1
|
Desai SA, Patel VP, Bhosle KP, Nagare SD, Thombare KC. The tumor microenvironment: shaping cancer progression and treatment response. J Chemother 2025; 37:15-44. [PMID: 38179655 DOI: 10.1080/1120009x.2023.2300224] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
The tumor microenvironment (TME) plays a crucial role in cancer progression and treatment response. It comprises a complex network of stromal cells, immune cells, extracellular matrix, and blood vessels, all of which interact with cancer cells and influence tumor behaviour. This review article provides an in-depth examination of the TME, focusing on stromal cells, blood vessels, signaling molecules, and ECM, along with commonly available therapeutic compounds that target these components. Moreover, we explore the TME as a novel strategy for discovering new anti-tumor drugs. The dynamic and adaptive nature of the TME offers opportunities for targeting specific cellular interactions and signaling pathways. We discuss emerging approaches, such as combination therapies that simultaneously target cancer cells and modulate the TME. Finally, we address the challenges and future prospects in targeting the TME. Overcoming drug resistance, improving drug delivery, and identifying new therapeutic targets within the TME are among the challenges discussed. We also highlight the potential of personalized medicine and the integration of emerging technologies, such as immunotherapy and nanotechnology, in TME-targeted therapies. This comprehensive review provides insights into the TME and its therapeutic implications. Understanding the TME's complexity and targeting its components offer promising avenues for the development of novel anti-tumor therapies and improved patient outcomes.
Collapse
Affiliation(s)
- Sharav A Desai
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| | - Vipul P Patel
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| | - Kunal P Bhosle
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| | - Sandip D Nagare
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| | - Kirti C Thombare
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Kopargaon, India
| |
Collapse
|
2
|
Shibata S, Yamada K, Kon S. Carnosic acid inhibits integrin expression and prevents pulmonary metastasis of melanoma. Biosci Biotechnol Biochem 2025; 89:284-293. [PMID: 39577858 DOI: 10.1093/bbb/zbae177] [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: 09/02/2024] [Accepted: 11/08/2024] [Indexed: 11/24/2024]
Abstract
Carnosic acid is a naturally occurring, plant-derived polyphenolic abietane diterpene with antitumor properties. However, its underlying mechanisms are still unclear. Therefore, we investigated the effects of carnosic acid on lung metastasis in a murine melanoma model. C57BL/6 mice were intravenously injected with B16-BL6 cells, followed by carnosic acid treatment. Lung weights were recorded, and tumor cell colonies were counted at the end of the experiment. Integrin expression was evaluated using flow cytometry and cell adhesion assays. Lung weights were significantly lower in the carnosic acid group than in the control group, indicating the suppression of metastasis. Carnosic acid suppressed α4 integrin expression in B16-BL6 cells and inhibited α4 and α9 integrin-dependent cell adhesion. Thus, our data suggest that carnosic acid prevents lung metastasis, possibly by suppressing integrin expression. Our findings support the clinical application of carnosic acid as a potential natural antitumor agent, offering a complementary approach to conventional therapies.
Collapse
Affiliation(s)
- Sachi Shibata
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Soja, Okayama, Japan
| | - Kohei Yamada
- Department of Molecular Immunology, Faculty of Pharmaceutical Sciences, Fukuyama University, Fukuyama, Hiroshima, Japan
| | - Shigeyuki Kon
- Department of Molecular Immunology, Faculty of Pharmaceutical Sciences, Fukuyama University, Fukuyama, Hiroshima, Japan
| |
Collapse
|
3
|
Limonta P, Marchesi S, Giannitti G, Casati L, Fontana F. The biological function of extracellular vesicles in prostate cancer and their clinical application as diagnostic and prognostic biomarkers. Cancer Metastasis Rev 2024; 43:1611-1627. [PMID: 39316264 PMCID: PMC11554767 DOI: 10.1007/s10555-024-10210-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024]
Abstract
Prostate cancer (PCa) is one of the most commonly diagnosed malignancies and main causes of cancer-related deaths worldwide. It is characterized by high heterogeneity, ranging from slow-growing tumor to metastatic disease. Since both therapy selection and outcome strongly rely on appropriate patient stratification, it is crucial to differentiate benign from more aggressive conditions using new and improved diagnostic and prognostic biomarkers. Extracellular vesicles (EVs) are membrane-coated particles carrying a specific biological cargo composed of nucleic acids, proteins, and metabolites. Here, we provide an overview of the role of EVs in PCa, focusing on both their biological function and clinical value. Specifically, we summarize the oncogenic role of EVs in mediating the interactions with PCa microenvironment as well as the horizontal transfer of metastatic traits and drug resistance between PCa cells. Furthermore, we discuss the potential usage of EVs as innovative tools for PCa diagnosis and prognosis.
Collapse
Affiliation(s)
- Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Sara Marchesi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Gaia Giannitti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Lavinia Casati
- Department of Health Sciences, Università Degli Studi Di Milano, Milan, Italy
| | - Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy.
| |
Collapse
|
4
|
Piroozkhah M, Zabihi M, Jalali P, Salehi Z. Comprehensive Multi-Omics Analysis Reveals NPC2 and ITGAV Genes as Potential Prognostic Biomarkers in Gastrointestinal Cancers. Cancer Rep (Hoboken) 2024; 7:e70087. [PMID: 39690926 DOI: 10.1002/cnr2.70087] [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: 05/09/2024] [Revised: 10/23/2024] [Accepted: 12/03/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Gastrointestinal cancers (GICs) continue to dominate in terms of both incidence and mortality worldwide. Due to the absence of efficient and accurate prognostic biomarkers, the prognosis and treatment outcomes of many GICs are poor. Identifying biomarkers to predict individual clinical outcomes efficiently is a fundamental challenge in clinical oncology. Although several biomarkers have been continually discovered, their predictive accuracy is relatively modest, and their therapeutic use is restricted. In light of this, the discovery of reliable biomarkers for predicting prognosis and outcome in GIC is urgently required. MATERIALS AND METHODS We evaluated the Human Protein Atlas dataset and identified NPC Intracellular Cholesterol Transporter 2 (NPC2) and Integrin Subunit Alpha V (ITGAV) as probable poor predictive genes for these cancers. In addition, we used the GEPIA2, cBioPortal, UALCAN, LinkedOmics, STRING, Enrichr, TISDB, TIMER2.0, hTFTarget, miRTarBase, circBank, and drug-gene interaction database databases to conduct a comprehensive and systematic analysis of the NPC2 and ITGAV genes. RESULT Our results found high expression levels of NPC2 and ITGAV in most GICs. The aforementioned gene expressions were linked to several clinicopathological characteristics of GICs as well as poorer prognosis in LIHC and STAD. The most common alteration type of NPC2 was amplification, and for ITGAV was deep deletion. Significant promotor hypermethylation was also seen in NPC2 and ITGAV in PAAD and COAD, respectively. For the immunologic significance, NPC2 and ITGAV were positively correlated with the abundance of tumor-infiltrating lymphocytes and macrophages. Furthermore, various immunomodulators showed strong correlations with the expression of these genes. There were currently 10 small molecule drugs targeting ITGAV. CONCLUSION Consequently, our bioinformatics analysis showed that NPC2 and ITGAV might be used as potential biomarkers to determine the prognosis of various GICs and are also related to immune infiltration.
Collapse
Affiliation(s)
- Moein Piroozkhah
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Zabihi
- Institute of Biochemistry and Biophysics (IBB), Department of Bioinformatics, Laboratory of Complex Biological Systems and Bioinformatics (CBB), University of Tehran, Tehran, Iran
| | - Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Salehi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Fernandes DA. Comprehensive Review on Bubbles: Synthesis, Modification, Characterization and Biomedical Applications. Bioconjug Chem 2024; 35:1639-1686. [PMID: 39377727 DOI: 10.1021/acs.bioconjchem.4c00137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Accurate detection, treatment, and imaging of diseases are important for effective treatment outcomes in patients. In this regard, bubbles have gained much attention, due to their versatility. Bubbles usually 1 nm to 10 μm in size can be produced and loaded with a variety of lipids, polymers, proteins, and therapeutic and imaging agents. This review details the different production and loading methods for bubbles, for imaging and treatment of diseases/conditions such as cancer, tumor angiogenesis, thrombosis, and inflammation. Bubbles can also be used for perfusion measurements, important for diagnostic and therapeutic decision making in cardiac disease. The different factors important in the stability of bubbles and the different techniques for characterizing their physical and chemical properties are explained, for developing bubbles with advanced therapeutic and imaging features. Hence, the review provides important insights for researchers studying bubbles for biomedical applications.
Collapse
|
6
|
Bialkowska K, El Khalki L, Rana PS, Wang W, Lindner DJ, Parker Y, Languino LR, Altieri DC, Pluskota E, Sossey-Alaoui K, Plow EF. Role of Kindlin 2 in prostate cancer. Sci Rep 2024; 14:19809. [PMID: 39191802 PMCID: PMC11349918 DOI: 10.1038/s41598-024-70202-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024] Open
Abstract
Kindlin-2 is a cytoskeletal adapter protein that is present in many different cell types. By virtue of its interaction with multiple binding partners, Kindlin-2 intercalates into numerous signaling pathways and cytoskeletal nodes. A specific interaction of Kindlin-2 that is of paramount importance in many cellular responses is its direct binding to the cytoplasmic tails of integrins, an interaction that controls many of the adhesive, migratory and signaling responses mediated by members of the integrin family of cell-surface heterodimers. Kindlin-2 is highly expressed in many cancers and is particularly prominent in prostate cancer cells. CRISPR/cas9 was used as a primary approach to knockout expression of Kindlin-2 in both androgen-independent and dependent prostate cancer cell lines, and the effects of Kindlin-2 suppression on oncogenic properties of these prostate cancer cell lines was examined. Adhesion to extracellular matrix proteins was markedly blunted, consistent with the control of integrin function by Kindlin-2. Migration across matrices was also affected. Anchorage independent growth was markedly suppressed. These observations indicate that Kindlin-2 regulates hallmark features of prostate cancer cells. In androgen expressing cells, testosterone-stimulated adhesion was Kindlin-2-dependent. Furthermore, tumor growth of a prostate cancer cell line lacking Kindlin-2 and implanted into the prostate gland of immunocompromised mice was markedly blunted and was associated with suppression of angiogenesis in the developing tumor. These results establish a key role of Kindlin-2 in prostate cancer progression and suggest that Kindlin-2 represents an interesting therapeutic target for treatment of prostate cancer.
Collapse
Affiliation(s)
- Katarzyna Bialkowska
- Department of Cardiovascular Biology and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH, 44139, USA
| | - Lamyae El Khalki
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Priyanka S Rana
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Wei Wang
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Daniel J Lindner
- Case Comprehensive Cancer Center, Cleveland, OH, USA
- Translational Hematology and Oncology Research, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Yvonne Parker
- Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Lucia R Languino
- Department of Pharmacology, Physiology and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Dario C Altieri
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, USA
| | - Elzbieta Pluskota
- Department of Cardiovascular Biology and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH, 44139, USA
| | - Khalid Sossey-Alaoui
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, Cleveland, OH, USA.
- Division of Cancer Biology, MetroHealth Medical Center, Rammelkamp Center for Research, R457, 2500 MetroHealth Drive, Cleveland, OH, 44109, USA.
| | - Edward F Plow
- Department of Cardiovascular Biology and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH, 44139, USA.
| |
Collapse
|
7
|
Surman M, Wilczak M, Bzowska M, Tylko G, Przybyło M. The Proangiogenic Effects of Melanoma-Derived Ectosomes Are Mediated by αvβ5 Integrin Rather than αvβ3 Integrin. Cells 2024; 13:1336. [PMID: 39195226 PMCID: PMC11352487 DOI: 10.3390/cells13161336] [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: 06/28/2024] [Revised: 07/28/2024] [Accepted: 08/10/2024] [Indexed: 08/29/2024] Open
Abstract
Ectosomes are carriers of proangiogenic factors during cancer progression. This study investigated whether the proangiogenic effect exerted by melanoma-derived ectosomes on recipient endothelial cells is mediated by ectosomal αvβ3 and αvβ5 integrins. Ectosomes were isolated from the conditioned culture media of four melanoma cell lines and melanocytes. Changes in gene and protein expression of αvβ3 and αvβ5 integrins, as well as VEGF and TNF-α were assessed in ectosome-treated endothelial cells. To confirm the functional involvement of ectosomal integrins in functional tests (Alamar Blue, wound healing and tube formation assays), ectosomes were also pretreated with anti-integrin antibodies and integrin-blocking peptides echistatin and cilengitide. Melanoma-derived ectosomes induced changes in the expression of αvβ3 and αvβ5 integrins in recipient endothelial cells, leading to increased viability, migratory properties, and tube formation potential. The extent of proangiogenic stimulation varied depending on the types of cells releasing ectosomes and the recipient cells. The use of anti-integrin antibodies and integrin-blocking peptides revealed a more significant role for the αvβ5 integrin/VEGF than the αvβ3 integrin/TNF-α pathway in the interactions between ectosomes and endothelial cells. The study demonstrated the functional role of ectosomal αvβ3 and αvβ5 integrins. It also provided a baseline understanding of ectosome-mediated αvβ3 integrin/TNF-α and αvβ5 integrin/VEGF signaling in angiogenesis.
Collapse
Affiliation(s)
- Magdalena Surman
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland; (M.S.); (M.W.)
| | - Magdalena Wilczak
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland; (M.S.); (M.W.)
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348 Krakow, Poland
| | - Małgorzata Bzowska
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland;
| | - Grzegorz Tylko
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland;
| | - Małgorzata Przybyło
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland; (M.S.); (M.W.)
| |
Collapse
|
8
|
Shajari N, Baradaran B, Tohidkia MR, Nasiri H, Sepehri M, Setayesh S, Aghebati-Maleki L. Advancements in Melanoma Therapies: From Surgery to Immunotherapy. Curr Treat Options Oncol 2024; 25:1073-1088. [PMID: 39066854 DOI: 10.1007/s11864-024-01239-8] [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] [Accepted: 06/14/2024] [Indexed: 07/30/2024]
Abstract
OPINION STATEMENT Melanoma is defined as the most aggressive and deadly form of skin cancer. The treatment of melanoma depends on the disease stage, tumor location, and extent of its spread from its point of origin. Melanoma treatment has made significant advances, notably in the context of targeted and immunotherapies. Surgical resection is the main therapeutic option for earlystage melanoma, and it provides favourable outcomes. With disease metastasis, systemic treatments such as immunotherapy and targeted therapy become increasingly important. The identification of mutations that lead to melanoma has influenced treatment strategies. Targeted therapies focusing on these mutations offer improved response rates and fewer toxicities than conventional chemotherapy. Furthermore, developing immunotherapies, including checkpoint inhibitors and tumor-infiltrating lymphocyte (TIL) therapies, has demonstrated encouraging outcomes in effectively combating cancer cells. These therapeutic agents demonstrate superior effectiveness and a more tolerable side-effect profile, improving the quality of life for patients receiving treatment. The future of melanoma treatment may involve a multimodal approach consisting of a combination of surgery, targeted therapy, and immunotherapy adapted to each patient's profile. This approach may improve survival rates and health outcomes.
Collapse
Affiliation(s)
- Neda Shajari
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Tohidkia
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Nasiri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Sepehri
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Setayesh
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
9
|
Paulus J, Sewald N. Small molecule- and peptide-drug conjugates addressing integrins: A story of targeted cancer treatment. J Pept Sci 2024; 30:e3561. [PMID: 38382900 DOI: 10.1002/psc.3561] [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: 09/20/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 02/23/2024]
Abstract
Targeted cancer treatment should avoid side effects and damage to healthy cells commonly encountered during traditional chemotherapy. By combining small molecule or peptidic ligands as homing devices with cytotoxic drugs connected by a cleavable or non-cleavable linker in peptide-drug conjugates (PDCs) or small molecule-drug conjugates (SMDCs), cancer cells and tumours can be selectively targeted. The development of highly affine, selective peptides and small molecules in recent years has allowed PDCs and SMDCs to increasingly compete with antibody-drug conjugates (ADCs). Integrins represent an excellent target for conjugates because they are overexpressed by most cancer cells and because of the broad knowledge about native binding partners as well as the multitude of small-molecule and peptidic ligands that have been developed over the last 30 years. In particular, integrin αVβ3 has been addressed using a variety of different PDCs and SMDCs over the last two decades, following various strategies. This review summarises and describes integrin-addressing PDCs and SMDCs while highlighting points of great interest.
Collapse
Affiliation(s)
- Jannik Paulus
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Bielefeld, Germany
| | - Norbert Sewald
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Bielefeld, Germany
| |
Collapse
|
10
|
Guo X, Fu Y, Peng J, Fu Y, Dong S, Ding RB, Qi X, Bao J. Emerging anticancer potential and mechanisms of snake venom toxins: A review. Int J Biol Macromol 2024; 269:131990. [PMID: 38704067 DOI: 10.1016/j.ijbiomac.2024.131990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/13/2024] [Accepted: 04/28/2024] [Indexed: 05/06/2024]
Abstract
Animal-derived venom, like snake venom, has been proven to be valuable natural resources for the drug development. Previously, snake venom was mainly investigated in its pharmacological activities in regulating coagulation, vasodilation, and cardiovascular function, and several marketed cardiovascular drugs were successfully developed from snake venom. In recent years, snake venom fractions have been demonstrated with anticancer properties of inducing apoptotic and autophagic cell death, restraining proliferation, suppressing angiogenesis, inhibiting cell adhesion and migration, improving immunity, and so on. A number of active anticancer enzymes and peptides have been identified from snake venom toxins, such as L-amino acid oxidases (LAAOs), phospholipase A2 (PLA2), metalloproteinases (MPs), three-finger toxins (3FTxs), serine proteinases (SPs), disintegrins, C-type lectin-like proteins (CTLPs), cell-penetrating peptides, cysteine-rich secretory proteins (CRISPs). In this review, we focus on summarizing these snake venom-derived anticancer components on their anticancer activities and underlying mechanisms. We will also discuss their potential to be developed as anticancer drugs in the future.
Collapse
Affiliation(s)
- Xijun Guo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Yuanfeng Fu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Junbo Peng
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Ying Fu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Shuai Dong
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Ren-Bo Ding
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Xingzhu Qi
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China.
| | - Jiaolin Bao
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China.
| |
Collapse
|
11
|
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
|
12
|
Chambers C, Chitwood B, Smith CJ, Miao Y. Elevating theranostics: The emergence and promise of radiopharmaceutical cell-targeting heterodimers in human cancers. IRADIOLOGY 2024; 2:128-155. [PMID: 38708130 PMCID: PMC11067702 DOI: 10.1002/ird3.62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 01/30/2024] [Indexed: 05/07/2024]
Abstract
Optimal therapeutic and diagnostic efficacy is essential for healthcare's global mission of advancing oncologic drug development. Accurate diagnosis and detection are crucial prerequisites for effective risk stratification and personalized patient care in clinical oncology. A paradigm shift is emerging with the promise of multi-receptor-targeting compounds. While existing detection and staging methods have demonstrated some success, the traditional approach of monotherapy is being reevaluated to enhance therapeutic effectiveness. Heterodimeric site-specific agents are a versatile solution by targeting two distinct biomarkers with a single theranostic agent. This review describes the innovation of dual-targeting compounds, examining their design strategies, therapeutic implications, and the promising path they present for addressing complex diseases.
Collapse
Affiliation(s)
- Claudia Chambers
- Molecular Imaging and Theranostics Center, Columbia, Missouri, USA
- Research Division, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri, USA
- Department of Chemistry, University of Missouri, Columbia, Missouri, USA
| | - Broc Chitwood
- Molecular Imaging and Theranostics Center, Columbia, Missouri, USA
| | - Charles J. Smith
- Molecular Imaging and Theranostics Center, Columbia, Missouri, USA
- Research Division, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri, USA
- Department of Radiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- University of Missouri Research Reactor Center, University of Missouri, Columbia, Missouri, USA
| | - Yubin Miao
- Department of Radiology, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
13
|
Xu L, Barrett JG, Peng J, Li S, Messadi D, Hu S. ITGAV Promotes the Progression of Head and Neck Squamous Cell Carcinoma. Curr Oncol 2024; 31:1311-1322. [PMID: 38534932 PMCID: PMC10969037 DOI: 10.3390/curroncol31030099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/01/2024] [Accepted: 02/13/2024] [Indexed: 05/26/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) refers to the malignancy of squamous cells in the head and neck region. Ranked as the seventh most common cancer worldwide, HNSCC has a very low survival rate, highlighting the importance of finding therapeutic targets for the disease. Integrins are cell surface receptors that play a crucial role in mediating cellular interactions with the extracellular matrix (ECM). Within this protein family, Integrin αV (ITGAV) has received attention for its important functional role in cancer progression. In this study, we first demonstrated the upregulation of ITGAV expression in HNSCC, with higher ITGAV expression levels correlating with significantly lower overall survival, based on TCGA (the Cancer Genome Atlas) and GEO datasets. Subsequent in vitro analyses revealed an overexpression of ITGAV in highly invasive HNSCC cell lines UM1 and UMSCC-5 in comparison to low invasive HNSCC cell lines UM2 and UMSCC-6. In addition, knockdown of ITGAV significantly inhibited the migration, invasion, viability, and colony formation of HNSCC cells. In addition, chromatin immunoprecipitation (ChIP) assays indicated that SOX11 bound to the promoter of ITGAV gene, and SOX11 knockdown resulted in decreased ITGAV expression in HNSCC cells. In conclusion, our studies suggest that ITGAV promotes the progression of HNSCC cells and may be regulated by SOX11 in HNSCC cells.
Collapse
Affiliation(s)
- Lingyi Xu
- School of Dentistry, University of California, Los Angeles, CA 90095, USA; (L.X.); (J.G.B.); (J.P.); (D.M.)
| | - Jeremy G Barrett
- School of Dentistry, University of California, Los Angeles, CA 90095, USA; (L.X.); (J.G.B.); (J.P.); (D.M.)
| | - Jiayi Peng
- School of Dentistry, University of California, Los Angeles, CA 90095, USA; (L.X.); (J.G.B.); (J.P.); (D.M.)
| | - Suk Li
- School of Dentistry, University of California, Los Angeles, CA 90095, USA; (L.X.); (J.G.B.); (J.P.); (D.M.)
| | - Diana Messadi
- School of Dentistry, University of California, Los Angeles, CA 90095, USA; (L.X.); (J.G.B.); (J.P.); (D.M.)
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90024, USA
| | - Shen Hu
- School of Dentistry, University of California, Los Angeles, CA 90095, USA; (L.X.); (J.G.B.); (J.P.); (D.M.)
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90024, USA
| |
Collapse
|
14
|
Wendong Y, Jiali J, Qiaomei F, Yayun W, Xianze X, Zheng S, Wei H. Biomechanical forces and force-triggered drug delivery in tumor neovascularization. Biomed Pharmacother 2024; 171:116117. [PMID: 38171243 DOI: 10.1016/j.biopha.2023.116117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/25/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024] Open
Abstract
Tumor angiogenesis is one of the typical hallmarks of tumor occurrence and development, and tumor neovascularization also exhibits distinct characteristics from normal blood vessels. As the number of cells and matrix inside the tumor increases, the biomechanical force is enhanced, specifically manifested as solid stress, fluid stress, stiffness, and topology. This mechanical microenvironment also provides shelter for tumors and intensifies angiogenesis, providing oxygen and nutritional support for tumor progression. During tumor development, the biomechanical microenvironment also emerges, which in turn feeds back to regulate the tumor progression, including tumor angiogenesis, and biochemical and biomechanical signals can regulate tumor angiogenesis. Blood vessels possess inherent sensitivity to mechanical stimuli, but compared to the extensive research on biochemical signal regulation, the study of the regulation of tumor neovascularization by biomechanical signals remains relatively scarce. Biomechanical forces can affect the phenotypic characteristics and mechanical signaling pathways of tumor blood vessels, directly regulating angiogenesis. Meanwhile, they can indirectly regulate tumor angiogenesis by causing an imbalance in angiogenesis signals and affecting stromal cell function. Understanding the regulatory mechanism of biomechanical forces in tumor angiogenesis is beneficial for better identifying and even taming the mechanical forces involved in angiogenesis, providing new therapeutic targets for tumor vascular normalization. Therefore, we summarized the composition of biomechanical forces and their direct or indirect regulation of tumor neovascularization. In addition, this review discussed the use of biomechanical forces in combination with anti-angiogenic therapies for the treatment of tumors, and biomechanical forces triggered delivery systems.
Collapse
Affiliation(s)
- Yao Wendong
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Jiang Jiali
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Fan Qiaomei
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Weng Yayun
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Xie Xianze
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China
| | - Shi Zheng
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China.
| | - Huang Wei
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310005, China.
| |
Collapse
|
15
|
Carrera-Aguado I, Marcos-Zazo L, Carrancio-Salán P, Guerra-Paes E, Sánchez-Juanes F, Muñoz-Félix JM. The Inhibition of Vessel Co-Option as an Emerging Strategy for Cancer Therapy. Int J Mol Sci 2024; 25:921. [PMID: 38255995 PMCID: PMC10815934 DOI: 10.3390/ijms25020921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Vessel co-option (VCO) is a non-angiogenic mechanism of vascularization that has been associated to anti-angiogenic therapy. In VCO, cancer cells hijack the pre-existing blood vessels and use them to obtain oxygen and nutrients and invade adjacent tissue. Multiple primary tumors and metastases undergo VCO in highly vascularized tissues such as the lungs, liver or brain. VCO has been associated with a worse prognosis. The cellular and molecular mechanisms that undergo VCO are poorly understood. Recent studies have demonstrated that co-opted vessels show a quiescent phenotype in contrast to angiogenic tumor blood vessels. On the other hand, it is believed that during VCO, cancer cells are adhered to basement membrane from pre-existing blood vessels by using integrins, show enhanced motility and a mesenchymal phenotype. Other components of the tumor microenvironment (TME) such as extracellular matrix, immune cells or extracellular vesicles play important roles in vessel co-option maintenance. There are no strategies to inhibit VCO, and thus, to eliminate resistance to anti-angiogenic therapy. This review summarizes all the molecular mechanisms involved in vessel co-option analyzing the possible therapeutic strategies to inhibit this process.
Collapse
Affiliation(s)
- Iván Carrera-Aguado
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (I.C.-A.); (L.M.-Z.); (P.C.-S.); (E.G.-P.); (F.S.-J.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Laura Marcos-Zazo
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (I.C.-A.); (L.M.-Z.); (P.C.-S.); (E.G.-P.); (F.S.-J.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Patricia Carrancio-Salán
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (I.C.-A.); (L.M.-Z.); (P.C.-S.); (E.G.-P.); (F.S.-J.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Elena Guerra-Paes
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (I.C.-A.); (L.M.-Z.); (P.C.-S.); (E.G.-P.); (F.S.-J.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Fernando Sánchez-Juanes
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (I.C.-A.); (L.M.-Z.); (P.C.-S.); (E.G.-P.); (F.S.-J.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - José M. Muñoz-Félix
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (I.C.-A.); (L.M.-Z.); (P.C.-S.); (E.G.-P.); (F.S.-J.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
16
|
Thapa K, Khan H, Kaur G, Kumar P, Singh TG. Therapeutic targeting of angiopoietins in tumor angiogenesis and cancer development. Biochem Biophys Res Commun 2023; 687:149130. [PMID: 37944468 DOI: 10.1016/j.bbrc.2023.149130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 11/12/2023]
Abstract
The formation and progression of tumors in humans are linked to the abnormal development of new blood vessels known as neo-angiogenesis. Angiogenesis is a broad word that encompasses endothelial cell migration, proliferation, tube formation, and intussusception, as well as peri-EC recruitment and extracellular matrix formation. Tumor angiogenesis is regulated by angiogenic factors, out of which some of the most potent angiogenic factors such as vascular endothelial growth factor and Angiopoietins (ANGs) in the body are produced by macrophages and other immune cells within the tumor microenvironment. ANGs have a distinct function in tumor angiogenesis and behavior. ANG1, ANG 2, ANG 3, and ANG 4 are the family members of ANG out of which ANG2 has been extensively investigated owing to its unique role in modifying angiogenesis and its tight association with tumor progression, growth, and invasion/metastasis, which makes it an excellent candidate for therapeutic intervention in human malignancies. ANG modulators have demonstrated encouraging outcomes in the treatment of tumor development, either alone or in conjunction with VEGF inhibitors. Future development of more ANG modulators targeting other ANGs is needed. The implication of ANG1, ANG3, and ANG4 as probable therapeutic targets for anti-angiogenesis treatment in tumor development should be also evaluated. The article has described the role of ANG in tumor angiogenesis as well as tumor growth and the treatment strategies modulating ANGs in tumor angiogenesis as demonstrated in clinical studies. The pharmacological modulation of ANGs and ANG-regulated pathways that are responsible for tumor angiogenesis and cancer development should be evaluated for the development of future molecular therapies.
Collapse
Affiliation(s)
- Komal Thapa
- Chitkara School of Pharmacy, Chitkara University, 174103, Himachal Pradesh, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | - Gagandeep Kaur
- Chitkara School of Pharmacy, Chitkara University, 174103, Himachal Pradesh, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda, 151401, Bathinda, India
| | | |
Collapse
|
17
|
Agnihotri TG, Salave S, Shinde T, Srikanth I, Gyanani V, Haley JC, Jain A. Understanding the role of endothelial cells in brain tumor formation and metastasis: a proposition to be explored for better therapy. JOURNAL OF THE NATIONAL CANCER CENTER 2023; 3:222-235. [PMID: 39035200 PMCID: PMC11256543 DOI: 10.1016/j.jncc.2023.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 07/23/2024] Open
Abstract
Glioblastoma is one of the most devastating central nervous system disorders. Being a highly vascular brain tumor, it is distinguished by aberrant vessel architecture. This lends credence to the idea that endothelial cells (ECs) linked with glioblastoma vary fundamentally from ECs seen in the healthy human brain. To effectively design an antiangiogenic treatment, it is crucial to identify the functional and phenotypic characteristics of tumor-associated ECs. The ECs associated with glioblastoma are less prone to apoptosis than control cells and are resistant to cytotoxic treatments. Additionally, ECs associated with glioblastoma migrate more quickly than control ECs and naturally produce large amounts of growth factors such as endothelin-1, interleukin-8, and vascular endothelial growth factor (VEGF). For designing innovative antiangiogenic drugs that particularly target tumor-related ECs in gliomas, it is critical to comprehend these distinctive features of ECs associated with gliomas. This review discusses the process of angiogenesis, other factors involved in the genesis of tumors, and the possibility of ECs as a potential target in combating glioblastoma. It also sheds light on the association of tumor microenvironment and ECs with immunotherapy. This review, thus gives us the hope that neuro endothelial targeting with growth factors and angiogenesis regulators combined with gene therapy would open up new doorways and change our traditional perspective of treating cancer.
Collapse
Affiliation(s)
- Tejas Girish Agnihotri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, India
| | - Sagar Salave
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, India
| | - Tanuja Shinde
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, India
| | - Induri Srikanth
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, India
| | - Vijay Gyanani
- Long Acting Drug Delivery, Celanese Corporation, Irving, United States
| | - Jeffrey C. Haley
- Long Acting Drug Delivery, Celanese Corporation, Irving, United States
| | - Aakanchha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, India
| |
Collapse
|
18
|
Tsai CC, Yang YCSH, Chen YF, Huang LY, Yang YN, Lee SY, Wang WL, Lee HL, Whang-Peng J, Lin HY, Wang K. Integrins and Actions of Androgen in Breast Cancer. Cells 2023; 12:2126. [PMID: 37681860 PMCID: PMC10486718 DOI: 10.3390/cells12172126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/16/2023] [Accepted: 08/19/2023] [Indexed: 09/09/2023] Open
Abstract
Androgen has been shown to regulate male physiological activities and cancer proliferation. It is used to antagonize estrogen-induced proliferative effects in breast cancer cells. However, evidence indicates that androgen can stimulate cancer cell growth in estrogen receptor (ER)-positive and ER-negative breast cancer cells via different types of receptors and different mechanisms. Androgen-induced cancer growth and metastasis link with different types of integrins. Integrin αvβ3 is predominantly expressed and activated in cancer cells and rapidly dividing endothelial cells. Programmed death-ligand 1 (PD-L1) also plays a vital role in cancer growth. The part of integrins in action with androgen in cancer cells is not fully mechanically understood. To clarify the interactions between androgen and integrin αvβ3, we carried out molecular modeling to explain the potential interactions of androgen with integrin αvβ3. The androgen-regulated mechanisms on PD-L1 and its effects were also addressed.
Collapse
Affiliation(s)
- Chung-Che Tsai
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (C.-C.T.); (Y.-F.C.)
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Chen S. H. Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei 11031, Taiwan;
| | - Yi-Fong Chen
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (C.-C.T.); (Y.-F.C.)
| | - Lin-Yi Huang
- Department of Pediatrics, E-DA Hospital, I-Shou University, Kaohsiung 82445, Taiwan; (L.-Y.H.); (Y.-N.Y.)
| | - Yung-Ning Yang
- Department of Pediatrics, E-DA Hospital, I-Shou University, Kaohsiung 82445, Taiwan; (L.-Y.H.); (Y.-N.Y.)
- School of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Sheng-Yang Lee
- Dentistry, Wan-Fang Medical Center, Taipei Medical University, Taipei 11031, Taiwan;
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Wen-Long Wang
- Department of Life Science, Fu Jen Catholic University, New Taipei City 242, Taiwan;
| | - Hsin-Lun Lee
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110, Taiwan
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan
| | | | - Hung-Yun Lin
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (C.-C.T.); (Y.-F.C.)
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan;
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA
| | - Kuan Wang
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
19
|
Yin L, Li X, Wang R, Zeng Y, Zeng Z, Xie T. Recent Research Progress of RGD Peptide–Modified Nanodrug Delivery Systems in Tumor Therapy. Int J Pept Res Ther 2023; 29:53. [DOI: 10.1007/s10989-023-10523-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2023] [Indexed: 01/06/2025]
Abstract
AbstractThere have been great advancements in targeted nanodrug delivery systems for tumor therapy. Liposomes, polymeric nanoparticles, and inorganic nanoparticles are commonly employed as nanocarriers for drug delivery, and it has been found that arginine glycine aspartic acid (RGD) peptides and their derivatives can be used as ligands of integrin receptors to enhance the direct targeting ability. In this paper, we review the recent applications of RGD-modified liposomes, polymeric nanoparticles, and inorganic nanocarriers in cancer diagnosis and treatment, discuss the current challenges and prospects, and examine the progress made by the latest research on RGD peptide–modified nano delivery systems in cancer therapy. In recent years, RGD peptide–modified nanodrug delivery systems have been proven to have great potential in tumor therapy. Finally, we provide an overview of the current limitations and future directions of RGD peptide–modified nano-drug delivery systems for cancer therapy. This review aims to elucidate the contribution of RGD peptide–modified nanodrug delivery systems in the field of tumor therapy.
Collapse
|
20
|
Dzobo K, Dandara C. The Extracellular Matrix: Its Composition, Function, Remodeling, and Role in Tumorigenesis. Biomimetics (Basel) 2023; 8:146. [PMID: 37092398 PMCID: PMC10123695 DOI: 10.3390/biomimetics8020146] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/25/2023] Open
Abstract
The extracellular matrix (ECM) is a ubiquitous member of the body and is key to the maintenance of tissue and organ integrity. Initially thought to be a bystander in many cellular processes, the extracellular matrix has been shown to have diverse components that regulate and activate many cellular processes and ultimately influence cell phenotype. Importantly, the ECM's composition, architecture, and stiffness/elasticity influence cellular phenotypes. Under normal conditions and during development, the synthesized ECM constantly undergoes degradation and remodeling processes via the action of matrix proteases that maintain tissue homeostasis. In many pathological conditions including fibrosis and cancer, ECM synthesis, remodeling, and degradation is dysregulated, causing its integrity to be altered. Both physical and chemical cues from the ECM are sensed via receptors including integrins and play key roles in driving cellular proliferation and differentiation and in the progression of various diseases such as cancers. Advances in 'omics' technologies have seen an increase in studies focusing on bidirectional cell-matrix interactions, and here, we highlight the emerging knowledge on the role played by the ECM during normal development and in pathological conditions. This review summarizes current ECM-targeted therapies that can modify ECM tumors to overcome drug resistance and better cancer treatment.
Collapse
Affiliation(s)
- Kevin Dzobo
- Medical Research Council, SA Wound Healing Unit, Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
| | - Collet Dandara
- Division of Human Genetics and Institute of Infectious Disease and Molecular Medicine, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
- The South African Medical Research Council-UCT Platform for Pharmacogenomics Research and Translation, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
| |
Collapse
|
21
|
Navarro-Becerra JA, Castillo JI, Di Ruzza F, Borden MA. Monodispersity Increases Adhesion Efficiency and Specificity for Ultrasound-Targeted Microbubbles. ACS Biomater Sci Eng 2023; 9:991-1001. [PMID: 36153974 DOI: 10.1021/acsbiomaterials.2c00528] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ultrasound molecular imaging with targeted microbubbles (MBs) can be used to noninvasively diagnose, monitor, and study the progression of different endothelial-associated diseases. Acoustic radiation force (Frad) can initiate and enhance MB adhesion at the target site. The goal of this study was to elucidate the effects of various MB parameters on Frad targeting. Monodisperse or polydisperse MBs with the immune-stealth cloaked (buried)-ligand architecture were conjugated with targeting RGD or nonspecific isotype control RAD peptides and then pumped through an αvβ3 integrin-coated microvessel phantom at a wall shear stress of 3.5 dyn/cm2. Targeting was assessed by measuring MB attachment for varying Frad time and frequency, as well as MB concentration and size distribution. We first confirmed that primary Frad is necessary to target the cloaked-ligand MBs. MB targeting increased monotonically with αvβ3 integrin density and Frad time. MB attachment and, to a lesser extent specificity, also increased when driven by Frad near resonance. MB targeting increased with MB concentration, although a shift in behavior was observed with increasing MB-MB interactions and aggregations forming from secondary Frad effects as MB concentration was increased. These secondary Frad effects reduced targeting specificity. Finally, after having validated our approach by testing different parameters with the appropriate controls, we then determined the effects of monodispersity on adhesion efficiency and specific targeting. We observed that both MB targeting efficiency and specificity were greatly enhanced for monodisperse vs polydisperse MBs. Analysis of videomicroscopy images indicated that secondary Frad effects may have disproportionally inhibited targeting of polydisperse MBs. In conclusion, our in vitro results indicate that monodisperse MBs driven near resonance and at a low concentration (∼106 MB/mL) can be used to maximize the adhesion efficiency (up to 88%) and specificity of RGD-MB targeting.
Collapse
Affiliation(s)
- J Angel Navarro-Becerra
- Mechanical Engineering Department, University of Colorado Boulder, Boulder, Colorado 80309-0427, United States
| | - Jair I Castillo
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, Colorado 80309-0427, United States
| | - Federico Di Ruzza
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, Colorado 80309-0427, United States.,Chemical Science and Technology Department, University of Rome Tor Vergata, Roma 00133, Italy
| | - Mark A Borden
- Mechanical Engineering Department, University of Colorado Boulder, Boulder, Colorado 80309-0427, United States.,Biomedical Engineering Program, University of Colorado Boulder, Boulder, Colorado 80309-0427, United States
| |
Collapse
|
22
|
Mana G, Valdembri D, Askari JA, Li Z, Caswell P, Zhu C, Humphries MJ, Ballestrem C, Serini G. The βI domain promotes active β1 integrin clustering into mature adhesion sites. Life Sci Alliance 2023; 6:e202201388. [PMID: 36410791 PMCID: PMC9679427 DOI: 10.26508/lsa.202201388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/22/2022] Open
Abstract
Modulation of integrin function is required in many physiological and pathological settings, such as angiogenesis and cancer. Integrin allosteric changes, clustering, and trafficking cooperate to regulate cell adhesion and motility on extracellular matrix proteins via mechanisms that are partly defined. By exploiting four monoclonal antibodies recognizing distinct conformational epitopes, we show that in endothelial cells (ECs), the extracellular βI domain, but not the hybrid or I-EGF2 domain of active β1 integrins, promotes their FAK-regulated clustering into tensin 1-containing fibrillar adhesions and impairs their endocytosis. In this regard, the βI domain-dependent clustering of active β1 integrins is necessary to favor fibronectin-elicited directional EC motility, which cannot be effectively promoted by β1 integrin conformational activation alone.
Collapse
Affiliation(s)
- Giulia Mana
- Candiolo Cancer Institute - FPO, IRCCS, Candiolo (TO), Italy
- Department of Oncology, University of Torino School of Medicine, Candiolo (TO), Italy
| | - Donatella Valdembri
- Candiolo Cancer Institute - FPO, IRCCS, Candiolo (TO), Italy
- Department of Oncology, University of Torino School of Medicine, Candiolo (TO), Italy
| | - Janet A Askari
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Zhenhai Li
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Patrick Caswell
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Cheng Zhu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Martin J Humphries
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Christoph Ballestrem
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Guido Serini
- Candiolo Cancer Institute - FPO, IRCCS, Candiolo (TO), Italy
- Department of Oncology, University of Torino School of Medicine, Candiolo (TO), Italy
| |
Collapse
|
23
|
Shi Q, Ji T, Tang X, Guo W. The role of tumor-platelet interplay and micro tumor thrombi during hematogenous tumor metastasis. Cell Oncol (Dordr) 2023; 46:521-532. [PMID: 36652166 DOI: 10.1007/s13402-023-00773-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND In addition to their pivotal roles in coagulation and thrombosis, platelets are crucial in tumor progression, with plenty of clinical and experimental data demonstrating that the interplay of platelets and tumor cells is essential for hematogenous tumor metastasis. After detach from primary sites, tumor cells intravasate into the blood circulation becoming circulating tumor cells and induce platelet activation, aggregation and encasement around tumor cells to form micro tumor thrombi, which create a permissive tumor microenvironment for metastasis. Platelets in micro tumor thrombi protect tumor cells from immune surveillance and anoikis (detachment-triggered apoptosis) through various pathways, which are significant for tumor cell survival in the bloodstream. Moreover, platelets can facilitate tumor metastasis by expediting epithelial-mesenchymal transition (EMT), adhesion to the endothelium, angiogenesis, tumor proliferation processes and platelet-derived microvesicle (PMV) formation. CONCLUSIONS Here, we provide a synopsis of the current understanding of the formation of micro tumor thrombi and the role of micro tumor thrombi in tumor hematogenous metastasis based on the tumor-platelet interplay. We also highlight potential therapeutic strategies targeting platelets for tumor treatment, including cancer-associated platelet-targeted nanomedicines.
Collapse
Affiliation(s)
- Qianyu Shi
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, 100044, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Tao Ji
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, 100044, Beijing, China.
- Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China.
| | - Xiaodong Tang
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, 100044, Beijing, China
| | - Wei Guo
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, 100044, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| |
Collapse
|
24
|
Moreira HR, Rodrigues DB, Freitas-Ribeiro S, da Silva LP, Morais ADS, Jarnalo M, Horta R, Reis RL, Pirraco RP, Marques AP. Spongy-like hydrogels prevascularization with the adipose tissue vascular fraction delays cutaneous wound healing by sustaining inflammatory cell influx. Mater Today Bio 2022; 17:100496. [PMID: 36420053 PMCID: PMC9677215 DOI: 10.1016/j.mtbio.2022.100496] [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: 07/28/2022] [Revised: 11/03/2022] [Accepted: 11/13/2022] [Indexed: 11/16/2022] Open
Abstract
In vitro prevascularization is one of the most explored approaches to foster engineered tissue vascularization. We previously demonstrated a benefit in tissue neovascularization by using integrin-specific biomaterials prevascularized by stromal vascular fraction (SVF) cells, which triggered vasculogenesis in the absence of extrinsic growth factors. SVF cells are also associated to biological processes important in cutaneous wound healing. Thus, we aimed to investigate whether in vitro construct prevascularization with SVF accelerates the healing cascade by fostering early vascularization vis-à-vis SVF seeding prior to implantation. Prevascularized constructs delayed re-epithelization of full-thickness mice wounds compared to both non-prevascularized and control (no SVF) groups. Our results suggest this delay is due to a persistent inflammation as indicated by a significantly lower M2(CD163+)/M1(CD86+) macrophage subtype ratio. Moreover, a slower transition from the inflammatory to the proliferative phase of the healing was confirmed by reduced extracellular matrix deposition and increased presence of thick collagen fibers from early time-points, suggesting the prevalence of fiber crosslinking in relation to neodeposition. Overall, while prevascularization potentiates inflammatory cell influx, which negatively impacts the cutaneous wound healing cascade, an effective wound healing was guaranteed in non-prevascularized SVF cell-containing spongy-like hydrogels confirming that the SVF can have enhanced efficacy.
Collapse
Affiliation(s)
- Helena R. Moreira
- 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 – Zona Industrial da Gandra, Guimaraes, 4805-017, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga/Guimaraes, 4805-017, Portugal
| | - 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 – Zona Industrial da Gandra, Guimaraes, 4805-017, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga/Guimaraes, 4805-017, Portugal
| | - Sara Freitas-Ribeiro
- 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 – Zona Industrial da Gandra, Guimaraes, 4805-017, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga/Guimaraes, 4805-017, Portugal
| | - Lucília P. da Silva
- 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 – Zona Industrial da Gandra, Guimaraes, 4805-017, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga/Guimaraes, 4805-017, Portugal
| | - Alain da S. Morais
- 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 – Zona Industrial da Gandra, Guimaraes, 4805-017, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga/Guimaraes, 4805-017, Portugal
| | - Mariana Jarnalo
- Department of Plastic and Reconstructive Surgery, And Burn Unity, Centro Hospitalar de São João, Porto, Portugal
- Faculty of Medicine - University of Porto, Portugal
| | - Ricardo Horta
- Department of Plastic and Reconstructive Surgery, And Burn Unity, Centro Hospitalar de São João, Porto, Portugal
- Faculty of Medicine - University of Porto, 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 – Zona Industrial da Gandra, Guimaraes, 4805-017, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga/Guimaraes, 4805-017, 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 – Zona Industrial da Gandra, Guimaraes, 4805-017, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga/Guimaraes, 4805-017, Portugal
| | - Alexandra P. Marques
- 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 – Zona Industrial da Gandra, Guimaraes, 4805-017, Portugal
- ICVS/3B's – PT Government Associate Laboratory, Braga/Guimaraes, 4805-017, Portugal
| |
Collapse
|
25
|
Cationic RGD peptidomimetic nanoconjugates as effective tumor targeting gene delivery vectors with antimicrobial potential. Bioorg Chem 2022; 129:106197. [DOI: 10.1016/j.bioorg.2022.106197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/23/2022] [Accepted: 10/04/2022] [Indexed: 11/22/2022]
|
26
|
Ethanolic Extract of Ocimum sanctum Linn. Inhibits Cell Migration of Human Lung Adenocarcinoma Cells (A549) by Downregulation of Integrin αvβ3, α5β1, and VEGF. Sci Pharm 2022. [DOI: 10.3390/scipharm90040069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Adenocarcinoma lung cancer is a type of non-small cell lung carcinoma (NSCLC), which accounts for 85% of lung cancer incidence globally. The therapies that are being applied, both conventional therapies and antibody-based treatments, are still found to have side effects. Several previous studies have demonstrated the ability of the ethanolic extract of Ocimum sanctum Linn. (EEOS) as an ethnomedicine with anti-tumor properties. The aim of this study was to determine the effect of Ocimum sanctum Linn. ethanolic extract in inhibiting the proliferation, angiogenesis, and migration of A549 cells (NSCLC). The adhesion as well as the migration assay was performed. Furthermore, enzyme-linked immunosorbent assay (ELISA) was used to measure the expression of αvβ3 integrins, α5β1 integrins, and VEGF. The cells were divided into the following treatment groups: control (non-treated/NT), positive control (AP3/inhibitor β3 80 µg/mL), cisplatin (9 µg/mL), and EEOS at concentrations of 50, 70, 100, and 200 µg/mL. The results showed that EEOS inhibits the adhesion ability and migration of A549 cells, with an optimal concentration of 200 µg/mL. ELISA testing showed that the group of A549 cells given EEOS 200 µg/mL presented a decrease in the optimal expression of integrin α5β1, integrin αvβ3, and VEGF.
Collapse
|
27
|
Wen X, Chen S, Chen X, Qiu H, Wang W, Zhang N, Liu W, Wang T, Ding X, Zhang L. ITGB5 promotes innate radiation resistance in pancreatic adenocarcinoma by promoting DNA damage repair and the MEK/ERK signaling pathway. Front Oncol 2022; 12:887068. [PMID: 36249018 PMCID: PMC9563233 DOI: 10.3389/fonc.2022.887068] [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: 03/14/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is one of the most aggressive digestive system tumors in the world, with a low early diagnosis rate and a high mortality. Integrin beta 5 (ITGB5) is demonstrated to be a potent tumor promoter in several carcinomas. However, it is unknown whether ITGB5 participates in the occurrence and development of PAAD. In this study, we confirmed a high expression of ITGB5 in PAAD and its role in promoting invasiveness and transitivity in PAAD. Besides, the knockdown of ITGB5 increased cell sensitivity to radiation by promoting DNA damage repair and the MEK/ERK signaling pathway. Collectively, these results show that ITGB5 plays an essential role in pancreatic cancer growth and survival.
Collapse
Affiliation(s)
- Xin Wen
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Cancer Institute of Xuzhou Medical University, Xuzhou, China
| | - Si Chen
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Department of Radiation Oncology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xueting Chen
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hui Qiu
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wei Wang
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Nie Zhang
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wanming Liu
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Tingting Wang
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xin Ding
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Longzhen Zhang
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Cancer Institute of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou, China
| |
Collapse
|
28
|
Integrin-specific hydrogels for growth factor-free vasculogenesis. NPJ Regen Med 2022; 7:57. [PMID: 36167724 PMCID: PMC9515164 DOI: 10.1038/s41536-022-00253-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022] Open
Abstract
Integrin-binding biomaterials have been extensively evaluated for their capacity to enable de novo formation of capillary-like structures/vessels, ultimately supporting neovascularization in vivo. Yet, the role of integrins as vascular initiators in engineered materials is still not well understood. Here, we show that αvβ3 integrin-specific 3D matrices were able to retain PECAM1+ cells from the stromal vascular fraction (SVF) of adipose tissue, triggering vasculogenesis in vitro in the absence of extrinsic growth factors. Our results suggest that αvβ3-RGD-driven signaling in the formation of capillary-like structures prevents the activation of the caspase 8 pathway and activates the FAK/paxillin pathway, both responsible for endothelial cells (ECs) survival and migration. We also show that prevascularized αvβ3 integrin-specific constructs inosculate with the host vascular system fostering in vivo neovascularization. Overall, this work demonstrates the ability of the biomaterial to trigger vasculogenesis in an integrin-specific manner, by activating essential pathways for EC survival and migration within a self-regulatory growth factor microenvironment. This strategy represents an improvement to current vascularization routes for Tissue Engineering constructs, potentially enhancing their clinical applicability.
Collapse
|
29
|
Attenuated Salmonella Typhimurium with truncated LPS and outer membrane-displayed RGD peptide for cancer therapy. Biomed Pharmacother 2022; 155:113682. [PMID: 36095964 DOI: 10.1016/j.biopha.2022.113682] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/01/2022] [Accepted: 09/07/2022] [Indexed: 11/21/2022] Open
Abstract
Gram-negative, facultatively anaerobic bacteria Salmonella Typhimurium is a candidate agent or delivery vector for cancer therapy. Effective targeted therapies in addition to radiotherapy, chemotherapy and surgery have been urgently needed as an alternative or supplement. This study expected to further improve the tumor-targeting ability of Salmonella bacteria through genetic modifications. Based on an auxotrophic Salmonella bacterial strain (D2), we constructed Salmonella mutants with altered LPS length to facilitate displaying the RGD4C targeting peptide on the outer membrane surface of Salmonella. The expression of RGD4C peptide in fusion with OmpA was identified by outer membrane protein extraction and WB detection in different mutant strains. However, flow cytometry analysis following immunofluorescence staining demonstrated that the extracellular length of Salmonella LPS did affect the surface display of RGD4C peptide. The strain D2-RGD4C that synthesized intact LPS including lipid A, core oligosaccharides and O antigen polysaccharides could hardly display RGD4C peptide, showing the same fluorescence signal intensity as the strains not expressing RGD4C peptide. Among different strains, D2 ∆rfaJ-RGD4C that synthesized truncated LPS including lipid A and partial core oligosaccharides was capable of displaying RGD4C peptide most efficiently and showed the highest ability to target HUVECs expressing αV integrin and tumor tissue with abundant neovascularization. Animal experiments also demonstrated that this tumor-targeting attenuated Salmonella strain to simultaneously deliver endostatin and TRAIL, two agents with different anti-tumor activities, could significantly inhibit tumor growth and prolong mouse survival. Thus, our studies revealed that Salmonella could be genetically engineered to improve its tumor targeting via the truncation of LPS and surface display of targeting peptides, thereby eliciting superior anti-tumor effects through targeted delivery of drug molecules.
Collapse
|
30
|
Morris K, Schnoor B, Papa AL. Platelet cancer cell interplay as a new therapeutic target. Biochim Biophys Acta Rev Cancer 2022; 1877:188770. [DOI: 10.1016/j.bbcan.2022.188770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 10/16/2022]
|
31
|
Wang M, Cai W, Yang AJ, Wang CY, Zhang CL, Liu W, Xie XF, Gong YY, Zhao YY, Wu WC, Zhou Q, Zhao CY, Dong JF, Li M. Gastric cancer cell-derived extracellular vesicles disrupt endothelial integrity and promote metastasis. Cancer Lett 2022; 545:215827. [PMID: 35842018 DOI: 10.1016/j.canlet.2022.215827] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 07/05/2022] [Accepted: 07/10/2022] [Indexed: 11/26/2022]
Abstract
The endothelium is the critical barrier that controls transendothelial communications. Blood vessels in cancer tissue are poorly developed and highly permeable. However, it is poorly understood how circulating cancer cells released through these "leaky" vessels break the intact vasculature of remote organs to metastasize. We investigated the roles of cancer cell-derived extracellular vesicles (CEVs) in regulating cancer metastasis by analyzing samples from gastric cancer patients, performing in vitro experiments, and studying mouse models. We made several novel observations. First, the rate of metastasis was closely associated with plasma levels of CEVs in patients with gastric cancer. Second, cultured endothelial cells endocytosed CEVs, resulting in cytoskeletal rearrangement, low expression of the junction proteins cadherin and CD31, and forming large intercellular gaps to allow the transendothelial migration of cancer cells. The dynamin inhibitor Dynasore prevented these CEV-induced changes of endothelial cells by blocking CEVs endocytosis. Third, CEVs disrupted the endothelial barrier of cancer-bearing mice to promote cancer metastasis. Finally, lactadherin promoted the clearance of circulating CEVs to reduce metastasis. These results demonstrate the essential role of CEVs in promoting the metastasis of gastric cancer.
Collapse
Affiliation(s)
- Min Wang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; Institute of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| | - Wei Cai
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; Gansu Provincial Hospital, Lanzhou, China.
| | - Ai-Jun Yang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| | - Chen-Yu Wang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| | - Chen-Li Zhang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| | - Wei Liu
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| | - Xiao-Feng Xie
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; School of Medicine, Northwest MinZu University, Lanzhou, China.
| | - Yuan-Yuan Gong
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; Institute of Pathology, Department of Basic Medical Sciences, Fenyang College of Shanxi Medical University, Fenyang, China.
| | - Ying-Ying Zhao
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; Institute of Pathology, Department of Basic Medical Sciences, Fenyang College of Shanxi Medical University, Fenyang, China.
| | - Wen-Cheng Wu
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| | - Quan Zhou
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| | - Chan-Yuan Zhao
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| | - Jing-Fei Dong
- Bloodworks Research Institute, Seattle, WA, USA; Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| | - Min Li
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; Institute of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, China.
| |
Collapse
|
32
|
Cyclic Peptides for the Treatment of Cancers: A Review. Molecules 2022; 27:molecules27144428. [PMID: 35889301 PMCID: PMC9317348 DOI: 10.3390/molecules27144428] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/03/2022] [Accepted: 07/07/2022] [Indexed: 02/04/2023] Open
Abstract
Cyclic peptides have been widely reported to have therapeutic abilities in the treatment of cancer. This has been proven through in vitro and in vivo studies against breast, lung, liver, colon, and prostate cancers, among others. The multitude of data available in the literature supports the potential of cyclic peptides as anticancer agents. This review summarizes the findings from previously reported studies and discusses the different cyclic peptide compounds, the sources, and their modes of action as anticancer agents. The prospects and future of cyclic peptides will also be described to give an overview on the direction of cyclic peptide development for clinical applications.
Collapse
|
33
|
Kim I, Choi S, Yoo S, Lee M, Kim IS. Cancer-Associated Fibroblasts in the Hypoxic Tumor Microenvironment. Cancers (Basel) 2022; 14:cancers14143321. [PMID: 35884382 PMCID: PMC9320406 DOI: 10.3390/cancers14143321] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/30/2022] [Accepted: 07/05/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Cancers have regions of low oxygen concentration where hypoxia-related signaling pathways are activated. The hypoxic tumor microenvironment has been widely accepted as a hallmark of cancer and shown to be a critical factor in the crosstalk between cancer and stromal cells. Fibroblasts are one of the most abundant cellular components in the tumor stroma and are also significantly affected by oxygen deprivation. In this case, we discuss the molecular and cellular mechanisms that regulate fibroblasts under hypoxic conditions and their effect on cancer development and progression. Unraveling these regulatory mechanisms could be exploited in developing potential fibroblast-specific therapeutics for cancer. Abstract Solid cancers are composed of malignant cells and their surrounding matrix components. Hypoxia plays a critical role in shaping the tumor microenvironment that contributes to cancer progression and treatment failure. Cancer-associated fibroblasts (CAFs) are one of the most prominent components of the tumor microenvironment. CAFs are highly sensitive to hypoxia and participates in the crosstalk with cancer cells. Hypoxic CAFs modulate several mechanisms that induce cancer malignancy, such as extracellular matrix (ECM) remodeling, immune evasion, metabolic reprogramming, angiogenesis, metastasis, and drug resistance. Key signaling molecules regulating CAFs in hypoxia include transforming growth factor (TGF-β) and hypoxia-inducible factors (HIFs). In this article, we summarize the mechanisms underlying the hypoxic regulation of CAFs and how hypoxic CAFs affect cancer development and progression. We also discuss the potential therapeutic strategies focused on targeting CAFs in the hypoxic tumor microenvironment.
Collapse
Affiliation(s)
- Iljin Kim
- Department of Pharmacology and Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, Korea; (S.C.); (S.Y.)
- Correspondence: (I.K.); (I.-S.K.)
| | - Sanga Choi
- Department of Pharmacology and Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, Korea; (S.C.); (S.Y.)
| | - Seongkyeong Yoo
- Department of Pharmacology and Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, Korea; (S.C.); (S.Y.)
| | - Mingyu Lee
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute Science and Technology, Seoul 02792, Korea
- Correspondence: (I.K.); (I.-S.K.)
| |
Collapse
|
34
|
Abstract
Multiple myeloma (MM) accounts for 0.9% of cancer diagnoses, and incidence and mortality rate have increased in previous years. 18F-fluorodeoxyglucose (FDG) PET-computed tomography (CT) is an established modality for MM evaluation. MR imaging is helpful where 18F-FDG PET-CT is lacking. To standardize PET reporting, methods like Italian Myeloma Criteria for PET Use and Deauville criteria have been studied. Tracers like 11C-acetate and 11C-choline/18F-fluoromethylcholine (FCH) have shown higher sensitivity and detected more focal lesions and diffuse involvement than 18F-FDG PET-CT. 18F-FCH showed higher maximum standardized uptake value than 18FDG. 11C-methionine appears to be the best radiopharmaceutical, apart from 18F-FDG, for evaluating MM.
Collapse
Affiliation(s)
- Angel Hemrom
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Avinash Tupalli
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Abass Alavi
- Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Rakesh Kumar
- Diagnostic Nuclear Medicine Division, Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
35
|
Yamada Y, Onda T, Hagiuda A, Kan R, Matsunuma M, Hamada K, Kikkawa Y, Nomizu M. RGDX 1 X 2 motif regulates integrin αvβ5 binding for pluripotent stem cell adhesion. FASEB J 2022; 36:e22389. [PMID: 35657599 DOI: 10.1096/fj.202200317r] [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: 02/24/2022] [Revised: 04/28/2022] [Accepted: 05/23/2022] [Indexed: 11/11/2022]
Abstract
The arginine-glycine-aspartic acid (RGD) motif is a cell adhesion sequence that binds to integrins. Some RGD-containing peptides promote adhesion of both embryonic stem cells and induced pluripotent stem cells (iPSCs); however, not all such RGD-containing peptides are active. In this study, we elucidated the role of RGD-neighboring sequences on iPSC adhesion using diverse synthetic peptides and recombinant proteins. Our results indicate that iPSC adhesion requires RGDX1 X2 sequences, such as RGDVF and RGDNY, and that the X1 X2 residues are essential for the adhesion via integrin αvβ5 but not αvβ3. iPSCs express integrin αvβ5 but not αvβ3; therefore, iPSC adhesion requires the RGDX1 X2 -containing sequences. The importance of the X1 X2 residues was confirmed with both HeLa and A549 cells, which express integrin αvβ5 but not αvβ3. Analysis of RGD-neighboring sequences provides important insights into ligand-binding specificity of integrins. Identification of integrin αvβ5-binding motifs is potentially useful in drug development, drug delivery, cell culture, and tissue engineering.
Collapse
Affiliation(s)
- Yuji Yamada
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Toru Onda
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Ayami Hagiuda
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Ryuji Kan
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Masumi Matsunuma
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Keisuke Hamada
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Yamato Kikkawa
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Motoyoshi Nomizu
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| |
Collapse
|
36
|
Novel Functions of Integrins as Receptors of CD154: Their Role in Inflammation and Apoptosis. Cells 2022; 11:cells11111747. [PMID: 35681441 PMCID: PMC9179867 DOI: 10.3390/cells11111747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/18/2022] [Accepted: 05/24/2022] [Indexed: 12/16/2022] Open
Abstract
CD154, an inflammatory mediator also known as CD40 ligand, has been identified as a novel binding partner for some members of the integrin family. The αIIbβ3, specifically expressed on platelets, was the first integrin to be described as a receptor for CD154 after CD40. Its interaction with soluble CD154 (sCD154) highly contributes to thrombus formation and stability. Identifying αIIbβ3 opened the door for investigating other integrins as partners of CD154. The αMβ2 expressed on myeloid cells was shown capable of binding CD154 and contributing as such to cell activation, adhesion, and release of proinflammatory mediators. In parallel, α5β1 communicates with sCD154, inducing pro-inflammatory responses. Additional pathogenic effects involving apoptosis-preventing functions were exhibited by the CD154–α5β1 dyad in T cells, conferring a role for such interaction in the survival of malignant cells, as well as the persistence of autoreactive T cells. More recently, CD154 receptors integrated two new integrin members, αvβ3 and α4β1, with little known as to their biological significance in this context. This article provides an overview of the novel role of integrins as receptors of CD154 and as critical players in pro-inflammatory and apoptotic responses.
Collapse
|
37
|
Abdel Sater AH, Bouferraa Y, Amhaz G, Haibe Y, Lakkiss AE, Shamseddine A. From Tumor Cells to Endothelium and Gut Microbiome: A Complex Interaction Favoring the Metastasis Cascade. Front Oncol 2022; 12:804983. [PMID: 35600385 PMCID: PMC9117727 DOI: 10.3389/fonc.2022.804983] [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: 10/29/2021] [Accepted: 04/12/2022] [Indexed: 11/30/2022] Open
Abstract
Metastasis is a complicated process through which tumor cells disseminate to distant organs and adapt to novel tumor microenvironments. This multi-step cascade relies on the accumulation of genetic and epigenetic alterations within the tumor cells as well as the surrounding non-tumor stromal cells. Endothelial cells constitute a major player in promoting metastasis formation either by inducing the growth of tumor cells or by directing them towards dissemination in the blood or lymph. In fact, the direct and indirect interactions between tumor and endothelial cells were shown to activate several mechanisms allowing cancer cells’ invasion and extravasation. On the other side, gastrointestinal cancer development was shown to be associated with the disruption of the gut microbiome. While several proposed mechanisms have been investigated in this regard, gut and tumor-associated microbiota were shown to impact the gut endothelial barrier, increasing the dissemination of bacteria through the systemic circulation. This bacterial dislocation allows the formation of an inflammatory premetastatic niche in the distant organs promoting the metastatic cascade of primary tumors. In this review, we discuss the role of the endothelial cells in the metastatic cascade of tumors. We will focus on the role of the gut vascular barrier in the regulation metastasis. We will also discuss the interaction between this vascular barrier and the gut microbiota enhancing the process of metastasis. In addition, we will try to elucidate the different mechanisms through which this bacterial dislocation prepares the favorable metastatic niche at distant organs allowing the dissemination and successful deposition of tumor cells in the new microenvironments. Finally, and given the promising results of the studies combining immune checkpoint inhibitors with either microbiota alterations or anti-angiogenic therapy in many types of cancer, we will elaborate in this review the complex interaction between these 3 factors and their possible therapeutic combination to optimize response to treatment.
Collapse
Affiliation(s)
- Ali H Abdel Sater
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Youssef Bouferraa
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ghid Amhaz
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Yolla Haibe
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ahmed El Lakkiss
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ali Shamseddine
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
38
|
Translating Molecules into Imaging—The Development of New PET Tracers for Patients with Melanoma. Diagnostics (Basel) 2022; 12:diagnostics12051116. [PMID: 35626272 PMCID: PMC9139963 DOI: 10.3390/diagnostics12051116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/22/2022] [Accepted: 04/27/2022] [Indexed: 01/27/2023] Open
Abstract
Melanoma is a deadly disease that often exhibits relentless progression and can have both early and late metastases. Recent advances in immunotherapy and targeted therapy have dramatically increased patient survival for patients with melanoma. Similar advances in molecular targeted PET imaging can identify molecular pathways that promote disease progression and therefore offer physiological information. Thus, they can be used to assess prognosis, tumor heterogeneity, and identify instances of treatment failure. Numerous agents tested preclinically and clinically demonstrate promising results with high tumor-to-background ratios in both primary and metastatic melanoma tumors. Here, we detail the development and testing of multiple molecular targeted PET-imaging agents, including agents for general oncological imaging and those specifically for PET imaging of melanoma. Of the numerous radiopharmaceuticals evaluated for this purpose, several have made it to clinical trials and showed promising results. Ultimately, these agents may become the standard of care for melanoma imaging if they are able to demonstrate micrometastatic disease and thus provide more accurate information for staging. Furthermore, these agents provide a more accurate way to monitor response to therapy. Patients will be able to receive treatment based on tumor uptake characteristics and may be able to be treated earlier for lesions that with traditional imaging would be subclinical, overall leading to improved outcomes for patients.
Collapse
|
39
|
Li Y, Xie M, Jones JB, Zhang Z, Wang Z, Dang T, Wang X, Lipowska M, Mao H. Targeted Delivery of DNA Topoisomerase Inhibitor SN38 to Intracranial Tumors of Glioblastoma Using Sub-5 Ultrafine Iron Oxide Nanoparticles. Adv Healthc Mater 2022; 11:e2102816. [PMID: 35481625 DOI: 10.1002/adhm.202102816] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 04/07/2022] [Indexed: 11/09/2022]
Abstract
Effectively delivering therapeutics for treating brain tumors is hindered by the physical and biological barriers in the brain. Even with the compromised blood-brain barrier and highly angiogenic blood-tumor barrier seen in glioblastoma (GBM), most drugs, including nanomaterial-based formulations, hardly reach intracranial tumors. This work investigates sub-5 nm ultrafine iron oxide nanoparticles (uIONP) with 3.5 nm core diameter as a carrier for delivering DNA topoisomerase inhibitor 7-ethyl-10-hydroxyl camptothecin (SN38) to treat GBM. Given a higher surface-to-volume ratio, uIONP shows one- or three-folds higher SN38 loading efficiency (48.3 ± 6.1%, mg/mg Fe) than those with core sizes of 10 or 20 nm. SN38 encapsulated in the coating polymer exhibits pH sensitive release with <10% over 48 h at pH 7.4, but 86% at pH 5, thus being protected from converting to inactive glucuronide by UDP-glucuronosyltransferase 1A1. Conjugating αv β3 -integrin-targeted cyclo(Arg-Gly-Asp-D-Phe-Cys) (RGD) as ligands, RGD-uIONP/SN38 demonstrates targeted cytotoxicity to αv β3 -integrin-overexpressed U87MG GBM cells with a half-maximal inhibitory concentration (IC50 ) of 30.9 ± 2.2 nm. The efficacy study using an orthotopic mouse model of GBM reveals tumor-specific delivery of 11.5% injected RGD-uIONP/SN38 (10 mg Fe kg-1 ), significantly prolonging the survival in mice by 41%, comparing to those treated with SN38 alone (p < 0.001).
Collapse
Affiliation(s)
- Yuancheng Li
- Department of Radiology and Imaging Sciences Emory University Atlanta GA 30329 USA
- 5M Biomed, LLC Atlanta GA 30303 USA
| | - Manman Xie
- Department of Radiology and Imaging Sciences Emory University Atlanta GA 30329 USA
| | - Joshua B. Jones
- Department of Radiology and Imaging Sciences Emory University Atlanta GA 30329 USA
| | - Zhaobin Zhang
- Department of Neurosurgery Emory University Atlanta GA 30329 USA
| | - Zi Wang
- Department of Radiology and Imaging Sciences Emory University Atlanta GA 30329 USA
| | - Tu Dang
- Division of Research Philadelphia College of Osteopathic Medicine – Georgia Campus Suwanee GA 30024 USA
| | - Xinyu Wang
- Department of Pharmaceutical Sciences Philadelphia College of Osteopathic Medicine – Georgia Campus Suwanee GA 30024 USA
| | - Malgorzata Lipowska
- Department of Radiology and Imaging Sciences Emory University Atlanta GA 30329 USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences Emory University Atlanta GA 30329 USA
| |
Collapse
|
40
|
Kummer D, Steinbacher T, Thölmann S, Schwietzer MF, Hartmann C, Horenkamp S, Demuth S, Peddibhotla SS, Brinkmann F, Kemper B, Schnekenburger J, Brandt M, Betz T, Liashkovich I, Kouzel IU, Shahin V, Corvaia N, Rottner K, Tarbashevich K, Raz E, Greune L, Schmidt MA, Gerke V, Ebnet K. A JAM-A-tetraspanin-αvβ5 integrin complex regulates contact inhibition of locomotion. J Biophys Biochem Cytol 2022; 221:213070. [PMID: 35293964 PMCID: PMC8931538 DOI: 10.1083/jcb.202105147] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 12/16/2021] [Accepted: 01/21/2022] [Indexed: 12/30/2022] Open
Abstract
Contact inhibition of locomotion (CIL) is a process that regulates cell motility upon collision with other cells. Improper regulation of CIL has been implicated in cancer cell dissemination. Here, we identify the cell adhesion molecule JAM-A as a central regulator of CIL in tumor cells. JAM-A is part of a multimolecular signaling complex in which tetraspanins CD9 and CD81 link JAM-A to αvβ5 integrin. JAM-A binds Csk and inhibits the activity of αvβ5 integrin-associated Src. Loss of JAM-A results in increased activities of downstream effectors of Src, including Erk1/2, Abi1, and paxillin, as well as increased activity of Rac1 at cell-cell contact sites. As a consequence, JAM-A-depleted cells show increased motility, have a higher cell-matrix turnover, and fail to halt migration when colliding with other cells. We also find that proper regulation of CIL depends on αvβ5 integrin engagement. Our findings identify a molecular mechanism that regulates CIL in tumor cells and have implications on tumor cell dissemination.
Collapse
Affiliation(s)
- Daniel Kummer
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany,Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany
| | - Tim Steinbacher
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Sonja Thölmann
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Mariel Flavia Schwietzer
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Christian Hartmann
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Simone Horenkamp
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Sabrina Demuth
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Swetha S.D. Peddibhotla
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Frauke Brinkmann
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany
| | - Björn Kemper
- Biomedical Technology Center, Medical Faculty, University of Münster, Münster, Germany
| | - Jürgen Schnekenburger
- Biomedical Technology Center, Medical Faculty, University of Münster, Münster, Germany
| | - Matthias Brandt
- Institute-associated Research Group “Mechanics of Cellular Systems”, Institute of Cell Biology, ZMBE, University of Münster, Münster, Germany
| | - Timo Betz
- Institute-associated Research Group “Mechanics of Cellular Systems”, Institute of Cell Biology, ZMBE, University of Münster, Münster, Germany
| | - Ivan Liashkovich
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Ivan U. Kouzel
- Sars International Centre for Marine Molecular Biology University of Bergen Thormøhlensgt, Bergen, Norway
| | - Victor Shahin
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Nathalie Corvaia
- Centre d’Immunologie Pierre Fabre (CIPF), Saint-Julien-en-Genevois, France
| | - Klemens Rottner
- Divison of Molecular Cell Biology, Zoological Institute, Technical University Braunschweig, Braunschweig, Germany,Molecular Cell Biology Group, Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Erez Raz
- Institute of Cell Biology, ZMBE, University of Münster, Münster, Germany,Cells-in-Motion Cluster of Excellence (EXC 1003—CiM), University of Münster, 48419 Münster, Germany
| | - Lilo Greune
- Institute of Infectiology, ZMBE, University of Münster, Münster, Germany
| | | | - Volker Gerke
- Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany,Cells-in-Motion Cluster of Excellence (EXC 1003—CiM), University of Münster, 48419 Münster, Germany
| | - Klaus Ebnet
- Institute-associated Research Group “Cell Adhesion and Cell Polarity”, Münster, Germany,Institute of Medical Biochemistry, ZMBE, University of Münster, Münster, Germany,Interdisciplinary Clinical Research Center (IZKF), University of Münster, Münster, Germany,Cells-in-Motion Cluster of Excellence (EXC 1003—CiM), University of Münster, 48419 Münster, Germany
| |
Collapse
|
41
|
Tamijani SMS, Valian N, Heravi M, Ahmadiani A, Beirami E, Dargahi L. Implication of thyroid hormone receptors in methamphetamine neurocognitive effects. Neurotoxicology 2022; 90:130-135. [PMID: 35301009 DOI: 10.1016/j.neuro.2022.03.003] [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: 10/16/2021] [Revised: 02/09/2022] [Accepted: 03/10/2022] [Indexed: 11/27/2022]
Abstract
Methamphetamine (MA) induces neurocognitive effects via several mechanisms. In the present study, we investigated the alteration of thyroid hormone receptor's expression in the context of MA-induced memory impairment and explored the protective effects of exogenous thyroid hormones (THs). Male wistar rats, received increasing regime of MA (1-10mg/kg, intraperitoneal, twice a day for 10 days), were treated with T3 (40μg/rat/day; intranasal, 2.5μl/nostril) or T4 (20µg/kg/day; intraperitoneal) for 7 days after MA cessation. All rats were subjected to novel object recognition memory test and then the mRNA levels of TH nuclear receptors (TRα1 and TRβ1) and seladin-1, an anti-apoptotic factor, and the protein level of TH cell surface receptor (integrin αvβ3) were measured in the hippocampus of rats. Our results showed that MA-induced memory impairment is concomitant with decreased level of TRα1 mRNA. T3 or T4 treatment significantly alleviated MA-induced memory impairment, but had no significant effect on the mRNA levels of TH nuclear receptors. However, T4 treatment significantly increased the protein level of cell surface receptor (av subunit) in MA-treated rats. These findings suggest that MA neurocognitive effects can be associated with impaired TH signaling in the brain and introduce this pathway as a promising therapeutic approach against MA-induced memory impairment.
Collapse
Affiliation(s)
| | - Neda Valian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mansooreh Heravi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elmira Beirami
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Leila Dargahi
- NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
42
|
Chen YF, Yang YN, Chu HR, Huang TY, Wang SH, Chen HY, Li ZL, Yang YCSH, Lin HY, Hercbergs A, Whang-Peng J, Wang K, Davis PJ. Role of Integrin αvβ3 in Doxycycline-Induced Anti-Proliferation in Breast Cancer Cells. Front Cell Dev Biol 2022; 10:829788. [PMID: 35237605 PMCID: PMC8884148 DOI: 10.3389/fcell.2022.829788] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/24/2022] [Indexed: 12/13/2022] Open
Abstract
Doxycycline, an antibiotic, displays the inhibition of different signal transduction pathways, such as anti-inflammation and anti-proliferation, in different types of cancers. However, the anti-cancer mechanisms of doxycycline via integrin αvβ3 are incompletely understood. Integrin αvβ3 is a cell-surface anchor protein. It is the target for estrogen, androgen, and thyroid hormone and plays a pivotal role in the proliferation, migration, and angiogenic process in cancer cells. In our previous study, thyroxine hormones can interact with integrin αvβ3 to activate the extracellular signal-regulated kinase 1/2 (ERK1/2), and upregulate programmed death-ligand 1 (PD-L1) expression. In the current study, we investigated the inhibitory effects of doxycycline on proliferation in two breast cancer cell lines, MCF-7 and MDA-MB-231 cells. Doxycycline induces concentration-dependent anti-proliferation in both breast cancer cell lines. It regulates gene expressions involved in proliferation, pro-apoptosis, and angiogenesis. Doxycycline suppresses cell cyclin D1 (CCND1) and c-Myc which play crucial roles in proliferation. It also inhibits PD-L1 gene expression. Our findings show that modulation on integrin αvβ3 binding activities changed both thyroxine- and doxycycline-induced signal transductions by an integrin αvβ3 inhibitor (HSDVHK-NH2). Doxycycline activates phosphorylation of focal adhesion kinase (FAK), a downstream of integrin, but inhibits the ERK1/2 phosphorylation. Regardless, doxycycline-induced FAK phosphorylation is blocked by HSDVHK-NH2. In addition, the specific mechanism of action associated with pERK1/2 inhibition via integrin αvβ3 is unknown for doxycycline treatment. On the other hand, our findings indicated that inhibiting ERK1/2 activation leads to suppression of PD-L1 expression by doxycycline treatment. Furthermore, doxycycline-induced gene expressions are disturbed by a specific integrin αvβ3 inhibitor (HSDVHK-NH2) or a mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinases (ERK) kinase (MAPK/ERK, MEK) inhibitor (PD98059). The results imply that doxycycline may interact with integrin αvβ3 and inhibits ERK1/2 activation, thereby regulating cell proliferation and downregulating PD-L1 gene expression in estrogen receptor (ER)-negative breast cancer MDA-MB-231 cells.
Collapse
Affiliation(s)
- Yi-Fong Chen
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Yung-Ning Yang
- School of Medicine, I-Shou University, Kaohsiung, Taiwan.,Department of Pediatrics, E-DA Hospital, Kaohsiung, Taiwan
| | - Hung-Ru Chu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Tung-Yung Huang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Shwu-Huey Wang
- Core Facility Center, Department of Research Development, Taipei Medical University, Taipei, Taiwan
| | - Han-Yu Chen
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Zi-Lin Li
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chen S H Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Hung-Yun Lin
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.,Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.,Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY, United States
| | - Aleck Hercbergs
- Department of Radiation Oncology, Cleveland Clinic, Cleveland, OH, United States
| | | | - Kuan Wang
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Paul J Davis
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY, United States.,Department of Medicine, Albany Medical College, Albany, NY, United States
| |
Collapse
|
43
|
Liang K, Zhang R, Luo H, Zhang J, Tian Z, Zhang X, Zhang Y, Ali MK, Kong Q. Optimized Attenuated Salmonella Typhimurium Suppressed Tumor Growth and Improved Survival in Mice. Front Microbiol 2022; 12:774490. [PMID: 35003007 PMCID: PMC8733734 DOI: 10.3389/fmicb.2021.774490] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/02/2021] [Indexed: 01/03/2023] Open
Abstract
The gram-negative facultative anaerobic bacteria Salmonella enterica serovar Typhimurium (hereafter S. Typhimurium) has always been considered as one candidate of anti-tumor agents or vectors for delivering drug molecules. In this study, we compared several widely studied S. Typhimurium strains in their anti-tumor properties aiming to screen out the best one for further optimization and use in cancer therapy. In terms of the motility, virulence and anti-tumor efficacy, the three strains 14028, SL1344, and UK-1 were similar and obviously better than LT-2, and UK-1 showed the best phenotypes among them. Therefore, the strain UK-1 (D) was selected for the following studies. Its auxotrophic mutant strain (D1) harboring ∆aroA and ∆purM mutations was further optimized through the modification of lipid A structure, generating a new strain named D2 with stronger immunostimulatory activity. Finally, the ∆asd derivative of D2 was utilized as one live vector to deliver anti-tumor molecules including the angiogenesis inhibitor endostatin and apoptosis inducer TRAIL and the therapeutic and toxic-side effects were evaluated in mouse models of colon carcinoma and melanoma. After intraperitoneal infection, engineered Salmonella bacteria equipped with endostatin and/or TRAIL significantly suppressed the tumor growth and prolonged survival of tumor-bearing mice compared to PBS or bacteria carrying the empty plasmid. Consistently, immunohistochemical studies confirmed the colonization of Salmonella bacteria and the expression of anti-tumor molecules inside tumor tissue, which were accompanied by the increase of cell apoptosis and suppression of tumor angiogenesis. These results demonstrated that the beneficial anti-tumor efficacy of attenuated S. Typhimurium bacteria could be improved through delivery of drug molecules with powerful anti-tumor activities.
Collapse
Affiliation(s)
- Kang Liang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Rui Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Haiyan Luo
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Jinlong Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Zhenyuan Tian
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xiaofen Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Yulin Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Md Kaisar Ali
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Qingke Kong
- College of Veterinary Medicine, Southwest University, Chongqing, China
| |
Collapse
|
44
|
Santoni G, Amantini C, Nabissi M, Arcella A, Maggi F, Santoni M, Morelli MB. Functional In Vitro Assessment of VEGFA/NOTCH2 Signaling Pathway and pRB Proteasomal Degradation and the Clinical Relevance of Mucolipin TRPML2 Overexpression in Glioblastoma Patients. Int J Mol Sci 2022; 23:ijms23020688. [PMID: 35054871 PMCID: PMC8775570 DOI: 10.3390/ijms23020688] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 12/30/2021] [Accepted: 01/06/2022] [Indexed: 12/28/2022] Open
Abstract
Glioblastoma (GBM) is the most malignant glioma with an extremely poor prognosis. It is characterized by high vascularization and its growth depends on the formation of new blood vessels. We have previously demonstrated that TRPML2 mucolipin channel expression increases with the glioma pathological grade. Herein by ddPCR and Western blot we found that the silencing of TRPML2 inhibits expression of the VEGFA/Notch2 angiogenic pathway. Moreover, the VEGFA/Notch2 expression increased in T98 and U251 cells stimulated with the TRPML2 agonist, ML2-SA1, or by enforced-TRPML2 levels. In addition, changes in TRPML2 expression or ML2-SA1-induced stimulation, affected Notch2 activation and VEGFA release. An increased invasion capability, associated with a reduced VEGF/VEGFR2 expression and increased vimentin and CD44 epithelial-mesenchymal transition markers in siTRPML2, but not in enforced-TRPML2 or ML2-SA1-stimulated glioma cells, was demonstrated. Furthermore, an increased sensitivity to Doxorubicin cytotoxicity was demonstrated in siTRPML2, whereas ML2-SA1-treated GBM cells were more resistant. The role of proteasome in Cathepsin B-dependent and -independent pRB degradation in siTRPML2 compared with siGLO cells was studied. Finally, through Kaplan-Meier analysis, we found that high TRPML2 mRNA expression strongly correlates with short survival in GBM patients, supporting TRPML2 as a negative prognostic factor in GBM patients.
Collapse
Affiliation(s)
- Giorgio Santoni
- School of Pharmacy, Section of Experimental Medicine, University of Camerino, 62032 Camerino, Italy;
- Correspondence: (G.S.); (M.B.M.); Tel.: +39-0737403319 (G.S.); +39-0737403312 (M.B.M.)
| | - Consuelo Amantini
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (C.A.); (F.M.)
| | - Massimo Nabissi
- School of Pharmacy, Section of Experimental Medicine, University of Camerino, 62032 Camerino, Italy;
| | - Antonietta Arcella
- Neuropathology Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico Neuromed, 86077 Pozzilli, Italy;
| | - Federica Maggi
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (C.A.); (F.M.)
- Department of Molecular Medicine, Sapienza University, 00185 Rome, Italy
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, 62100 Macerata, Italy;
| | - Maria Beatrice Morelli
- School of Pharmacy, Section of Experimental Medicine, University of Camerino, 62032 Camerino, Italy;
- Correspondence: (G.S.); (M.B.M.); Tel.: +39-0737403319 (G.S.); +39-0737403312 (M.B.M.)
| |
Collapse
|
45
|
Charabati M, Grasmuck C, Ghannam S, Bourbonnière L, Fournier AP, Lécuyer MA, Tastet O, Kebir H, Rébillard RM, Hoornaert C, Gowing E, Larouche S, Fortin O, Pittet C, Filali-Mouhim A, Lahav B, Moumdjian R, Bouthillier A, Girard M, Duquette P, Cayrol R, Peelen E, Quintana FJ, Antel JP, Flügel A, Larochelle C, Arbour N, Zandee S, Prat A. DICAM promotes T H17 lymphocyte trafficking across the blood-brain barrier during autoimmune neuroinflammation. Sci Transl Med 2022; 14:eabj0473. [PMID: 34985970 DOI: 10.1126/scitranslmed.abj0473] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Marc Charabati
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Camille Grasmuck
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Soufiane Ghannam
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Lyne Bourbonnière
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - Antoine P Fournier
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Marc-André Lécuyer
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada.,Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen D-37073, Germany
| | - Olivier Tastet
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - Hania Kebir
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Rose-Marie Rébillard
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Chloé Hoornaert
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Elizabeth Gowing
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Sandra Larouche
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - Olivier Fortin
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - Camille Pittet
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - Ali Filali-Mouhim
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - Boaz Lahav
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Multiple Sclerosis Clinic, Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec H2L 4M1, Canada
| | - Robert Moumdjian
- Division of Neurosurgery, Université de Montréal and CHUM, Montreal, Quebec H2L 4M1, Canada
| | - Alain Bouthillier
- Division of Neurosurgery, Université de Montréal and CHUM, Montreal, Quebec H2L 4M1, Canada
| | - Marc Girard
- Multiple Sclerosis Clinic, Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec H2L 4M1, Canada
| | - Pierre Duquette
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Multiple Sclerosis Clinic, Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec H2L 4M1, Canada
| | - Romain Cayrol
- Department of Pathology, Université de Montréal and CHUM, Montreal, Quebec H2L 4M1, Canada
| | - Evelyn Peelen
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Francisco J Quintana
- Ann Romney Carter for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute and Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Alexander Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen D-37073, Germany
| | - Catherine Larochelle
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada.,Multiple Sclerosis Clinic, Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec H2L 4M1, Canada
| | - Nathalie Arbour
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Stephanie Zandee
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Alexandre Prat
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada.,Multiple Sclerosis Clinic, Division of Neurology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec H2L 4M1, Canada
| |
Collapse
|
46
|
Morbidelli L, Donnini S. Introduction. ANTIANGIOGENIC DRUGS AS CHEMOSENSITIZERS IN CANCER THERAPY 2022:1-28. [DOI: 10.1016/b978-0-323-90190-1.00018-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
47
|
Varrone F, Mandrich L, Caputo E. Melanoma Immunotherapy and Precision Medicine in the Era of Tumor Micro-Tissue Engineering: Where Are We Now and Where Are We Going? Cancers (Basel) 2021; 13:5788. [PMID: 34830940 PMCID: PMC8616100 DOI: 10.3390/cancers13225788] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/05/2021] [Accepted: 11/12/2021] [Indexed: 11/16/2022] Open
Abstract
Malignant melanoma still remains a cancer with very poor survival rates, although it is at the forefront of personalized medicine. Most patients show partial responses and disease progressed due to adaptative resistance mechanisms, preventing long-lasting clinical benefits to the current treatments. The response to therapies can be shaped by not only taking into account cancer cell heterogeneity and plasticity, but also by its structural context as well as the cellular component of the tumor microenvironment (TME). Here, we review the recent development in the field of immunotherapy and target-based therapy and how, in the era of tumor micro-tissue engineering, ex-vivo assays could help to enhance our melanoma biology knowledge in its complexity, translating it in the development of successful therapeutic strategies, as well as in the prediction of therapeutic benefits.
Collapse
Affiliation(s)
| | - Luigi Mandrich
- Research Institute on Terrestrial Ecosystem—IRET-CNR Via Pietro Castellino 111, I-80131 Naples, Italy;
| | - Emilia Caputo
- Institute of Genetics and Biophysics—IGB-CNR, “A. Buzzati-Traverso”, Via Pietro Castellino 111, I-80131 Naples, Italy
| |
Collapse
|
48
|
Hennigs JK, Matuszcak C, Trepel M, Körbelin J. Vascular Endothelial Cells: Heterogeneity and Targeting Approaches. Cells 2021; 10:2712. [PMID: 34685692 PMCID: PMC8534745 DOI: 10.3390/cells10102712] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 01/18/2023] Open
Abstract
Forming the inner layer of the vascular system, endothelial cells (ECs) facilitate a multitude of crucial physiological processes throughout the body. Vascular ECs enable the vessel wall passage of nutrients and diffusion of oxygen from the blood into adjacent cellular structures. ECs regulate vascular tone and blood coagulation as well as adhesion and transmigration of circulating cells. The multitude of EC functions is reflected by tremendous cellular diversity. Vascular ECs can form extremely tight barriers, thereby restricting the passage of xenobiotics or immune cell invasion, whereas, in other organ systems, the endothelial layer is fenestrated (e.g., glomeruli in the kidney), or discontinuous (e.g., liver sinusoids) and less dense to allow for rapid molecular exchange. ECs not only differ between organs or vascular systems, they also change along the vascular tree and specialized subpopulations of ECs can be found within the capillaries of a single organ. Molecular tools that enable selective vascular targeting are helpful to experimentally dissect the role of distinct EC populations, to improve molecular imaging and pave the way for novel treatment options for vascular diseases. This review provides an overview of endothelial diversity and highlights the most successful methods for selective targeting of distinct EC subpopulations.
Collapse
Affiliation(s)
- Jan K. Hennigs
- ENDomics Lab, Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Christiane Matuszcak
- ENDomics Lab, Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Martin Trepel
- Department of Hematology and Medical Oncology, University Medical Center Augsburg, 86156 Augsburg, Germany;
| | - Jakob Körbelin
- ENDomics Lab, Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| |
Collapse
|
49
|
Aspriţoiu VM, Stoica I, Bleotu C, Diaconu CC. Epigenetic Regulation of Angiogenesis in Development and Tumors Progression: Potential Implications for Cancer Treatment. Front Cell Dev Biol 2021; 9:689962. [PMID: 34552922 PMCID: PMC8451900 DOI: 10.3389/fcell.2021.689962] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022] Open
Abstract
Angiogenesis is a multi-stage process of new blood vessel development from pre-existing vessels toward an angiogenic stimulus. The process is essential for tissue maintenance and homeostasis during embryonic development and adult life as well as tumor growth. Under normal conditions, angiogenesis is involved in physiological processes, such as wound healing, cyclic regeneration of the endometrium, placental development and repairing certain cardiac damage, in pathological conditions, it is frequently associated with cancer development and metastasis. The control mechanisms of angiogenesis in carcinogenesis are tightly regulated at the genetic and epigenetic level. While genetic alterations are the critical part of gene silencing in cancer cells, epigenetic dysregulation can lead to repression of tumor suppressor genes or oncogene activation, becoming an important event in early development and the late stages of tumor development, as well. The global alteration of the epigenetic spectrum, which includes DNA methylation, histone modification, chromatin remodeling, microRNAs, and other chromatin components, is considered one of the hallmarks of cancer, and the efforts are concentrated on the discovery of molecular epigenetic markers that identify cancerous precursor lesions or early stage cancer. This review aims to highlight recent findings on the genetic and epigenetic changes that can occur in physiological and pathological angiogenesis and analyze current knowledge on how deregulation of epigenetic modifiers contributes to tumorigenesis and tumor maintenance. Also, we will evaluate the clinical relevance of epigenetic markers of angiogenesis and the potential use of "epi-drugs" in modulating the responsiveness of cancer cells to anticancer therapy through chemotherapy, radiotherapy, immunotherapy and hormone therapy as anti-angiogenic strategies in cancer.
Collapse
Affiliation(s)
| | - Ileana Stoica
- Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Coralia Bleotu
- Faculty of Biology, University of Bucharest, Bucharest, Romania.,Romanian Academy, Stefan S. Nicolau Institute of Virology, Bucharest, Romania
| | | |
Collapse
|
50
|
Mahadik N, Bhattacharya D, Padmanabhan A, Sakhare K, Narayan KP, Banerjee R. Targeting steroid hormone receptors for anti-cancer therapy-A review on small molecules and nanotherapeutic approaches. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1755. [PMID: 34541822 DOI: 10.1002/wnan.1755] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/11/2022]
Abstract
The steroid hormone receptors (SHRs) among nuclear hormone receptors (NHRs) are steroid ligand-dependent transcription factors that play important roles in the regulation of transcription of genes promoted via hormone responsive elements in our genome. Aberrant expression patterns and context-specific regulation of these receptors in cancer, have been routinely reported by multiple research groups. These gave an window of opportunity to target those receptors in the context of developing novel, targeted anticancer therapeutics. Besides the development of a plethora of SHR-targeting synthetic ligands and the availability of their natural, hormonal ligands, development of many SHR-targeted, anticancer nano-delivery systems and theranostics, especially based on small molecules, have been reported. It is intriguing to realize that these cytoplasmic receptors have become a hot target for cancer selective delivery. This is in spite of the fact that these receptors do not fall in the category of conventional, targetable cell surface bound or transmembrane receptors that enjoy over-expression status. Glucocorticoid receptor (GR) is one such exciting SHR that in spite of it being expressed ubiquitously in all cells, we discovered it to behave differently in cancer cells, thus making it a truly druggable target for treating cancer. This review selectively accumulates the knowledge generated in the field of SHR-targeting as a major focus for cancer treatment with various anticancer small molecules and nanotherapeutics on progesterone receptor, mineralocorticoid receptor, and androgen receptor while selectively emphasizing on GR and estrogen receptor. This review also briefly highlights lipid-modification strategy to convert ligands into SHR-targeted cancer nanotherapeutics. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Biology-Inspired Nanomaterials > Lipid-Based Structures Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Namita Mahadik
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| | - Dwaipayan Bhattacharya
- Department of Biological Sciences, Birla Institute of Technology Pilani, Hyderabad, India
| | - Akshaya Padmanabhan
- Department of Biological Sciences, Birla Institute of Technology Pilani, Hyderabad, India
| | - Kalyani Sakhare
- Department of Biological Sciences, Birla Institute of Technology Pilani, Hyderabad, India
| | - Kumar Pranav Narayan
- Department of Biological Sciences, Birla Institute of Technology Pilani, Hyderabad, India
| | - Rajkumar Banerjee
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|