1
|
Guo F, Ai Y, Chen Y, Wu G, Wu H, Lin J, Jiang Y, Zhang J, Zhang T. Discovery of 3-phenyl-[1,2,4]triazolo[4,3-a]pyridine derivatives as potent smoothened inhibitors against colorectal carcinoma. Bioorg Med Chem 2025; 125:118214. [PMID: 40318542 DOI: 10.1016/j.bmc.2025.118214] [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: 12/10/2024] [Revised: 04/07/2025] [Accepted: 04/25/2025] [Indexed: 05/07/2025]
Abstract
Smoothened (SMO) in Hedgehog (Hh) signaling pathway is intricately associated with the genesis of colorectal carcinoma (CRC), but SMO inhibitor Vismodegib lacks of therapeutic benefits. Based on the principles of preserving critical interactions and ring substitution, a series of 3-phenyl-[1,2,4]triazolo[4,3-a]pyridine derivatives were designed and synthesized. Among them, compound A11 exhibited significant inhibitory activity against SMOWT (IC50 = 0.27 ± 0.06 µM) and SMOD473H (IC50 = 0.84 ± 0.12 µM), respectively, while displayed negligible toxicity towards normal cells. Furthermore, A11 demonstrated superior antiproliferative activity against CRC cells compared to Vismodegib. In additions, A11 competitively bound to SMO and inhibit its downstream signaling pathways. Molecular modeling studies suggested that the triazolopyridine ring of A11 may contribute to the important interaction for binding to SMO. In sum, these results suggest that A11 deserves further investigation as a SMO inhibitor for CRC treatment.
Collapse
Affiliation(s)
- Fengqiu Guo
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Yangcheng Ai
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China; Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Yongxin Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Guowu Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Huanxian Wu
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jingyun Lin
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Ying Jiang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan 442000, China.
| | - Jiajie Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China.
| | - Tingting Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, People's Republic of China.
| |
Collapse
|
2
|
Zhu M, Liao J, Wei M, Huang S, Xu J, Li Q, Pan X. A comparison of the survival outcome of paclitaxel liposome-based chemoradiotherapy with or without rhEndostatin for unresectable esophageal squamous cell carcinoma: a retrospective study. Discov Oncol 2025; 16:925. [PMID: 40418300 DOI: 10.1007/s12672-025-02711-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 05/14/2025] [Indexed: 05/27/2025] Open
Abstract
OBJECTIVES This study aimed to compare the survival outcomes of paclitaxel liposome-based chemoradiotherapy, with or without rhEndostatin, in patients with unresectable locally advanced esophageal squamous cell carcinoma (ESCC). METHODS Patients with ESCC treated with paclitaxel liposome-based definitive chemoradiotherapy (dCRT), with or without rhEndostatin (E), between February 2015 and June 2020 were included. Patients received induction chemotherapy followed by concurrent chemoradiotherapy, with or without rhEndostatin. The chemotherapy regimen consisted of platinum-based doublet (paclitaxel liposome + cisplatin/nedaplatin). RhEndostatin was administered at a dose of 30 mg/d from day 1 to day 5 of each chemotherapy cycle. Total radiotherapy dose was 66-68 Gy, delivered in fractions of 2.0-2.2 Gy/d. Follow-up continued until December 2023. The primary endpoints were 3-year progression-free survival (PFS) rate. Secondary endpoints included 3-year overall survival (OS) rate, objective response rate (ORR), disease control rate (DCR), and toxicity. RESULTS A total of 80 patients were included, with 34 in the dCRT group and 46 in the E + dCRT group. The 3-year PFS was 26.47% (95% confidence interval [CI] 13.19-41.81) in the dCRT group and 56.29% (95% CI 40.79-69.20) in the E + dCRT group (Hazard ratio (HR), 0.50; 95% CI 0.28-0.89, P = 0.012). Patients in the E + dCRT group had a superior 3-year OS compared to those in the dCRT group (80.44% [95% CI 65.77-89.30] vs. 47.06% [95% CI 29.83-62.52]; HR, 0.40; 95% CI 0.21-0.72; P = 0.003). The ORR was 91.18% in the dCRT group and 95.65% in the E + dCRT group. The most common grade 3-4 toxicities were leukopenia, neutropenia, and thrombocytopenia. CONCLUSION The addition of rhEndostatin to paclitaxel liposome-based dCRT may improve clinical outcomes for patients with unresectable ESCC while maintaining manageable toxicities. However, further prospective randomized controlled studies are necessary to confirm the survival benefits of this treatment strategy.
Collapse
Affiliation(s)
- Mengyuan Zhu
- Department of Oncology, The Seventh Affiliated Hospital, Sun Yat-Sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, 518107, China
| | - Jiehao Liao
- Department of Oncology, The Seventh Affiliated Hospital, Sun Yat-Sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, 518107, China
| | - Min Wei
- Department of Oncology, The Seventh Affiliated Hospital, Sun Yat-Sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, 518107, China
| | - Shan Huang
- Department of Oncology, The Seventh Affiliated Hospital, Sun Yat-Sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, 518107, China
| | - Junjie Xu
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, No.651, East Gongfeng Road, Yuexiu District, Guangzhou, China.
| | - Qun Li
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, No.651, East Gongfeng Road, Yuexiu District, Guangzhou, China.
| | - Xiaofen Pan
- Department of Oncology, The Seventh Affiliated Hospital, Sun Yat-Sen University, No.628, Zhenyuan Road, Guangming District, Shenzhen, 518107, China.
| |
Collapse
|
3
|
Zhang S, Zhang T, Kinsella GK, Curtin JF. A review of the efficacy of prostate cancer therapies against castration-resistant prostate cancer. Drug Discov Today 2025; 30:104384. [PMID: 40409404 DOI: 10.1016/j.drudis.2025.104384] [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: 02/11/2025] [Revised: 05/05/2025] [Accepted: 05/16/2025] [Indexed: 05/25/2025]
Abstract
The standard treatments for prostate cancer (PCa) include chemotherapy, hormone therapy, targeted therapies based on androgen receptor (AR) and/or gonadotropin-releasing hormone (GnRH) receptor antagonists, and radiation therapy. But PCa therapeutic resistance remains an unsolved challenge, leading to progression to castration-resistant prostate cancer (CRPC). Emerging PCa therapies - including poly(ADP-ribose) polymerase (PARP) inhibitors, AR crosstalk signalling pathway inhibitors, B-cell lymphoma 2 (BCL-2) inhibitors, cyclin-dependent kinase 4 (CDK4)/CDK6 inhibitors, CRISPR/Cas9, epigenetic inhibitors, and nanotechnology-based drug-delivery approaches - provide promising targeted solutions. Targeted protein degradation therapy, particularly AR degradation therapies, effectively inhibits resistance at its source. This review summarises the established and emerging PCa therapies, focusing on discussing their efficacy in terms of PCa resistance with supporting experimental findings and the mechanisms of PCa drug resistance.
Collapse
Affiliation(s)
- Shengxin Zhang
- School of Food Science and Environmental Health, Technological University Dublin, Grangegorman, Dublin D07 ADY7, Ireland; Sustainability and Health Research Hub (SHRH), Technological University Dublin, Grangegorman, Dublin D07 H6K8, Ireland
| | - Tao Zhang
- School of Food Science and Environmental Health, Technological University Dublin, Grangegorman, Dublin D07 ADY7, Ireland; Sustainability and Health Research Hub (SHRH), Technological University Dublin, Grangegorman, Dublin D07 H6K8, Ireland
| | - Gemma K Kinsella
- School of Food Science and Environmental Health, Technological University Dublin, Grangegorman, Dublin D07 ADY7, Ireland; Sustainability and Health Research Hub (SHRH), Technological University Dublin, Grangegorman, Dublin D07 H6K8, Ireland.
| | - James F Curtin
- School of Food Science and Environmental Health, Technological University Dublin, Grangegorman, Dublin D07 ADY7, Ireland; Sustainability and Health Research Hub (SHRH), Technological University Dublin, Grangegorman, Dublin D07 H6K8, Ireland
| |
Collapse
|
4
|
Chen Y, Ohara T, Hamada Y, Wang Y, Tian M, Noma K, Tazawa H, Fujisawa M, Yoshimura T, Matsukawa A. HIF-PH inhibitors induce pseudohypoxia in T cells and suppress the growth of microsatellite stable colorectal cancer by enhancing antitumor immune responses. Cancer Immunol Immunother 2025; 74:192. [PMID: 40343532 PMCID: PMC12064516 DOI: 10.1007/s00262-025-04067-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 04/18/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND Recent studies have revealed that CD8+ T cells can be activated via genetic upregulation of HIF-1α, thereby augmenting antitumor effector functions. HIF-1α upregulation can be attained by inhibiting HIF-prolyl hydroxylase (HIF-PH) under normoxic conditions, termed pseudohypoxia. This study investigated whether pseudohypoxia induced by HIF-PH inhibitors suppresses Microsatellite stable (MSS) colorectal cancer (CRC) by affecting tumor immune response. METHODS The HIF-PH inhibitors Roxadustat and Vadadustat were utilized in this study. In vitro, we assessed the effects of HIF-PH inhibitors on human and murine colon cancer cell lines (SW480, HT29, Colon26) and murine T cells. In vivo experiments were performed with mice bearing Colon26 tumors to evaluate the effect of these inhibitors on tumor immune responses. Tumor and spleen samples were analyzed using immunohistochemistry, RT-qPCR, and flow cytometry to elucidate potential mechanisms. RESULTS HIF-PH inhibitors demonstrated antitumor effects in vivo but not in vitro. These inhibitors enhanced the tumor immune response by increasing the infiltration of CD8+ and CD4+ tumor-infiltrating lymphocytes (TILs). HIF-PH inhibitors induced IL-2 production in splenic and intratumoral CD4+ T cells, promoting T cell proliferation, differentiation, and immune responses. Roxadustat synergistically enhanced the efficacy of anti-PD-1 antibody for MSS cancer by increasing the recruitment of TILs and augmenting effector-like CD8+ T cells. CONCLUSION Pseudohypoxia induced by HIF-PH inhibitors activates antitumor immune responses, at least in part, through the induction of IL-2 secretion from CD4+ T cells in the spleen and tumor microenvironment, thereby enhancing immune efficacy against MSS CRC.
Collapse
Affiliation(s)
- Yuehua Chen
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Toshiaki Ohara
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| | - Yusuke Hamada
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Yuze Wang
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Miao Tian
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Kazuhiro Noma
- Department of Gastroenterological Surgery, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Hiroshi Tazawa
- Department of Gastroenterological Surgery, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, Japan
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Masayoshi Fujisawa
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Teizo Yoshimura
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Akihiro Matsukawa
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| |
Collapse
|
5
|
Wu Y, Chen Z, Shi M, Qiu S, Zhang Y. Nimotuzumab and bevacizumab combined with temozolomide and radiotherapy in patients with newly diagnosed glioblastoma multiforme: a retrospective single-arm study. J Neurooncol 2025; 172:429-436. [PMID: 39760795 DOI: 10.1007/s11060-024-04932-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 12/26/2024] [Indexed: 01/07/2025]
Abstract
PURPOSE Glioblastoma (GBM), the most common malignant tumor of the central nervous system (CNS) in adults, continues to result in poor survival rates despite standard treatment. Advancements in understanding GBM's molecular complexity have increased interest in targeted therapeutic approaches. This retrospective, single-center, single-arm study combined nimotuzumab and bevacizumab with radiotherapy (RT) and temozolomide (TMZ) for the treatment of newly diagnosed GBM. The objectives were to determine the efficacy of this treatment combination and the associated toxicity. METHODS A retrospective analysis of clinical data of GBM patients treated at our institution from September 2021 to May 2023 with postoperative combination therapy of nimotuzumab, bevacizumab, and TMZ concurrent with RT, as well as maintenance therapy with bevacizumab and TMZ. Follow-ups were performed every 3 to 6 months via hospital visits and telephone interviews. The primary endpoints were overall survival (OS) and progression-free survival (PFS). The secondary endpoint was the incidence of adverse events (AEs). RESULTS A total of 18 patients were included. The median follow-up time was 23 months. The one-year PFS rate was 77.8%, and the one-year OS rate was 94.4%. The median PFS was 18 months (95%CI, 15.9-20.1), and the median OS was 28 months (95%CI, 18.9-37.1). All AEs were controllable. CONCLUSION The combination of nimotuzumab and bevacizumab with TMZ and RT appears to demonstrate efficacy and safety in newly diagnosed GBM patients, providing a reference for clinical treatment. Further prospective studies are needed to confirm our results.
Collapse
Affiliation(s)
- Yaping Wu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, China
| | - Zhiying Chen
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, China
| | - Mingtao Shi
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, China
| | - Shuo Qiu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, China
| | - Yongchun Zhang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, China.
| |
Collapse
|
6
|
Cai J, Wang W, Cong D, Bai Y, Zhang W. Development of treatment strategies for advanced HER2-positive gastric cancer: Insights from clinical trials. Crit Rev Oncol Hematol 2025; 207:104617. [PMID: 39805409 DOI: 10.1016/j.critrevonc.2025.104617] [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: 04/05/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
HER2-positive gastric cancer (GC), a unique molecular subtype, has garnered significant interest in recent years. Here, we review clinical trial data on advanced HER2-positive GC from the past 15 years. Trastuzumab plus standard chemotherapy remain the first-line treatment. The initial survival benefits conferred by immune checkpoint inhibitors plus trastuzumab and standard chemotherapy are encouraging. The combination of ramucirumab and mono-chemotherapy, as well as the antibody conjugated drug trastuzumab deruxtecan, is the recommended second-line regimen. Treatment with immune checkpoint inhibitors plus ramucirumab and mono-chemotherapy shows promise. Despite the limited treatment options for third line and beyond, development of novel therapeutic strategies is expected. Although clinical cure of advanced HER2-positive GC is unlikely, current clinical studies offer valuable insight into regimens that prolong survival.
Collapse
Affiliation(s)
- Jing Cai
- Department of Pediatrics, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Wanning Wang
- Department of Nephrology, the First Hospital of Jilin University, Changchun 130021, China
| | - Dan Cong
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Yuansong Bai
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Wenlong Zhang
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
7
|
Li X, Yang W, Dai C, Qiu Z, Luan X, Zhang X, Zhang L. Integrative multi-Omics and network pharmacology reveal angiogenesis promotion by Quan-Du-Zhong Capsule via VEGFA/PI3K-Akt pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119222. [PMID: 39647590 DOI: 10.1016/j.jep.2024.119222] [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: 09/27/2024] [Revised: 11/25/2024] [Accepted: 12/05/2024] [Indexed: 12/10/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Quan-du-zhong capsule (QDZ), derived from the whole plant extract of Eucommiaulmoides Oliv., is a traditional Chinese herbal medicine used in treating vascular-related diseases, including hypertension and osteoporosis. Despite its established uses, its pro-angiogenic effects and underlying mechanisms require further investigation. AIM OF THIS STUDY This study aims to investigate the pro-angiogenic effects of QDZ and explore the underlying mechanisms. MATERIALS AND METHODS The chemical compositions of QDZ, including its absorbed prototypes in rats, were analyzed using UHPLC-Q Exactive-Orbitrap-MS. The pro-angiogenic activities of QDZ were evaluated in human umbilical vein endothelial cells (HUVECs) through various assays, including CCK-8, migration, scratch, tubule formation, and 3D sprouting assays. Additionally, the pro-angiogenic effects of QDZ were further assessed invivo through the matrigel plug assay and a hindlimb ischemia-reperfusion model, with three-dimensional blood flow visualized via micro-CT. A comprehensive approach involving network pharmacology, molecular docking, transcriptomics, and proteomics was utilized to explore the pro-angiogenic mechanism of QDZ, with validation by Western blot analysis. RESULTS QDZ significantly promoted the proliferation, migration, and tubule formation of HUVECs. The matrigel plug assay further confirmed its pro-angiogenic potential. Invivo, QDZ-treated mice displayed enhanced vascular distribution and faster blood flow recovery post-ischemia-reperfusion. Chemical analysis identified 49 compounds in QDZ, with 16 absorbed prototypes detected in rat plasma. Mechanistic investigations through network pharmacology, transcriptomics, and proteomics suggested that QDZ's pro-angiogenic effects were mediated through the VEGFA/PI3K-Akt signaling pathway, with increased phosphorylation of angiogenesis-related proteins such as PI3K, Akt, FAK, and Src. CONCLUSIONS This study demonstrates that QDZ promotes angiogenesis via activating the VEGFA and its downstream PI3K-Akt signaling pathway, shedding light on the mechanisms that underpin its traditional medicinal use in vascular health.
Collapse
Affiliation(s)
- Xiaofeng Li
- School of Pharmacy, Fudan University, Shanghai, 200120, China
| | - Wanyue Yang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chunlan Dai
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ziyang Qiu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xin Luan
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xuemei Zhang
- School of Pharmacy, Fudan University, Shanghai, 200120, China.
| | - Lijun Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
8
|
Elkaeed EB, Elkady H, Khattab AM, Yousef RG, Al-ghulikah HA, Husein DZ, Ibrahim IM, Elkady MA, Metwaly AM, Eissa IH. Integrated in silico and in vitro exploration of the anti-VEGFR-2 activities of a semisynthetic xanthine alkaloid inhibiting breast cancer. PLoS One 2025; 20:e0316146. [PMID: 39869618 PMCID: PMC11771932 DOI: 10.1371/journal.pone.0316146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 12/05/2024] [Indexed: 01/29/2025] Open
Abstract
This study presents T-1-NBAB, a new compound derived from the natural xanthine alkaloid theobromine, aimed at inhibiting VEGFR-2, a crucial protein in angiogenesis. T-1-NBAB's potential to interacts with and inhibit the VEGFR-2 was indicated using in silico techniques like molecular docking, MD simulations, MM-GBSA, PLIP, essential dynamics, and bi-dimensional projection experiments. DFT experiments was utilized also to study the structural and electrostatic properties of T-1-NBAB. Computational analysis was performed to predict the ADME-Tox profiles of T-1-NBAB. After semisynthesis, the in vitro results showed that T-1-NBAB effectively inhibits VEGFR-2, with an IC50 of 0.115 μM, compared to sorafenib's 0.0591 μM. In vitro tests also demonstrated significant activity of T-1-NBAB against breast cancer cell lines MCF7 and T47D, with IC50 values of 16.88 μM and 61.17 μM, respectively, and high selectivity. Importantly, T-1-NBAB induced early and late apoptosis in MCF7 cells, indicating its potential as a strong anticancer agent. Additionally, T-1-NBAB reduced the migration and healing abilities of MCF7 cells, suggesting it could be a promising anti-angiogenic agent. Overall, these findings suggest that T-1-NBAB is a promising lead compound for further research as a potential treatment for breast cancer.
Collapse
Affiliation(s)
- Eslam B. Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia
| | - Hazem Elkady
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Ahmed M. Khattab
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Reda G. Yousef
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Merit University, Sohag, Egypt
| | - Hanan A. Al-ghulikah
- Department of Chemistry, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Dalal Z. Husein
- Chemistry Department, Faculty of Science, New Valley University, El-Kharja, Egypt
| | - Ibrahim M. Ibrahim
- Biophysics Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Mohamed A. Elkady
- Biochemistry & Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Ahmed M. Metwaly
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Ibrahim H. Eissa
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
9
|
Wu J, Yang R. Peptide Biomarkers - An Emerging Diagnostic Tool and Current Applicable Assay. Curr Protein Pept Sci 2025; 26:167-184. [PMID: 39323336 DOI: 10.2174/0113892037315736240907131856] [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: 04/15/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 09/27/2024]
Abstract
In the past few decades, impressive progress achieved in technology development and improvement has accelerated the application of peptides as diagnostic biomarkers for various diseases. We outline the advantages of peptides as good diagnostic targets, since they serve as molecular surrogates of enzyme activities, much more specific biomarkers than proteins, and also play vital roles in many biological processes. On the basis of an extensive literature survey, peptide markers with high specificity and sensitivity that are currently applied in clinical tests, as well as recently identified, are summarized for the following four major categories of diseases: neurodegenerative disease, heart failure, infectious disease, and cancer. In addition, we summarize a few prevalent techniques used in peptide biomarker discovery and analysis, such as immunoassays, nanopore-based and nanoparticle-based peptide detection, and also MS-based peptide analysis techniques, and their pros and cons. Currently, there are plenty of analytical technologies available to achieve fast, sensitive and reliable peptide analyses, benefiting from the developments of hardware and instrumentation, as well as data analysis software and databases. Thus, with peptides emerging as sensitive, specific and reliable biomarkers for early detection of diseases, therapeutic monitoring, clinical treatment decisions and disease prognosis, the medical need for peptide biomarkers will increase strongly in the future.
Collapse
Affiliation(s)
- Jing Wu
- Department of Clinical Laboratory, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin Institute of Hepatobiliary Disease, Artificial Cell Engineering Technology Research Center, The Third Central Hospital of Tianjin, Tianjin, 300170, P.R. China
| | - Rui Yang
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, P.R. China
| |
Collapse
|
10
|
Esfahani SA, Ma L, Krishna S, Ma H, Raheem SJ, Shuvaev S, Rotile NJ, Weigand-Whittier J, Boice AT, Borges N, Treaba CA, Deffler C, Diyabalanage H, Humblet V, Sosnovik DE, Mahmood U, Heidari P, Shih A, Catana C, Strickland MR, Klempner SJ, Caravan P. Development of a fibrin-targeted theranostic for gastric cancer. Sci Transl Med 2024; 16:eadn7218. [PMID: 39661705 DOI: 10.1126/scitranslmed.adn7218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 08/18/2024] [Accepted: 11/20/2024] [Indexed: 12/13/2024]
Abstract
Patients with advanced gastric cancer (GCa) have limited treatment options, and alternative treatment approaches are necessary to improve their clinical outcomes. Because fibrin is abundant in gastric tumors but not in healthy tissues, we hypothesized that fibrin could be used as a high-concentration depot for a high-energy beta-emitting cytotoxic radiopharmaceutical delivered to tumor cells. We showed that fibrin is present in 64 to 75% of primary gastric tumors and 50 to 100% of metastatic gastric adenocarcinoma cores. First-in-human 64Cu-FBP8 fibrin-targeted positron emission tomography (PET) imaging in seven patients with gastric or gastroesophageal junction cancer showed high probe uptake in all target lesions with tumor-to-background (muscle) uptake ratios of 9.9 ± 6.6 in primary (n = 7) and 11.2 ± 6.6 in metastatic (n = 45) tumors. Using two mouse models of human GCa, one fibrin-high (SNU-16) and one fibrin-low (NCI-N87), we showed that PET imaging with a related fibrin-specific peptide, CM500, labeled with copper-64 (64Cu-CM500) specifically bound to and precisely quantified tumor fibrin in both models. We then labeled the fibrin-specific peptide CM600 with yttrium-90 and showed that 90Y-CM600 effectively decreased tumor growth in these mouse models. Mice carrying fibrin-high SNU-16 tumors experienced tumor growth inhibition and prolonged survival in response to either a single high dosage or fractionated lower dosage of 90Y-CM600, whereas mice carrying fibrin-low NCI-N87 tumors experienced prolonged survival in response to a fractionated lower dosage of 90Y-CM600. These results lay the foundation for a fibrin-targeted theranostic that may expand options for patients with advanced GCa.
Collapse
Affiliation(s)
- Shadi A Esfahani
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Li Ma
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Shriya Krishna
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Hua Ma
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Shvan J Raheem
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Sergey Shuvaev
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Nicholas J Rotile
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Jonah Weigand-Whittier
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Avery T Boice
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Nicholas Borges
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Constantina A Treaba
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Caitlin Deffler
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | | | - David E Sosnovik
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Umar Mahmood
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Pedram Heidari
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Angela Shih
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ciprian Catana
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Matthew R Strickland
- Division of Hematology-Oncology, Mass General Cancer Center and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Samuel J Klempner
- Division of Hematology-Oncology, Mass General Cancer Center and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
- Institute for Innovation in Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| |
Collapse
|
11
|
Lv X, Niu W, Zhang B, Chen J, Yang S, Xue Y, Dong Y, Yuan P, Pan Y, Tan J, Yang YY, Ding X, Zhao X. Self-Assembled Peptide Hydrogels Loaded with Umbilical Cord-Derived Mesenchymal Stem Cells Repairing Injured Endometrium and Restoring Fertility. Adv Healthc Mater 2024; 13:e2400524. [PMID: 39148284 DOI: 10.1002/adhm.202400524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/20/2024] [Indexed: 08/17/2024]
Abstract
Endometrial injury is a major cause of infertility and recurrent miscarriage. However, no clinically available methods currently exist to effectively repair the damaged endometrium. Mesenchymal stem cells (MSCs) have emerged as a promising therapeutic approach for promoting tissue regeneration, yet a biocompatible scaffold capable of delivering MSCs and supporting their growth is needed. Herein, the study reports a peptide hydrogel scaffold, self-assembled from a peptide IVK8-RGD consisting of an ionic complementary peptide sequence IEVEIRVK and a bioactive sequence RGD, to load umbilical cord-derived mesenchymal stem cells (UC-MSCs). This peptide forms a hydrogel under the physiological condition through self-assembly, and the peptide hydrogel exhibits injectability and adhesiveness to uterus, making it suitable for endometrial repair. Importantly, this hydrogel supports the adhesion and proliferation of UC-MSCs in a 3D environment. In vivo experiments using rats with endometrial injury have shown that treatment with IVK8-RGD hydrogel loaded with UC-MSCs effectively restores endometrial thickness, inhibits fibrosis, and facilitates angiogenesis through activating Raf/MEK/ERK pathway, leading to significantly improved fertility and live birth rate. These findings demonstrate the potential of the UC-MSCs-loaded hydrogel in repairing damaged endometrium and may address the unmet clinical needs of treating recurrent miscarriage and infertility induced by endometrial damage.
Collapse
Affiliation(s)
- Xue Lv
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Wenjing Niu
- Department of Reproductive Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Bin Zhang
- Department of Gynecology and Obstetrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Jingbo Chen
- Department of Reproductive Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Shicong Yang
- Department of Pathology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuhuan Xue
- Department of Reproductive Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Yutian Dong
- Department of Reproductive Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Yue Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jeremy Tan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| | - Yi Yan Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Way, Centros #06-01, Singapore, 138668, Singapore
| | - Xin Ding
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
- School of Medicine, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Xiaomiao Zhao
- Department of Reproductive Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan 2nd Road, Guangzhou, 510080, China
| |
Collapse
|
12
|
Syaj S, Saeed A. Profile of Fruquintinib in the Management of Advanced Refractory Metastatic Colorectal Cancer: Design, Development and Potential Place in Therapy. Drug Des Devel Ther 2024; 18:5203-5210. [PMID: 39568782 PMCID: PMC11577260 DOI: 10.2147/dddt.s388577] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/09/2024] [Indexed: 11/22/2024] Open
Abstract
Colorectal cancer (CRC) is a prevalent and deadly cancer, with metastatic CRC (mCRC) often leading to poor outcomes despite advancements in screening and chemotherapy. Anti-angiogenic agents targeting vascular endothelial growth factor (VEGF) pathways have become essential in mCRC treatment. Bevacizumab, a VEGF inhibitor, was the first agent used in this context. However, drug resistance prompted the development of more selective inhibitors, such as fruquintinib, a tyrosine kinase inhibitor (TKI) that targets VEGFR-1, -2, and -3. Fruquintinib has shown promise in clinical trials, particularly for third-line mCRC treatment. The Phase III FRESCO trial in China demonstrated its efficacy, significantly improving overall survival (OS) and progression-free survival (PFS) compared to placebo, with manageable safety concerns like hypertension and hand-foot skin reactions. The FRESCO-2 trial extended these findings to European and North American populations, leading to a recent FDA approval for previously treated mCRC patients. The pharmacodynamic profile of fruquintinib includes potent inhibition of VEGFR, angiogenesis, and lymphangiogenesis. It has shown synergistic effects when combined with other treatments like chemotherapy and immune checkpoint inhibitors (ICIs). Current research focuses on exploring fruquintinib's combination with ICIs, such as PD-1 inhibitors, to enhance treatment efficacy, especially in microsatellite stable (MSS) CRC. Ongoing trials are investigating Fruquintinib's potential in combination with other therapies and its use in earlier lines of treatment. While promising, further studies are required to optimize its place in therapy and identify predictive biomarkers for better patient selection.
Collapse
Affiliation(s)
- Sebawe Syaj
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anwaar Saeed
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
13
|
Sui L, Wang J, Jiang WG, Song X, Ye L. Molecular mechanism of bone metastasis in breast cancer. Front Oncol 2024; 14:1401113. [PMID: 39605887 PMCID: PMC11599183 DOI: 10.3389/fonc.2024.1401113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Bone metastasis is a debilitating complication that frequently occurs in the advanced stages of breast cancer. However, the underlying molecular and cellular mechanisms of the bone metastasis remain unclear. Here, we elucidate how bone metastasis arises from tumor cells that detach from the primary lesions and infiltrate into the surrounding tissue, as well as how these cells disseminate to distant sites. Specifically, we elaborate how tumor cells preferentially grow within the bone micro-environment and interact with bone cells to facilitate bone destruction, characterized as osteoclastic bone metastasis, as well as new bone matrix deposition, characterized as osteoblastic bone metastasis. We also updated the current understanding of the molecular mechanisms underlying bone metastasis and reasons for relapse in breast cancer, and also opportunities of developing novel diagnostic approaches and treatment.
Collapse
Affiliation(s)
- Laijian Sui
- Department of Orthopedics, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Jing Wang
- Department of Intensive Care Unit, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Wen G. Jiang
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Xicheng Song
- Department of Otorhinolaryngol and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Lin Ye
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| |
Collapse
|
14
|
Micheli L, Nobili S, Lucarini E, Toti A, Margiotta F, Ciampi C, Venturi D, Di Cesare Mannelli L, Ghelardini C. New insights in the mechanisms of opioid analgesia and tolerance: Ultramicronized palmitoylethanolamide down-modulates vascular endothelial growth factor-A in the nervous system. Pharmacol Res 2024; 209:107472. [PMID: 39448045 DOI: 10.1016/j.phrs.2024.107472] [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: 04/22/2024] [Revised: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024]
Abstract
Growing evidence suggests that opioid analgesics modulate angiogenesis during pathophysiological processes. Vascular endothelial growth factor-A (VEGF-A) was recently proposed to be involved in pain development. To date, no anti-angiogenic drug is used for pain management. When administered in a bioavailable formulation, (i.e., ultramicronized) N-palmitoylethanolamine (PEA) delays the onset of morphine tolerance, improves morphine analgesic activity and reduces angiogenesis in in vivo models. This study aimed at investigating whether VEGF-A is involved in PEA-induced delay of morphine tolerance. The anti-VEGF-A monoclonal antibody bevacizumab was used as a reference drug. Preemptive and concomitant treatment with ultramicronized PEA delayed morphine tolerance and potentiated the analgesic effect of morphine, while counteracting morphine-induced increase of VEGF-A in the nervous system. Similar results were obtained when bevacizumab was administered together with morphine. Of note, bevacizumab showed an analgesic effect per se. In equianalgesic treatment regimens (obtained through increasing morphine doses and associating PEA), PEA resulted in lower expression of VEGF-A in dorsal root ganglia (DRG) and spinal cord compared to morphine alone. Similar results were observed for plasma levels of the soluble VEGF receptor 1 (sFLT-1). Moreover, in morphine-treated animals, two pain-related genes (i.e., Serpina3n and Eaat2) showed a more than 3-fold increase in their expression at spinal cord and DRG level, with the increase being significantly counteracted by PEA treatment. This study supports the hypothesis that the effects of PEA on morphine analgesia and tolerance may be mediated by the down-modulation of VEGF-A and sFLT-1 in the nervous system and plasma, respectively.
Collapse
Affiliation(s)
- Laura Micheli
- Department of Neuroscience, Psychology, DrugResearch and Child Health - NEUROFARBA - Section of Pharmacology andToxicology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy.
| | - Stefania Nobili
- Department of Neuroscience, Psychology, DrugResearch and Child Health - NEUROFARBA - Section of Pharmacology andToxicology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy
| | - Elena Lucarini
- Department of Neuroscience, Psychology, DrugResearch and Child Health - NEUROFARBA - Section of Pharmacology andToxicology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy
| | - Alessandra Toti
- Department of Neuroscience, Psychology, DrugResearch and Child Health - NEUROFARBA - Section of Pharmacology andToxicology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy
| | - Francesco Margiotta
- Department of Neuroscience, Psychology, DrugResearch and Child Health - NEUROFARBA - Section of Pharmacology andToxicology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy
| | - Clara Ciampi
- Department of Neuroscience, Psychology, DrugResearch and Child Health - NEUROFARBA - Section of Pharmacology andToxicology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy
| | - Daniel Venturi
- Department of Neuroscience, Psychology, DrugResearch and Child Health - NEUROFARBA - Section of Pharmacology andToxicology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, DrugResearch and Child Health - NEUROFARBA - Section of Pharmacology andToxicology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, DrugResearch and Child Health - NEUROFARBA - Section of Pharmacology andToxicology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy
| |
Collapse
|
15
|
Tsang ES, Aggarwal RR, Bergsland EK, Calabrese S, Rozie A, Chaudhuri S, Dhawan MS, Pawlowska N, Grabowsky J, Thomas S, Munster PN. Updated Survival Follow-Up for Phase Ib Trial of the Histone Deacetylase Inhibitor Abexinostat With Pazopanib in Patients With Solid Tumor Malignancies. JCO Precis Oncol 2024; 8:e2400328. [PMID: 39509670 DOI: 10.1200/po.24.00328] [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: 05/22/2024] [Revised: 08/15/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024] Open
Abstract
PURPOSE Histone deacetylase (HDAC) inhibition downregulates hypoxia-inducible factor-1α and modulates multiple metabolomic pathways relevant in cancer. Here we report a potential novel biomarker to predict exceptional responders (>3 years) in patients receiving HDAC and vascular endothelial growth factor (VEGF) inhibition. PATIENTS AND METHODS Patients with solid tumor malignancies were enrolled in this phase Ib trial of abexinostat (4/7 ×21 days) and pazopanib (28/28 days), with a dose expansion in renal cell carcinoma (RCC). Plasma was analyzed for metabolomics and peripheral blood mononuclear cells (PBMCs) for VEGF and HDAC2 expression levels. RESULTS Fifty-one patients were enrolled: n = 36 patients in dose escalation and n = 15 in dose expansion. After the initial report in 2017, six patients had remained on study: four with RCC and one each with medullary thyroid and thymic neuroendocrine carcinoma. One patient with RCC remains on treatment for >11 years after progression on five systemic therapies. Overall, the median duration of therapy measured 5.6 (1-133) months. The median duration of therapy in exceptional responders measured 44.1 (39.8-133+) months. The median overall survival in patients with high PBMC HDAC2 expression versus low HDAC2 was 32.3 versus 9.2 months (P = .004) for all patients and 43.3 versus 25.1 months for patients with RCC (P = .09). Exceptional responders had lower kynurenine levels both pre- and post-treatment (P = .002, P < .001, respectively). HDAC2 and kynurenine expression levels were inversely correlated (P = .02). CONCLUSION Abexinostat added to pazopanib shows extended tolerability and long-term responses and survival. PBMC HDAC2 levels, the abexinostat target, are relevant predictors of response. In addition, metabolomic assessment points to kynurenine as a predictor for exceptional response to combined VEGF plus HDAC inhibition.
Collapse
Affiliation(s)
- Erica S Tsang
- Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Rahul R Aggarwal
- Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA
| | - Emily K Bergsland
- Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA
| | - Susan Calabrese
- Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA
| | - Alexandrine Rozie
- Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA
| | - Sibapriya Chaudhuri
- Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA
| | - Mallika S Dhawan
- Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA
| | - Nela Pawlowska
- Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA
| | - Jennifer Grabowsky
- Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA
| | - Scott Thomas
- Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA
| | - Pamela N Munster
- Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA
| |
Collapse
|
16
|
Mosalmanzadeh N, Pence BD. Oxidized Low-Density Lipoprotein and Its Role in Immunometabolism. Int J Mol Sci 2024; 25:11386. [PMID: 39518939 PMCID: PMC11545486 DOI: 10.3390/ijms252111386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/04/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Modified cholesterols such as oxidized low-density lipoprotein (OxLDL) contribute to atherosclerosis and other disorders through the promotion of foam cell formation and inflammation. In recent years, it has become evident that immune cell responses to inflammatory molecules such as OxLDLs depend on cellular metabolic functions. This review examines the known effects of OxLDL on immunometabolism and immune cell responses in atherosclerosis and several other diseases. We additionally provide context on the relationship between OxLDL and aging/senescence and identify gaps in the literature and our current understanding in these areas.
Collapse
Affiliation(s)
| | - Brandt D. Pence
- College of Health Sciences and Center for Nutraceutical and Dietary Supplement Research, University of Memphis, Memphis, TN 38111, USA
| |
Collapse
|
17
|
Vecchiotti D, Clementi L, Cornacchia E, Di Vito Nolfi M, Verzella D, Capece D, Zazzeroni F, Angelucci A. Evidence of the Link between Stroma Remodeling and Prostate Cancer Prognosis. Cancers (Basel) 2024; 16:3215. [PMID: 39335188 PMCID: PMC11430343 DOI: 10.3390/cancers16183215] [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: 08/11/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Prostate cancer (PCa), the most commonly diagnosed cancer in men worldwide, is particularly challenging for oncologists when a precise prognosis needs to be established. Indeed, the entire clinical management in PCa has important drawbacks, generating an intense debate concerning the possibility to individuate molecular biomarkers able to avoid overtreatment in patients with pathological indolent cancers. To date, the paradigmatic change in the view of cancer pathogenesis prompts to look for prognostic biomarkers not only in cancer epithelial cells but also in the tumor microenvironment. PCa ecology has been defined with increasing details in the last few years, and a number of promising key markers associated with the reactive stroma are now available. Here, we provide an updated description of the most biologically significant and cited prognosis-oriented microenvironment biomarkers derived from the main reactive processes during PCa pathogenesis: tissue adaptations, inflammatory response and metabolic reprogramming. Proposed biomarkers include factors involved in stromal cell differentiation, cancer-normal cell crosstalk, angiogenesis, extracellular matrix remodeling and energy metabolism.
Collapse
Affiliation(s)
- Davide Vecchiotti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Letizia Clementi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Emanuele Cornacchia
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Mauro Di Vito Nolfi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Daniela Verzella
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Daria Capece
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Francesca Zazzeroni
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Adriano Angelucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| |
Collapse
|
18
|
Liu Y, Wu Y, Li Z, Wan D, Pan J. Targeted Drug Delivery Strategies for the Treatment of Hepatocellular Carcinoma. Molecules 2024; 29:4405. [PMID: 39339402 PMCID: PMC11434448 DOI: 10.3390/molecules29184405] [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: 08/21/2024] [Revised: 09/12/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Hepatocellular carcinoma (HCC) ranks among the most prevalent malignant tumors, exhibiting a high incidence rate that presents a substantial threat to human health. The use of sorafenib and lenvatinib, commonly employed as single-agent targeted inhibitors, complicates the treatment process due to the absence of definitive targeting. Nevertheless, the advent of nanotechnology has injected new optimism into the domain of liver cancer therapy. Nanocarriers equipped with active targeting or passive targeting mechanisms have demonstrated the capability to deliver drugs to tumor cells with high efficiency. This approach not only facilitates precise delivery to the affected site but also enables targeted drug release, thereby enhancing therapeutic efficacy. As medical technology progresses, there is an increasing call for innovative treatment modalities, including novel chemotherapeutic agents, gene therapy, phototherapy, immunotherapy, and combinatorial treatments for HCC. These emerging therapies are anticipated to yield improved clinical outcomes for patients, while minimizing systemic toxicity and adverse effects. Consequently, the application of nanotechnology is poised to significantly improve HCC treatment. This review focused on targeted strategies for HCC and the application of nanotechnology in this area.
Collapse
Affiliation(s)
- Yonghui Liu
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
| | - Yanan Wu
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
| | - Zijian Li
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China
| | - Dong Wan
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China
| | - Jie Pan
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
| |
Collapse
|
19
|
Zhang X, Kong H, Liu X, Li Q, Fang X, Wang J, Qin Z, Hu N, Tian J, Cui H, Zhang L. Nomograms for predicting recurrence of HER2-positive breast cancer with different HR status based on ultrasound and clinicopathological characteristics. Cancer Med 2024; 13:e70146. [PMID: 39248049 PMCID: PMC11381954 DOI: 10.1002/cam4.70146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 09/10/2024] Open
Abstract
PURPOSE This study aimed to identify ultrasound and clinicopathological characteristics related to recurrence in HER2-positive (HER2+) breast cancer, and to develop nomograms for predicting recurrence. METHODS In this dual-center study, we retrospectively enrolled 570 patients with HER2+ breast cancer. The ultrasound and clinicopathological characteristics of hormone receptor (HR)-/HER2+ patients and HR+/HER2+ patients were analyzed separately according to HR status. Eighty percent of the original samples from HR-/HER2+ and HR+/HER2+ patients were extracted by bootstrap sampling as the training cohorts, while the remaining 20% were used as the external validation cohorts. Informative characteristics were screened through univariate and multivariable Cox regression in the training cohorts and used to develop nomograms for predicting recurrence. The predictive accuracy was calculated using Harrell's C-index and calibration curves. RESULTS Three informative characteristics (axillary nodal status, calcification, and Adler degree) were identified in HR-/HER2+ patients, and another three (histological grade, axillary nodal status, and echogenic halo) in HR+/HER2+ patients. Based on these, two separate nomograms were constructed to assess recurrence risk. In the training cohorts, the C-index was 0.740 (95% CI: 0.667-0.811) for HR-/HER2+ nomogram, and 0.749 (95% CI: 0.679-0.820) for HR+/HER2+ nomogram. In the validation cohorts, the C-index was 0.708 (95% CI: 0.540-0.877) for HR-/HER2+ group, and 0.705 (95% CI: 0.557-0.853) for HR+/HER2+ group. The calibration curves also indicated the excellent accuracy of the nomograms. CONCLUSIONS Ultrasound performance of HER2+ breast cancers with different HR status was significantly different. Nomograms integrating ultrasound and clinicopathological characteristics exhibited favorable performance and have the potential to serve as a reliable method for predicting recurrence in heterogeneous breast cancer.
Collapse
Affiliation(s)
- Xudong Zhang
- Department of Abdominal Ultrasoundthe First Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Hanqing Kong
- Department of Ultrasound Medicinethe Second Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Xiaoxue Liu
- Department of Ultrasound Medicinethe Second Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Qingxiang Li
- Department of Ultrasound Medicinethe Second Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Xinran Fang
- Department of Ultrasound Medicinethe Second Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Junjia Wang
- Department of Ultrasound Medicinethe Second Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Zihao Qin
- Department of Ultrasound Medicinethe Second Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Nana Hu
- Department of Ultrasound Medicinethe Second Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Jiawei Tian
- Department of Ultrasound Medicinethe Second Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
- Ultrasound molecular imaging Joint laboratory of Heilongjiang province (International Cooperation)HarbinHeilongjiangChina
| | - Hao Cui
- Department of Ultrasound Medicinethe Second Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
- Ultrasound molecular imaging Joint laboratory of Heilongjiang province (International Cooperation)HarbinHeilongjiangChina
| | - Lei Zhang
- Department of Abdominal Ultrasoundthe First Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
- Ultrasound molecular imaging Joint laboratory of Heilongjiang province (International Cooperation)HarbinHeilongjiangChina
| |
Collapse
|
20
|
Cozma A, Sitar-Tăuț AV, Orășan OH, Briciu V, Leucuța D, Sporiș ND, Lazăr AL, Mălinescu TV, Ganea AM, Sporiș BM, Vlad CV, Lupșe M, Țâru MG, Procopciuc LM. VEGF Polymorphisms ( VEGF-936 C/T, VEGF-634 G/C and VEGF-2578 C/A) and Cardiovascular Implications in Long COVID Patients. Int J Mol Sci 2024; 25:8667. [PMID: 39201353 PMCID: PMC11354396 DOI: 10.3390/ijms25168667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
The COVID-19 pandemic has raised awareness of the virus's long-term non-pulmonary consequences. This study examined the relationship between genetic polymorphisms of VEGF and cardiac dysfunction and subclinical atherosclerosis in patients recovering from COVID-19. This study included 67 patients previously diagnosed with COVID-19. VEGF-936C/T, VEGF-634G/C, and VEGF-2578C/A statuses were determined. Conventional echocardiography and arterial parameters assessments were performed at inclusion and at six months after the first assessment. For VEGF-936C/T, dominant and over-dominant models showed a significant increase in ejection fraction at six months after COVID (p = 0.044 and 0.048) and was also a predictive independent factor for the augmentation index (β = 3.07; p = 0.024). The dominant model showed a rise in RV-RA gradient (3.702 mmHg) (p = 0.028 95% CI: 0.040-7.363), with the over-dominant model indicating a greater difference (4.254 mmHg) (p = 0.025 95% CI: 0.624-7.884). The findings for VEGF-634G/C were not statistically significant, except for a difference in TAPSE during initial evaluation, using the codominant model. For VEGF-2578C/A, a difference in ventricular filling pressure (E/E'ratio) was best described under the recessive model. Our research suggests that the VEG-936C/T genotype may impact the baseline level and subsequent changes in cardiac function and subclinical atherosclerosis. These findings offer valuable insights into the complex correlation between genetic polymorphisms and cardiovascular disfunction in long COVID patients.
Collapse
Affiliation(s)
- Angela Cozma
- Department of Internal Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Adela Viviana Sitar-Tăuț
- Department of Internal Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Olga Hilda Orășan
- Department of Internal Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Violeta Briciu
- Department of Infectious Diseases and Epidemiology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400348 Cluj-Napoca, Romania
| | - Daniel Leucuța
- Department of Medical Informatics and Biostatistics, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Nicolae-Dan Sporiș
- Department of Medical Oncology, Prof. Dr. I. Chiricuța Oncology Institute, 400015 Cluj-Napoca, Romania
| | - Andrada-Luciana Lazăr
- Department of Dermatology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Toma-Vlad Mălinescu
- Department of Internal Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Andreea-Maria Ganea
- Department of Cardiology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Bianca Mihaela Sporiș
- Department of Gastroenterology, Regional Institute of Gastroenterology “Prof. Dr. Octavian Fodor”, 400394 Cluj-Napoca, Romania
| | - Călin Vasile Vlad
- Department of Internal Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Mihaela Lupșe
- Department of Internal Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Mădălina-Gabriela Țâru
- Department of Gastroenterology, Regional Institute of Gastroenterology “Prof. Dr. Octavian Fodor”, 400394 Cluj-Napoca, Romania
| | - Lucia Maria Procopciuc
- Department of Medical Biochemistry, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|
21
|
Zhang JL, Fan DG, Yin W, Hu B. CM082 suppresses hypoxia-induced retinal neovascularization in larval zebrafish. Front Pharmacol 2024; 15:1336249. [PMID: 39135806 PMCID: PMC11317304 DOI: 10.3389/fphar.2024.1336249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
Retinal neovascularization is a common feature of several ocular neovascular diseases, which are the leading cause of blindness in the world. Current treatments are administered through invasive intravitreal injections, leading to poor patient compliance, serious ocular complications and heavy economic burdens. Thus, an alternative less or non-invasive therapeutic strategy is in demand. Here, a non-invasive oral tyrosine kinase inhibitor, CM082, was evaluated in a retinal neovascularization model induced by hypoxia in zebrafish larvae. We found that CM082 effectively suppressed retinal neovascularization, rescued cell loss in the retinal ganglion cell layer, and rescued the visual function deficiency. Our results elucidated that CM082 mediated its therapeutic efficacy primarily through the inhibition of Vegfr2 phosphorylation. The findings demonstrated that CM082 possessed strong antiangiogenic effects and may serve as a potential treatment for angiogenesis in ocular neovascular diseases.
Collapse
Affiliation(s)
- Jun-long Zhang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Ding-gang Fan
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wu Yin
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Anhui Province Key Laboratory of Geriatric Immunology and Nutrition Therapy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Bing Hu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
22
|
Yan L, Shi J, Zhu J. Cellular and molecular events in colorectal cancer: biological mechanisms, cell death pathways, drug resistance and signalling network interactions. Discov Oncol 2024; 15:294. [PMID: 39031216 PMCID: PMC11265098 DOI: 10.1007/s12672-024-01163-1] [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] [Received: 05/27/2024] [Accepted: 07/15/2024] [Indexed: 07/22/2024] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related deaths worldwide, affecting millions each year. It emerges from the colon or rectum, parts of the digestive system, and is closely linked to both genetic and environmental factors. In CRC, genetic mutations such as APC, KRAS, and TP53, along with epigenetic changes like DNA methylation and histone modifications, play crucial roles in tumor development and treatment responses. This paper delves into the complex biological underpinnings of CRC, highlighting the pivotal roles of genetic alterations, cell death pathways, and the intricate network of signaling interactions that contribute to the disease's progression. It explores the dysregulation of apoptosis, autophagy, and other cell death mechanisms, underscoring the aberrant activation of these pathways in CRC. Additionally, the paper examines how mutations in key molecular pathways, including Wnt, EGFR/MAPK, and PI3K, fuel CRC development, and how these alterations can serve as both diagnostic and prognostic markers. The dual function of autophagy in CRC, acting as a tumor suppressor or promoter depending on the context, is also scrutinized. Through a comprehensive analysis of cellular and molecular events, this research aims to deepen our understanding of CRC and pave the way for more effective diagnostics, prognostics, and therapeutic strategies.
Collapse
Affiliation(s)
- Lei Yan
- Medical Department, The Central Hospital of Shaoyang Affiliated to University of South China, Shaoyang, China
| | - Jia Shi
- Department of Obstetrics and Gynecology, The Central Hospital of Shaoyang Affiliated to University of South China, Shaoyang, China
| | - Jiazuo Zhu
- Department of Oncology, Xuancheng City Central Hospital, No. 117 Tong Road, Xuancheng, Anhui, China.
| |
Collapse
|
23
|
Hheidari A, Mohammadi J, Ghodousi M, Mahmoodi M, Ebrahimi S, Pishbin E, Rahdar A. Metal-based nanoparticle in cancer treatment: lessons learned and challenges. Front Bioeng Biotechnol 2024; 12:1436297. [PMID: 39055339 PMCID: PMC11269265 DOI: 10.3389/fbioe.2024.1436297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024] Open
Abstract
Cancer, being one of the deadliest diseases, poses significant challenges despite the existence of traditional treatment approaches. This has led to a growing demand for innovative pharmaceutical agents that specifically target cancer cells for effective treatment. In recent years, the use of metal nanoparticles (NPs) as a promising alternative to conventional therapies has gained prominence in cancer research. Metal NPs exhibit unique properties that hold tremendous potential for various applications in cancer treatment. Studies have demonstrated that certain metals possess inherent or acquired anticancer capabilities through their surfaces. These properties make metal NPs an attractive focus for therapeutic development. In this review, we will investigate the applicability of several distinct classes of metal NPs for tumor targeting in cancer treatment. These classes may include gold, silver, iron oxide, and other metals with unique properties that can be exploited for therapeutic purposes. Additionally, we will provide a comprehensive summary of the risk factors associated with the therapeutic application of metal NPs. Understanding and addressing these factors will be crucial for successful clinical translation and to mitigate any potential challenges or failures in the translation of metal NP-based therapies. By exploring the therapeutic potential of metal NPs and identifying the associated risk factors, this review aims to contribute to the advancement of cancer treatment strategies. The anticipated outcome of this review is to provide valuable insights and pave the way for the advancement of effective and targeted therapies utilizing metal NPs specifically for cancer patients.
Collapse
Affiliation(s)
- Ali Hheidari
- Department of Mechanical Engineering, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Javad Mohammadi
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Maryam Ghodousi
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, United States
| | - Mohammadreza Mahmoodi
- Bio-microfluidics Lab, Department of Electrical Engineering and Information Technology, Iranian Research Organization for Science and Technology, Tehran, Iran
| | - Sina Ebrahimi
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Esmail Pishbin
- Bio-microfluidics Lab, Department of Electrical Engineering and Information Technology, Iranian Research Organization for Science and Technology, Tehran, Iran
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol, Iran
| |
Collapse
|
24
|
Aloss K, Hamar P. Augmentation of the EPR effect by mild hyperthermia to improve nanoparticle delivery to the tumor. Biochim Biophys Acta Rev Cancer 2024; 1879:189109. [PMID: 38750699 DOI: 10.1016/j.bbcan.2024.189109] [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: 02/12/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/20/2024]
Abstract
The clinical translation of the nanoparticle (NP)-based anticancer therapies is still unsatisfactory due to the heterogeneity of the enhanced permeability and retention (EPR) effect. Despite the promising preclinical outcome of the pharmacological EPR enhancers, their systemic toxicity can limit their clinical application. Hyperthermia (HT) presents an efficient tool to augment the EPR by improving tumor blood flow (TBF) and vascular permeability, lowering interstitial fluid pressure (IFP), and disrupting the structure of the extracellular matrix (ECM). Furthermore, the HT-triggered intravascular release approach can overcome the EPR effect. In contrast to pharmacological approaches, HT is safe and can be focused to cancer tissues. Moreover, HT conveys direct anti-cancer effects, which improve the efficacy of the anti-cancer agents encapsulated in NPs. However, the clinical application of HT is challenging due to the heterogeneous distribution of temperature within the tumor, the length of the treatment and the complexity of monitoring.
Collapse
Affiliation(s)
- Kenan Aloss
- Institute of Translational Medicine - Semmelweis University - 1094, Tűzoltó utca, 37-49, Budapest, Hungary
| | - Péter Hamar
- Institute of Translational Medicine - Semmelweis University - 1094, Tűzoltó utca, 37-49, Budapest, Hungary.
| |
Collapse
|
25
|
Amin N, Abbasi IN, Wu F, Shi Z, Sundus J, Badry A, Yuan X, Zhao BX, Pan J, Mi XD, Luo Y, Geng Y, Fang M. The Janus face of HIF-1α in ischemic stroke and the possible associated pathways. Neurochem Int 2024; 177:105747. [PMID: 38657682 DOI: 10.1016/j.neuint.2024.105747] [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: 10/30/2023] [Revised: 03/01/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
Stroke is the most devastating disease, causing paralysis and eventually death. Many clinical and experimental trials have been done in search of a new safe and efficient medicine; nevertheless, scientists have yet to discover successful remedies that are also free of adverse effects. This is owing to the variability in intensity, localization, medication routes, and each patient's immune system reaction. HIF-1α represents the modern tool employed to treat stroke diseases due to its functions: downstream genes such as glucose metabolism, angiogenesis, erythropoiesis, and cell survival. Its role can be achieved via two downstream EPO and VEGF strongly related to apoptosis and antioxidant processes. Recently, scientists paid more attention to drugs dealing with the HIF-1 pathway. This review focuses on medicines used for ischemia treatment and their potential HIF-1α pathways. Furthermore, we discussed the interaction between HIF-1α and other biological pathways such as oxidative stress; however, a spotlight has been focused on certain potential signalling contributed to the HIF-1α pathway. HIF-1α is an essential regulator of oxygen balance within cells which affects and controls the expression of thousands of genes related to sustaining homeostasis as oxygen levels fluctuate. HIF-1α's role in ischemic stroke strongly depends on the duration and severity of brain damage after onset. HIF-1α remains difficult to investigate, particularly in ischemic stroke, due to alterations in the acute and chronic phases of the disease, as well as discrepancies between the penumbra and ischemic core. This review emphasizes these contrasts and analyzes the future of this intriguing and demanding field.
Collapse
Affiliation(s)
- Nashwa Amin
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Department of Zoology, Faculty of Science, Aswan University, Egypt; Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Irum Naz Abbasi
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Wu
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zongjie Shi
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Javaria Sundus
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Azhar Badry
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xia Yuan
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing-Xin Zhao
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jie Pan
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xiao-Dan Mi
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuhuan Luo
- Department of Pediatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Geng
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Marong Fang
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China; Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
26
|
Hua C, Guo Z, Dai M, Zhou J, Ge H, Xue G, Xu F, Ru L, Lv K, Zhang G, Zheng L, Wang M, Teng Y, Yu W, Guo W. Lumbrokinase Extracted from Earthworms Synergizes with Bevacizumab and Chemotherapeutics in Treating Non-Small Cell Lung Cancer by Targeted Inactivation of BPTF/VEGF and NF-κB/COX-2 Signaling. Biomolecules 2024; 14:741. [PMID: 39062456 PMCID: PMC11274885 DOI: 10.3390/biom14070741] [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: 04/29/2024] [Revised: 06/11/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
As a kind of proteolytic enzyme extracted from earthworms, lumbrokinase has been used as an antithrombotic drug clinically. Nevertheless, its potential in anti-cancer, especially in anti-non-small cell lung cancer (NSCLC), as a single form of treatment or in combination with other therapies, is still poorly understood. In this study, we explored the anti-tumor role and the responsive molecular mechanisms of lumbrokinase in suppressing tumor angiogenesis and chemoresistance development in NSCLC and its clinical potential in combination with bevacizumab and chemotherapeutics. Lumbrokinase was found to inhibit cell proliferation in a concentration-dependent manner and caused metastasis suppression and apoptosis induction to varying degrees in NSCLC cells. Lumbrokinase enhanced the anti-angiogenesis efficiency of bevacizumab by down-regulating BPTF expression, decreasing its anchoring at the VEGF promoter region and subsequent VEGF expression and secretion. Furthermore, lumbrokinase treatment reduced IC50 values of chemotherapeutics and improved their cytotoxicity in parental and chemo-resistant NSCLC cells via inactivating the NF-κB pathway, inhibiting the expression of COX-2 and subsequent secretion of PGE2. LPS-induced NF-κB activation reversed its inhibition on NSCLC cell proliferation and its synergy with chemotherapeutic cytotoxicity, while COX-2 inhibitor celecoxib treatment boosted such effects. Lumbrokinase combined with bevacizumab, paclitaxel, or vincristine inhibited the xenograft growth of NSCLC cells in mice more significantly than a single treatment. In conclusion, lumbrokinase inhibited NSCLC survival and sensitized NSCLC cells to bevacizumab or chemotherapeutics treatment by targeted down-regulation of BPTF/VEGF signaling and inactivation of NF-κB/COX-2 signaling, respectively. The combinational applications of lumbrokinase with bevacizumab or chemotherapeutics are expected to be developed as promising candidate therapeutic strategies to improve the efficacy of the original monotherapy in anti-NSCLC.
Collapse
Affiliation(s)
- Chunyu Hua
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Ziyue Guo
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Meng Dai
- Dalian Municipal Central Hospital, Dalian University of Technology, Dalian 116044, China;
| | - Jie Zhou
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Hanxiao Ge
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Guoqing Xue
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Fahui Xu
- The Second Clinical College, Dalian Medical University, Dalian 116044, China;
| | - Liyuan Ru
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Kuan Lv
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Guohui Zhang
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Lina Zheng
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Meiyi Wang
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Yun Teng
- The Second Affiliated Hospital, Dalian Medical University, Dalian 116044, China;
| | - Wendan Yu
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| | - Wei Guo
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China; (C.H.); (Z.G.); (J.Z.); (H.G.); (G.X.); (L.R.); (K.L.); (G.Z.); (L.Z.); (M.W.)
| |
Collapse
|
27
|
K Karunakar K, Cheriyan BV, R K, M G, B A. "Therapeutic advancements in nanomedicine: The multifaceted roles of silver nanoparticles". BIOTECHNOLOGY NOTES (AMSTERDAM, NETHERLANDS) 2024; 5:64-79. [PMID: 39416696 PMCID: PMC11446369 DOI: 10.1016/j.biotno.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 10/19/2024]
Abstract
Nanotechnology has the advantages of enhanced bioactivity, reduced toxicity, target specificity, and sustained release and NPs can penetrate cell membranes. The small size of silver nanoparticles, AgNPs, large surface area, and unique physicochemical properties contribute to cell lysis and increased permeability of cell membranes used in the field of biomedicine. Functional precursors integrate with phytochemicals to create distinctive therapeutic properties and the stability of the nanoparticles can be enhanced by Surface coatings and encapsulation methods, The current study explores the various synthesis methods and characterization techniques of silver nanoparticles (AgNPs) and highlights their intrinsic activity in therapeutic applications, Anti-cancer activity noted at a concentration range of 5-50 μg/ml and angiogenesis is mitigated at a dosage range of 10-50 μg/ml, Diabetes is controlled within the same concentration. Wound healing is improved at concentrations of 10-50 μg/ml and with a typical range of 10-08 μg/ml for bacteria with antimicrobial capabilities. Advancement of silver nanoparticles with a focus on the future use of AgNPs-coated wound dressings and medical devices to decrease the risk of infection. Chemotherapeutic drugs can be administered by AgNPs, which reduces adverse effects and an improvement in treatment outcomes. AgNPs have been found to improve cell proliferation and differentiation, making them beneficial for tissue engineering and regenerative medicine. Our study highlights emerging patterns and developments in the field of medicine, inferring potential future paths.
Collapse
Affiliation(s)
- Karthik K Karunakar
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, TN, India
| | - Binoy Varghese Cheriyan
- Department of Pharmaceutical Chemistry, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, TN, India
| | - Krithikeshvaran R
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, TN, India
| | - Gnanisha M
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, TN, India
| | - Abinavi B
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, TN, India
| |
Collapse
|
28
|
Fadaly WAA, Nemr MTM, Kahk NM. Discovery of novel pyrazole based Urea/Thiourea derivatives as multiple targeting VEGFR-2, EGFR WT, EGFR T790M tyrosine kinases and COX-2 Inhibitors, with anti-cancer and anti-inflammatory activities. Bioorg Chem 2024; 147:107403. [PMID: 38691909 DOI: 10.1016/j.bioorg.2024.107403] [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: 02/14/2024] [Revised: 04/15/2024] [Accepted: 04/25/2024] [Indexed: 05/03/2024]
Abstract
A novel series of pyrazole derivatives with urea/thiourea scaffolds 16a-l as hybrid sorafenib/erlotinib/celecoxib analogs was designed, synthesized and tested for its VEGFR-2, EGFRWT, EGFRT790M tyrosine kinases and COX-2, pro-inflammatory cytokines TNF-α and IL-6 inhibitory activities. All the tested compounds showed excellent COX-2 selectivity index in range of 18.04-47.87 compared to celecoxib (S.I. = 26.17) and TNF-α and IL-6 inhibitory activities (IC50 = 5.0-7.50, 6.23-8.93 respectively, compared to celecoxib IC50 = 8.40 and 8.50, respectively). Screening was carried out against 60 human cancer cell lines by National Cancer Institute (NCI), compounds 16a, 16c, 16d and 16 g were the most potent inhibitors with GI% ranges of 100 %, 99.63-87.02 %, 98.98-43.10 % and 98.68-23.62 % respectively, and with GI50 values of 1.76-15.50 µM, 1.60-5.38 µM, 1.68-7.39 µM and 1.81-11.0 µM respectively, in addition, of showing good safety profile against normal cell line (F180). Moreover, compounds 16a, 16c, 16d and 16 g had cell cycle arrest at G2/M phase with induced necrotic percentage compared to sorafenib of 2.06 %, 2.47 %, 1.57 %, 0.88 % and 1.83 % respectively. Amusingly, compounds 16a, 16c, 16d and 16 g inhibited VEGFR-2 with IC50 of 25 nM, 52 nM, 324 nM and 110 nM respectively, compared to sorafenib (IC50 = 85 nM), and had excellent EGFRWT and EGFRT790M kinase inhibitory activities (IC50 = 94 nM, 128 nM, 160 nM, 297 nM), (10 nM, 25 nM, 36 nM and 48 nM) respectively, compared to both erlotinib and osimertinib (IC50 = 114 nM, 56 nM) and (70 nM, 37 nM) respectively and showed (EGFRwt/EGFRT790M S.I.) of (range: 4.44-9.40) compared to erlotinib (2.03) and osmertinib (1.89).
Collapse
Affiliation(s)
- Wael A A Fadaly
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Mohamed T M Nemr
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Eini street 11562, Cairo, Egypt.
| | - Nesma M Kahk
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| |
Collapse
|
29
|
Maruya Y, Akazawa Y, Norimatsu K, Sailaubekova Y, Zhumagazhiyeva N, Kobayashi S, Higashi M, Hashiguchi K, Yamaguchi N, Nakashima M, Nakao K, Kanetaka K, Eguchi S. Long-term prognosis and DNA damage status after oral mucosal epithelial cell sheet transplantation following esophageal endoscopic submucosal dissection for squamous cell carcinoma: A case series. Regen Ther 2024; 26:557-563. [PMID: 39228905 PMCID: PMC11369366 DOI: 10.1016/j.reth.2024.08.007] [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: 12/21/2023] [Revised: 07/04/2024] [Accepted: 08/02/2024] [Indexed: 09/05/2024] Open
Abstract
Autologous oral mucosal epithelial cell sheet (AOMECS) transplantation has recently been applied in human patients to prevent postprocedural stenosis following endoscopic submucosal dissection (ESD) for esophageal squamous cell carcinoma. However, the long-term safety of AOMECS transplantation remains unclear. We evaluated the long-term outcomes of 10 patients who participated in a clinical trial of AOMECS transplantation after esophageal ESD. Additionally, we assessed the local DNA damage response in the esophageal epithelium using p53 binding protein 1 (53BP1) immunofluorescence in post-AOMECS biopsy specimens. The median follow-up period was 118.5 months (range: 46-130 months). Two patients developed primary esophageal cancer near the AOMECS site and successfully underwent additional ESD. One patient developed lymph node metastasis and underwent chemotherapy. None of the patients died from the original disease, although one patient died from unrelated causes. The rate of abnormal 53BP1 nuclear foci, indicative of increased genome instability, increased with the progression of neoplasia in patients post AOMECS. Our case series suggests that AOMECS transplantation provides an acceptable long-term prognosis and 53BP1 foci may serve as a useful marker for assessing DNA instability in the post-AOMECS esophageal epithelium.
Collapse
Affiliation(s)
- Yasuhiro Maruya
- Tissue Engineering and Regenerative Therapeutics in Gastrointestinal Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yuko Akazawa
- Department of Histology and Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kiyuu Norimatsu
- Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Yerkezhan Sailaubekova
- Department of Histology and Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Nazigul Zhumagazhiyeva
- Department of Histology and Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shinichiro Kobayashi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Miki Higashi
- Tissue Engineering and Regenerative Therapeutics in Gastrointestinal Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Keiichi Hashiguchi
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Naoyuki Yamaguchi
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Masahiro Nakashima
- Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Kazuhiko Nakao
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kengo Kanetaka
- Tissue Engineering and Regenerative Therapeutics in Gastrointestinal Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Susumu Eguchi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
30
|
Zhai J, Liu Y, Ji W, Huang X, Wang P, Li Y, Li H, Wong AHH, Zhou X, Chen P, Wang L, Yang N, Chen C, Chen H, Mak PI, Deng CX, Martins R, Yang M, Ho TY, Yi S, Yao H, Jia Y. Drug screening on digital microfluidics for cancer precision medicine. Nat Commun 2024; 15:4363. [PMID: 38778087 PMCID: PMC11111680 DOI: 10.1038/s41467-024-48616-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Drug screening based on in-vitro primary tumor cell culture has demonstrated potential in personalized cancer diagnosis. However, the limited number of tumor cells, especially from patients with early stage cancer, has hindered the widespread application of this technique. Hence, we developed a digital microfluidic system for drug screening using primary tumor cells and established a working protocol for precision medicine. Smart control logic was developed to increase the throughput of the system and decrease its footprint to parallelly screen three drugs on a 4 × 4 cm2 chip in a device measuring 23 × 16 × 3.5 cm3. We validated this method in an MDA-MB-231 breast cancer xenograft mouse model and liver cancer specimens from patients, demonstrating tumor suppression in mice/patients treated with drugs that were screened to be effective on individual primary tumor cells. Mice treated with drugs screened on-chip as ineffective exhibited similar results to those in the control groups. The effective drug identified through on-chip screening demonstrated consistency with the absence of mutations in their related genes determined via exome sequencing of individual tumors, further validating this protocol. Therefore, this technique and system may promote advances in precision medicine for cancer treatment and, eventually, for any disease.
Collapse
Affiliation(s)
- Jiao Zhai
- State Key Laboratory of Analog and Mixed-Signal VLSI, Institute of Microelectronics, University of Macau, Macau SAR, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, China
| | - Yingying Liu
- State Key Laboratory of Analog and Mixed-Signal VLSI, Institute of Microelectronics, University of Macau, Macau SAR, China
- Faculty of Science and Technology, University of Macau, Macau SAR, China
| | - Weiqing Ji
- School of Computer and Communication Engineering, University of Science and Technology Beijing, Beijing, China
| | - Xinru Huang
- Liver Transplantation Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ping Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yunyi Li
- State Key Laboratory of Analog and Mixed-Signal VLSI, Institute of Microelectronics, University of Macau, Macau SAR, China
| | - Haoran Li
- State Key Laboratory of Analog and Mixed-Signal VLSI, Institute of Microelectronics, University of Macau, Macau SAR, China
- Faculty of Science and Technology, University of Macau, Macau SAR, China
| | - Ada Hang-Heng Wong
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China
| | - Xiong Zhou
- State Key Laboratory of Analog and Mixed-Signal VLSI, Institute of Microelectronics, University of Macau, Macau SAR, China
- College of electrical and information engineering, Hunan University, Changsha, China
| | - Ping Chen
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Lianhong Wang
- College of electrical and information engineering, Hunan University, Changsha, China
| | - Ning Yang
- State Key Laboratory of Analog and Mixed-Signal VLSI, Institute of Microelectronics, University of Macau, Macau SAR, China
- Department of Electronic Information Engineering, Jiangsu University, Zhenjiang, China
| | - Chi Chen
- Liver Transplantation Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Haitian Chen
- Liver Transplantation Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Pui-In Mak
- State Key Laboratory of Analog and Mixed-Signal VLSI, Institute of Microelectronics, University of Macau, Macau SAR, China
- Faculty of Science and Technology, University of Macau, Macau SAR, China
| | - Chu-Xia Deng
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Rui Martins
- State Key Laboratory of Analog and Mixed-Signal VLSI, Institute of Microelectronics, University of Macau, Macau SAR, China
- Faculty of Science and Technology, University of Macau, Macau SAR, China
- On leave from Instituto Superior Tecnico, Universidade de Lisboa, Lisboa, Portugal
| | - Mengsu Yang
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, China
| | - Tsung-Yi Ho
- Department of Compute Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Shuhong Yi
- Liver Transplantation Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Hailong Yao
- School of Computer and Communication Engineering, University of Science and Technology Beijing, Beijing, China.
| | - Yanwei Jia
- State Key Laboratory of Analog and Mixed-Signal VLSI, Institute of Microelectronics, University of Macau, Macau SAR, China.
- Faculty of Science and Technology, University of Macau, Macau SAR, China.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China.
| |
Collapse
|
31
|
Sur D, Coroama CI, Audisio A, Fazio R, Coroama M, Lungulescu CV. Clinical Outcome of Colorectal Cancer Patients with Concomitant Hypertension: A Systematic Review and Meta-Analysis. J Pers Med 2024; 14:520. [PMID: 38793102 PMCID: PMC11122181 DOI: 10.3390/jpm14050520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 05/08/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Arterial hypertension is regarded as a possible biomarker of treatment efficacy in colorectal cancer. Also, extended anti-angiogenic use in the metastatic treatment of the colorectal neoplasm may result in elevated blood pressure. We carried out a systematic review and meta-analysis to assess the clinical outcome of colorectal cancer patients with concomitant hypertension (HTN). METHODS We conducted a systematic search on Embase, Web of Science, Scopus, PubMed (Medline), the Cochrane Library, and CINAHL from inception until October 2023 for articles that addressed the relationship between HTN and progressive free survival (PFS), overall survival (OS), and overall response rate (ORR) for the first and second line of systemic therapy in patients with metastatic colorectal cancer. RESULTS Eligibility criteria were met by 16 articles out of 802 screened studies. Pooled analysis showed that HTN was associated with significantly improved PFS (HR: 0.507, 95% CI: 0.460-0.558, p ≤ 0.001) and OS (HR: 0.677, 95% CI: 0.592-0.774, p ≤ 0.001) in patients with metastatic colorectal cancer. In addition, the pooled RR of HTN for the ORR (RR: 1.28, 95% CI: 1.108-1.495, p = 0.001) suggests that HTN could be a predictive factor of ORR in patients with metastatic colorectal cancer. CONCLUSIONS Elevated blood pressure is associated with better clinical outcomes in patients with metastatic colorectal cancer.
Collapse
Affiliation(s)
- Daniel Sur
- Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Haţieganu”, 400012 Cluj-Napoca, Romania;
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuţă”, 400015 Cluj-Napoca, Romania
| | - Constantin Ionut Coroama
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuţă”, 400015 Cluj-Napoca, Romania
| | - Alessandro Audisio
- Department of Digestive Oncology, Institute Jules Bordet, The Brussels University Hospital, 1070 Brussels, Belgium; (A.A.); (R.F.)
| | - Roberta Fazio
- Department of Digestive Oncology, Institute Jules Bordet, The Brussels University Hospital, 1070 Brussels, Belgium; (A.A.); (R.F.)
| | - Maria Coroama
- Department of Pathophysiology, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 4–6 Victor Babeş Street, 400012 Cluj-Napoca, Romania;
- Department of Cardiology, “Niculae Stancioiu” Heart Institute, 400001 Cluj-Napoca, Romania
| | | |
Collapse
|
32
|
Xue J, Deng J, Qin H, Yan S, Zhao Z, Qin L, Liu J, Wang H. The interaction of platelet-related factors with tumor cells promotes tumor metastasis. J Transl Med 2024; 22:371. [PMID: 38637802 PMCID: PMC11025228 DOI: 10.1186/s12967-024-05126-6] [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: 01/16/2024] [Accepted: 03/22/2024] [Indexed: 04/20/2024] Open
Abstract
Platelets not only participate in thrombosis and hemostasis but also interact with tumor cells and protect them from mechanical damage caused by hemodynamic shear stress and natural killer cell lysis, thereby promoting their colonization and metastasis to distant organs. Platelets can affect the tumor microenvironment via interactions between platelet-related factors and tumor cells. Metastasis is a key event in cancer-related death and is associated with platelet-related factors in lung, breast, and colorectal cancers. Although the factors that promote platelet expression vary slightly in terms of their type and mode of action, they all contribute to the overall process. Recognizing the correlation and mechanisms between these factors is crucial for studying the colonization of distant target organs and developing targeted therapies for these three types of tumors. This paper reviews studies on major platelet-related factors closely associated with metastasis in lung, breast, and colorectal cancers.
Collapse
Affiliation(s)
- Jie Xue
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, China
- Department of Blood Transfusion, The Central Hospital of Qingdao Jiaozhou, 99 Yunxi River South Road, Qingdao, 266300, Shandong, China
| | - Jianzhao Deng
- Clinical Laboratory, The Central Hospital of Qingdao Jiaozhou, 99 Yunxi River South Road, Qingdao, 266300, Shandong, China
| | - Hongwei Qin
- Department of Blood Transfusion, The Central Hospital of Qingdao Jiaozhou, 99 Yunxi River South Road, Qingdao, 266300, Shandong, China
| | - Songxia Yan
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, China
| | - Zhen Zhao
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, China
| | - Lifeng Qin
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, China
| | - Jiao Liu
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, China
| | - Haiyan Wang
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, China.
| |
Collapse
|
33
|
Chen P, Wang Y, Zhu X, Huang Y, Chen J, Sun H, Wang Y, Zhao S, You Y, Wu Y, Yang T, Wei T, Duan X, Zhao T, Jia H, Ren J. SiRNA-HIF-1α delivered by attenuated Salmonella enhances the efficacy of Lenvatinib against hepatocellular carcinoma. Int Immunopharmacol 2024; 130:111728. [PMID: 38430801 DOI: 10.1016/j.intimp.2024.111728] [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: 09/07/2023] [Revised: 02/03/2024] [Accepted: 02/16/2024] [Indexed: 03/05/2024]
Abstract
The treatment of hepatocellular carcinoma (HCC) remains a major challenge in the medical field. Lenvatinib, a multi-target tyrosine kinase inhibitor, has demonstrated anti-HCC effects by targeting and inhibiting pathways such as vascular endothelial growth factor receptor 1-3 (VEGFR1-3). However, the therapeutic efficacy of Lenvatinib is subject to various influences, with the hypoxic microenvironment of the tumor being a pivotal factor. Consequently, altering the hypoxic milieu of the tumor emerges as a viable strategy to augment the efficacy of Lenvatinib. Hypoxia-inducible factor-1α (HIF-1α), synthesized by tumor cells in response to oxygen-deprived conditions, regulates the expression of resistance genes, promotes tumor angiogenesis and cell proliferation, enhances tumor cell invasion, and confers resistance to radiotherapy and chemotherapy. Thus, we constructed a self-designed siRNA targeting HIF-1α to suppress its expression and improve the efficacy of Lenvatinib in treating HCC. The therapeutic efficacy of siRNA-HIF-1α in combination with Lenvatinib on HCC were evaluated through in vivo and in vitro experiments. The results showed that the recombinant Salmonella delivering siRNA-HIF-1α in combination with Lenvatinib effectively inhibited tumor growth and prolonged the survival of tumor-bearing mice. This treatment approach reduced cell proliferation and angiogenesis in HCC tissues while promoting tumor cell apoptosis. Additionally, this combined therapy significantly increased the infiltration of T lymphocytes and M1 macrophages within the tumor microenvironment, as well as elevated the proportion of immune cells in the spleen, thereby potentiating the host's immune response against the tumor.
Collapse
Affiliation(s)
- Pengfei Chen
- Department of Interventional Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Yanling Wang
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, PR China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453003, PR China; Chinese Medicine Hospital of Puyang, Puyang, Henan 457001, PR China
| | - Xingshu Zhu
- Department of Interventional Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Yujing Huang
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, PR China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453003, PR China
| | - Jinwei Chen
- Department of Interventional Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Hao Sun
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, PR China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453003, PR China
| | - Yang Wang
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, PR China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453003, PR China
| | - Shenning Zhao
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, PR China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453003, PR China
| | - Yiqing You
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, PR China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453003, PR China
| | - Yufei Wu
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, PR China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453003, PR China
| | - Tongguo Yang
- Department of Interventional Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Tian Wei
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453003, PR China
| | - Xuhua Duan
- Department of Interventional Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Tiesuo Zhao
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453003, PR China
| | - Huijie Jia
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, PR China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453003, PR China.
| | - Jianzhuang Ren
- Department of Interventional Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China.
| |
Collapse
|
34
|
Yang Z, Zhang X, Bai X, Xi X, Liu W, Zhong W. Anti-angiogenesis in colorectal cancer therapy. Cancer Sci 2024; 115:734-751. [PMID: 38233340 PMCID: PMC10921012 DOI: 10.1111/cas.16063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/16/2023] [Accepted: 12/16/2023] [Indexed: 01/19/2024] Open
Abstract
The morbidity of colorectal cancer (CRC) has risen to third place among malignant tumors worldwide. In addition, CRC is a common cancer in China whose incidence increases annually. Angiogenesis plays an important role in the development of tumors because it can bring the nutrients that cancer cells need and take away metabolic waste. Various mechanisms are involved in the formation of neovascularization, and vascular endothelial growth factor is a key mediator. Meanwhile, angiogenesis inhibitors and drug resistance (DR) are challenges to consider when formulating treatment strategies for patients with different conditions. Thus, this review will discuss the molecules, signaling pathways, microenvironment, treatment, and DR of angiogenesis in CRC.
Collapse
Affiliation(s)
- Zhenni Yang
- Department of Gastroenterology and HepatologyGeneral Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive DiseasesTianjinChina
- Department of Gastroenterology and HepatologyXing'an League People's HospitalXing'an LeagueChina
| | - Xuqian Zhang
- Department of Gastroenterology and HepatologyGeneral Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive DiseasesTianjinChina
- Department of Gastroenterology and HepatologyChina Aerospace Science and Industry CorporationBeijingChina
| | - Xiaozhe Bai
- Department of Gastroenterology and HepatologyXing'an League People's HospitalXing'an LeagueChina
| | - Xiaonan Xi
- State Key Laboratory of Medicinal Chemical Biology and College of PharmacyNankai UniversityTianjinChina
| | - Wentian Liu
- Department of Gastroenterology and HepatologyGeneral Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive DiseasesTianjinChina
| | - Weilong Zhong
- Department of Gastroenterology and HepatologyGeneral Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive DiseasesTianjinChina
| |
Collapse
|
35
|
Onyeukwu C, Williams A, Seyboth B, Muñoz L, Scaria G, Kent P. Partial Response to Naxitamab for Brain Metastasis in Neuroblastoma. J Pediatr Hematol Oncol 2024; 46:e188-e190. [PMID: 38189408 DOI: 10.1097/mph.0000000000002807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 11/20/2023] [Indexed: 01/09/2024]
Abstract
Neuroblastoma (NBL) is a common pediatric tumor arising from sympathetic ganglion cells. High-risk NBL is based on age, stage, histology, and MYCN amplification, and is associated with a high mortality rate. The combination of naxitamab (NAX) and granulocyte-macrophage (cerebrospinal fluid) is a new treatment for high-risk and relapsed NBL approved for bone or bone marrow disease. NAX is a monoclonal antibody directed against anti-disialoganglioside, which is overexpressed in neuroblastoma. Under normal circumstances, monoclonal antibodies, such as NAX, cannot cross the blood-brain barrier due to size. We present the case of a patient with high-risk NBL treated with NAX for multiple bony relapses. Unexpectedly, her brain metastasis responded clinically, histologically, and by imaging to the treatment. We believe this is the first documented case of NBL of the brain responding to NAX.
Collapse
Affiliation(s)
| | | | | | | | | | - Paul Kent
- Medical Director, FibroFighters Foundation, Temecula, CA
| |
Collapse
|
36
|
Clissa PB, Della-Casa MS, Zychar BC, Sanabani SS. The Role of Snake Venom Disintegrins in Angiogenesis. Toxins (Basel) 2024; 16:127. [PMID: 38535794 PMCID: PMC10974740 DOI: 10.3390/toxins16030127] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/18/2024] [Accepted: 02/08/2024] [Indexed: 01/03/2025] Open
Abstract
Angiogenesis, the formation of new blood vessels, plays a critical role in various physiological and pathological conditions. Snake venom disintegrins (SVDs) have been identified as significant regulators of this process. In this review, we explore the dual roles of SVD in angiogenesis, both as antiangiogenic agents by inhibiting integrin binding and interfering with vascular endothelial growth factors and as proangiogenic agents by enhancing integrin binding, stimulating cell migration and proliferation, and inducing neoangiogenesis. Studies in vitro and in animal models have demonstrated these effects and offer significant therapeutic opportunities. The potential applications of SVD in diseases related to angiogenesis, such as cancer, ocular diseases, tissue regeneration, wound healing, and cardiovascular diseases, are also discussed. Overall, SVDs are promising potential therapeutics, and further advances in this field could lead to innovative treatments for diseases related to angiogenesis.
Collapse
Affiliation(s)
| | | | | | - Sabri Saeed Sanabani
- Laboratory of Medical Investigation LIM-56, Division of Dermatology, Medical School, University of São Paulo, Sao Paulo 05508-220, Brazil
| |
Collapse
|
37
|
Kim J, Bose D, Araínga M, Haque MR, Fennessey CM, Caddell RA, Thomas Y, Ferrell DE, Ali S, Grody E, Goyal Y, Cicala C, Arthos J, Keele BF, Vaccari M, Lorenzo-Redondo R, Hope TJ, Villinger F, Martinelli E. TGF-β blockade drives a transitional effector phenotype in T cells reversing SIV latency and decreasing SIV reservoirs in vivo. Nat Commun 2024; 15:1348. [PMID: 38355731 PMCID: PMC10867093 DOI: 10.1038/s41467-024-45555-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
HIV-1 persistence during ART is due to the establishment of long-lived viral reservoirs in resting immune cells. Using an NHP model of barcoded SIVmac239 intravenous infection and therapeutic dosing of anti-TGFBR1 inhibitor galunisertib (LY2157299), we confirm the latency reversal properties of in vivo TGF-β blockade, decrease viral reservoirs and stimulate immune responses. Treatment of eight female, SIV-infected macaques on ART with four 2-weeks cycles of galunisertib leads to viral reactivation as indicated by plasma viral load and immunoPET/CT with a 64Cu-DOTA-F(ab')2-p7D3-probe. Post-galunisertib, lymph nodes, gut and PBMC exhibit lower cell-associated (CA-)SIV DNA and lower intact pro-virus (PBMC). Galunisertib does not lead to systemic increase in inflammatory cytokines. High-dimensional cytometry, bulk, and single-cell (sc)RNAseq reveal a galunisertib-driven shift toward an effector phenotype in T and NK cells characterized by a progressive downregulation in TCF1. In summary, we demonstrate that galunisertib, a clinical stage TGF-β inhibitor, reverses SIV latency and decreases SIV reservoirs by driving T cells toward an effector phenotype, enhancing immune responses in vivo in absence of toxicity.
Collapse
Affiliation(s)
- Jinhee Kim
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Deepanwita Bose
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, USA
| | - Mariluz Araínga
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, USA
| | - Muhammad R Haque
- Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Christine M Fennessey
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Rachel A Caddell
- Division of Immunology, Tulane National Primate Research Center, Covington, LA, USA
| | - Yanique Thomas
- Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Douglas E Ferrell
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, USA
| | - Syed Ali
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, USA
| | - Emanuelle Grody
- Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Synthetic Biology, Northwestern University, Chicago, IL, USA
| | - Yogesh Goyal
- Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Synthetic Biology, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Claudia Cicala
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - James Arthos
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Monica Vaccari
- Division of Immunology, Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ramon Lorenzo-Redondo
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL, USA
| | - Thomas J Hope
- Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Francois Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, USA
| | - Elena Martinelli
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
38
|
Gibbons T, Rahmioglu N, Zondervan KT, Becker CM. Crimson clues: advancing endometriosis detection and management with novel blood biomarkers. Fertil Steril 2024; 121:145-163. [PMID: 38309818 DOI: 10.1016/j.fertnstert.2023.12.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 02/05/2024]
Abstract
Endometriosis is an inflammatory condition affecting approximately 10% of the female-born population. Despite its prevalence, the lack of noninvasive biomarkers has contributed to an established global diagnostic delay. The intricate pathophysiology of this enigmatic disease may leave signatures in the blood, which, when detected, can be used as noninvasive biomarkers. This review provides an update on how investigators are utilizing the established disease pathways and innovative methodologies, including genome-wide association studies, next-generation sequencing, and machine learning, to unravel the clues left in the blood to develop blood biomarkers. Many blood biomarkers show promise in the discovery phase, but because of a lack of standardized and robust methodologies, they rarely progress to the development stages. However, we are now seeing biomarkers being validated with high diagnostic accuracy and improvements in standardization protocols, providing promise for the future of endometriosis blood biomarkers.
Collapse
Affiliation(s)
- Tatjana Gibbons
- Oxford Endometriosis CaRe Centre, Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom.
| | - Nilufer Rahmioglu
- Oxford Endometriosis CaRe Centre, Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom; Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Krina T Zondervan
- Oxford Endometriosis CaRe Centre, Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom; Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Christian M Becker
- Oxford Endometriosis CaRe Centre, Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
39
|
McEneaney LJ, Vithayathil M, Khan S. Screening, Surveillance, and Prevention of Hepatocellular Carcinoma. GASTROINTESTINAL ONCOLOGY ‐ A CRITICAL MULTIDISCIPLINARY TEAM APPROACH 2E 2024:271-290. [DOI: 10.1002/9781119756422.ch16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
40
|
Kong L, Li J, Bai Y, Xu S, Zhang L, Chen W, Gao L, Wang F. Inhibition of soluble epoxide hydrolase enhances the dentin-pulp complex regeneration mediated by crosstalk between vascular endothelial cells and dental pulp stem cells. J Transl Med 2024; 22:61. [PMID: 38229161 PMCID: PMC10790489 DOI: 10.1186/s12967-024-04863-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 01/06/2024] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Revascularization and restoration of normal pulp-dentin complex are important for tissue-engineered pulp regeneration. Recently, a unique periodontal tip-like endothelial cells subtype (POTCs) specialized to dentinogenesis was identified. We have confirmed that TPPU, a soluble epoxide hydrolase (sEH) inhibitor targeting epoxyeicosatrienoic acids (EETs) metabolism, promotes bone growth and regeneration by angiogenesis and osteogenesis coupling. We hypothesized that TPPU could also promote revascularization and induce POTCs to contribute to pulp-dentin complex regeneration. Here, we in vitro and in vivo characterized the potential effect of TPPU on the coupling of angiogenesis and odontogenesis and investigated the relevant mechanism, providing new ideas for pulp-dentin regeneration by targeting sEH. METHODS In vitro effects of TPPU on the proliferation, migration, and angiogenesis of dental pulp stem cells (DPSCs), human umbilical vein endothelial cells (HUVECs) and cocultured DPSCs and HUVECs were detected using cell counting kit 8 (CCK8) assay, wound healing, transwell, tube formation and RT-qPCR. In vivo, Matrigel plug assay was performed to outline the roles of TPPU in revascularization and survival of grafts. Then we characterized the VEGFR2 + POTCs around odontoblast layer in the molar of pups from C57BL/6 female mice gavaged with TPPU. Finally, the root segments with DPSCs mixed with Matrigel were implanted subcutaneously in BALB/c nude mice treated with TPPU and the root grafts were isolated for histological staining. RESULTS In vitro, TPPU significantly promoted the migration and tube formation capability of cocultured DPSCs and HUVECs. ALP and ARS staining and RT-qPCR showed that TPPU promoted the osteogenic and odontogenic differentiation of cultured cells, treatment with an anti-TGF-β blocking antibody abrogated this effect. Knockdown of HIF-1α in HUVECs significantly reversed the effect of TPPU on the expression of angiogenesis, osteogenesis and odontogenesis-related genes in cocultured cells. Matrigel plug assay showed that TPPU increased VEGF/VEGFR2-expressed cells in transplanted grafts. TPPU contributed to angiogenic-odontogenic coupling featured by increased VEGFR2 + POTCs and odontoblast maturation during early dentinogenesis in molar of newborn pups from C57BL/6 female mice gavaged with TPPU. TPPU induced more dental pulp-like tissue with more vessels and collagen fibers in transplanted root segment. CONCLUSIONS TPPU promotes revascularization of dental pulp regeneration by enhancing migration and angiogenesis of HUVECs, and improves odontogenic differentiation of DPSCs by TGF-β. TPPU boosts the angiogenic-odontogenic coupling by enhancing VEGFR2 + POTCs meditated odontoblast maturation partly via upregulating HIF-1α, which contributes to increasing pulp-dentin complex for tissue-engineered pulp regeneration.
Collapse
Affiliation(s)
- Lingwenyao Kong
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Juanjuan Li
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Yuwen Bai
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Shaoyang Xu
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Lin Zhang
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Weixian Chen
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China
| | - Lu Gao
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China.
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China.
- The Affiliated Stomatological Hospital of Dalian Medical University, Dalian, China.
| | - Fu Wang
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian, 116044, People's Republic of China.
- Academician Laboratory of Immune and Oral Development & Regeneration, Dalian Medical University, Dalian, China.
- The Affiliated Stomatological Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
41
|
Guo C, Cui Y, Jiao M, Yao J, Zhao J, Tian Y, Dong J, Liao L. Crosstalk between proximal tubular epithelial cells and other interstitial cells in tubulointerstitial fibrosis after renal injury. Front Endocrinol (Lausanne) 2024; 14:1256375. [PMID: 38260142 PMCID: PMC10801024 DOI: 10.3389/fendo.2023.1256375] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/22/2023] [Indexed: 01/24/2024] Open
Abstract
The energy needs of tubular epithelial components, especially proximal tubular epithelial cells (PTECs), are high and they heavily depend on aerobic metabolism. As a result, they are particularly vulnerable to various injuries caused by factors such as ischemia, proteinuria, toxins, and elevated glucose levels. Initial metabolic and phenotypic changes in PTECs after injury are likely an attempt at survival and repair. Nevertheless, in cases of recurrent or prolonged injury, PTECs have the potential to undergo a transition to a secretory state, leading to the generation and discharge of diverse bioactive substances, including transforming growth factor-β, Wnt ligands, hepatocyte growth factor, interleukin (IL)-1β, lactic acid, exosomes, and extracellular vesicles. By promoting fibroblast activation, macrophage recruitment, and endothelial cell loss, these bioactive compounds stimulate communication between epithelial cells and other interstitial cells, ultimately worsening renal damage. This review provides a summary of the latest findings on bioactive compounds that facilitate the communication between these cellular categories, ultimately leading to the advancement of tubulointerstitial fibrosis (TIF).
Collapse
Affiliation(s)
- Congcong Guo
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yuying Cui
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- First Clinical Medical College, Shandong University of Traditional Chinese Medicin, Jinan, Shandong, China
| | - Mingwen Jiao
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Jinming Yao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Junyu Zhao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Yutian Tian
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Jianjun Dong
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lin Liao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- First Clinical Medical College, Shandong University of Traditional Chinese Medicin, Jinan, Shandong, China
| |
Collapse
|
42
|
Zhang C, Sui Y, Liu S, Yang M. Bioinformatic Analysis and Computer-Aided Study to Investigate the Potential Application of a Bioflavonoid Compound Bilobetin in Liver Cancer Treatment. FRONT BIOSCI-LANDMRK 2023; 28:360. [PMID: 38179744 DOI: 10.31083/j.fbl2812360] [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: 06/09/2023] [Revised: 07/21/2023] [Accepted: 08/18/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC/LIHC) is the most common type of primary liver cancer, which is a leading cause of cancer death worldwide. Patients with HCC have a short survival time after diagnosis. Unfortunately, there are no effective treatments for advanced or aggressive HCC. Thus, the rapid development of new therapeutic drugs or treatment methods for HCC is urgently needed. METHODS Bioinformatic tools and computer-aided predictions advance the processes of drug development. In this study, we incorporated bioinformatic analyses and computer-aided drug development processes to investigate the potential application of bilobetin, a bioactive compound of bioflavonoid, as a therapeutic agent for HCC treatment. RESULTS Our results revealed that 4 out of 20 predicted hub target genes of bilobetin displayed functional importance in cancer-related signaling pathways in different cancers, including HCC. Importantly, the mRNA expression levels of these four key hub genes (VEGFA, SRC, MMP9, and CDK1) were significantly different between normal and HCC tumor samples. Their expression levels were significantly associated with the clinical survival outcomes of HCC patients, as well as the immune cell infiltration levels in the HCC tumor microenvironment. In addition, these four genes showed significant co-expression correlated with immune checkpoint genes, including CD274, PDCD1, CTLA4, and CD47. Furthermore, we used computer-aided approaches to investigate the binding affinity and potential binding mechanisms between bilobetin and target proteins encoded by four key hub genes. CONCLUSIONS In conclusion, our study shed light on the potential application of the bioactive bioflavonoid molecule bilobetin in LIHC treatment by regulating four key hub genes.
Collapse
Affiliation(s)
- Chunye Zhang
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65212, USA
| | - Yuxiang Sui
- School of Life Science, Shanxi Normal University, 041004 Linfen, Shanxi, China
| | - Shuai Liu
- The First Affiliated Hospital, Zhejiang University, 310006 Hangzhou, Zhejiang, China
| | - Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
43
|
Raafat SN, El Wahed SA, Badawi NM, Saber MM, Abdollah MR. Enhancing the anticancer potential of metformin: fabrication of efficient nanospanlastics, in vitro cytotoxic studies on HEP-2 cells and reactome enhanced pathway analysis. Int J Pharm X 2023; 6:100215. [PMID: 38024451 PMCID: PMC10630776 DOI: 10.1016/j.ijpx.2023.100215] [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: 05/14/2023] [Revised: 10/17/2023] [Accepted: 10/21/2023] [Indexed: 12/01/2023] Open
Abstract
Metformin (MET), an oral antidiabetic drug, was reported to possess promising anticancer effects. We hypothesized that MET encapsulation in unique nanospanlastics would enhance its anticancer potential against HEP-2 cells. Our results showed the successful fabrication of Nano-MET spanlastics (d = 232.10 ± 0.20 nm; PDI = 0.25 ± 0.11; zeta potential = (-) 44.50 ± 0.96; drug content = 99.90 ± 0.11 and entrapment efficiency = 88.01 ± 2.50%). MTT assay revealed the enhanced Nano-MET cytotoxicity over MET with a calculated IC50 of 50 μg/mL and > 500 μg/mL, respectively. Annexin V/PI apoptosis assay showed that Nano-MET significantly decreased the percentage of live cells from 95.49 to 93.70 compared to MET and increased the percentage of cells arrested in the G0/G1 phase by 8.38%. Moreover, Nano-MET downregulated BCL-2 and upregulated BAX protein levels by 1.57 and 1.88 folds, respectively. RT-qPCR revealed that Nano-MET caused a significant 13.75, 4.15, and 2.23-fold increase in caspase-3, -8, and - 9 levels as well as a 100 and 43.47-fold decrease in cyclin D1 and mTOR levels, respectively. The proliferation marker Ki67 immunofluorescent staining revealed a 3-fold decrease in positive cells in Nano-MET compared to the control. Utilizing the combined Pathway-Enrichment Analysis (PEA) and Reactome analysis indicated high enrichment of certain pathways including nucleotides metabolism, Nudix-type hydrolase enzymes, carbon dioxide hydration, hemostasis, and the innate immune system. In summary, our results confirm MET cytotoxicity enhancement by its encapsulation in nanospanlastics. We also highlight, using PEA, that MET can modulate multiple pathways implicated in carcinogenesis.
Collapse
Affiliation(s)
- Shereen Nader Raafat
- Department of Pharmacology, Faculty of Dentistry, The British University in Egypt, Cairo, Egypt
- Stem Cells and Tissue Culture Hub (CIDS), Faculty of Dentistry, The British University in Egypt, Cairo, Egypt
| | - Sara Abd El Wahed
- Department of Oral Pathology, Faculty of Dentistry, The British University in Egypt, Cairo, Egypt
| | - Noha M. Badawi
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
- Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El Sherouk City, Egypt
| | - Mona M. Saber
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Giza, Egypt
| | - Maha R.A. Abdollah
- Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El Sherouk City, Egypt
- Department of Pharmacology, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| |
Collapse
|
44
|
He Q, He Y, Li C, Wang J, Xia T, Xiong X, Xu J, Liu L. Downregulated BIRC5 inhibits proliferation and metastasis of melanoma through the β-catenin/HIF-1α/VEGF/MMPs pathway. J Cancer Res Clin Oncol 2023; 149:16797-16809. [PMID: 37728702 DOI: 10.1007/s00432-023-05425-3] [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: 07/14/2023] [Accepted: 09/12/2023] [Indexed: 09/21/2023]
Abstract
PURPOSE Melanoma is a malignant skin tumor caused by melanocytes and associated with high mortality rates. This study aims to investigate the specific mechanism of ZWZ-3 in melanoma proliferation and metastasis. METHODS RNA sequencing was performed to identify the effect of ZWZ-3 on gene expression. siRNA was used to inhibit BIRC5 gene expression in the B16F10 cell line. A zebrafish tumor model was used to assess the therapeutic effect of ZWZ-3 in vivo. Mechanistic insights into the inhibition of tumor metastasis by ZWZ-3 were obtained through analysis of tumor tissue sections in mice. RESULTS Our findings demonstrated that ZWZ-3 suppressed melanoma cell proliferation and migration. We performed RNA sequencing in melanoma cells after the treatment with ZWZ-3 and found that Birc5, which is closely associated with tumor metastasis, was significantly down-regulated. Bioinformatics analysis and the immuno-histochemical results of tissue chips for melanoma further confirmed the high expression of BIRC5 in melanoma and its effect on disease progression. Moreover, Birc5 knock-down significantly inhibited melanoma cell proliferation and metastasis, which was correlated with the β-catenin/HIF-1α/VEGF/MMPs pathway. Additionally, ZWZ-3 significantly inhibited tumor growth in the zebrafish tumor model without any evident side effects. Histological and immuno-histochemical analyses revealed that ZWZ-3 inhibited tumor cell metastasis by down-regulating HIF-1α, VEGF, and MMP9. CONCLUSION Our findings revealed that ZWZ-3 could downregulate BIRC5 and inhibit melanoma proliferation and metastasis through the β-catenin/HIF-1α/VEGF/MMPs pathway. Therefore, BIRC5 represents a promising therapeutic target for the treatment of melanoma.
Collapse
Affiliation(s)
- Qingqing He
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yijing He
- Department of Science and Technology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Changqiang Li
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Jianv Wang
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Tong Xia
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Xia Xiong
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Jixiang Xu
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Li Liu
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
45
|
Dai W, Chen Y, Xue Y, Wan M, Mao C, Zhang K. Progress in the Treatment of Peritoneal Metastatic Cancer and the Application of Therapeutic Nanoagents. ACS APPLIED BIO MATERIALS 2023; 6:4518-4548. [PMID: 37916787 DOI: 10.1021/acsabm.3c00662] [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] [Indexed: 11/03/2023]
Abstract
Peritoneal metastatic cancer is a cancer caused by the direct growth of cancer cells from the primary site through the bloodstream, lymph, or peritoneum, which is a difficult part of current clinical treatment. In the abdominal cavity of patients with metastatic peritoneal cancer, there are usually nodules of various sizes and malignant ascites. Among them, nodules of different sizes can obstruct intestinal movement and form intestinal obstruction, while malignant ascites can cause abdominal distension and discomfort, and even cause patients to have difficulty in breathing. The pathology and physiology of peritoneal metastatic cancer are complex and not fully understood. The main hypothesis is "seed" and "soil"; i.e., cells from the primary tumor are shed and implanted in the peritoneal cavity (peritoneal metastasis). In the last two decades, the main treatment modalities used clinically are cytoreductive surgery (CRS), systemic chemotherapy, intraperitoneal chemotherapy, and combined treatment, all of which help to improve patient survival and quality of life (QOL). However, the small-molecule chemotherapeutic drugs used clinically still have problems such as rapid drug metabolism and systemic toxicity. With the rapid development of nanotechnology in recent years, therapeutic nanoagents for the treatment of peritoneal metastatic cancer have been gradually developed, which has improved the therapeutic effect and reduced the systemic toxicity of small-molecule chemotherapeutic drugs to a certain extent. In addition, nanomaterials have been developed not only as therapeutic agents but also as imaging agents to guide peritoneal tumor CRS. In this review, we describe the etiology and pathological features of peritoneal metastatic cancer, discuss in detail the clinical treatments that have been used for peritoneal metastatic cancer, and analyze the advantages and disadvantages of the different clinical treatments and the QOL of the treated patients, followed by a discussion focusing on the progress, obstacles, and challenges in the use of therapeutic nanoagents in peritoneal metastatic cancer. Finally, therapeutic nanoagents and therapeutic tools that may be used in the future for the treatment of peritoneal metastatic cancer are prospected.
Collapse
Affiliation(s)
- Wenjun Dai
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Yidan Chen
- Department of Radiation Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yunxin Xue
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Ke Zhang
- Department of Radiation Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| |
Collapse
|
46
|
Qu FJ, Zhou Y, Wang H. Oral Anlotinib Maintenance Therapy for an Advanced Malignant Peritoneal Mesothelioma Diagnosed by Laparoscopy After Initial Misdiagnosis to Obtain Longer Progression-Free Survival: Case Report and Literature Review. Onco Targets Ther 2023; 16:961-972. [PMID: 38021445 PMCID: PMC10657764 DOI: 10.2147/ott.s430190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023] Open
Abstract
Malignant peritoneal mesothelioma (MPeM) is a rare and highly invasive malignant tumor with a lack of specificity in clinical manifestations, which can easily lead to misdiagnosis and missed diagnosis. Due to the difficulty of early diagnosis, most patients are already in the advanced stage when diagnosed, and the prognosis is poor. At present, there is no standard treatment strategy, and the existing treatment methods are not effective, the duration of remission is short, which cannot meet the clinical needs. Here we describe a patient with advanced MPeM, initially misdiagnosed as ovarian cancer, who responded to treatment with bevacizumab in combination with albumin-bound paclitaxel and cisplatin. In preparation for cytoreductive surgery (CRS), MPeM was confirmed by laparoscopic peritoneal nodule biopsy combined with histological and immunohistochemical results. Subsequently, due to intolerable neurotoxicity after chemotherapy, she received oral anlotinib therapy on April 25, 2022, and remained stable disease (SD) with the medication, having achieved more than 14 months of progression-free survival (PFS) as of the date of our manuscript submission. The patient's total treatment time was over 19 months. These treatments delayed tumor progression, reduced drug side effects, maintained a good quality of life, and further extended overall survival (OS). Our experience is that on the one hand, it is necessary to increase the clinician's understanding of the disease, and make full use of tissue samples and immunohistochemical staining to reduce the occurrence of misdiagnosis. On the other hand, based on preliminary evidence, we found that oral anlotinib offers a viable maintenance treatment strategy for patients with advanced mesothelioma, which needs to be further explored in future studies.
Collapse
Affiliation(s)
- Fan-Jie Qu
- Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian, Liaoning Province, 116033, People’s Republic of China
| | - Yi Zhou
- Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian, Liaoning Province, 116033, People’s Republic of China
| | - Hai Wang
- Department of Pathology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian, Liaonin Provinceg, 116033, People’s Republic of China
| |
Collapse
|
47
|
Adebayo AS, Agbaje K, Adesina SK, Olajubutu O. Colorectal Cancer: Disease Process, Current Treatment Options, and Future Perspectives. Pharmaceutics 2023; 15:2620. [PMID: 38004598 PMCID: PMC10674471 DOI: 10.3390/pharmaceutics15112620] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/28/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Colorectal cancer (CRC) is one of the deadliest malignancies in the US, ranking fourth after lung, prostate, and breast cancers, respectively, in general populations. It continues to be a menace, and the incidence has been projected to more than double by 2035, especially in underdeveloped countries. This review seeks to provide some insights into the disease progression, currently available treatment options and their challenges, and future perspectives. Searches were conducted in the PubMed search engine in the university's online library. The keywords were "Colorectal Cancer" AND "disease process" OR "disease mechanisms" OR "Current Treatment" OR "Prospects". Selection criteria were original articles published primarily during the period of 2013 through 2023. Abstracts, books and documents, and reviews/systematic reviews were filtered out. Of over 490 thousand articles returned, only about 800 met preliminary selection criteria, 200 were reviewed in detail, but 191 met final selection criteria. Fifty-one other articles were used due to cross-referencing. Although recently considered a disease of lifestyle, CRC incidence appears to be rising in countries with low, low-medium, and medium social demographic indices. CRC can affect all parts of the colon and rectum but is more fatal with poor disease outcomes when it is right-sided. The disease progression usually takes between 7-10 years and can be asymptomatic, making early detection and diagnosis difficult. The CRC tumor microenvironment is made up of different types of cells interacting with each other to promote the growth and proliferation of the tumor cells. Significant advancement has been made in the treatment of colorectal cancer. Notable approaches include surgery, chemotherapy, radiation therapy, and cryotherapy. Chemotherapy, including 5-fluorouracil, irinotecan, oxaliplatin, and leucovorin, plays a significant role in the management of CRC that has been diagnosed at advanced stages. Two classes of monoclonal antibody therapies have been approved by the FDA for the treatment of colorectal cancer: the vascular endothelial growth factor (VEGF) inhibitor, e.g., bevacizumab (Avastin®), and the epidermal growth factor receptor (EGFR) inhibitor, e.g., cetuximab (Erbitux®) and panitumumab (Verbitix®). However, many significant problems are still being experienced with these treatments, mainly off-target effects, toxic side effects, and the associated therapeutic failures of small molecular drugs and the rapid loss of efficacy of mAb therapies. Other novel delivery strategies continue to be investigated, including ligand-based targeting of CRC cells.
Collapse
Affiliation(s)
- Amusa S. Adebayo
- College of Pharmacy, Howard University, 2400 6th St NW, Washington, DC 20059, USA; (K.A.); (S.K.A.); (O.O.)
| | | | | | | |
Collapse
|
48
|
Choi Y, Jung K. Normalization of the tumor microenvironment by harnessing vascular and immune modulation to achieve enhanced cancer therapy. Exp Mol Med 2023; 55:2308-2319. [PMID: 37907742 PMCID: PMC10689787 DOI: 10.1038/s12276-023-01114-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/07/2023] [Accepted: 08/12/2023] [Indexed: 11/02/2023] Open
Abstract
Solid tumors are complex entities that actively shape their microenvironment to create a supportive environment for their own growth. Angiogenesis and immune suppression are two key characteristics of this tumor microenvironment. Despite attempts to deplete tumor blood vessels using antiangiogenic drugs, extensive vessel pruning has shown limited efficacy. Instead, a targeted approach involving the judicious use of drugs at specific time points can normalize the function and structure of tumor vessels, leading to improved outcomes when combined with other anticancer therapies. Additionally, normalizing the immune microenvironment by suppressing immunosuppressive cells and activating immunostimulatory cells has shown promise in suppressing tumor growth and improving overall survival. Based on these findings, many studies have been conducted to normalize each component of the tumor microenvironment, leading to the development of a variety of strategies. In this review, we provide an overview of the concepts of vascular and immune normalization and discuss some of the strategies employed to achieve these goals.
Collapse
Affiliation(s)
- Yechan Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Keehoon Jung
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, 03080, Republic of Korea.
| |
Collapse
|
49
|
Martinez-Espinosa I, Serrato JA, Ortiz-Quintero B. The Role of Exosome-Derived microRNA on Lung Cancer Metastasis Progression. Biomolecules 2023; 13:1574. [PMID: 38002256 PMCID: PMC10669807 DOI: 10.3390/biom13111574] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023] Open
Abstract
The high mortality from lung cancer is mainly attributed to the presence of metastases at the time of diagnosis. Despite being the leading cause of lung cancer death, the underlying molecular mechanisms driving metastasis progression are still not fully understood. Recent studies suggest that tumor cell exosomes play a significant role in tumor progression through intercellular communication between tumor cells, the microenvironment, and distant organs. Furthermore, evidence shows that exosomes release biologically active components to distant sites and organs, which direct metastasis by preparing metastatic pre-niche and stimulating tumorigenesis. As a result, identifying the active components of exosome cargo has become a critical area of research in recent years. Among these components are microRNAs, which are associated with tumor progression and metastasis in lung cancer. Although research into exosome-derived microRNA (exosomal miRNAs) is still in its early stages, it holds promise as a potential target for lung cancer therapy. Understanding how exosomal microRNAs promote metastasis will provide evidence for developing new targeted treatments. This review summarizes current research on exosomal miRNAs' role in metastasis progression mechanisms, focusing on lung cancer.
Collapse
Affiliation(s)
| | | | - Blanca Ortiz-Quintero
- Department of Molecular Biomedicine and Translational Research, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City CP 14080, Mexico; (I.M.-E.); (J.A.S.)
| |
Collapse
|
50
|
Li K, Zeng X, Liu P, Zeng X, Lv J, Qiu S, Zhang P. The Role of Inflammation-Associated Factors in Head and Neck Squamous Cell Carcinoma. J Inflamm Res 2023; 16:4301-4315. [PMID: 37791117 PMCID: PMC10544098 DOI: 10.2147/jir.s428358] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/16/2023] [Indexed: 10/05/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC), which originates in the head or neck tissues, is characterized by high rates of recurrence and metastasis. Inflammation is important in HNSCC prognosis. Inflammatory cells and their secreted factors contribute to the various stages of HNSCC development through multiple mechanisms. In this review, the mechanisms through which inflammatory factors, signaling pathways, and cells contribute to the initiation and progression of HNSCC have been discussed in detail. Furthermore, the diagnostic and therapeutic potential of targeting inflammation in HNSCC has been discussed to gain new insights into improving patient prognosis.
Collapse
Affiliation(s)
- Kang Li
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong, People’s Republic of China
- Department of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital & Shenzhen Key Laboratory of Otorhinolaryngology, Shenzhen Institute of Otorhinolaryngology, Shenzhen, Guangdong, People’s Republic of China
| | - Xianhai Zeng
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong, People’s Republic of China
- Department of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital & Shenzhen Key Laboratory of Otorhinolaryngology, Shenzhen Institute of Otorhinolaryngology, Shenzhen, Guangdong, People’s Republic of China
| | - Peng Liu
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong, People’s Republic of China
- Department of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital & Shenzhen Key Laboratory of Otorhinolaryngology, Shenzhen Institute of Otorhinolaryngology, Shenzhen, Guangdong, People’s Republic of China
| | - Xiaoxia Zeng
- Department of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital & Shenzhen Key Laboratory of Otorhinolaryngology, Shenzhen Institute of Otorhinolaryngology, Shenzhen, Guangdong, People’s Republic of China
| | - Jie Lv
- School of Computer Science and Engineering, Yulin Normal University, Yulin, Guangxi, People’s Republic of China
| | - Shuqi Qiu
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong, People’s Republic of China
- Department of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital & Shenzhen Key Laboratory of Otorhinolaryngology, Shenzhen Institute of Otorhinolaryngology, Shenzhen, Guangdong, People’s Republic of China
| | - Peng Zhang
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong, People’s Republic of China
- Department of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital & Shenzhen Key Laboratory of Otorhinolaryngology, Shenzhen Institute of Otorhinolaryngology, Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|