancer Stemness and Metastasis by Suppressing MEIS1 Expression. Int J Mol Sci 2021; 22:ijms222413413. [PMID: 34948208 PMCID: PMC8704261 DOI: 10.3390/ijms222413413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 02/07/2023] Open
Abstract
Recurrence and metastasis remain major obstacles in colorectal cancer (CRC) treatment. Recent studies suggest that a small subpopulation of cells with a self-renewal ability, called cancer stem-like cells (CSCs), promotes recurrence and metastasis in CRC. Unfortunately, no CSC inhibitor has been demonstrated to be more effective than existing chemotherapeutic drugs, resulting in a significant unmet need for effective CRC therapies. In this study, transcriptomic profiling of metastatic tumors from CRC patients revealed significant upregulation in the Wnt pathway and stemness genes. Thus, we examined the therapeutic effect of the small-molecule Wnt inhibitor ICG-001 on cancer stemness and metastasis. The ICG-001 treatment efficiently attenuated self-renewal activity and metastatic potential. Mechanistically, myeloid ecotropic viral insertion site 1 (MEIS1) was identified as a target gene of ICG-001 that is transcriptionally regulated by Wnt signaling. A series of functional analyses revealed that MEIS1 enhanced the CSC behavior and metastatic potential of the CRC cells. Collectively, our findings suggest that ICG-001 efficiently inhibits CRC stemness and metastasis by suppressing MEIS1 expression. These results provide a basis for the further clinical investigation of ICG-001 as a targeted therapy for CSCs, opening a new avenue for the development of novel Wnt inhibitors for the treatment of CRC metastasis.
Collapse
|
21
|
Giordano M, Decio A, Battistini C, Baronio M, Bianchi F, Villa A, Bertalot G, Freddi S, Lupia M, Jodice MG, Ubezio P, Colombo N, Giavazzi R, Cavallaro U. L1CAM promotes ovarian cancer stemness and tumor initiation via FGFR1/SRC/STAT3 signaling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:319. [PMID: 34645505 PMCID: PMC8513260 DOI: 10.1186/s13046-021-02117-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/26/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Cancer stem cells (CSC) have been implicated in tumor progression. In ovarian carcinoma (OC), CSC drive tumor formation, dissemination and recurrence, as well as drug resistance, thus contributing to the high death-to-incidence ratio of this disease. However, the molecular basis of such a pathogenic role of ovarian CSC (OCSC) has been elucidated only to a limited extent. In this context, the functional contribution of the L1 cell adhesion molecule (L1CAM) to OC stemness remains elusive. METHODS The expression of L1CAM was investigated in patient-derived OCSC. The genetic manipulation of L1CAM in OC cells provided gain and loss-of-function models that were then employed in cell biological assays as well as in vivo tumorigenesis experiments to assess the role of L1CAM in OC cell stemness and in OCSC-driven tumor initiation. We applied antibody-mediated neutralization to investigate L1CAM druggability. Biochemical approaches were then combined with functional in vitro assays to study the molecular mechanisms underlying the functional role of L1CAM in OCSC. RESULTS We report that L1CAM is upregulated in patient-derived OCSC. Functional studies showed that L1CAM promotes several stemness-related properties in OC cells, including sphere formation, tumor initiation and chemoresistance. These activities were repressed by an L1CAM-neutralizing antibody, pointing to L1CAM as a druggable target. Mechanistically, L1CAM interacted with and activated fibroblast growth factor receptor-1 (FGFR1), which in turn induced the SRC-mediated activation of STAT3. The inhibition of STAT3 prevented L1CAM-dependent OC stemness and tumor initiation. CONCLUSIONS Our study implicate L1CAM in the tumorigenic function of OCSC and point to the L1CAM/FGFR1/SRC/STAT3 signaling pathway as a novel driver of OC stemness. We also provide evidence that targeting this pathway can contribute to OC eradication.
Collapse
Affiliation(s)
- Marco Giordano
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy
| | - Alessandra Decio
- Laboratory of Tumor Metastasis Therapeutics, Mario Negri Institute for Pharmacological Research - IRCCS, Milan, Italy
| | - Chiara Battistini
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy
| | - Micol Baronio
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy
| | - Fabrizio Bianchi
- Cancer Biomarkers Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013, San Giovanni Rotondo, FG, Italy
| | - Alessandra Villa
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy.,Philochem AG, Otelfingen, Switzerland
| | - Giovanni Bertalot
- Department of Experimental Oncology, European Institute of Oncology IRCSS, Milan, Italy.,Division of Anatomical Pathology, Santa Chiara Hospital, Trento, Italy
| | - Stefano Freddi
- Department of Experimental Oncology, European Institute of Oncology IRCSS, Milan, Italy
| | - Michela Lupia
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy
| | - Maria Giovanna Jodice
- Department of Experimental Oncology, European Institute of Oncology IRCSS, Milan, Italy
| | - Paolo Ubezio
- Laboratory of Tumor Metastasis Therapeutics, Mario Negri Institute for Pharmacological Research - IRCCS, Milan, Italy
| | - Nicoletta Colombo
- Division of Gynecologic Oncology, European Institute of Oncology IRCSS, Milan, Italy.,University of Milan-Bicocca, Milan, Italy
| | - Raffaella Giavazzi
- Laboratory of Tumor Metastasis Therapeutics, Mario Negri Institute for Pharmacological Research - IRCCS, Milan, Italy
| | - Ugo Cavallaro
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy.
| |
Collapse
|
22
|
Stalin J, Imhof BA, Coquoz O, Jeitziner R, Hammel P, McKee TA, Jemelin S, Poittevin M, Pocard M, Matthes T, Kaci R, Delorenzi M, Rüegg C, Miljkovic-Licina M. Targeting OLFML3 in Colorectal Cancer Suppresses Tumor Growth and Angiogenesis, and Increases the Efficacy of Anti-PD1 Based Immunotherapy. Cancers (Basel) 2021; 13:cancers13184625. [PMID: 34572851 PMCID: PMC8464773 DOI: 10.3390/cancers13184625] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 12/24/2022] Open
Abstract
The role of the proangiogenic factor olfactomedin-like 3 (OLFML3) in cancer is unclear. To characterize OLFML3 expression in human cancer and its role during tumor development, we undertook tissue expression studies, gene expression analyses of patient tumor samples, in vivo studies in mouse cancer models, and in vitro coculture experiments. OLFML3 was expressed at high levels, mainly in blood vessels, in multiple human cancers. We focused on colorectal cancer (CRC), as elevated expression of OLFML3 mRNA correlated with shorter relapse-free survival, higher tumor grade, and angiogenic microsatellite stable consensus molecular subtype 4 (CMS4). Treatment of multiple in vivo tumor models with OLFML3-blocking antibodies and deletion of the Olfml3 gene from mice decreased lymphangiogenesis, pericyte coverage, and tumor growth. Antibody-mediated blockade of OLFML3 and deletion of host Olfml3 decreased the recruitment of tumor-promoting tumor-associated macrophages and increased infiltration of the tumor microenvironment by NKT cells. Importantly, targeting OLFML3 increased the antitumor efficacy of anti-PD-1 checkpoint inhibitor therapy. Taken together, the results demonstrate that OLFML3 is a promising candidate therapeutic target for CRC.
Collapse
Affiliation(s)
- Jimmy Stalin
- Department of Pathology and Immunology, University of Geneva Medical School, Rue Michel Servet 1, CH-1211 Geneva, Switzerland; (B.A.I.); (P.H.); (S.J.); (M.P.); (M.M.-L.)
- Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 18, PER17, CH-1700 Fribourg, Switzerland; (O.C.); (C.R.)
- Correspondence: ; Tel.: +41-26-300-8658
| | - Beat A. Imhof
- Department of Pathology and Immunology, University of Geneva Medical School, Rue Michel Servet 1, CH-1211 Geneva, Switzerland; (B.A.I.); (P.H.); (S.J.); (M.P.); (M.M.-L.)
- Medicity Research Laboratory, University of Turku, Tykistökatu 6A, 20520 Turku, Finland
| | - Oriana Coquoz
- Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 18, PER17, CH-1700 Fribourg, Switzerland; (O.C.); (C.R.)
| | - Rachel Jeitziner
- Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland; (R.J.); (M.D.)
| | - Philippe Hammel
- Department of Pathology and Immunology, University of Geneva Medical School, Rue Michel Servet 1, CH-1211 Geneva, Switzerland; (B.A.I.); (P.H.); (S.J.); (M.P.); (M.M.-L.)
| | - Thomas A. McKee
- Division of Clinical Pathology, Geneva University Hospital, Rue Michel Servet 1, CH-1211 Geneva, Switzerland;
| | - Stephane Jemelin
- Department of Pathology and Immunology, University of Geneva Medical School, Rue Michel Servet 1, CH-1211 Geneva, Switzerland; (B.A.I.); (P.H.); (S.J.); (M.P.); (M.M.-L.)
| | - Marine Poittevin
- Department of Pathology and Immunology, University of Geneva Medical School, Rue Michel Servet 1, CH-1211 Geneva, Switzerland; (B.A.I.); (P.H.); (S.J.); (M.P.); (M.M.-L.)
| | - Marc Pocard
- CAP Paris-Tech, Université de Paris Diderot, INSERM U1275, 49 Boulevard de la Chapelle, CEDEX 10, F-75475 Paris, France; (M.P.); (R.K.)
- Department of Oncologic and Digestive Surgery, AP-HP, Hôpital Lariboisière, 2 Rue Ambroise Paré, CEDEX 10, F-75475 Paris, France
| | - Thomas Matthes
- Department of Oncology, Hematology Service, Geneva University Hospital, Rue Michel Servet 1, CH-1211 Geneva, Switzerland;
- Department of Diagnostics, Clinical Pathology Service, Geneva University Hospital, Rue Michel Servet 1, CH-1211 Geneva, Switzerland
- Translational Research Centre in Oncohaematology, University of Geneva Medical School, Rue Michel Servet 1, CH-1211 Geneva, Switzerland
| | - Rachid Kaci
- CAP Paris-Tech, Université de Paris Diderot, INSERM U1275, 49 Boulevard de la Chapelle, CEDEX 10, F-75475 Paris, France; (M.P.); (R.K.)
- Department of Anatomopathology, AP-HP, Hôpital Lariboisière, 2 Rue Ambroise Paré, CEDEX 10, F-75475 Paris, France
| | - Mauro Delorenzi
- Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland; (R.J.); (M.D.)
- Department of Oncology, University Lausanne, CH-1011 Lausanne, Switzerland
| | - Curzio Rüegg
- Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 18, PER17, CH-1700 Fribourg, Switzerland; (O.C.); (C.R.)
| | - Marijana Miljkovic-Licina
- Department of Pathology and Immunology, University of Geneva Medical School, Rue Michel Servet 1, CH-1211 Geneva, Switzerland; (B.A.I.); (P.H.); (S.J.); (M.P.); (M.M.-L.)
- Department of Oncology, Hematology Service, Geneva University Hospital, Rue Michel Servet 1, CH-1211 Geneva, Switzerland;
- Department of Diagnostics, Clinical Pathology Service, Geneva University Hospital, Rue Michel Servet 1, CH-1211 Geneva, Switzerland
- Translational Research Centre in Oncohaematology, University of Geneva Medical School, Rue Michel Servet 1, CH-1211 Geneva, Switzerland
| |
Collapse
|
23
|
Arboretto P, Cillo M, Leonardi A. New Insights into Cancer Targeted Therapy: Nodal and Cripto-1 as Attractive Candidates. Int J Mol Sci 2021; 22:ijms22157838. [PMID: 34360603 PMCID: PMC8345935 DOI: 10.3390/ijms22157838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
The transforming growth factor beta (TGF-β) signaling is fundamental for correct embryonic development. However, alterations of this pathway have been correlated with oncogenesis, tumor progression and sustaining of cancer stem cells (CSCs). Cripto-1 (CR-1) and Nodal are two embryonic proteins involved in TGF-β signaling. Their expression is almost undetectable in terminally differentiated cells, but they are often re-expressed in tumor cells, especially in CSCs. Moreover, cancer cells that show high levels of CR-1 and/or Nodal display more aggressive phenotypes in vitro, while in vivo their expression correlates with a worse prognosis in several human cancers. The ability to target CSCs still represents an unmet medical need for the complete eradication of certain types of tumors. Given the prognostic role and the selective expression of CR-1 and Nodal on cancer cells, they represent archetypes for targeted therapy. The aim of this review is to clarify the role of CR-1 and Nodal in cancer stem populations and to summarize the current therapeutic strategy to target CSCs using monoclonal antibodies (mAbs) or other molecular tools to interfere with these two proteins.
Collapse
|
24
|
Metformin and Niclosamide Synergistically Suppress Wnt and YAP in APC-Mutated Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13143437. [PMID: 34298652 PMCID: PMC8308039 DOI: 10.3390/cancers13143437] [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: 05/04/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 01/09/2023] Open
Abstract
Simple Summary Hyperactivation of the canonical Wnt and inactivation of the Hippo pathway are well-known genetic backgrounds for familial adenomatosis polyposis (FAP) and colorectal cancer (CRC), although the reciprocal regulation between those pathways is not yet clear. In this study, we found that Axin2, a bona fide downstream target of canonical Wnt, activates the Hippo pathway in APC-mutated CRC, limiting the therapeutic potential of niclosamide on advanced CRC through the inactivation of the Hippo pathway. To overcome the limitation, we combined niclosamide with AMPK activator metformin to activate Hippo and found that this combination synergistically suppressed canonical Wnt and activated Hippo in APC-mutated CRC. Using patient-derived cancer organoid and an APC-MIN mice model, we found the combinatory approach to be effective for APC-mutated CRC. Our results provide not only the reciprocal link between Wnt and Hippo in APC-mutated CRC, but they also provide an effective therapeutic approach with clinically available drugs for FAP and CRC patients. Abstract The Wnt and Hippo pathways are tightly coordinated and understanding their reciprocal regulation may provide a novel therapeutic strategy for cancer. Anti-helminthic niclosamide is an effective inhibitor of Wnt and is now in a phase II trial for advanced colorectal cancer (CRC) patients. We found that Axin2, an authentic target gene of canonical Wnt, acts as aYAP phosphorylation activator in APC-mutated CRC. While niclosamide effectively suppresses Wnt, it also inhibits Hippo, limiting its therapeutic potential for CRC. To overcome this limitation, we utilized metformin, a clinically available AMPK activator. This combinatory approach not only suppresses canonical Wnt activity, but also inhibits YAP activity in CRC cancer cells and in patient-derived cancer organoid through the suppression of cancer stemness. Further, combinatory oral administration suppressed in vivo tumorigenesis and the cancer progression of APC-MIN mice models. Our observations provide not only a reciprocal link between Wnt and Hippo, but also clinically available novel therapeutics that are able to target Wnt and YAP in APC-mutated CRC.
Collapse
|