1
|
Gupta S, Singh A, Deorah S, Tomar A. Immunotherapy in OSCC: Current trend and challenges. Crit Rev Oncol Hematol 2025; 209:104672. [PMID: 39993651 DOI: 10.1016/j.critrevonc.2025.104672] [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: 01/28/2025] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 02/26/2025] Open
Abstract
OBJECTIVES Oral Cancer is one of the most prevalent malignant tumors of the head and neck. The three primary clinical treatments available till now for oral cancer are chemotherapy, radiation, and surgery. The goal of this review was to outline the basic principles of immunotherapy along with various immunotherapeutic agents on Oral Squamous Cell Carcinoma. MATERIALS AND METHODS A comprehensive search in PubMed, Scopus, and Google Scholar was performed using relevant keywords. All the articles, both English as well as non-English were included also with inclusion data from high-incidence countries (South-east Asia) and the compilation was ten done after getting the data reviewed from two pathologists who were blinded to the data. RESULTS All the data has been compiled and the various sections in the manuscript provides an insight into the current trends in immunotherapy. CONCLUSIONS Advanced research studies are needed to counteract the hurdles associated with immunotherapy so that a greater proportion of patients can be treated. CLINICAL RELEVANCE One of the more recent developments that is promising is immunotherapy, which can be quite beneficial when used as a monotherapy or an adjuvant treatment. This more recent treatment approach could serve as the fourth pillar in cancer care, alongside radiation, chemotherapy, and surgery. Because immunotherapy relies on the patient's immunological environment, careful patient selection is essential to its effectiveness.
Collapse
Affiliation(s)
- Shalini Gupta
- Department of Oral Pathology and Microbiology, King George's Medical University, Lucknow 226003, India.
| | - Akanchha Singh
- Department of Oral Pathology and Microbiology, King George's Medical University, Lucknow 226003, India
| | - Sakshi Deorah
- Department of Oral Pathology and Microbiology, King George's Medical University, Lucknow 226003, India
| | - Arushi Tomar
- Department of Oral Pathology and Microbiology, King George's Medical University, Lucknow 226003, India
| |
Collapse
|
2
|
Mottaghi S, Abbaszadeh H, Valizadeh A, Hafezi K. The polyphenolic compound, α-conidendrin, exerts anti-colon cancer and anti-angiogenic effects by targeting several signaling molecules. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04090-2. [PMID: 40208320 DOI: 10.1007/s00210-025-04090-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/21/2025] [Indexed: 04/11/2025]
Abstract
Our previous study indicated that α-conidendrin had considerable anti-proliferative activities against breast cancer cell lines. The present study aimed to evaluate the anti-colon cancer and anti-angiogenic influences of α-conidendrin as well as its molecular mechanisms. The findings of the current study demonstrate that α-conidendrin possesses potent anti-colon cancer and anti-angiogenic effects. α-Conidendrin significantly inhibited the proliferation of colon cancer cells. This polyphenolic compound induced caspase-mediated apoptosis in HT-29 cells by modulating the PTEN/PI3K/Akt/mTOR signaling pathway. α-Conidendrin markedly upregulated the protein expression of PTEN and downregulated the protein expression of p-PI3K, p-AKt, and p-mTOR. The protein expression of caspase-3 and caspase-9 enhanced in colon cancer cells following treatment with α-conidendrin. This study also revealed the anti-angiogenic activities of α-conidendrin in the ex vivo and in vitro models. α-Conidendrin significantly downregulated the mRNA expression of HIF-1α, VEGF, MMP-2, and MMP-9 in endothelial cells. These data highlight that α-conidendrin can act as a novel and promising anti-cancer and anti-angiogenic agent for treatment of colon cancer.
Collapse
Affiliation(s)
- Sayeh Mottaghi
- Department of Pediatrics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hassan Abbaszadeh
- Department of Pharmacology, Faculty of Pharmacy, Medicinal Plants Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Armita Valizadeh
- Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Katayoon Hafezi
- Department of Pharmacology, Faculty of Pharmacy, Medicinal Plants Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
3
|
Saloni, Sachan M, Rahul, Verma RS, Patel GK. SOXs: Master architects of development and versatile emulators of oncogenesis. Biochim Biophys Acta Rev Cancer 2025; 1880:189295. [PMID: 40058508 DOI: 10.1016/j.bbcan.2025.189295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/22/2025]
Abstract
Transcription factors regulate a variety of events and maintain cellular homeostasis. Several transcription factors involved in embryonic development, has been shown to be closely associated with carcinogenesis when deregulated. Sry-like high mobility group box (SOX) proteins are potential transcription factors which are evolutionarily conserved. They regulate downstream genes to determine cell fate, via various signaling pathways and cellular processes essential for tissue and organ development. Dysregulation of SOXs has been reported to promote or suppress tumorigenesis by modulating cellular reprogramming, growth, proliferation, angiogenesis, metastasis, apoptosis, immune modulation, lineage plasticity, maintenance of the stem cell pool, therapy resistance and cancer relapse. This review provides a crucial understanding of the molecular mechanism by which SOXs play multifaceted roles in embryonic development and carcinogenesis. It also highlights their potential in advancing therapeutic strategies aimed at targeting SOXs and their downstream effectors in various malignancies.
Collapse
Affiliation(s)
- Saloni
- Cancer and Stem Cell Laboratory, Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India
| | - Manisha Sachan
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India
| | - Rahul
- Department of Surgical Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow 226014, India
| | - Rama Shanker Verma
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India.
| | - Girijesh Kumar Patel
- Cancer and Stem Cell Laboratory, Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj 211004, India.
| |
Collapse
|
4
|
Swallah MS, Bondzie-Quaye P, Yu X, Fetisoa MR, Shao CS, Huang Q. Elucidating the protective mechanism of ganoderic acid DM on breast cancer based on network pharmacology and in vitro experimental validation. Biotechnol Appl Biochem 2025; 72:415-436. [PMID: 39318248 DOI: 10.1002/bab.2673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 09/08/2024] [Indexed: 09/26/2024]
Abstract
Ganoderma lucidum, a popular medicinal fungus, has been utilized to treat a variety of diseases. It possesses a unique therapeutic and pharmacological reputation in suppressing cancer/tumor progression, especially breast cancer, due to its embedded rich bioactive chemical constituents, mainly triterpenoids (ganoderic acids). The most prevalent malignant tumor in women with a high mortality and morbidity rate is breast cancer. Ganoderic acids A, D, DM, F, and H are evidenced in previous research to have breast cancer-preventive properties by exhibiting autophagic and apoptosis, anti-proliferative, and anti-angiogenesis effects. However, the anti-breast cancer mechanism remains unclear. The putative targets of the ganoderic acids were further determined using bioinformatics techniques and molecular docking calculation. Finally, the key targets were verified in vitro. A total of 53 potential target proteins associated with 202 pathways were predicted to be related to breast cancer. The potential targets were narrowed down to six key targets (AKT1, PIK3CA, epidermal growth factor receptor [EGFR], STAT1, ESR1, and CTNNB1), using different algorithms of the CytoHubba plugin, which were further validated using molecular docking analysis. The ganoderic acid DM (GADM) and the targets (PIK3CA and EGFR) with the strongest interactions were validated via MDA-MB-231 and MCF7 cells. The expression level of PIK3CA in both MDA-MB-231 and MCF7 cells was dose-dependently suppressed by GADM, whereas EGFR expression was unexpectedly increased, which warrants further investigation. These data indicated that the network pharmacology-based prediction of GADM targets for treating human breast cancer could be reliable.
Collapse
Affiliation(s)
- Mohammed Sharif Swallah
- CAS Key Laboratory of High Magnetic Field and Iron Beam Physical Biology, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, China
| | - Precious Bondzie-Quaye
- CAS Key Laboratory of High Magnetic Field and Iron Beam Physical Biology, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, China
| | - Xin Yu
- CAS Key Laboratory of High Magnetic Field and Iron Beam Physical Biology, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, China
| | - Monia Ravelonandrasana Fetisoa
- CAS Key Laboratory of High Magnetic Field and Iron Beam Physical Biology, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, China
| | - Chang-Sheng Shao
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Qing Huang
- CAS Key Laboratory of High Magnetic Field and Iron Beam Physical Biology, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, China
| |
Collapse
|
5
|
Vonica RC, Butuca A, Morgovan C, Pumnea M, Cipaian RC, Frum A, Dobrea CM, Vonica-Tincu AL, Pacnejer AM, Ghibu S, Batar F, Gligor FG. Bevacizumab-Insights from EudraVigilance Database on the Assessments of the Safety Profile of Monoclonal Antibodies Used as Targeted Cancer Treatment. Pharmaceuticals (Basel) 2025; 18:501. [PMID: 40283938 PMCID: PMC12030381 DOI: 10.3390/ph18040501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/22/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Worldwide, colon cancer is a major cause of cancer-related mortality, with an increasing incidence influenced by genetic, environmental, and lifestyle factors. Despite advances in diagnosis and personalized treatments, challenges remain in improving patient prognosis, particularly in metastatic colorectal cancer (mCRC). Bevacizumab (BEV), a monoclonal antibody, is widely used in colorectal cancer treatment. This study aimed to analyze adverse events associated with BEV compared with other therapies based on data from the EudraVigilance (EV) database. Methods: A descriptive and disproportionality analysis was conducted on signals reported in the EV database related to BEV. The study included comparisons with other antineoplastic treatments, such as chemotherapy, targeted therapy, and immunotherapy. Patient demographics, severity of adverse drug reactions (ADRs), and distribution patterns were analyzed to assess the safety profile of BEV in colorectal cancer treatment. Results: The majority of the signals for BEV were from patients aged 18-64 years (39.42%) and 65-85 years (34.08%). Hypertension, thromboembolism, proteinuria, and gastrointestinal disorders have been the most frequently reported. Serious ADRs, including gastrointestinal perforations, hemorrhage, and arterial thromboembolism, were observed in 93.74% of Individual Case Safety Reports. BEV was associated with a higher likelihood of vascular and endocrine disorders compared with chemotherapy and other targeted therapies. Immunotherapy was linked to increased immunological ADRs, while BEV demonstrated fewer immune-related toxicities. Conclusions: Continuous monitoring is necessary to optimize patient management, particularly in elderly patients or those with cardiovascular comorbidities. Understanding BEV's safety profile allows for better personalization of treatment strategies, minimizing risks while enhancing therapeutic outcomes.
Collapse
Affiliation(s)
- Razvan Constantin Vonica
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Anca Butuca
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Claudiu Morgovan
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Manuela Pumnea
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Remus Calin Cipaian
- Clinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania;
- County Clinical Emergency Hospital of Sibiu, 2–4 Corneliu Coposu Str., 550245 Sibiu, Romania
| | - Adina Frum
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Carmen Maximiliana Dobrea
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Andreea Loredana Vonica-Tincu
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Aliteia-Maria Pacnejer
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timişoara, Romania
| | - Steliana Ghibu
- Department of Pharmacology, Physiology and Pathophysiology, Faculty of Pharmacy, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Florina Batar
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Felicia Gabriela Gligor
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| |
Collapse
|
6
|
Chitoran E, Rotaru V, Stefan DC, Gullo G, Simion L. Blocking Tumoral Angiogenesis VEGF/VEGFR Pathway: Bevacizumab-20 Years of Therapeutic Success and Controversy. Cancers (Basel) 2025; 17:1126. [PMID: 40227654 PMCID: PMC11988089 DOI: 10.3390/cancers17071126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/15/2025] Open
Abstract
The "angiogenesis switch"-defined as the active process by which solid tumors develop their own circulation-plays an important role in both tumoral growth and propagation. As the malignant tumor grows and reaches a critical size, the metabolic needs as a function of an ever-increasing distance to the nearest emergent blood vessel, can no longer be covered by the microenvironment of the peritumoral tissue. Although a relatively discrete process, the "angiogenic switch" acts as a limiting stage of tumoral development present from the avascular hyperplasia phase to the vascularized neoplastic phase, providing support for tumor expansion and metastasis. Over time, research has focused on blocking the angiogenetic pathways (such as VEGF/VEGFR signaling axis) leading to the development of targeted therapeutic agents such as Bevacizumab. Objectives: We conducted a review of the molecular principles of tumoral angiogenesis and we tried to follow the history of Bevacizumab from its first approval for human usage 20 years ago to current days, focusing on the impact this agent had in solid tumor therapy. A comprehensive review of clinical trials pertaining to Bevacizumab (from the era of the preclinic trials leading to approval for human usage, to the more recent randomized trial focusing on combination targeted therapy) further details the role of this drug. We aimed to establish if this ancient drug continues to have a place in modern oncology. Conclusions: Bevacizumab, one of the first drugs targeting tumoral microenvironment, remains one of the most important oncologic agents blocking the VEGF/VEGFR angiogenic pathway. otherwise, history of 20 years marked by numerous controversies (ranging from methodological errors of clinical trials to withdrawal of approval for human usage in breast cancer patients, from discussions about severe side effects to resistance to therapy and limited efficacity), Bevacizumab continues to provide an optimal therapeutic option for many solid tumors that previously had little to no means of treatment, improving otherwise bleak outcomes. Even in the era of personalized precision oncology, Bevacizumab continues to be a key element in many therapeutic regimens both as monotherapy and in combination with newer targeted agents.
Collapse
Affiliation(s)
- Elena Chitoran
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Vlad Rotaru
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Daniela-Cristina Stefan
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Giuseppe Gullo
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, University of Palermo, 90146 Palermo, Italy
| | - Laurentiu Simion
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| |
Collapse
|
7
|
Rana JN, Mumtaz S. Prunin: An Emerging Anticancer Flavonoid. Int J Mol Sci 2025; 26:2678. [PMID: 40141319 PMCID: PMC11942023 DOI: 10.3390/ijms26062678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Despite the substantial advances in cancer therapies, developing safe and effective treatment methodologies is critical. Natural (plant-derived compounds), such as flavonoids, might be crucial in developing a safe treatment methodology without toxicity toward healthy tissues. Prunin is a flavonoid with the potential to be used in biomedical applications. Prunin has yet to undergo thorough scientific research, and its precise molecular mechanisms of action remain largely unexplored. This review summarizes the therapeutic potential of prunin for the first time, focusing on its underlying mechanisms as an anticancer compound. Prunin has gained significant attention due to its antioxidant, anti-inflammatory, and anticancer effects. This review aims to unlock how prunin functions at the molecular level to exert its anticancer effects, primarily modulating key cellular pathways. Furthermore, we have discussed the prunin's potential as an adjunctive therapy with conventional treatments, highlighting its ability to strengthen treatment responses while decreasing drug resistance. Moreover, the discussion probes into innovative delivery methods, particularly nanoformulations, that might address prunin's bioavailability, solubility, and stability limitations and optimize its therapeutic application. By providing a comprehensive analysis of prunin's properties, this review aims to stimulate further exploration of using prunin as an anticancer agent, thereby progressing the development of targeted, selective, safe, and effective therapeutic methods.
Collapse
Affiliation(s)
- Juie Nahushkumar Rana
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| | - Sohail Mumtaz
- Department of Chemical and Biological Engineering, Gachon University, 1342 Seongnamdaero, Sujeong-gu, Seongnam-si 13120, Republic of Korea
| |
Collapse
|
8
|
Kotsifaki A, Kalouda G, Maroulaki S, Foukas A, Armakolas A. The Genetic and Biological Basis of Pseudoarthrosis in Fractures: Current Understanding and Future Directions. Diseases 2025; 13:75. [PMID: 40136615 PMCID: PMC11941250 DOI: 10.3390/diseases13030075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/27/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Pseudoarthrosis-the failure of normal fracture healing-remains a significant orthopedic challenge affecting approximately 10-15% of long bone fractures, and is associated with significant pain, prolonged disability, and repeated surgical interventions. Despite extensive research into the pathophysiological mechanisms of bone healing, diagnostic approaches remain reliant on clinical findings and radiographic evaluations, with little innovation in tools to predict or diagnose non-union. The present review evaluates the current understanding of the genetic and biological basis of pseudoarthrosis and highlights future research directions. Recent studies have highlighted the potential of specific molecules and genetic markers to serve as predictors of unsuccessful fracture healing. Alterations in mesenchymal stromal cell (MSC) function, including diminished osteogenic potential and increased cellular senescence, are central to pseudoarthrosis pathogenesis. Molecular analyses reveal suppressed bone morphogenetic protein (BMP) signaling and elevated levels of its inhibitors, such as Noggin and Gremlin, which impair bone regeneration. Genetic studies have uncovered polymorphisms in BMP, matrix metalloproteinase (MMP), and Wnt signaling pathways, suggesting a genetic predisposition to non-union. Additionally, the biological differences between atrophic and hypertrophic pseudoarthrosis, including variations in vascularity and inflammatory responses, emphasize the need for targeted approaches to management. Emerging biomarkers, such as circulating microRNAs (miRNAs), cytokine profiles, blood-derived MSCs, and other markers (B7-1 and PlGF-1), have the potential to contribute to early detection of at-risk patients and personalized therapeutic approaches. Advancing our understanding of the genetic and biological underpinnings of pseudoarthrosis is essential for the development of innovative diagnostic tools and therapeutic strategies.
Collapse
Affiliation(s)
- Amalia Kotsifaki
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.K.); (G.K.); (S.M.)
| | - Georgia Kalouda
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.K.); (G.K.); (S.M.)
| | - Sousanna Maroulaki
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.K.); (G.K.); (S.M.)
| | - Athanasios Foukas
- Third Department of Orthopaedic Surgery, “KAT” General Hospital of Athens, 2, Nikis Street, 14561 Kifissia, Greece;
| | - Athanasios Armakolas
- Physiology Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.K.); (G.K.); (S.M.)
| |
Collapse
|
9
|
Tamas F, Tamas CI, Suciu BA, Balasa AF. Extracellular Vesicle-Associated Angiopoietin-2 and Cell Migration-Inducing Protein in Lung Cancer Progression and Brain Metastases. Cureus 2025; 17:e80200. [PMID: 40190907 PMCID: PMC11972550 DOI: 10.7759/cureus.80200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Angiopoietin-2 (ANGPT2) and cell migration-inducing protein (CEMIP) are key regulators of angiogenesis, extracellular matrix remodeling, and metastatic progression in various cancers, including lung cancer (LC). The presence of these biomarkers in extracellular vesicles (EVs) may offer valuable insights into the molecular mechanisms underlying LC progression and metastasis. Extracellular vesicles play a critical role in LC by enhancing intercellular communication and supporting processes such as angiogenesis, immune evasion, and metastasis, transferring key molecules like vascular endothelial growth factor (VEGF) and pro-angiogenic microRNAs (miRNAs). METHODS This study aimed to investigate the presence and quantification of ANGPT2 and CEMIP in the cargo of EVs isolated from plasma samples obtained from the peripheral venous blood of patients with localized lung cancer (LLC group), lung cancer with brain metastases (LCM group), and healthy controls (HC group). EVs were isolated using the density gradient ultracentrifugation method, and their characterization was performed through scanning and transmission electron microscopy as well as flow cytometry. Western blot analysis was used to identify ANGPT2 and CEMIP in EV cargo, and band intensity from western blot images was quantified using specialized software. RESULTS The expression of ANGPT2 and CEMIP in EV cargo was significantly higher in the oncologic groups (LLC and LCM combined) compared to the HC group. Notably, EV CEMIP levels were, on average, 59% higher in patients with brain metastases than in those with localized lung cancer. Following surgical resection, postoperative EV ANGPT2 and EV CEMIP levels decreased by 36% and 8.5%, respectively, in the LLC group, and by 40% and 4.6%, respectively, in the LCM group. CONCLUSION These findings emphasize the potential of ANGPT2 and CEMIP as biomarkers for LC progression and prognosis. To our knowledge, no previous study has evaluated the presence and quantification of ANGPT2 and CEMIP in EV cargo from lung cancer patients. To further validate their role in cancer progression, functional studies should explore the mechanistic effects of EV-associated ANGPT2 and CEMIP on angiogenesis, immune modulation, cell migration, extracellular matrix remodeling, and tumor progression in lung cancer models.
Collapse
Affiliation(s)
- Flaviu Tamas
- Neurosurgery, Doctoral School of Medicine and Pharmacy, "George Emil Palade" University of Medicine, Pharmacy, Science and Technology, Târgu Mureș, ROU
- Neurosurgery, Emergency Clinical County Hospital, Târgu Mureș, ROU
| | - Corina I Tamas
- Neurosurgery, Doctoral School of Medicine and Pharmacy, "George Emil Palade" University of Medicine, Pharmacy, Science and Technology, Târgu Mureș, ROU
- Neurosurgery, Emergency Clinical County Hospital, Târgu Mureș, ROU
| | - Bogdan A Suciu
- Thoracic Surgery, Emergency Clinical County Hospital, Târgu Mureș, ROU
| | - Adrian F Balasa
- Neurosurgery, Emergency Clinical County Hospital, Târgu Mureș, ROU
| |
Collapse
|
10
|
Wang R, Li W, Cao H, Zhang L. Decoding the Tumor-Associated Microbiota: From Origins to Nanomedicine Applications in Cancer Therapy. BIOLOGY 2025; 14:243. [PMID: 40136500 PMCID: PMC11940167 DOI: 10.3390/biology14030243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
Growing evidence reveals that the tumor microbiome-comprising distinct microbial communities within neoplastic tissues-exerts a profound influence on cancer initiation, progression, and therapeutic response. These microbes actively reshape the tumor microenvironment (TME) through metabolite secretion, the modulation of immune pathways, and direct interactions with host cells, thereby affecting tumor biology and therapeutic outcomes. Despite substantial heterogeneity among cancer types, recent insights underscore the tumor microbiome's potential as both a diagnostic/prognostic biomarker and a targetable component for innovative treatments. In this review, we synthesize emerging knowledge on the mechanistic roles of tumor-associated microbiota in shaping the TME, with a focus on how these discoveries can guide novel therapeutic strategies. We further explore interdisciplinary advances, including the convergence of microbiomics and nanotechnology, to enhance drug delivery, circumvent resistance, and foster TME remodeling. By highlighting these cutting-edge developments, our review underscores the transformative potential of integrating tumor microbiome research into precision oncology and advancing more personalized cancer therapies.
Collapse
Affiliation(s)
- Ruiqi Wang
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (R.W.); (W.L.)
| | - Weizheng Li
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (R.W.); (W.L.)
| | - Hongqian Cao
- Department of Health Inspection and Quarantine, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lei Zhang
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (R.W.); (W.L.)
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| |
Collapse
|
11
|
Mullick Chowdhury S, Hong F, Rolfo C, Li Z, He K, Wesolowski R, Mortazavi A, Meng L. CNPY2 in Solid Tumors: Mechanisms, Biomarker Potential, and Therapeutic Implications. BIOLOGY 2025; 14:214. [PMID: 40001982 PMCID: PMC11851889 DOI: 10.3390/biology14020214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/10/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025]
Abstract
Canopy FGF signaling regulator 2 (CNPY2) has emerged as a crucial player in cancer development by promoting cell proliferation, tissue repair, and angiogenesis. This review synthesizes the current understanding of CNPY2's role in solid tumors, particularly renal cell carcinoma, prostate cancer, hepatocellular carcinoma, and non-small-cell lung cancer. CNPY2 modulates key pathways such as p53, MYLIP, NF-κB, and AKT/GSK3β, thereby driving tumor growth and progression. In renal cell carcinoma, CNPY2 paradoxically promotes tumor growth through p53 upregulation, while in hepatocellular carcinoma, CNPY2 drives cell cycle progression via p53 destabilization. In prostate cancer, it enhances tumor progression by stabilizing androgen receptors through MYLIP interaction, and in non-small-cell lung cancer, it contributes to chemoresistance and metastasis through NF-κB and AKT/GSK3β signaling. Additionally, CNPY2 influences the tumor microenvironment, impacting immune function and metastatic potential. As a potential biomarker, CNPY2 shows promise for cancer detection and prognosis, particularly when used in combination with other markers. Early therapeutic strategies, including siRNA and miRNA approaches, are under exploration, though challenges remain due to CNPY2's expression in normal tissues and potential off-target effects. This review underscores the need for further research to fully elucidate CNPY2's oncogenic mechanisms and develop targeted therapies. Improved understanding of CNPY2's diverse roles may lead to novel diagnostic and therapeutic approaches in solid tumors.
Collapse
Affiliation(s)
- Sayan Mullick Chowdhury
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
| | - Feng Hong
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Christian Rolfo
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
| | - Zihai Li
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Kai He
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Robert Wesolowski
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
| | - Lingbin Meng
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
| |
Collapse
|
12
|
Fatima R, Batool A, Ijaz T, Khan SU, Akhtar Y, Ercisli S, Almutairi SM, Elshikh MS, Saleem A, Javed MA. Growth suppressing effect of Fagonia arabica extracts on cancerous cell line. Cytotechnology 2025; 77:36. [PMID: 39764423 PMCID: PMC11700075 DOI: 10.1007/s10616-024-00663-w] [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: 06/06/2024] [Accepted: 10/16/2024] [Indexed: 03/08/2025] Open
Abstract
Homeostasis of tissues requires a complex balance between cell proliferation and cell death. The disruption of this balance leads to tumors. Cancer is a mortal disease that spreads all over the body, it is an irregular cell growth. All the tumors are not cancerous, benign tumors do not spread to all body parts. Cancer is caused when cells start to grow out of control, known as increased proliferation. Cancer is of many kinds and the reason for cancer is the uncontrolled growth of cells that can affect any tissue of the body Conventional plants are precious sources of novel cytotoxic agents and are still in a better role in health concerns. The study aimed to evaluate the effect of in vitro anti-proliferative activity of Fagonia arabica plant extracts against HeLa and HepG2 cell lines and compared with normal BHK cells. Fagonia arabica plant (seed, leaves, fruit peel, fruit pulp, stem, and root) is extracted in their solvent's ethanol, ethyl acetate, and petroleum ether. For the estimation of anti-proliferation, cell viability, and cell death in HeLa, HepG2, BHK, MTT assay was done. The angiogenic potential was checked via Immunocytochemistry and ELISA of VEGF. Immunocytochemistry and ELISA of Annexin-V were performed for the estimation of apoptosis in HeLa cells and BHK. Furthermore, Immunocytochemistry and ELISA for p53 were also performed. Cancer cells (HeLa, HepG2) showed reduced angiogenesis, low proliferation, increased apoptotic level, reduced viability, and increased cell death. It is found that Fagonia arabica plant extract induces apoptosis along with inhibition of proliferation and angiogenesis may strongly have profound effects on growth suppression of HeLa and HepG2 cell lines.
Collapse
Affiliation(s)
- Rida Fatima
- Faculty of Allied Health Sciences, The Superior University, Lahore, 55150 Pakistan
| | - Aqsa Batool
- Department of Chemistry, Government College University, Lahore, 54000 Pakistan
| | - Tabinda Ijaz
- Department of Clinical and Molecular Medicines, University of Palermo, 90133 Palermo, Italy
| | - Sajjad Ullah Khan
- Faculty of Allied Health Sciences, The University of Lahore, Lahore, 54810 Pakistan
| | - Yasmin Akhtar
- Department of Chemistry, Government College University, Lahore, 54000 Pakistan
| | - Sezai Ercisli
- Department of Horticulture, Agricultural Faculty, Ataturk University, 25240 Erzurum, Turkey
| | - Saeedah Musaed Almutairi
- Department of Botany and Microbiology, College of Science, King Saud University, 11451 Riyadh, Saudi Arabia
| | - Mohamed Soliman Elshikh
- Department of Botany and Microbiology, College of Science, King Saud University, 11451 Riyadh, Saudi Arabia
| | - Aroona Saleem
- Department of Microbiology, Dr. Ikram-Ul-Haq Institute of Industrial Biotechnology (IIIB), Government College University, Lahore, 54000 Pakistan
| | - Muhammad Ammar Javed
- Department of Microbiology, Dr. Ikram-Ul-Haq Institute of Industrial Biotechnology (IIIB), Government College University, Lahore, 54000 Pakistan
| |
Collapse
|
13
|
Kustiati U, Nugrahaningsih DAA, Kusindarta DL, Wihadmadyatami H. Lung cancer: Animal model of lung cancer, molecular carcinogenesis of lung cancer, and antitumor effect of Ocimum sanctum against lung cancer. Open Vet J 2025; 15:482-503. [PMID: 40201854 PMCID: PMC11974298 DOI: 10.5455/ovj.2025.v15.i2.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 12/03/2024] [Indexed: 04/10/2025] Open
Abstract
Lung cancer is the leading cause of fatalities related to cancer globally. There are numerous ways to treat lung cancer, including surgery, chemotherapy, and radiation. Since these treatments have not yet shown satisfactory results, more research into the underlying mechanisms and different approaches to therapy and prevention are needed. Animal models are essential to the study of lung cancer because they offer priceless information about the etiology, course, and possible treatments for the illness. The therapeutic application of phytochemicals and medicinal plants to treat cancer-related compounds has gained attention subsequently. In addition to discussing the molecular carcinogenic and antitumor effects of the herbal treatment Ocimum sanctum (OS) in connection to lung cancer, this review will address the current awareness regarding lung cancer in animal models. The multitude of animal models used in lung cancer research-such as genetically modified mice, carcinogen-induced models, and xenograft induction-provides a solid foundation for understanding the illness. By easing the examination of the environmental and genetic factors involved and enhancing the analysis of possibilities for treatment, these models eventually assist in the further development of lung cancer therapy. Additionally, using the herb plant OS is essential for both treating and preventing lung cancer. Standardizing dosages and enforcing laws on the use of herbal medications require more in-depth investigation.
Collapse
Affiliation(s)
- Ulayatul Kustiati
- Post-Graduate School of Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Laboratory of Pharmacology, Faculty of Veterinary Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Dwi Aris Agung Nugrahaningsih
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Dwi Liliek Kusindarta
- Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Hevi Wihadmadyatami
- Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
14
|
Abd Elhameed AA, Ali AR, Ghabbour HA, Bayomi SM, El-Gohary NS. Probing structural requirements for thiazole-based mimetics of sunitinib as potent VEGFR-2 inhibitors. RSC Med Chem 2025:d4md00754a. [PMID: 39850549 PMCID: PMC11753467 DOI: 10.1039/d4md00754a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/29/2024] [Indexed: 01/25/2025] Open
Abstract
Novel thiazole analogs 3a, 3b, 4, 5, 6a-g, 8a, 8b, 9a-c, 10a-d and 11 were designed and synthesized as molecular mimetics of sunitinib. In vitro antitumor activity of the obtained compounds was investigated against HepG2, HCT-116, MCF-7, HeP-2 and HeLa cancer cell lines. The obtained data showed that compounds 3b and 10c are the most potent members toward HepG2, HCT-116, MCF-7 and HeLa cells. Moreover, compounds 3a, 3b, 6g, 8a and 10c were assessed for their in vitro VEGFR-2 inhibitory activity. Results proved that compound 10c exhibited outstanding VEGFR-2 inhibition (IC50 = 0.104 μM) compared to sunitinib. Compound 10c paused the G0-G1 phase of the cell cycle in HCT-116 and MCF-7 cells and the S phase in HeLa cells. Additionally, compound 10c elevated caspase-3/9 levels in HCT-116 and HeLa cells, leading to cancer cell death via apoptosis. Furthermore, compound 10c showed a significant reduction in tumor volume in Swiss albino female mice as an in vivo breast cancer model. Docking results confirmed the tight binding interactions of compound 10c with the VEGFR-2 binding site, with its binding energy surpassing that of sunitinib. In silico PK studies predicted compound 10c to have good oral bioavailability and a good drug score with low human toxicity risks.
Collapse
Affiliation(s)
- Alaa A Abd Elhameed
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University Mansoura 35516 Egypt
| | - Ahmed R Ali
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University Mansoura 35516 Egypt
| | - Hazem A Ghabbour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University Mansoura 35516 Egypt
| | - Said M Bayomi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University Mansoura 35516 Egypt
| | - Nadia S El-Gohary
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University Mansoura 35516 Egypt
| |
Collapse
|
15
|
Jafri Z, Li Y, Zhang J, O’Meara CH, Khachigian LM. Jun, an Oncological Foe or Friend? Int J Mol Sci 2025; 26:555. [PMID: 39859271 PMCID: PMC11766113 DOI: 10.3390/ijms26020555] [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: 11/28/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 01/27/2025] Open
Abstract
Jun/JUN is a basic leucine zipper (bZIP) protein and a prototypic member of the activator protein-1 (AP-1) family of transcription factors that can act as homo- or heterodimers, interact with DNA elements and co-factors, and regulate gene transcription. Jun is expressed by both immune and inflammatory cells. Jun is traditionally seen as an oncoprotein that regulates processes involved in transformation and oncogenesis in human tumours. This article examines the traditional view that Jun plays a permissive role in cancer development and progression, whilst exploring emerging evidence supporting Jun's potential to prevent immune cell exhaustion and promote anti-tumour efficacy.
Collapse
Affiliation(s)
- Zuhayr Jafri
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Yue Li
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jingwen Zhang
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Connor H. O’Meara
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Division of Head & Neck Oncology and Microvascular Reconstruction, Department of Otolaryngology, Head & Neck Surgery, University of Virginia Health Services, Charlottesville, VA 22903, USA
- Department of Otolaryngology, Head & Neck Surgery, Australian National University, Acton, ACT 0200, Australia
| | - Levon M. Khachigian
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
16
|
Huang X, Ye H, Hu Y, Lei Y, Tian Y, Huang X, Zhang J, Zhang Y, Gui B, Liu Q, Zhang G, Deng Q. Ultrasound super-resolution imaging for non-invasive assessment of microvessel in prostate lesion. Cancer Imaging 2025; 25:1. [PMID: 39773358 PMCID: PMC11706184 DOI: 10.1186/s40644-024-00819-z] [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: 09/04/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Prostate cancer (PCa) is the leading cause of cancer-related morbidity and mortality in men worldwide. An early and accurate diagnosis is crucial for effective treatment and prognosis. Traditional invasive procedures such as image-guided prostate biopsy often cause discomfort and complications, deterring some patients from undergoing these necessary tests. This study aimed to explore the feasibility and clinical value of using ultrasound super-resolution imaging (US SRI) for non-invasively assessing the microvessel characteristics of prostate lesion. METHODS This study included 127 patients with prostate lesion who presented at Renmin Hospital of Wuhan University between November 2023 and June 2024 were included in this study. All the patients underwent transrectal US (TRUS), contrast-enhanced US (CEUS), and US SRI. CEUS parameters of time-intensity curve (TIC): arrival time (AT), rising time (RT), time to peak (TTP), peak intensity (PKI), falling time (FT), mean transit time (MTT), ascending slope (AS), descending slope (DS), D/A slope ratio (SR), and area under the TIC (AUC). US SRI parameters: microvessel density (MVD), microvessel diameter (D), microvessel velocity (V), microvessel tortuosity (T), and fractal number (FN), were analyzed and compared between prostate benign and malignant lesion. RESULTS The tumor markers of prostate in the malignant group were all higher than those in the benign group, and the differences were statistically significant (P < 0.001). The TIC parameters of CEUS revealed that the PKI, AS, DS, and AUC were significantly higher in the malignant group than in the benign group (P < 0.001), whereas the RT, TTP and FT in the malignant group were significantly lower (P < 0.001). Malignant lesion exhibited significantly higher MVD, larger D, faster V, greater T, and more complex FN than benign lesion (P < 0.001). CONCLUSIONS US SRI is a promising non-invasive imaging modality that can provide detailed microvessel characteristics of prostate lesion, offering an advancement in the differential diagnosis for prostate lesion. And, US SRI may be a valuable tool in clinical practice with its ability to display and quantify microvessel with high precision.
Collapse
Affiliation(s)
- Xin Huang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Huarong Ye
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Yugang Hu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yumeng Lei
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Yi Tian
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Xingyue Huang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jun Zhang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yao Zhang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bin Gui
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qianhui Liu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ge Zhang
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China.
- Physics for Medicine Paris, Inserm U1273, ESPCI Paris, PSL University, Paris, 75015, France.
| | - Qing Deng
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
17
|
Guo Y, Yang P, Wu Z, Zhang S, You F. Mechanisms of Astragalus membranaceus (Fisch.) Bge. var. mongholicus (Bge.) Hsiao (huang qi) and Angelica sinensis (Oliv.) Diels (dang gui) in Ameliorating Hypoxia and Angiogenesis to Delay Pulmonary Nodule Malignant Transformation. Integr Cancer Ther 2025; 24:15347354241311917. [PMID: 39882753 PMCID: PMC11780663 DOI: 10.1177/15347354241311917] [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: 09/19/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025] Open
Abstract
Screening for pulmonary nodules (PN) using low-dose CT has proven effective in reducing lung cancer (LC) mortality. However, current treatments relying on follow-up and surgical excision fail to fully address clinical needs. Pathological angiogenesis plays a pivotal role in supplying oxygen necessary for the progression of PN to LC. The interplay between hypoxia and angiogenesis establishes a vicious cycle, rendering anti-angiogenesis therapy alone insufficient to prevent PN to LC transformation. In traditional Chinese medicine (TCM), PN is referred to as "Feiji," which is mainly attributed to Qi and blood deficiency, correspondingly, the most commonly prescribed medicines are Astragalus membranaceus (Fisch.) Bge. var. mongholicus (Bge.) Hsiao (huang qi) (AR) and Angelica sinensis (Oliv.) Diels (dang gui) (ARS). Modern pharmacological studies have demonstrated that AR and ARS possess immune-enhancing, anti-tumor, anti-inflammatory, and anti-angiogenic properties. However, the precise mechanisms through which AR and ARS exert anti-angiogenic effects to delay PN progression to LC remain inadequately understood. This review explores the critical roles of hypoxia and angiogenesis in the transition from PN to LC. It emphasizes that, compared to therapies targeting angiogenic growth factors alone, AR, ARS, and their compound-based prescriptions offer additional benefits. These include ameliorating hypoxia by restoring blood composition, enhancing vascular structure, accelerating circulation, promoting vascular normalization, and blocking or inhibiting various pro-angiogenic expressions and receptor interactions. Collectively, these actions inhibit angiogenesis and delay the PN-to-LC transformation. Finally, this review summarizes recent advancements in related research, identifies existing limitations and gaps in knowledge, and proposes potential strategies and recommendations to address these challenges.
Collapse
Affiliation(s)
- Ying Guo
- Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peng Yang
- Chengdu Fifth People’s Hospital, Chengdu, China
| | - Zihong Wu
- Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sanyin Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chengdu Integrated TCM & Western Medicine Hospital, Chengdu, China
| | - Fengming You
- Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
18
|
Pandey P, Nautiyal G, Purohit D, Lata S, Kumar V, Makhija M, Manchanda D, Minocha N, Kumar S, Kaushik D. Role of Nanoformulations in the Treatment of Lung Cancer. RECENT PATENTS ON NANOTECHNOLOGY 2025; 19:407-433. [PMID: 38321901 DOI: 10.2174/0118722105264531231205042817] [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: 07/14/2023] [Revised: 10/19/2023] [Accepted: 11/06/2023] [Indexed: 02/08/2024]
Abstract
Lung cancer is the second deadliest disease in the world. A major portion of deaths related to cancer are due to lung cancer in both males and females. Interestingly, unbelievable advances have occurred in recent years through the use of nanotechnology and development in both the diagnosis and treatment of lung cancer. Due to their in vivo stability, the nanotechnology-based pharmacological system gained huge attractiveness, solubility, absorption from the intestine, pharmacological effectiveness, etc. of various anticancer agents. However, this field needs to be utilized more to get maximum results in the treatment of lung cancer, along with wider context medicines. In the present review, authors have tried to concentrate their attention on lung cancer`s difficulties along with the current pharmacological and diagnostic situation, and current advancements in approaches based on nanotechnology for the treatment and diagnosis of lung cancer. While nanotechnology offers these promising avenues for lung cancer diagnosis and treatment, it is important to acknowledge the need for careful evaluation of safety, efficacy, and regulatory approval. With continued research and development, nanotechnology holds tremendous potential to revolutionize the management of lung cancer and improve patient outcomes. The review also highlights the involvement of endocrine systems, especially estrogen in lung cancer proliferation. Some of the recent clinical trials and patents on nanoparticle-based formulations that have applications in the treatment and diagnosis of lung cancer are also discussed.
Collapse
Affiliation(s)
- Parijat Pandey
- Department of Pharmaceutical Sciences, Gurugram University, Gurugram, 122018, Haryana, India
| | - Gunjan Nautiyal
- Department of Pharmaceutical Sciences, Gurugram University, Gurugram, 122018, Haryana, India
| | - Deepika Purohit
- Department of Pharmaceutical Sciences, Indira Gandhi University, Meerpur, Rewari, 123401, Haryana, India
| | - Sneh Lata
- Department of Pharmaceutical Sciences, Indira Gandhi University, Meerpur, Rewari, 123401, Haryana, India
| | - Virender Kumar
- College of Pharmacy, Pandit Bhagwat Dayal Sharma University of Health Sciences, Rohtak, 124001, Haryana, India
| | - Manish Makhija
- Department of Pharmaceutical Sciences, Indira Gandhi University, Meerpur, Rewari, 123401, Haryana, India
| | - Deeksha Manchanda
- Department of Pharmaceutical Sciences, Indira Gandhi University, Meerpur, Rewari, 123401, Haryana, India
| | - Neha Minocha
- Amity Institute of Pharmacy, Amity University, Gurugram, 122413, Haryana, India
- Chitkara University School of Pharmacy, Chitkara University, Himachal Pradesh, 174103, India
| | - Sunil Kumar
- Department of Pharmaceutical Sciences, Indira Gandhi University, Meerpur, Rewari, 123401, Haryana, India
| | - Deepak Kaushik
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| |
Collapse
|
19
|
Yang L, Shen A, Wang R, Zheng Z. S100A16 stabilizes the ITGA3‑mediated ECM‑receptor interaction pathway to drive the malignant properties of lung adenocarcinoma cells via binding MOV10. Mol Med Rep 2025; 31:11. [PMID: 39450567 PMCID: PMC11541165 DOI: 10.3892/mmr.2024.13376] [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: 05/11/2024] [Accepted: 08/30/2024] [Indexed: 10/26/2024] Open
Abstract
Lung adenocarcinoma (LUAD) is highly associated with lung cancer‑associated mortality. Notably, S100 calcium‑binding protein A16 (S100A16) has been increasingly considered to have prognostic value in LUAD; however, the underlying mechanism remains unknown. In the present study, S100A16 expression levels in LUAD tissues and cells were respectively analyzed by the UALCAN database and western blotting. Cell Counting Kit‑8 and 5‑ethynyl‑2'‑deoxyuridine assays were used to examine cell proliferation, whereas wound healing, Transwell and tube formation assays were used to assess cell migration, invasion and angiogenesis, respectively. Western blotting was also used to examine the expression levels of proteins associated with metastasis, angiogenesis, focal adhesion and the extracellular matrix (ECM)‑receptor interaction pathways. The relationship between S100A16 and Mov10 RNA helicase (MOV10) was predicted by bioinformatics tools, and was verified using a co‑immunoprecipitation assay. Furthermore, the interaction between MOV10 and integrin α3 (ITGA3) was verified by RNA immunoprecipitation assay, and the actinomycin D assay was used to detect ITGA3 mRNA stability. The results demonstrated that S100A16 expression was increased in LUAD tissues and cell lines, and was associated with unfavorable outcomes. Knocking down S100A16 expression hindered the proliferation, migration, invasion and angiogenesis of LUAD cells. Furthermore, S100A16 was shown to bind to MOV10 and positively modulate MOV10 expression in LUAD cells, while MOV10 overexpression partially reversed the suppressive role of S100A16 knockdown on the aggressive phenotypes of LUAD cells. Furthermore, it was demonstrated that S100A16 regulated the stability of ITGA3 mRNA via MOV10 to mediate ECM‑receptor interactions. In conclusion, S100A16 may bind to MOV10 to stabilize ITGA3 mRNA and regulate ECM‑receptor interactions, hence contributing to the malignant progression of LUAD.
Collapse
Affiliation(s)
- Lianren Yang
- Department of Medical Oncology, Taihe County People's Hospital, Fuyang, Anhui 236600, P.R. China
| | - Ajuan Shen
- Department of Medical Oncology, Taihe County People's Hospital, Fuyang, Anhui 236600, P.R. China
| | - Rujun Wang
- Department of Medical Oncology, Taihe County People's Hospital, Fuyang, Anhui 236600, P.R. China
| | - Zhihui Zheng
- Department of Medical Oncology, Taihe County People's Hospital, Fuyang, Anhui 236600, P.R. China
| |
Collapse
|
20
|
Chauhan P, Pandey P, Ramniwas S, Khan F, Maqsood R. Deciphering the Correlation between the Emergence of Lung Carcinoma Associated with Tuberculosis-related Inflammation. Endocr Metab Immune Disord Drug Targets 2025; 25:291-299. [PMID: 38831573 DOI: 10.2174/0118715303301146240522095638] [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: 01/24/2024] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 06/05/2024]
Abstract
Lung cancer and tuberculosis (TB) are classified as the second-most life-threatening diseases globally. They both are exclusively represented as major public health risks and might exhibit similar symptoms, occasionally diagnosed simultaneously. Several epidemiological studies suggest that TB is a significant risk factor for the progression of lung cancer. The staggering mortality rates of pulmonary disorders are intrinsically connected to lung cancer and TB. Numerous factors play a pivotal role in the development of TB and may promote lung carcinogenesis, particularly among the geriatric population. Understanding the intricacies involved in the association between lung carcinogenesis and TB has become a crucial demand of current research. Consequently, this study aims to comprehensively review current knowledge on the relationship between tuberculosis-related inflammation and the emergence of lung carcinoma, highlighting the impact of persistent inflammation on lung tissue, immune modulation, fibrosis, aspects of reactive oxygen species, and an altered microenvironment that are linked to the progression of tuberculosis and subsequently trigger lung carcinoma.
Collapse
Affiliation(s)
- Prashant Chauhan
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, Uttar Pradesh, India
| | - Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, Uttar Pradesh, India
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Seema Ramniwas
- University Centre of Research and Development, University Institute of Biotechnology, Chandigarh University Gharuan, Mohali, Punjab, India
| | - Fahad Khan
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, Uttar Pradesh, India
| | - Ramish Maqsood
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
21
|
Elbadawi M, Efferth T. In Vivo and Clinical Studies of Natural Products Targeting the Hallmarks of Cancer. Handb Exp Pharmacol 2025; 287:95-121. [PMID: 38797749 DOI: 10.1007/164_2024_716] [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: 05/29/2024]
Abstract
Despite more than 200 approved anticancer agents, cancer remains a leading cause of death worldwide due to disease complexity, tumour heterogeneity, drug toxicity, and the emergence of drug resistance. Accordingly, the development of chemotherapeutic agents with higher efficacy, a better safety profile, and the capability of bypassing drug resistance would be a cornerstone in cancer therapy. Natural products have played a pivotal role in the field of drug discovery, especially for the pharmacotherapy of cancer, infectious, and chronic diseases. Owing to their distinctive structures and multiple mechanistic activities, natural products and their derivatives have been utilized for decades in cancer treatment protocols. In this review, we delve into the potential of natural products as anticancer agents by targeting cancer's hallmarks, including sustained proliferative signalling, evading growth suppression, resisting apoptosis and cell death, enabling replicative immortality, inducing angiogenesis, and activating invasion and metastasis. We highlight the molecular mechanisms of some natural products, in vivo studies, and promising clinical trials. This review emphasizes the significance of natural products in fighting cancer and the need for further studies to uncover their fully therapeutic potential.
Collapse
Affiliation(s)
- Mohamed Elbadawi
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
22
|
Avolio E, Olivito I, Leo A, De Matteo C, Guarnieri L, Bosco F, Mahata SK, Minervini D, Alò R, De Sarro G, Citraro R, Facciolo RM. Vasostatin-1 restores autistic disorders in an idiopathic autism model (BTBR T+ Itpr3 tf/J mice) by decreasing hippocampal neuroinflammation. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111131. [PMID: 39209101 DOI: 10.1016/j.pnpbp.2024.111131] [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: 04/10/2024] [Revised: 08/15/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Chromogranin A (CgA), a ∼ 49 kDa acidic secretory protein, is ubiquitously distributed in endocrine and neuroendocrine cells and neurons. As a propeptide, CgA is proteolytically cleaved to generate several peptides of biological importance, including pancreastatin (PST: hCgA250-301), Vasostatin 1 (VS1: hCgA1-76), and catestatin (CST: CgA 352-372). VS1 represents the most conserved fragment of CgA. A 20 amino acid domain within VS1 (CgA 47-66) exhibits potent antimicrobial and anti-inflammatory activities. Autism is known to be associated with inflammation. Therefore, we seek to test the hypothesis that VS1 modulates autism behaviors by reducing inflammation in the hippocampus. Treatment of C57BL/6 (B6) and BTBR (a mouse model of idiopathic autism) mice with VS1 revealed the following: BTBR mice showed a significant decrease in chamber time in the presence of a stranger or a novel object. Treatment with VS1 significantly increased chamber time in both cases, underscoring a crucial role for VS1 in improving behavioral deficits in BTBR mice. In contrast to chamber time, sniffing time in BTBR mice in the presence of a stranger was less compared to B6 control mice. VS1 did not improve this latter parameter. Surprisingly, sniffing time in BTBR mice in the presence of a novel object was comparable with B6 mice. Proinflammatory cytokines such as IL-6 and IL-1b, as well as other inflammatory markers, were elevated in BTBR mice, which were dramatically reduced after supplementation with VS1. Interestingly, even Beclin-1/p62, pAKT/AKT, and p-p70-S6K/p70-S6K ratios were notably reduced by VS1. We conclude that VS1 plays a crucial role in restoring autistic spectrum disorders (ASD) plausibly by attenuating neuroinflammation.
Collapse
Affiliation(s)
- Ennio Avolio
- Comparative Neuroanatomy Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, Ponte Pietro Bucci 4B, Arcavacata di Rende, 87030 Cosenza, Italy
| | - Ilaria Olivito
- Comparative Neuroanatomy Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, Ponte Pietro Bucci 4B, Arcavacata di Rende, 87030 Cosenza, Italy
| | - Antonio Leo
- Department of Health Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; System and Applied Pharmacology@University Magna Grecia, 88100 Catanzaro, Italy.
| | - Claudia De Matteo
- Department of Health Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; System and Applied Pharmacology@University Magna Grecia, 88100 Catanzaro, Italy
| | - Lorenza Guarnieri
- Department of Health Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; System and Applied Pharmacology@University Magna Grecia, 88100 Catanzaro, Italy
| | - Francesca Bosco
- Department of Health Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; System and Applied Pharmacology@University Magna Grecia, 88100 Catanzaro, Italy.
| | - Sushil K Mahata
- VA San Diego Healthcare System, San Diego, CA, USA; University of California San Diego, La Jolla, CA 92093, United States of America
| | - Damiana Minervini
- Comparative Neuroanatomy Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, Ponte Pietro Bucci 4B, Arcavacata di Rende, 87030 Cosenza, Italy
| | - Raffaella Alò
- Comparative Neuroanatomy Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, Ponte Pietro Bucci 4B, Arcavacata di Rende, 87030 Cosenza, Italy
| | - Giovambattista De Sarro
- Department of Health Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; System and Applied Pharmacology@University Magna Grecia, 88100 Catanzaro, Italy
| | - Rita Citraro
- Department of Health Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; System and Applied Pharmacology@University Magna Grecia, 88100 Catanzaro, Italy
| | - Rosa Maria Facciolo
- Comparative Neuroanatomy Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, Ponte Pietro Bucci 4B, Arcavacata di Rende, 87030 Cosenza, Italy
| |
Collapse
|
23
|
Ciupei D, Colişar A, Leopold L, Stănilă A, Diaconeasa ZM. Polyphenols: From Classification to Therapeutic Potential and Bioavailability. Foods 2024; 13:4131. [PMID: 39767073 PMCID: PMC11675957 DOI: 10.3390/foods13244131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/14/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Though ubiquitous in nature, polyphenols gained scientific prominence only after the pioneering work of researchers like E. Fischer and K. Freudenberg, who demonstrated their potential beyond traditional applications, such as in the leather industry. Today, these bioactive compounds are recognized for their diverse therapeutic roles, including their use as adjuvants in cancer treatment, cancer prevention, and their anti-inflammatory and antioxidant properties. Additionally, polyphenols have demonstrated benefits in managing obesity, cardiovascular diseases, and neuromodulation. Their synthesis is influenced by environmental and genetic factors, with their concentrations varying based on the intensity of these variables, as well as the stage of ripening. This review provides a comprehensive overview of polyphenols, covering their classification, chemical structures, and bioavailability. The mechanisms influencing bioavailability, bioaccessibility, and bioactivity are explored in detail, alongside an introduction to their bioactive effects and associated metabolic pathways. Specific examples, such as the bioavailability of polyphenols in coffee and various types of onions, are analyzed. Despite their promising biological activities, a significant limitation of polyphenols lies in their inherently low oral bioavailability. However, their systemic circulation and the bioactive by-products formed during digestion present exciting opportunities for further research and application.
Collapse
Affiliation(s)
- Daria Ciupei
- Life Science Institute, University of Agricultural Sciences and Veterinary Medicine, Manastur 3-5, 400372 Cluj-Napoca, Romania;
| | - Alexandru Colişar
- Faculty of Forestry and Cadastre, University of Agricultural Sciences and Veterinary Medicine, Manastur 3-5, 400372 Cluj-Napoca, Romania;
| | - Loredana Leopold
- Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine, Manastur 3-5, 400372 Cluj-Napoca, Romania; (L.L.); (A.S.)
| | - Andreea Stănilă
- Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine, Manastur 3-5, 400372 Cluj-Napoca, Romania; (L.L.); (A.S.)
| | - Zorița M. Diaconeasa
- Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine, Manastur 3-5, 400372 Cluj-Napoca, Romania; (L.L.); (A.S.)
| |
Collapse
|
24
|
Qin L, Zeng X, Qiu X, Chen X, Liu S. The role of N6-methyladenosine modification in tumor angiogenesis. Front Oncol 2024; 14:1467850. [PMID: 39691597 PMCID: PMC11649548 DOI: 10.3389/fonc.2024.1467850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 11/11/2024] [Indexed: 12/19/2024] Open
Abstract
Tumor angiogenesis is a characteristics of malignant cancer progression that facilitates cancer cell growth, diffusion and metastasis, and has an indispensable role in cancer development. N6-methyladenosine (m6A) is among the most prevalent internal modifications in eukaryotic RNAs, and has considerable influence on RNA metabolism, including its transcription, splicing, localization, translation, recognition, and degradation. The m6A modification is generated by m6A methyltransferases ("writers"), removed by m6A demethylases ("erasers"), and recognized by m6A-binding proteins ("readers"). There is accumulating evidence that abnormal m6A modification is involved in the pathogenesis of multiple diseases, including cancers, and promotes cancer occurrence, development, and progression through its considerable impact on oncoprotein expression. Furthermore, increasing studies have demonstrated that m6A modification can influence angiogenesis in cancers through multiple pathways to regulate malignant processes. In this review, we elaborate the role of m6A modification in tumor angiogenesis-related molecules and pathways in detail, providing insights into the interactions between m6A and tumor angiogenesis. Moreover, we describe how targeting m6A modification in combination with anti-angiogenesis drugs is expected to be a promising anti-tumor treatment strategy, with potential value for addressing the challenge of drug resistance.
Collapse
Affiliation(s)
| | | | | | | | - Shiquan Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical
University, Nanning, Guangxi, China
| |
Collapse
|
25
|
Kędzierska M, Bańkosz M. Role of Proteins in Oncology: Advances in Cancer Diagnosis, Prognosis, and Targeted Therapy-A Narrative Review. J Clin Med 2024; 13:7131. [PMID: 39685591 DOI: 10.3390/jcm13237131] [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: 10/27/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Modern oncology increasingly relies on the role of proteins as key components in cancer diagnosis, prognosis, and targeted therapy. This review examines advancements in protein biomarkers across several cancer types, including breast cancer, lung cancer, ovarian cancer, and hepatocellular carcinoma. These biomarkers have proven critical for early detection, treatment response monitoring, and tailoring personalized therapeutic strategies. The article highlights the utility of targeted therapies, such as tyrosine kinase inhibitors and monoclonal antibodies, in improving treatment efficacy while minimizing systemic toxicity. Despite these advancements, challenges like tumor resistance, variability in protein expression, and diagnostic heterogeneity persist, complicating universal application. The review underscores future directions, including the integration of artificial intelligence, advanced protein analysis technologies, and the development of combination therapies to overcome these barriers and refine personalized cancer treatment.
Collapse
Affiliation(s)
- Magdalena Kędzierska
- Department of Chemotherapy, Medical University of Lodz, Copernicus Memorial Hospital of Lodz, 90-549 Lodz, Poland
| | - Magdalena Bańkosz
- CUT Doctoral School, Faculty of Materials Engineering and Physics, Department of Material Engineering, Cracow University of Technology, 37 Jana Pawla II Av., 31-864 Krakow, Poland
| |
Collapse
|
26
|
Ren W, Liang H, Sun J, Cheng Z, Liu W, Wu Y, Shi Y, Zhou Z, Chen C. TNFAIP2 promotes HIF1α transcription and breast cancer angiogenesis by activating the Rac1-ERK-AP1 signaling axis. Cell Death Dis 2024; 15:821. [PMID: 39532855 PMCID: PMC11557851 DOI: 10.1038/s41419-024-07223-2] [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: 07/02/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Angiogenesis is well known to play a critical role in breast cancer. We previously reported that TNFAIP2 activates Rac1 to promote triple-negative breast cancer (TNBC) cell proliferation, migration, and chemoresistance. However, the potential contribution of TNFAIP2 to tumor angiogenesis remains unknown. In this study, we demonstrated that TNFAIP2 promotes TNBC angiogenesis by activating the Rac1-ERK-AP1-HIF1α signaling axis. Under hypoxia, TNFAIP2 activates Rac1 and ERK sequentially. Following that, ERK activates the AP-1 (c-Jun/Fra1) transcription factor. By employing chromatin immunoprecipitation and luciferase reporter assays, we showed that AP-1 directly interacts with the HIF1α gene promoter, thereby enhancing its transcription. The combined application of ERK inhibitors, U0126 or trametinib, with the VEGFR inhibitor Apatinib, additively suppresses angiogenesis and tumor growth of HCC1806 in nude mice. These findings provide new therapeutic strategies for TNBC.
Collapse
MESH Headings
- Humans
- Animals
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- rac1 GTP-Binding Protein/metabolism
- rac1 GTP-Binding Protein/genetics
- Female
- Cell Line, Tumor
- Mice, Nude
- Triple Negative Breast Neoplasms/genetics
- Triple Negative Breast Neoplasms/pathology
- Triple Negative Breast Neoplasms/metabolism
- Triple Negative Breast Neoplasms/drug therapy
- Mice
- Signal Transduction
- Transcription Factor AP-1/metabolism
- Pyrimidinones/pharmacology
- Pyridines/pharmacology
- Cell Proliferation
- Gene Expression Regulation, Neoplastic
- Transcription, Genetic/drug effects
- Pyridones/pharmacology
- Mice, Inbred BALB C
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Nitriles/pharmacology
- MAP Kinase Signaling System/drug effects
- Human Umbilical Vein Endothelial Cells/metabolism
- Angiogenesis
Collapse
Affiliation(s)
- Wenlong Ren
- School of Life Science, University of Science & Technology of China, Hefei, Anhui, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Huichun Liang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jian Sun
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Zhuo Cheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Wenjing Liu
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Yingying Wu
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yujie Shi
- Department of Pathology, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China.
| | - Zhongmei Zhou
- The School of Continuing Education, Kunming Medical University, Kunming, China.
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China.
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Academy of Biomedical Engineering, Kunming Medical University, Kunming, ChinaAcademy of Biomedical Engineering, Kunming Medical University, Kunming, China.
| |
Collapse
|
27
|
Chen H, Zhou Y, Tang Y, Lan J, Lin C, Chen Q, Kuang H. Neutrophil extracellular traps in tumor progression of gynecologic cancers. Front Immunol 2024; 15:1421889. [PMID: 39555072 PMCID: PMC11563837 DOI: 10.3389/fimmu.2024.1421889] [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: 04/23/2024] [Accepted: 09/30/2024] [Indexed: 11/19/2024] Open
Abstract
This article delves into the intricate interplay between tumors, particularly gynecologic malignancies, and neutrophil extracellular traps (NETs). The relationship between tumors, specifically gynecologic malignancies, and NETs is a multifaceted and pivotal area of study. Neutrophils, pivotal components of the immune system, are tasked with combating foreign invaders. NETs, intricate structures released by neutrophils, play a vital role in combating systemic infections but also play a role in non-infectious conditions such as inflammation, autoimmune diseases, and cancer. Cancer cells have the ability to attract neutrophils, creating tumor-associated neutrophils, which then stimulate the release of NETs into the tumor microenvironment. The impact of NETs within the tumor microenvironment is profound and intricate. They play a significant role in influencing cancer development and metastasis, as well as modulating tumor immune responses. Through the release of proteases and pro-inflammatory cytokines, NETs directly alter the behavior of tumor cells, increasing invasiveness and metastatic potential. Additionally, NETs can trigger epithelial-mesenchymal transition in tumor cells, a process associated with increased invasion and metastasis. The interaction between tumors and NETs is particularly critical in gynecologic malignancies such as ovarian, cervical, and endometrial cancer. Understanding the mechanisms through which NETs operate in these tumors can offer valuable insights for the development of targeted therapeutic interventions. Researchers are actively working towards harnessing this interaction to impede tumor progression and metastasis, opening up new avenues for future treatment modalities. As our understanding of the interplay between tumors and NETs deepens, it is anticipated that novel treatment strategies will emerge, potentially leading to improved outcomes for patients with gynecologic malignancies. This article provides a comprehensive overview of the latest research findings on the interaction between NETs and cancer, particularly in gynecologic tumors, serving as a valuable resource for future exploration in this field.
Collapse
Affiliation(s)
- Hong Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Ying Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Yaling Tang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jianfa Lan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Chao Lin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Qionghua Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Hongying Kuang
- The Second Department of Gynecology, The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
28
|
Almeida GDO, Cintra ACO, Silva TA, de Oliveira IS, Correia LIV, Torquato RJS, Ferreira Junior RS, Arantes EC, Sampaio SV. Moojecin: The first disintegrin from Bothrops moojeni venom and its antitumor activity in acute myeloid leukemia. Int J Biol Macromol 2024; 279:135066. [PMID: 39197621 DOI: 10.1016/j.ijbiomac.2024.135066] [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: 05/14/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
Disintegrins are a class of peptides found in snake venom that inhibit the activity of integrins, which are essential cell adhesion receptors in tumor progression and development. In this work, moojecin, a RGD disintegrin, was isolated from Bothrops moojeni snake venom, and its antitumor potential in acute myeloid leukemia (AML) HL-60 and THP-1 cells was characterized. The isolation was performed using a C18 reverse-phase column in two chromatographic steps, and its molecular mass is 7417.84 Da. N-terminal and de novo sequencing was performed to identify moojecin. Moojecin did not show cytotoxic or antiproliferative activity in THP-1 and HL-60 at tested concentrations, but it exhibited significant antimigratory activity in both cell lines, as well as inhibition of angiogenesis in the tube formation assay on Matrigel in a dose-dependent manner. A stronger interaction with integrin αVβ3 was shown in integrin interaction assays compared to α5β1, and the platelet aggregation assay indicated an IC50 of 5.039 μg/mL. Preliminary evaluation of disintegrin toxicity revealed no incidence of hemolysis or cytotoxic effects on peripheral blood mononuclear cells (PBMCs) across the tested concentrations. Thus, this is the first study to report the isolation, functional and structural characterization of a disintegrin from B. moojeni venom and bring a new perspective to assist in AML treatment.
Collapse
Affiliation(s)
- Gabriela de Oliveira Almeida
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Adélia Cristina Oliveira Cintra
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Thiago Abrahão Silva
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Isadora Sousa de Oliveira
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | | | - Rui Seabra Ferreira Junior
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Eliane Candiani Arantes
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Suely Vilela Sampaio
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
29
|
Wang C, Feng Q, Shi S, Qin Y, Lu H, Zhang P, Liu J, Chen B. The Rational Engineered Bacteria Based Biohybrid Living System for Tumor Therapy. Adv Healthc Mater 2024; 13:e2401538. [PMID: 39051784 DOI: 10.1002/adhm.202401538] [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: 04/26/2024] [Revised: 07/16/2024] [Indexed: 07/27/2024]
Abstract
Living therapy based on bacterial cells has gained increasing attention for their applications in tumor treatments. Bacterial cells can naturally target to tumor sites and active the innate immunological responses. The intrinsic advantages of bacteria attribute to the development of biohybrid living carriers for targeting delivery toward hypoxic environments. The rationally engineered bacterial cells integrate various functions to enhance the tumor therapy and reduce toxic side effects. In this review, the antitumor effects of bacteria and their application are discussed as living therapeutic agents across multiple antitumor platforms. The various kinds of bacteria used for cancer therapy are first introduced and demonstrated the mechanism of antitumor effects as well as the immunological effects. Additionally, this study focused on the genetically modified bacteria for the production of antitumor agents as living delivery system to treat cancer. The combination of living bacterial cells with functional nanomaterials is then discussed in the cancer treatments. In brief, the rational design of living therapy based on bacterial cells highlighted a rapid development in tumor therapy and pointed out the potentials in clinical applications.
Collapse
Affiliation(s)
- Chen Wang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Qiliner Feng
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Si Shi
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Yuxuan Qin
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Hongli Lu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Peng Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Jie Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Baizhu Chen
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, China
| |
Collapse
|
30
|
Azevedo T, Ferreira T, Peña‐Corona SI, Cortes H, Silva‐Reis R, da Costa RMG, Faustino‐Rocha AI, Oliveira PA, Calina D, Cardoso SM, Büsselberg D, Leyva‐Gómez G, Sharifi‐Rad J, Cho WC. Natural products‐based antiangiogenic agents: New frontiers in cancer therapy. FOOD FRONTIERS 2024; 5:2423-2466. [DOI: 10.1002/fft2.466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
AbstractAngiogenesis, vital for tumor growth and metastasis, is a promising target in cancer therapy. Natural compounds offer potential as antiangiogenic agents with reduced toxicity. This review provides a comprehensive overview of natural product‐based antiangiogenic therapies, focusing on molecular mechanisms and therapeutic potential. A systematic search identified relevant articles from 2019 to 2023. Various natural compounds, including polyphenols, terpenes, alkaloids, cannabinoids, omega‐3 fatty acids, polysaccharides, proteins, and carotenoids, were investigated for their antiangiogenic properties. Challenges such as dose standardization, routes of administration, and potential side effects remain. Further studies, including in‐depth animal models and human epidemiological studies, must elucidate clinical efficacy and safety. Synergistic effects with current antiangiogenic therapies, such as bevacizumab and tyrosine kinase inhibitors, should be explored. Additionally, the potential hormone‐dependent effects of compounds like genistein highlight the need for safety evaluation. In conclusion, natural products hold promise as adjunctive therapies to conventional antineoplastic drugs in modulating angiogenesis in cancer. However, robust clinical trials are needed to validate preclinical findings and ensure safety and efficacy.
Collapse
Affiliation(s)
- Tiago Azevedo
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
| | - Tiago Ferreira
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
| | - Sheila I. Peña‐Corona
- Departamento de Farmacia, Facultad de Química Universidad Nacional Autónoma de México Ciudad de México Mexico
| | - Hernán Cortes
- Laboratorio de Medicina Genómica, Departamento de Genómica Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Ciudad de México Mexico
| | - Rita Silva‐Reis
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
- LAQV‐REQUIMTE, Department of Chemistry University of Aveiro Aveiro Portugal
| | - Rui M. Gil da Costa
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI‐IPOP)/RISE@CI‐IPOP (Health Research Network) Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center (Porto. CCC) Porto Portugal
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering University of Porto Porto Portugal
- Associate Laboratory in Chemical Engineering (ALiCE), Faculty of Engineering University of Porto Porto Portugal
- Postgraduate Programme in Adult Health (PPGSAD), Department of Morphology Federal University of Maranhão (UFMA), UFMA University Hospital (HUUFMA) São Luís Brazil
| | - Ana I. Faustino‐Rocha
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
- Comprehensive Health Research Center, Department of Zootechnics, School of Sciences and Technology University of Évora Evora Portugal
| | - Paula A. Oliveira
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
| | - Daniela Calina
- Department of Clinical Pharmacy University of Medicine and Pharmacy of Craiova Craiova Romania
| | - Susana M. Cardoso
- LAQV‐REQUIMTE, Department of Chemistry University of Aveiro Aveiro Portugal
| | | | - Gerardo Leyva‐Gómez
- Departamento de Farmacia, Facultad de Química Universidad Nacional Autónoma de México Ciudad de México Mexico
| | - Javad Sharifi‐Rad
- Centro de Estudios Tecnológicos y Universitarios del Golfo Veracruz Mexico
- Department of Medicine, College of Medicine Korea University Seoul Republic of Korea
- Facultad de Medicina Universidad del Azuay Cuenca Ecuador
| | - William C. Cho
- Department of Clinical Oncology Queen Elizabeth Hospital Kowloon Hong Kong
| |
Collapse
|
31
|
Jiang RY, Zhu JY, Zhang HP, Yu Y, Dong ZX, Zhou HH, Wang X. STAT3: Key targets of growth-promoting receptor positive breast cancer. Cancer Cell Int 2024; 24:356. [PMID: 39468521 PMCID: PMC11520424 DOI: 10.1186/s12935-024-03541-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024] Open
Abstract
Breast cancer has become the malignant tumor with the first incidence and the second mortality among female cancers. Most female breast cancers belong to luminal-type breast cancer and HER2-positive breast cancer. These breast cancer cells all have different driving genes, which constantly promote the proliferation and metastasis of breast cancer cells. Signal transducer and activator of transcription 3 (STAT3) is an important breast cancer-related gene, which can promote the progress of breast cancer. It has been proved in clinical and basic research that over-expressed and constitutively activated STAT3 is involved in the progress, proliferation, metastasis and chemotherapy resistance of breast cancer. STAT3 is an important key target in luminal-type breast cancer and HER2-positive cancer, which has an important impact on the curative effect of related treatments. In breast cancer, the activation of STAT3 will change the spatial position of STAT3 protein and cause different phenotypic changes of breast cancer cells. In the current basic research and clinical research, small molecule inhibitors activated by targeting STAT3 can effectively treat breast cancer, and enhance the efficacy level of related treatment methods for luminal-type and HER2-positive breast cancers.
Collapse
Affiliation(s)
- Rui-Yuan Jiang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Jia-Yu Zhu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Huan-Ping Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Department of Graduate Student, Wenzhou Medical University, No.270, Xueyuan West Road, Lucheng District, Wenzhou, 325027, Zhejiang, China
| | - Yuan Yu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Zhi-Xin Dong
- Department of Oncology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, No.89-9, Dongge Road, Qingxiu District, Nanning, 530000, Guangxi, China
| | - Huan-Huan Zhou
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China.
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| | - Xiaojia Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
32
|
Lusardi M, Belvedere R, Petrella A, Iervasi E, Ponassi M, Brullo C, Spallarossa A. Novel tetrasubstituted 5-Arylamino pyrazoles able to interfere with angiogenesis and Ca 2+ mobilization. Eur J Med Chem 2024; 276:116715. [PMID: 39083983 DOI: 10.1016/j.ejmech.2024.116715] [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: 05/22/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
In the last years, 5-pyrazolyl ureas and 5-aminopyrazoles have been investigated for their antiangiogenetic properties and their potential interaction with the ubiquitous Ca2+ binding protein Calreticulin. Based on the structure of the active compounds I and GeGe-3, novel 5-arylamino pyrazoles 2 and 3 were synthesized through a stepwise procedure. In MTT assays, all the new derivatives proved to be non-cytotoxic against eight different tumor cell lines, normal fibroblasts, and endothelial cells. Furthermore, selected derivatives showed relevant antiangiogenetic properties, resulting more effective than reference molecules I and GeGe-3 in inhibiting HUVEC endothelial tube formation. 5-Arylamino pyrazoles 2a and 2d were identified as the most interesting compounds and significantly prevented tube formation of tumor secretome-stimulated HUVEC. Furthermore, the two compounds inhibited HUVEC migration in wound healing assay and altered cell invasion capability. Additionally, 2a and 2d strongly affected Ca2+ mobilization and cytoskeletal organization of HUVEC cells, being as active as the reference compound GeGe-3. Differently from previous studies, molecular docking simulations suggested a poor affinity of 2a towards Calreticulin, one of the interacting partners of the lead compound GeGe-3. Collectively, this new amino-pyrazole library further extends the structure-activity relationships of the previously prepared derivatives and confirmed the biological attractiveness of this chemical scaffold as antiangiogenetic agents.
Collapse
Affiliation(s)
- Matteo Lusardi
- Department of Pharmacy, Section of Medicinal Chemistry, University of Genoa, Viale Benedetto XV 3, I-16132, Genova, Italy
| | - Raffaella Belvedere
- Department of Pharmacy, University of Salerno, Viale Giovanni Paolo II, 84084, Salerno, Italy
| | - Antonello Petrella
- Department of Pharmacy, University of Salerno, Viale Giovanni Paolo II, 84084, Salerno, Italy
| | - Erika Iervasi
- IRCCS Ospedale Policlinico San Martino, Proteomics and Mass Spectrometry Unit, Largo. R. Benzi, 10, 16132, Genova, Italy
| | - Marco Ponassi
- IRCCS Ospedale Policlinico San Martino, Proteomics and Mass Spectrometry Unit, Largo. R. Benzi, 10, 16132, Genova, Italy
| | - Chiara Brullo
- Department of Pharmacy, Section of Medicinal Chemistry, University of Genoa, Viale Benedetto XV 3, I-16132, Genova, Italy
| | - Andrea Spallarossa
- Department of Pharmacy, Section of Medicinal Chemistry, University of Genoa, Viale Benedetto XV 3, I-16132, Genova, Italy.
| |
Collapse
|
33
|
Lessiak U, Melchert M, Walter I, Kummer S, Nell B, Tschulenk W, Pratscher B. Isolation-protocol, characterization, and in-vitro performance of equine umbilical vein endothelial cells. Front Vet Sci 2024; 11:1421946. [PMID: 39411390 PMCID: PMC11473255 DOI: 10.3389/fvets.2024.1421946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Angiogenesis plays a crucial role in various physiological and pathological conditions. However, research in equine angiogenesis is relative limited, necessitating the development of suitable in-vitro models. To effectively analyze angiogenesis in-vitro, it is essential to target the specific cells responsible for this process, namely endothelial cells. Human umbilical vein endothelial cells (HUVECs) are one of the most used in vitro models for studying angiogenesis in humans. Serving as an equivalent to HUVECs, we present a comprehensive isolation protocol for equine umbilical vein endothelial cells (EqUVECs) with relatively minimal requirements, thereby enhancing accessibility for researchers. Umbilical cords obtained from five foals were used to isolate endothelial cells, followed by morphological and immunohistochemical identification. Performance of the cells in various assays commonly used in angiogenesis research was studied. Additionally, EqUVEC expression of vascular endothelial growth factor (VEGF) was assessed using ELISA. EqUVECs exhibited endothelial characteristics, forming a homogeneous monolayer with distinctive morphology. Immunohistochemical staining confirmed positive expression of key endothelial markers including von Willebrand factor (vWF), CD31, and vascular endothelial growth factor receptor-2 (VEGFR-2). Furthermore, performance assessments in in-vitro assays demonstrated the viability, proliferation, migration, tube formation and VEGF-expression capabilities of EqUVECs. The findings suggest that EqUVECs are a promising in-vitro model for studying equine angiogenesis, offering a foundation for further investigations into equine-specific vascular processes and therapeutic interventions.
Collapse
Affiliation(s)
- Ulrike Lessiak
- Ophthalmology Unit, Department of Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Maria Melchert
- Centre for Animal Reproduction, Department of Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Ingrid Walter
- Department of Biomedical Science and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- VetCore Facility for Research, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Stefan Kummer
- VetCore Facility for Research, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Barbara Nell
- Ophthalmology Unit, Department of Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Waltraud Tschulenk
- Department of Biomedical Science and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Barbara Pratscher
- Research Unit Internal Medicine, Department of Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
34
|
Wang DX, Zhu M, Guo DH, Gu J, Xia L, Huang XW, Wang TL, Zhao CH. Safety of Endostar in combination with chemotherapy in patients with cancer. Indian J Cancer 2024; 61:694-702. [PMID: 39960696 DOI: 10.4103/ijc.ijc_1065_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 03/24/2021] [Indexed: 05/09/2025]
Abstract
BACKGROUND Several studies have indicated the benefits and safety of Endostar in combination with chemotherapy, but the exact real-life safety of Endostar is poorly known. This study aimed to assess the safety of Endostar in combination with chemotherapy in a real-life setting in China. MATERIALS AND METHODS This was a retrospective study of patients treated with Endostar combined with chemotherapy from January 1, 2006, to December 31, 2017. Data were obtained from the Hospital Information System (HIS). Laboratory abnormalities were evaluated according to Common Terminology Criteria for Adverse Events, Version 4.0. Bleeding events and wound complications after surgery associated with Endostar were evaluated. RESULTS Finally, 825 patients were included. No patients used Endostar alone. Anemia occurred in 74.5% of the patients, thrombocytopenia in 29.0%, abnormal white blood cell counts in 54.5%, abnormal liver function in 13.8%, and increased creatinine in 1.2%. No definite bleeding events and wound complications associated with Endostar were found. Most laboratory adverse effects (AEs) were found in Grades 1 and 2. Lung cancer, osteosarcoma, and doxorubicin-based chemotherapy were associated with an increased risk of Grade ≥3 abnormal white blood cell counts. The total dose of Endostar was not associated with severe (Grade ≥3) thrombocytopenia and abnormal white blood cell counts. CONCLUSION The occurrence of AEs during Endostar and chemotherapy treatment differed across different tumor types and chemotherapy regimens. No new unexpected Endostar-related AEs were observed. The total dose of Endostar was not associated with an increased risk of severe (Grade ≥3) thrombocytopenia and abnormal white blood cell counts when used in combination with chemotherapy in the real-life setting.
Collapse
Affiliation(s)
- Dong-Xiao Wang
- Department of Pharmacy, Peking University Shougang Hospital, Beijing, China
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Man Zhu
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Dai-Hong Guo
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Jin Gu
- Department of Pharmacy, Peking University Shougang Hospital, Beijing, China
| | - Lei Xia
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Xiao-Wu Huang
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Tian-Lin Wang
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Chang-Hong Zhao
- The State Key Laboratory of Translational Medicine and Innovative Drug Development, Nanjing, China
| |
Collapse
|
35
|
Figueroa L, Rosas M, Alvarez M, Aguilar E, Mateu V, Bonilla E. Interaction of Purine and its Derivatives with A1, A2-Adenosine Receptors and Vascular Endothelial Growth Factor Receptor-1 (Vegf-R1) as a Therapeutic Alternative to Treat Cancer. Drug Res (Stuttg) 2024; 74:379-393. [PMID: 39173673 DOI: 10.1055/a-2376-5771] [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: 08/24/2024]
Abstract
BACKGROUND There are several studies that indicate that cancer development may be conditioned by the activation of some biological systems that involve the interaction of different biomolecules, such as adenosine and vascular endothelial growth factor. These biomolecules have been targeted of some drugs for treat of cancer; however, there is little information on the interaction of purine derivatives with adenosine and vascular endothelial growth factor receptor (VEGF-R1). OBJECTIVE The aim of this research was to determine the possible interaction of purine (1: ) and their derivatives (2-31: ) with A1, A2-adenosine receptors, and VEGF-R1. METHODS Theoretical interaction of purine and their derivatives with A1, A2-adenosine receptors and VEGF-R1 was carried out using the 5uen, 5mzj and 3hng proteins as theoretical tools. Besides, adenosine, cgs-15943, rolofylline, cvt-124, wrc-0571, luf-5834, cvt-6883, AZD-4635, cabozantinib, pazopanib, regorafenib, and sorafenib drugs were used as controls. RESULTS The results showed differences in the number of aminoacid residues involved in the interaction of purine and their derivatives with 5uen, 5mzj and 3hng proteins compared with the controls. Besides, the inhibition constants (Ki) values for purine and their derivatives 5: , 9: , 10: , 14: , 15: , 16: , and 20: were lower compared with the controls CONCLUSIONS: Theoretical data suggest that purine and their derivatives 5: , 9: , 10: , 14: , 15: , 16: , and 20: could produce changes in cancer cell growth through inhibition of A1, A2-adenosine receptors and VEGFR-1 inhibition. These data indicate that these purine derivatives could be a therapeutic alternative to treat some types of cancer.
Collapse
Affiliation(s)
- Lauro Figueroa
- Laboratory of Pharmaco-Chemistry, Faculty of Chemical Biological Sciences, University Autonomous of Campeche, Campeche, Camp., México
| | - Marcela Rosas
- Faculty of Nutrition, University Veracruzana, Médicos y Odontologos, Unidad del Bosque Xalapa Veracruz, México
| | - Magdalena Alvarez
- Faculty of Nutrition, University Veracruzana, Médicos y Odontologos, Unidad del Bosque Xalapa Veracruz, México
| | - Emilio Aguilar
- Facultad de Medicina, Universidad Veracruzana, Médicos y Odontologos, Unidad del Bosque Xalapa Veracruz, México
| | - Virginia Mateu
- Faculty of Nutrition, University Veracruzana, Médicos y Odontologos, Unidad del Bosque Xalapa Veracruz, México
| | - Enrique Bonilla
- Faculty of Nutrition, University Veracruzana, Médicos y Odontologos, Unidad del Bosque Xalapa Veracruz, México
| |
Collapse
|
36
|
Yong GY, Muniandy N, Beishenaliev A, Lau BF, Kue CS. Anti-angiogenic and anti-tumour activities of Lignosus rhinocerus (Cooke) Ryvarden water extracts on HCT116 human colorectal carcinoma cells implanted in chick embryos. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118213. [PMID: 38636576 DOI: 10.1016/j.jep.2024.118213] [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: 10/10/2023] [Revised: 02/29/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The sclerotium of Lignosus rhinocerus (Cooke) Ryvarden is used by the local communities in Southeast Asia and China to treat cancer, asthma, fever, and other ailments based on traditional knowledge. The sclerotial water extracts were previously reported to exhibit cytotoxic, apoptotic, and immunomodulatory activities - providing a scientific basis for its use in treating cancer; however, there is still a lack of evidence on its potential anti-angiogenic activity. AIM OF THE STUDY This study aimed to investigate the toxicity, anti-angiogenic, and anti-tumour activities of the hot-water and cold-water extracts of L. rhinocerus using HCT116 human colorectal carcinoma cells implanted in the chick chorioallantoic membrane (CAM) model. MATERIALS AND METHODS The toxicity of L. rhinocerus extracts towards the chick embryos was determined 24 h post-treatment. The anti-angiogenic activity of the extracts was then investigated at 0.1-10 μg/embryo (6.7-670 μg/mL) at targeted blood vessels. The anti-tumour effect of selected extracts against the HCT116 human colorectal carcinoma cells xenografted onto the chick embryos was also studied. RESULTS The cold-water extracts of L. rhinocerus displayed strong in ovo toxicity (LC50: 1.2-37.7 μg/mL) while the hot-water extracts are non-toxic up to 670 μg/mL. Among the extracts, the hot-water extracts demonstrated the highest anti-angiogenic activity with 44.0 ± 17.7% reduction of capillary diameter (relative to the saline-treated control). Moreover, treatment of the HCT116 cells xenografted onto the chick embryos with the hot-water extracts resulted in smaller tumour size and lower number of blood vessels compared to the saline-treated control. CONCLUSIONS The hot-water extracts of L. rhinocerus sclerotium demonstrated anti-angiogenic and anti-tumour activities but most of the cold-water extracts at similar concentrations were devoid of that. Our findings provide further scientific validation of the medicinal use of the sclerotium in treating cancer and thus, expanding our knowledge on the possible mechanism of its anti-cancer effect apart from direct cytotoxicity, induction of apoptosis and immunomodulation that have been studied thus far.
Collapse
Affiliation(s)
- Gong Yi Yong
- Faculty of Health and Life Sciences, Management and Science University, Seksyen 13, 40100, Shah Alam, Selangor, Malaysia
| | - Nishalini Muniandy
- Faculty of Health and Life Sciences, Management and Science University, Seksyen 13, 40100, Shah Alam, Selangor, Malaysia
| | - Adilet Beishenaliev
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Beng Fye Lau
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Chin Siang Kue
- Faculty of Health and Life Sciences, Management and Science University, Seksyen 13, 40100, Shah Alam, Selangor, Malaysia.
| |
Collapse
|
37
|
Zuo WF, Pang Q, Zhu X, Yang QQ, Zhao Q, He G, Han B, Huang W. Heat shock proteins as hallmarks of cancer: insights from molecular mechanisms to therapeutic strategies. J Hematol Oncol 2024; 17:81. [PMID: 39232809 PMCID: PMC11375894 DOI: 10.1186/s13045-024-01601-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024] Open
Abstract
Heat shock proteins are essential molecular chaperones that play crucial roles in stabilizing protein structures, facilitating the repair or degradation of damaged proteins, and maintaining proteostasis and cellular functions. Extensive research has demonstrated that heat shock proteins are highly expressed in cancers and closely associated with tumorigenesis and progression. The "Hallmarks of Cancer" are the core features of cancer biology that collectively define a series of functional characteristics acquired by cells as they transition from a normal state to a state of tumor growth, including sustained proliferative signaling, evasion of growth suppressors, resistance to cell death, enabled replicative immortality, the induction of angiogenesis, and the activation of invasion and metastasis. The pivotal roles of heat shock proteins in modulating the hallmarks of cancer through the activation or inhibition of various signaling pathways has been well documented. Therefore, this review provides an overview of the roles of heat shock proteins in vital biological processes from the perspective of the hallmarks of cancer and summarizes the small-molecule inhibitors that target heat shock proteins to regulate various cancer hallmarks. Moreover, we further discuss combination therapy strategies involving heat shock proteins and promising dual-target inhibitors to highlight the potential of targeting heat shock proteins for cancer treatment. In summary, this review highlights how targeting heat shock proteins could regulate the hallmarks of cancer, which will provide valuable information to better elucidate and understand the roles of heat shock proteins in oncology and the mechanisms of cancer occurrence and development and aid in the development of more efficacious and less toxic novel anticancer agents.
Collapse
Affiliation(s)
- Wei-Fang Zuo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qiwen Pang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xinyu Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qian-Qian Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qian Zhao
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Gu He
- Department of Dermatology and Venereology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
38
|
Jacobs K, Docter D, de Smit L, Korfage HAM, Visser SC, Lobbezoo F, Hlushchuk R, de Bakker BS. High resolution imaging of human development: shedding light on contrast agents. Neuroradiology 2024; 66:1481-1493. [PMID: 38995394 PMCID: PMC11322402 DOI: 10.1007/s00234-024-03413-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/21/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND Visualizing (micro)vascular structures remains challenging for researchers and clinicians due to limitations in traditional radiological imaging methods. Exploring the role of vascular development in craniofacial malformations in experimental settings can enhance understanding of these processes, with the effectiveness of high-resolution imaging techniques being crucial for successful research in this field. Micro-CT imaging offers 3D microstructural insights, but requires contrast-enhancing staining agents (CESAs) for visualizing (micro)-vascular tissues, known as contrast-enhanced micro-CT (CECT). As effective contrast agents are crucial for optimal visualization, this review focuses on comparative studies investigating such agents for micro-vascular tissue imaging using micro-CT. Furthermore, we demonstrate the utilization of B-Lugol solution as a promising contrast agent for acquiring high-quality micro-CT images of (micro)vascular structures in human embryonic samples. METHOD This scoping review followed Preferred Reporting Items for Systematic Reviews and Meta-analysis Protocols. PubMed database provided relevant articles, screened initially by title and abstract. Inclusion and exclusion criteria defined outcomes of interest. RESULTS From an initial search, 273 records were identified, narrowed down to 9 articles after applying our criteria. Additionally, two articles were added through citation searching. This, a total of 11 articles were incorporated in this study. CONCLUSION This micro-CT contrast agent review underscores the need for tailored choices based on research goals. Both Barium sulfate and Iodine-based agents showing excellent results, providing high resolution (micro) vascular content, especially in ex-vivo specimens. However, careful consideration of protocols and tissue characteristics remains imperative for optimizing the effectiveness of micro-CT imaging for the study of cranio-facial vascular development.
Collapse
Affiliation(s)
- Karl Jacobs
- Department of Orofacial Pain and Disfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands.
- Department of Medical Biology, Section Clinical Anatomy & Embryology, Amsterdam UMC location AMC, University of Amsterdam, Meibergdreef 15, Amsterdam, The Netherlands.
- Amsterdam Reproduction and Development Research Institute, Meibergdreef 9, Amsterdam, The Netherlands.
| | - Daniel Docter
- Department of Medical Biology, Section Clinical Anatomy & Embryology, Amsterdam UMC location AMC, University of Amsterdam, Meibergdreef 15, Amsterdam, The Netherlands
| | - Lotte de Smit
- Department of Orofacial Pain and Disfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Hans A M Korfage
- Department of Orofacial Pain and Disfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Sophie C Visser
- Department of Medical Biology, Section Clinical Anatomy & Embryology, Amsterdam UMC location AMC, University of Amsterdam, Meibergdreef 15, Amsterdam, The Netherlands
| | - Frank Lobbezoo
- Department of Orofacial Pain and Disfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, The Netherlands
| | - Ruslan Hlushchuk
- Micro-CT Research Group, Institute of Anatomy, University of Bern, Baltzerstrasse 2, CH-3012, Bern, Switzerland
| | - Bernadette S de Bakker
- Amsterdam Reproduction and Development Research Institute, Meibergdreef 9, Amsterdam, The Netherlands
- Department of Obstetrics and Gynecology, Amsterdam UMC location AMC, University of Amsterdam, Amsterdam Reproduction & Development Research Institute, Meibergdreef 9, Amsterdam, The Netherlands
- Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Department of Pediatric Surgery, Rotterdam, The Netherlands
| |
Collapse
|
39
|
Prome AA, Robin TB, Ahmed N, Rani NA, Ahmad I, Patel H, Bappy MNI, Zinnah KMA. A reverse docking approach to explore the anticancer potency of natural compounds by interfering metastasis and angiogenesis. J Biomol Struct Dyn 2024; 42:7174-7189. [PMID: 37526218 DOI: 10.1080/07391102.2023.2240895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/14/2023] [Indexed: 08/02/2023]
Abstract
Angiogenesis, which results in the formation of new blood and lymph vessels, is required to serve metastatic cancer progression. Cancer medications may target these two interconnected pathways. Phytocompounds have emerged as promising options for treating cancer. In this study, we used a reverse docking strategy to find new candidate molecules for cancer treatment that target both pathways. Following a literature study, the important cancer-causing proteins vascular endothelial growth factor D (VEGF-D) and basic fibroblast growth factor (bFGF) for angiogenesis and matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9) for the metastatic pathway were targeted. Protein Data Bank was used to retrieve the structures of chosen proteins. 22 significant plant metabolites were identified as having anticancer activity. To determine the important protein binding residues, active site prediction was used. Using Lenvatinib and Withaferin A as reference ligands, the binding affinity of certain proteins for plant metabolites was determined by docking analysis. Homoharringtonine and viniferin, both have higher binding affinities when compared to reference ligands, with docking scores of -180.96 and -180.36 against the protein MMP-9, respectively. Moreover, Viniferin showed the highest binding affinity with both MMP-9 and MMP-2 proteins, which were then subjected to a 100-ns molecular dynamic simulation. where they were found to be significantly stable. In pharmacoinformatics investigations, the majority of our compounds were found to be non-toxic for the host. In this study, we suggested natural substances as cutting-edge anticancer treatments that target both angiogenesis and metastasis, which may aid in accelerating drug development and identifying viable therapeutic candidates.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Anindita Ash Prome
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Tanjin Barketullah Robin
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Nadim Ahmed
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Nurul Amin Rani
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, Prof. Ravindra Nikam College of Pharmacy, Dhule, Maharashtra, India
| | - Harun Patel
- Division of Computer Aided Drug Design, Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
| | - Md Nazmul Islam Bappy
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
- Department of Animal and Fish Biotechnology, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Kazi Md Ali Zinnah
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, Bangladesh
- Department of Animal and Fish Biotechnology, Sylhet Agricultural University, Sylhet, Bangladesh
| |
Collapse
|
40
|
Sharma V, Chaudhary AA, Bawari S, Gupta S, Mishra R, Khan SUD, Ali MAM, Shahid M, Srivastava S, Verma D, Gupta A, Kumar S, Kumar S. Unraveling cancer progression pathways and phytochemical therapeutic strategies for its management. Front Pharmacol 2024; 15:1414790. [PMID: 39246660 PMCID: PMC11377287 DOI: 10.3389/fphar.2024.1414790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/09/2024] [Indexed: 09/10/2024] Open
Abstract
Cancer prevention is currently envisioned as a molecular-based approach to prevent carcinogenesis in pre-cancerous stages, i.e., dysplasia and carcinoma in situ. Cancer is the second-leading cause of mortality worldwide, and a more than 61% increase is expected by 2040. A detailed exploration of cancer progression pathways, including the NF-kβ signaling pathway, Wnt-B catenin signaling pathway, JAK-STAT pathway, TNF-α-mediated pathway, MAPK/mTOR pathway, and apoptotic and angiogenic pathways and effector molecules involved in cancer development, has been discussed in the manuscript. Critical evaluation of these effector molecules through molecular approaches using phytomolecules can intersect cancer formation and its metastasis. Manipulation of effector molecules like NF-kβ, SOCS, β-catenin, BAX, BAK, VEGF, STAT, Bcl2, p53, caspases, and CDKs has played an important role in inhibiting tumor growth and its spread. Plant-derived secondary metabolites obtained from natural sources have been extensively studied for their cancer-preventing potential in the last few decades. Eugenol, anethole, capsaicin, sanguinarine, EGCG, 6-gingerol, and resveratrol are some examples of such interesting lead molecules and are mentioned in the manuscript. This work is an attempt to put forward a comprehensive approach to understanding cancer progression pathways and their management using effector herbal molecules. The role of different plant metabolites and their chronic toxicity profiling in modulating cancer development pathways has also been highlighted.
Collapse
Affiliation(s)
- Vikas Sharma
- Metro College of Health Sciences and Research, Greater Noida, India
- School of Pharmacy, Sharda University, Greater Noida, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Sweta Bawari
- Amity Institute of Pharmacy, Amity University, Noida, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, India
| | - Richa Mishra
- Department of Computer Engineering, Parul University, Vadodara, India
| | - Salah-Ud-Din Khan
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Mohamed A M Ali
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Mohammad Shahid
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | | | - Devvrat Verma
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand, India
| | - Arti Gupta
- Lloyd School of Pharmacy, Greater Noida, India
| | - Sanjay Kumar
- Biological and Bio-computational Laboratory, Department of Life Science, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, India
| | - Sandeep Kumar
- School of Pharmacy, Sharda University, Greater Noida, India
- DST-FIST Laboratory, Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, India
| |
Collapse
|
41
|
Valivand N, Aravand S, Lotfi H, Esfahani AJ, Ahmadpour-Yazdi H, Gheibi N. Propolis: a natural compound with potential as an adjuvant in cancer therapy - a review of signaling pathways. Mol Biol Rep 2024; 51:931. [PMID: 39177837 DOI: 10.1007/s11033-024-09807-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 07/18/2024] [Indexed: 08/24/2024]
Abstract
Propolis is a natural product used in cancer treatment, which is produced by bees via different sources. The chemical composition of Propolis is determined based on the climatic and geographical conditions, as well as harvesting time and method. This compound has been the subject of numerous investigational endeavors due to its expansive therapeutic capacity which includes antibacterial, anti-fungal, anti-inflammatory, anti-oxidant, anti-viral, and anti-cancer effects. The growing incidence rate of different cancers necessitates the need for developing novel preventive and therapeutic strategies. Chemotherapy, radiotherapy, and stem cell therapy have proved effective in cancer treatment, regardless of the adverse events associated with these modalities. Clinical application of natural compounds such as Propolis may confer promise as an adjuvant therapeutic intervention, particularly in certain subpopulations of patients that develop adverse events associated with anticancer regimens. The diverse biologically active compounds of propolis are believed to confer anti-cancer potential by modulation of critical signaling cascades such as caffeic acid phenethyl ester, Galangin, Artepillin C, Chrysin, Quercetin, Caffeic acid, Nymphaeols A and C, Frondoside A, Genistein, p-coumaric acid, and Propolin C. This review article aims to deliver a mechanistic account of anti-cancer effects of propolis and its components. Propolis can prevent angiogenesis by downregulating pathways involving Jun-N terminal kinase, ERK1/2, Akt and NF-ƘB, while counteracting metastatic progression of cancer by inhibiting Wtn2 and FAK, and MAPK and PI3K/AKT signaling pathways. Moreover, propolis or its main components show regulatory effects on cyclin D, CDK2/4/6, and their inhibitors. Additionally, propolis-induced up-regulation of p21 and p27 may result in cell cycle arrest at G2/M or G0/G1. The broad anti-apoptotic effects of propolis are mediated through upregulation of TRAIL, Bax, p53, and downregulation of the ERK1/2 signaling pathway. Considering the growing body of evidence regarding different anti-cancers effects of propolis and its active components, this natural compound could be considered an effective adjuvant therapy aimed at reducing related side effects associated with chemotherapy and radiotherapy.
Collapse
Affiliation(s)
- Nassim Valivand
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sara Aravand
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Hajie Lotfi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Azam Janati Esfahani
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Hossein Ahmadpour-Yazdi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Nematollah Gheibi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran.
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Qazvin University of Medical Sciences, Qazvin, Iran.
| |
Collapse
|
42
|
Pote MS, Singh D, M. A A, Suchita J, Gacche RN. Cancer metastases: Tailoring the targets. Heliyon 2024; 10:e35369. [PMID: 39170575 PMCID: PMC11336595 DOI: 10.1016/j.heliyon.2024.e35369] [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: 07/17/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Metastasis is an intricate and formidable pathophysiological process encompassing the dissemination of cancer cells from the primary tumour body to distant organs. It stands as a profound and devastating phenomenon that constitutes the primary driver of cancer-related mortality. Despite great strides of advancements in cancer research and treatment, tailored anti-metastasis therapies are either lacking or have shown limited success, necessitating a deeper understanding of the intrinsic elements driving cancer invasiveness. This comprehensive review presents a contemporary elucidation of pivotal facets within the realm of cancer metastasis, commencing with the intricate processes of homing and invasion. The process of angiogenesis, which supports tumour growth and metastasis, is addressed, along with the pre-metastatic niche, wherein the primary tumour prepares for a favorable microenvironment at distant sites for subsequent metastatic colonization. The landscape of metastasis-related genetic and epigenetic mechanisms, involvement of metastasis genes and metastasis suppressor genes, and microRNAs (miRNA) are also discussed. Furthermore, immune modulators' impact on metastasis and their potential as therapeutic targets are addressed. The interplay between cancer cells and the immune system, including immune evasion mechanisms employed by metastatic cells, is discussed, highlighting the importance of targeting immune modulation in arresting metastatic progression. Finally, this review presents promising treatment opportunities derived from the insights gained into the mechanisms of metastasis. Identifying novel therapeutic targets and developing innovative strategies to disrupt the metastatic cascade holds excellent potential for improving patient outcomes and ultimately reducing cancer-related mortality.
Collapse
Affiliation(s)
| | | | | | | | - Rajesh N. Gacche
- Department of Biotechnology, Savitribai Phule Pune University, Pune, Maharashtra, India
| |
Collapse
|
43
|
Chitoran E, Rotaru V, Ionescu SO, Gelal A, Capsa CM, Bohiltea RE, Mitroiu MN, Serban D, Gullo G, Stefan DC, Simion L. Bevacizumab-Based Therapies in Malignant Tumors-Real-World Data on Effectiveness, Safety, and Cost. Cancers (Basel) 2024; 16:2590. [PMID: 39061228 PMCID: PMC11274419 DOI: 10.3390/cancers16142590] [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: 06/30/2024] [Revised: 07/08/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Overall, it is estimated that more than 3,500,000 patients have received Bevacizumab as part of systemic oncologic treatment. Bevacizumab and its biosimilars are currently marketed in over 130 countries. Given the wide usage of Bevacizumab in current oncological practice, it is very important to compare the "real-world" results to those obtained in controlled clinical trials. This study aims to describe the clinical experience of using Bevacizumab in a large cohort of cancer patients in "non-controlled real-world" conditions with regard to effectiveness, safety, and cost of therapy. METHODS For this purpose, we conducted an open, observational, retrospective study involving all patients treated for solid malignant tumors in the Bucharest Institute of Oncology with "Prof. Dr. Al. Trestioreanu" with Bevacizumab-based systemic therapy, between 2017 and 2021. RESULTS The study consisted of 657 treatment episodes in 625 patients (F/B = 1.62/1, with a median age of 57.6 years) which were treated for malignant tumors (majority colorectal, non-small cell lung, ovarian, and breast cancer). First-line treatment was administered in 229 patients, and the rest received Bevacizumab as second or subsequent lines of treatment. The overall response rate to Bevacizumab-based therapies was around 60-65% across all indication except for subsequent treatment lines in colorectal and ovarian cancers, where lower values were recorded (27.1%, and 31.5% respectively). Median PFS for the entire cohort was 8.2 months (95% CI 6.8-9.6), and the median OS was 13.2 months (95% CI 11.5-14.9). Usual bevacizumab-related toxicities were observed, including bleeding, hypertension, wound-healing complications, gastrointestinal perforation, other types of fistulas, septic complications, and thromboembolic events. Although the clinical benefits are undeniable, the addition of Bevacizumab to standard chemotherapy increased the overall treatment cost by 213%. CONCLUSIONS Bevacizumab remains a high-cost therapy, but it can add to clinical benefits (like overall survival, progression-free survival, and response rate) when used in conjunction with standard chemotherapy. Similar results as those presented in various controlled trials are observable even on unselected cohorts of patients in the uncontrolled conditions of "real-world" oncological practice. Off-label usage is encountered in clinical practice, and this aspect should be monitored given the potential adverse effects of the therapy.
Collapse
Affiliation(s)
- Elena Chitoran
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Vlad Rotaru
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Sinziana-Octavia Ionescu
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Aisa Gelal
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Cristina-Mirela Capsa
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Radiology Department, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Roxana-Elena Bohiltea
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Obstetrics and Gynecology Department, “Filantropia” Clinical Hospital, 011132 Bucharest, Romania
| | - Madalina-Nicoleta Mitroiu
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Obstetrics and Gynecology Department, “Filantropia” Clinical Hospital, 011132 Bucharest, Romania
| | - Dragos Serban
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Surgery Department 4, Bucharest University Emergency Hospital, 050098 Bucharest, Romania
| | - Giuseppe Gullo
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, University of Palermo, 90146 Palermo, Italy
| | - Daniela-Cristina Stefan
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Laurentiu Simion
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| |
Collapse
|
44
|
Giménez-Bastida JA, Ávila-Gálvez MÁ, Martínez-López A, García-Moreno D, Espín JC, González-Sarrías A. ( R, S)-Equol 7-β-D-glucuronide, but not other circulating isoflavone metabolites, modulates migration and tubulogenesis in human aortic endothelial cells targeting the VEGF pathway. Food Funct 2024; 15:7387-7399. [PMID: 38078511 DOI: 10.1039/d3fo03946c] [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: 07/16/2024]
Abstract
Current knowledge indicates that the consumption of isoflavone-rich foodstuffs can have a beneficial impact on cardiovascular health. To what extent these isoflavones act as the main actors of that benefit is less clear. Genistein (GEN), daidzein (DAZ), and the DAZ-derived microbial metabolite equol (Eq) exhibit antiangiogenic effects in vitro, but their low bloodstream concentrations make it difficult to rationalize the in vivo effects. Their derived phase-II metabolites (glucuronides and sulfates) are major metabolites found in plasma, but their role as antiangiogenic molecules remains unexplored. We aimed here to first assess the anti-angiogenic activities of the main circulating isoflavone metabolites (glucuronides and sulfates) and compare them with their corresponding free forms at physiological concentrations (0.1-10 μM). The effects of the conjugated vs. free forms on tubulogenesis, cell migration, and VEGF-induced signalling were investigated in primary human aortic endothelial cells (HAECs). While (R,S)-equol 7-β-D-glucuronide (Eq 7-glur) exerted dose-dependent inhibition of tubulogenesis and endothelial migration comparable to that exerted by the free forms (GEN, DAZ, and Eq), the rest of the phase-II conjugates exhibited no significant effects. The underlying molecular mechanisms were independent of the bFGF but related to the modulation of the VEGF pathway. Besides, the observed dissimilar cellular metabolism (conjugation/deconjugation) places the phase-II metabolites as precursors of the free forms; however, the question of whether this metabolism impacts their biological activity requires additional studies. These new insights suggest that isoflavones and their circulating metabolites, including Eq 7-glur, may be involved in cardiovascular health (e.g., targeting angiogenesis).
Collapse
Affiliation(s)
- Juan Antonio Giménez-Bastida
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, Murcia, Spain.
| | - María Ángeles Ávila-Gálvez
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, Murcia, Spain.
- NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Alicia Martínez-López
- Center for Biomedical Research in Rare Diseases Network (CIBERER), Carlos III Health Institute, 28029, Madrid, Spain
- Biomedical Research Institute of Murcia (IMIB)-Pascual Parrilla, 30120, Murcia, Spain
| | - Diana García-Moreno
- Center for Biomedical Research in Rare Diseases Network (CIBERER), Carlos III Health Institute, 28029, Madrid, Spain
- Biomedical Research Institute of Murcia (IMIB)-Pascual Parrilla, 30120, Murcia, Spain
| | - Juan Carlos Espín
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, Murcia, Spain.
| | - Antonio González-Sarrías
- Laboratory of Food & Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, CEBAS-CSIC, Murcia, Spain.
| |
Collapse
|
45
|
Vitale DL, Parnigoni A, Viola M, Karousou E, Sevic I, Moretto P, Passi A, Alaniz L, Vigetti D. Deciphering Drug Resistance: Investigating the Emerging Role of Hyaluronan Metabolism and Signaling and Tumor Extracellular Matrix in Cancer Chemotherapy. Int J Mol Sci 2024; 25:7607. [PMID: 39062846 PMCID: PMC11276752 DOI: 10.3390/ijms25147607] [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: 06/07/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Hyaluronan (HA) has gained significant attention in cancer research for its role in modulating chemoresistance. This review aims to elucidate the mechanisms by which HA contributes to chemoresistance, focusing on its interactions within the tumor microenvironment. HA is abundantly present in the extracellular matrix (ECM) and binds to cell-surface receptors such as CD44 and RHAMM. These interactions activate various signaling pathways, including PI3K/Akt, MAPK, and NF-κB, which are implicated in cell survival, proliferation, and drug resistance. HA also influences the physical properties of the tumor stroma, enhancing its density and reducing drug penetration. Additionally, HA-mediated signaling contributes to the epithelial-mesenchymal transition (EMT), a process associated with increased metastatic potential and resistance to apoptosis. Emerging therapeutic strategies aim to counteract HA-induced chemoresistance by targeting HA synthesis, degradation, metabolism, or its binding to CD44. This review underscores the complexity of HA's role in chemoresistance and highlights the potential for HA-targeted therapies to improve the efficacy of conventional chemotherapeutics.
Collapse
Affiliation(s)
- Daiana L. Vitale
- Laboratorio de Microambiente Tumoral, Centro de Investigaciones Básicas y Aplicadas (CIBA), Universidad Nacional del Noroeste de la Provincia de Buenos Aires, Junín B6000, Argentina; (D.L.V.); (I.S.); (L.A.)
- Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires (CITNOBA), UNNOBA-UNSAdA-CONICET, Junín 6000, Argentina
| | - Arianna Parnigoni
- Department of Medical Biochemistry and Microbiology, Uppsala University, SE-751 23 Uppsala, Sweden;
| | - Manuela Viola
- Dipartimento di Medicina e Chirurgia, Universitá degli Studi dell’Insubria, 21100 Varese, Italy; (M.V.); (E.K.); (P.M.); (A.P.)
| | - Evgenia Karousou
- Dipartimento di Medicina e Chirurgia, Universitá degli Studi dell’Insubria, 21100 Varese, Italy; (M.V.); (E.K.); (P.M.); (A.P.)
| | - Ina Sevic
- Laboratorio de Microambiente Tumoral, Centro de Investigaciones Básicas y Aplicadas (CIBA), Universidad Nacional del Noroeste de la Provincia de Buenos Aires, Junín B6000, Argentina; (D.L.V.); (I.S.); (L.A.)
- Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires (CITNOBA), UNNOBA-UNSAdA-CONICET, Junín 6000, Argentina
| | - Paola Moretto
- Dipartimento di Medicina e Chirurgia, Universitá degli Studi dell’Insubria, 21100 Varese, Italy; (M.V.); (E.K.); (P.M.); (A.P.)
| | - Alberto Passi
- Dipartimento di Medicina e Chirurgia, Universitá degli Studi dell’Insubria, 21100 Varese, Italy; (M.V.); (E.K.); (P.M.); (A.P.)
| | - Laura Alaniz
- Laboratorio de Microambiente Tumoral, Centro de Investigaciones Básicas y Aplicadas (CIBA), Universidad Nacional del Noroeste de la Provincia de Buenos Aires, Junín B6000, Argentina; (D.L.V.); (I.S.); (L.A.)
- Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires (CITNOBA), UNNOBA-UNSAdA-CONICET, Junín 6000, Argentina
| | - Davide Vigetti
- Dipartimento di Medicina e Chirurgia, Universitá degli Studi dell’Insubria, 21100 Varese, Italy; (M.V.); (E.K.); (P.M.); (A.P.)
| |
Collapse
|
46
|
Rao PP, Shenoy Belle V, Nayak AG, Kumar N, Rao V, Cheruku SP, Prabhu K. Evaluation of the merit of ethanolic extract of Annona reticulata as an anti-cancer agent in human colon cancer cell lines (HCT-116). F1000Res 2024; 12:1571. [PMID: 39866203 PMCID: PMC11759889 DOI: 10.12688/f1000research.141542.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/04/2024] [Indexed: 01/28/2025] Open
Abstract
Background Colon cancer is the third most common cancer type worldwide. Novel alternative therapeutic anti-cancer drugs against colon cancer with less toxicity are to be explored . This study was aimed to explore the anti-proliferative and anti-migratory activity of various fractions of Annona reticulata ethanolic leaf extract on human colon cancer cell lines (HCT-116) and to explore the potential molecular targets from the most potent plant extract fraction. Methods After obtaining ethical clearance from the institutional ethics committee, the extract and fractions were prepared and a preliminary analysis of the phytochemical was done qualitatively. Total phenolic and flavonoids were determined. Ethanolic leaf extract and its fractions were subjected to cytotoxicity analysis using the sulforhodamine B assay and the most promising fraction which showed the highest viability was selected to study anti-migratory activity. The anti-migratory effect was studied using a scratch wound healing assay. Gas chromatography-mass spectrometry (GC-MS) was done to identify the major phytocompounds present in the fraction. The major five phytocompounds identified from the GC-MS were subjected to bioinformatics analysis. Result Among the four fractions, the petroleum ether fraction exhibited the highest anti-proliferative activity. The migration of colon cancer cells was significantly inhibited by the extract and petroleum ether fraction. The major phytocompounds identified from GC-MS were phytol (13.03%), 2,6-bis (3,4-methylenedioxyphenyl)-3,7-dioxabicyclo (3.3.0) octane (11.95%), gamma.-sitosterol (10.45%), alpha.-tocopherol-beta.-D-mannoside (7.50%) and 3-amino-4-piperonyl-5-pyrazolone (5.84%). The bioinformatics analysis of these phytochemicals showed a high potential to affect the levels of key proteins driving colon cancer progression, inhibiting the enzymes and proteins overexpressed in cancer. Conclusion The outcome of this study endorses the potential of phytochemicals of the petroleum ether fraction of ethanolic leaf extract of Annona reticulata for the development of a new chemotherapeutic agent in the treatment of colon cancer.
Collapse
Affiliation(s)
- Pooja Prakash Rao
- Department of Biochemistry, Kastubra Medical College Manipal, Maniapl Academy of Higher Education, Manipal, Karnataka, India
| | - Vijetha Shenoy Belle
- Department of Biochemistry, Kastubra Medical College Manipal, Maniapl Academy of Higher Education, Manipal, Karnataka, India
| | - Akshatha G Nayak
- Division of Biochemistry, Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Export Promotions Industrial Park (EPIP), Industrial Area Hajipur, Vaishali, Bihar, India
| | - Vanishree Rao
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sri Pragnya Cheruku
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Krishnananda Prabhu
- Department of Biochemistry, Kastubra Medical College Manipal, Maniapl Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
47
|
Eremia IA, Serban B, Popa M, Iancu A, Nica S, Cirstoiu C. Practical management of renal cell carcinoma: integrating current approaches with advances in bone metastasis treatment. EFORT Open Rev 2024; 9:488-502. [PMID: 38828980 PMCID: PMC11195343 DOI: 10.1530/eor-23-0178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/05/2024] Open
Abstract
Renal cell carcinoma (RCC) is a common type of tumor that can develop in the kidney. It is responsible for around one-third of all cases of neoplasms. RCC manifests itself in a variety of distinct subtypes. The most frequent of which is clear cell RCC, followed by papillary and chromophobe RCC. RCC has the potential for metastasis to a variety of organs; nevertheless, bone metastases are one of the most common and potentially fatal complications. These bone metastases are characterized by osteolytic lesions that can result in pathological fractures, hypercalcemia, and other complications, which can ultimately lead to a deterioration in quality of life and an increase morbidity. While nephrectomy remains a foundational treatment for RCC, emerging evidence suggests that targeted therapies, including tyrosine kinase inhibitors and T cell checkpoint inhibitors, may offer effective alternatives, potentially obviating the need for adjuvant nephrectomy in certain cases of metastatic RCC Bone metastases continue to be a difficult complication of RCC, which is why more research is required to enhance patient outcome.
Collapse
Affiliation(s)
- Irina-Anca Eremia
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Emergency Medicine, Emergency University Hospital, Bucharest, Romania
| | - Bogdan Serban
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Orthopaedics and Traumatology, Emergency University Hospital Bucharest, Romania
| | - Mihnea Popa
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Orthopaedics and Traumatology, Emergency University Hospital Bucharest, Romania
| | - Adela Iancu
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Silvia Nica
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Emergency Medicine, Emergency University Hospital, Bucharest, Romania
| | - Catalin Cirstoiu
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Orthopaedics and Traumatology, Emergency University Hospital Bucharest, Romania
| |
Collapse
|
48
|
Wang Y, He J, Zhao Q, Bo J, Zhou Y, Sun H, Ding B, Ren M. Evaluating the predictive value of angiogenesis-related genes for prognosis and immunotherapy response in prostate adenocarcinoma using machine learning and experimental approaches. Front Immunol 2024; 15:1416914. [PMID: 38817605 PMCID: PMC11137278 DOI: 10.3389/fimmu.2024.1416914] [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: 04/13/2024] [Accepted: 05/06/2024] [Indexed: 06/01/2024] Open
Abstract
Background Angiogenesis, the process of forming new blood vessels from pre-existing ones, plays a crucial role in the development and advancement of cancer. Although blocking angiogenesis has shown success in treating different types of solid tumors, its relevance in prostate adenocarcinoma (PRAD) has not been thoroughly investigated. Method This study utilized the WGCNA method to identify angiogenesis-related genes and assessed their diagnostic and prognostic value in patients with PRAD through cluster analysis. A diagnostic model was constructed using multiple machine learning techniques, while a prognostic model was developed employing the LASSO algorithm, underscoring the relevance of angiogenesis-related genes in PRAD. Further analysis identified MAP7D3 as the most significant prognostic gene among angiogenesis-related genes using multivariate Cox regression analysis and various machine learning algorithms. The study also investigated the correlation between MAP7D3 and immune infiltration as well as drug sensitivity in PRAD. Molecular docking analysis was conducted to assess the binding affinity of MAP7D3 to angiogenic drugs. Immunohistochemistry analysis of 60 PRAD tissue samples confirmed the expression and prognostic value of MAP7D3. Result Overall, the study identified 10 key angiogenesis-related genes through WGCNA and demonstrated their potential prognostic and immune-related implications in PRAD patients. MAP7D3 is found to be closely associated with the prognosis of PRAD and its response to immunotherapy. Through molecular docking studies, it was revealed that MAP7D3 exhibits a high binding affinity to angiogenic drugs. Furthermore, experimental data confirmed the upregulation of MAP7D3 in PRAD, correlating with a poorer prognosis. Conclusion Our study confirmed the important role of angiogenesis-related genes in PRAD and identified a new angiogenesis-related target MAP7D3.
Collapse
Affiliation(s)
| | | | | | | | | | | | - BeiChen Ding
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - MingHua Ren
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
49
|
Basar OY, Mohammed S, Qoronfleh MW, Acar A. Optimizing cancer therapy: a review of the multifaceted effects of metronomic chemotherapy. Front Cell Dev Biol 2024; 12:1369597. [PMID: 38813084 PMCID: PMC11133583 DOI: 10.3389/fcell.2024.1369597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/29/2024] [Indexed: 05/31/2024] Open
Abstract
Metronomic chemotherapy (MCT), characterized by the continuous administration of chemotherapeutics at a lower dose without prolonged drug-free periods, has garnered significant attention over the last 2 decades. Extensive evidence from both pre-clinical and clinical settings indicates that MCT induces distinct biological effects than the standard Maximum Tolerated Dose (MTD) chemotherapy. The low toxicity profile, reduced likelihood of inducing acquired therapeutic resistance, and low cost of MCT render it an attractive chemotherapeutic regimen option. One of the most prominent aspects of MCT is its anti-angiogenesis effects. It has been shown to stimulate the expression of anti-angiogenic molecules, thereby inhibiting angiogenesis. In addition, MCT has been shown to decrease the regulatory T-cell population and promote anti-tumor immune response through inducing dendritic cell maturation and increasing the number of cytotoxic T-cells. Combination therapies utilizing MCT along with oncolytic virotherapy, radiotherapy or other chemotherapeutic regimens have been studied extensively. This review provides an overview of the current status of MCT research and the established mechanisms of action of MCT treatment and also offers insights into potential avenues of development for MCT in the future.
Collapse
Affiliation(s)
- Oyku Yagmur Basar
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye
| | - Sawsan Mohammed
- Qatar University, QU Health, College of Medicine, Doha, Qatar
| | - M. Walid Qoronfleh
- Q3 Research Institute (QRI), Research and Policy Division, Ypsilanti, MI, United States
| | - Ahmet Acar
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye
| |
Collapse
|
50
|
Zech TJ, Wolf A, Hector M, Bischoff-Kont I, Krishnathas GM, Kuntschar S, Schmid T, Bracher F, Langmann T, Fürst R. 2-Desaza-annomontine (C81) impedes angiogenesis through reduced VEGFR2 expression derived from inhibition of CDC2-like kinases. Angiogenesis 2024; 27:245-272. [PMID: 38403816 PMCID: PMC11021337 DOI: 10.1007/s10456-024-09906-y] [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: 11/07/2023] [Accepted: 01/15/2024] [Indexed: 02/27/2024]
Abstract
Angiogenesis is a crucial process in the progression of various pathologies, like solid tumors, wet age-related macular degeneration, and chronic inflammation. Current anti-angiogenic treatments still have major drawbacks like limited efficacy in diseases that also rely on inflammation. Therefore, new anti-angiogenic approaches are sorely needed, and simultaneous inhibition of angiogenesis and inflammation is desirable. Here, we show that 2-desaza-annomontine (C81), a derivative of the plant alkaloid annomontine previously shown to inhibit endothelial inflammation, impedes angiogenesis by inhibiting CDC2-like kinases (CLKs) and WNT/β-catenin signaling. C81 reduced choroidal neovascularization in a laser-induced murine in vivo model, inhibited sprouting from vascular endothelial growth factor A (VEGF-A)-activated murine aortic rings ex vivo, and reduced angiogenesis-related activities of endothelial cells in multiple functional assays. This was largely phenocopied by CLK inhibitors and knockdowns, but not by inhibitors of the other known targets of C81. Mechanistically, CLK inhibition reduced VEGF receptor 2 (VEGFR2) mRNA and protein expression as well as downstream signaling. This was partly caused by a reduction of WNT/β-catenin pathway activity, as activating the pathway induced, while β-catenin knockdown impeded VEGFR2 expression. Surprisingly, alternative splicing of VEGFR2 was not detected. In summary, C81 and other CLK inhibitors could be promising compounds in the treatment of diseases that depend on angiogenesis and inflammation due to their impairment of both processes.
Collapse
Affiliation(s)
- T J Zech
- Faculty of Biochemistry, Chemistry and Pharmacy, Institute for Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany.
| | - A Wolf
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - M Hector
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - I Bischoff-Kont
- Faculty of Biochemistry, Chemistry and Pharmacy, Institute for Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
| | - G M Krishnathas
- Faculty of Biochemistry, Chemistry and Pharmacy, Institute for Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
| | - S Kuntschar
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - T Schmid
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - F Bracher
- Pharmaceutical Chemistry, Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - T Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - R Fürst
- Faculty of Biochemistry, Chemistry and Pharmacy, Institute for Pharmaceutical Biology, Goethe University Frankfurt, Frankfurt, Germany
- Pharmaceutical Biology, Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|