101
|
Amgalan B, Day CP, Przytycka TM. Exploring tumor-normal cross-talk with TranNet: Role of the environment in tumor progression. PLoS Comput Biol 2023; 19:e1011472. [PMID: 37721939 PMCID: PMC10538798 DOI: 10.1371/journal.pcbi.1011472] [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: 07/11/2023] [Revised: 09/28/2023] [Accepted: 08/23/2023] [Indexed: 09/20/2023] Open
Abstract
There is a growing awareness that tumor-adjacent normal tissues used as control samples in cancer studies do not represent fully healthy tissues. Instead, they are intermediates between healthy tissues and tumors. The factors that contribute to the deviation of such control samples from healthy state include exposure to the tumor-promoting factors, tumor-related immune response, and other aspects of tumor microenvironment. Characterizing the relation between gene expression of tumor-adjacent control samples and tumors is fundamental for understanding roles of microenvironment in tumor initiation and progression, as well as for identification of diagnostic and prognostic biomarkers for cancers. To address the demand, we developed and validated TranNet, a computational approach that utilizes gene expression in matched control and tumor samples to study the relation between their gene expression profiles. TranNet infers a sparse weighted bipartite graph from gene expression profiles of matched control samples to tumors. The results allow us to identify predictors (potential regulators) of this transition. To our knowledge, TranNet is the first computational method to infer such dependencies. We applied TranNet to the data of several cancer types and their matched control samples from The Cancer Genome Atlas (TCGA). Many predictors identified by TranNet are genes associated with regulation by the tumor microenvironment as they are enriched in G-protein coupled receptor signaling, cell-to-cell communication, immune processes, and cell adhesion. Correspondingly, targets of inferred predictors are enriched in pathways related to tissue remodelling (including the epithelial-mesenchymal Transition (EMT)), immune response, and cell proliferation. This implies that the predictors are markers and potential stromal facilitators of tumor progression. Our results provide new insights into the relationships between tumor adjacent control sample, tumor and the tumor environment. Moreover, the set of predictors identified by TranNet will provide a valuable resource for future investigations.
Collapse
Affiliation(s)
- Bayarbaatar Amgalan
- National Center for Biotechnology Information/National Library of Medicine, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Chi-Ping Day
- Laboratory of Cancer Biology and Genetics/Center for Cancer Research/National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Teresa M. Przytycka
- National Center for Biotechnology Information/National Library of Medicine, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
102
|
Hashemi M, Taheriazam A, Daneii P, Hassanpour A, Kakavand A, Rezaei S, Hejazi ES, Aboutalebi M, Gholamrezaie H, Saebfar H, Salimimoghadam S, Mirzaei S, Entezari M, Samarghandian S. Targeting PI3K/Akt signaling in prostate cancer therapy. J Cell Commun Signal 2023; 17:423-443. [PMID: 36367667 PMCID: PMC10409967 DOI: 10.1007/s12079-022-00702-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 05/26/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
Urological cancers have obtained much attention in recent years due to their mortality and morbidity. The most common and malignant tumor of urological cancers is prostate cancer that imposes high socioeconomic costs on public life and androgen-deprivation therapy, surgery, and combination of chemotherapy and radiotherapy are employed in its treatment. PI3K/Akt signaling is an oncogenic pathway responsible for migration, proliferation and drug resistance in various cancers. In the present review, the role of PI3K/Akt signaling in prostate cancer progression is highlighted. The activation of PI3K/Akt signaling occurs in prostate cancer, while PTEN as inhibitor of PI3K/Akt shows down-regulation. Stimulation of PI3K/Akt signaling promotes survival of prostate tumor cells and prevents apoptosis. The cell cycle progression and proliferation rate of prostate tumor cells increase by PI3K/Akt signaling induction. PI3K/Akt signaling stimulates EMT and enhances metastasis of prostate tumor cells. Silencing PI3K/Akt signaling impairs growth and metastasis of prostate tumor cells. Activation of PI3K/Akt signaling mediates drug resistance and reduces radio-sensitivity of prostate tumor cells. Anti-tumor compounds suppress PI3K/Akt signaling in impairing prostate tumor progression. Furthermore, upstream regulators such as miRNAs, lncRNAs and circRNAs regulate PI3K/Akt signaling and it has clinical implications for prostate cancer patients.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Pouria Daneii
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Aria Hassanpour
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amirabbas Kakavand
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Shamin Rezaei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elahe Sadat Hejazi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maryam Aboutalebi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hamidreza Gholamrezaie
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hamidreza Saebfar
- League of European Research Universities, European University Association, University of Milan, Milan, Italy
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
103
|
Chakraborty S, Mukherjee S, Basak U, Pati S, Dutta A, Dutta S, Dhar S, Sarkar T, Guin A, Sa G, Das T. Immune evasion by cancer stem cells ensures tumor initiation and failure of immunotherapy. EXPLORATION OF IMMUNOLOGY 2023:384-405. [DOI: 10.37349/ei.2023.00108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/12/2023] [Indexed: 01/04/2025]
Abstract
Cancer stem cells (CSCs) are a small subpopulation of cells that drive the formation and progression of tumors. However, during tumor initiation, how CSCs communicate with neighbouring immune cells to overcome the powerful immune surveillance barrier in order to form, spread, and maintain the tumor, remains poorly understood. It is, therefore, absolutely necessary to understand how a small number of tumor-initiating cells (TICs) survive immune attack during (a) the “elimination phase” of “tumor immune-editing”, (b) the establishment of regional or distant tumor after metastasis, and (c) recurrence after therapy. Mounting evidence suggests that CSCs suppress the immune system through a variety of distinct mechanisms that ensure the survival of not only CSCs but also non-stem cancer cells (NSCCs), which eventually form the tumor mass. In this review article, the mechanisms via which CSCs change the immune landscape of the tissue of origin, which contains macrophages, dendritic cells (DCs), myeloid-derived suppressor cells (MDSCs), natural killer (NK) cells, and tumor-infiltrating lymphocytes, in favour of tumorigenesis were discussed. The failure of cancer immunotherapy might also be explained by such interaction between CSCs and immune cells. This review will shed light on the critical role of CSCs in tumor immune evasion and emphasize the importance of CSC-targeted immunotherapy as a cutting-edge technique for battling cancer by restricting communication between immune cells and CSCs.
Collapse
Affiliation(s)
- Sourio Chakraborty
- Division of Molecular Medicine, Bose Institute, P-1/12, Calcutta Improvement Trust Scheme VII M, Kolkata 700054, India
| | - Sumon Mukherjee
- Division of Molecular Medicine, Bose Institute, P-1/12, Calcutta Improvement Trust Scheme VII M, Kolkata 700054, India
| | - Udit Basak
- Division of Molecular Medicine, Bose Institute, P-1/12, Calcutta Improvement Trust Scheme VII M, Kolkata 700054, India
| | - Subhadip Pati
- Division of Molecular Medicine, Bose Institute, P-1/12, Calcutta Improvement Trust Scheme VII M, Kolkata 700054, India
| | - Apratim Dutta
- Division of Molecular Medicine, Bose Institute, P-1/12, Calcutta Improvement Trust Scheme VII M, Kolkata 700054, India
| | - Saikat Dutta
- Division of Molecular Medicine, Bose Institute, P-1/12, Calcutta Improvement Trust Scheme VII M, Kolkata 700054, India
| | - Subhanki Dhar
- Division of Molecular Medicine, Bose Institute, P-1/12, Calcutta Improvement Trust Scheme VII M, Kolkata 700054, India
| | - Tania Sarkar
- Division of Molecular Medicine, Bose Institute, P-1/12, Calcutta Improvement Trust Scheme VII M, Kolkata 700054, India
| | - Aharna Guin
- Division of Molecular Medicine, Bose Institute, P-1/12, Calcutta Improvement Trust Scheme VII M, Kolkata 700054, India
| | - Gaurisankar Sa
- Division of Molecular Medicine, Bose Institute, P-1/12, Calcutta Improvement Trust Scheme VII M, Kolkata 700054, India
| | - Tanya Das
- Division of Molecular Medicine, Bose Institute, P-1/12, Calcutta Improvement Trust Scheme VII M, Kolkata 700054, India
| |
Collapse
|
104
|
Abraham A, Virdi S, Herrero N, Bryant I, Nwakama C, Jacob M, Khaparde G, Jordan D, McCuddin M, McKinley S, Taylor A, Peeples C, Ekpenyong A. Microfluidic Microcirculation Mimetic for Exploring Biophysical Mechanisms of Chemotherapy-Induced Metastasis. MICROMACHINES 2023; 14:1653. [PMID: 37763816 PMCID: PMC10536821 DOI: 10.3390/mi14091653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/18/2023] [Accepted: 08/19/2023] [Indexed: 09/29/2023]
Abstract
There is rapidly emerging evidence from pre-clinical studies, patient samples and patient subpopulations that certain chemotherapeutics inadvertently produce prometastatic effects. Prior to this, we showed that doxorubicin and daunorubicin stiffen cells before causing cell death, predisposing the cells to clogging and extravasation, the latter being a step in metastasis. Here, we investigate which other anti-cancer drugs might have similar prometastatic effects by altering the biophysical properties of cells. We treated myelogenous (K562) leukemic cancer cells with the drugs nocodazole and hydroxyurea and then measured their mechanical properties using a microfluidic microcirculation mimetic (MMM) device, which mimics aspects of blood circulation and enables the measurement of cell mechanical properties via transit times through the device. We also quantified the morphological properties of cells to explore biophysical mechanisms underlying the MMM results. Results from MMM measurements show that nocodazole- and hydroxyurea-treated K562 cells exhibit significantly altered transit times. Nocodazole caused a significant (p < 0.01) increase in transit times, implying a stiffening of cells. This work shows the feasibility of using an MMM to explore possible biophysical mechanisms that might contribute to chemotherapy-induced metastasis. Our work also suggests cell mechanics as a therapeutic target for much needed antimetastatic strategies in general.
Collapse
Affiliation(s)
- Ashley Abraham
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Sukhman Virdi
- Physics Department, Creighton University, Omaha, NE 68178, USA; (S.V.); (I.B.); (C.P.)
| | - Nick Herrero
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Israel Bryant
- Physics Department, Creighton University, Omaha, NE 68178, USA; (S.V.); (I.B.); (C.P.)
| | - Chisom Nwakama
- Chemistry Department, Creighton University, Omaha, NE 68178, USA;
| | - Megha Jacob
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Gargee Khaparde
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Destiny Jordan
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Mackenzie McCuddin
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Spencer McKinley
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Adam Taylor
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Conner Peeples
- Physics Department, Creighton University, Omaha, NE 68178, USA; (S.V.); (I.B.); (C.P.)
| | - Andrew Ekpenyong
- Physics Department, Creighton University, Omaha, NE 68178, USA; (S.V.); (I.B.); (C.P.)
| |
Collapse
|
105
|
Cao SJ, Zhang Y, Zhang YX, Zhao W, Pan LH, Sun XD, Jia Z, Ouyang W, Ye QS, Zhang FX, Guo YQ, Ai YQ, Zhao BJ, Yu JB, Liu ZH, Yin N, Li XY, Ma JH, Li HJ, Wang MR, Sessler DI, Ma D, Wang DX. Long-term survival in older patients given propofol or sevoflurane anaesthesia for major cancer surgery: follow-up of a multicentre randomised trial. Br J Anaesth 2023; 131:266-275. [PMID: 37474242 DOI: 10.1016/j.bja.2023.01.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/21/2022] [Accepted: 01/17/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND Experimental evidence indicates that i.v. anaesthesia might reduce cancer recurrence compared with volatile anaesthesia, but clinical information is observational only. We therefore tested the primary hypothesis that propofol-based anaesthesia improves survival over 3 or more years after potentially curative major cancer surgery. METHODS This was a long-term follow-up of a multicentre randomised trial in 14 tertiary hospitals in China. We enrolled 1228 patients aged 65-90 yr who were scheduled for major cancer surgery. They were randomised to either propofol-based i.v. anaesthesia or to sevoflurane-based inhalational anaesthesia. The primary endpoint was overall survival after surgery. Secondary endpoints included recurrence-free and event-free survival. RESULTS Amongst subjects randomised, 1195 (mean age 72 yr; 773 [65%] male) were included in the modified intention-to-treat analysis. At the end of follow-up (median 43 months), there were 188 deaths amongst 598 patients (31%) assigned to propofol-based anaesthesia compared with 175 deaths amongst 597 patients (29%) assigned to sevoflurane-based anaesthesia; adjusted hazard ratio 1.02; 95% confidence interval (CI): 0.83-1.26; P=0.834. Recurrence-free survival was 223/598 (37%) in patients given propofol anaesthesia vs 206/597 (35%) given sevoflurane anaesthesia; adjusted hazard ratio 1.07; 95% CI: 0.89-1.30; P=0.465. Event-free survival was 294/598 (49%) in patients given propofol anaesthesia vs 274/597 (46%) given sevoflurane anaesthesia; adjusted hazard ratio 1.09; 95% CI 0.93 to 1.29; P=0.298. CONCLUSIONS Long-term survival after major cancer surgery was similar with i.v. and volatile anaesthesia. Propofol-based iv. anaesthesia should not be used for cancer surgery with the expectation that it will improve overall or cancer-specific survival. CLINICAL TRIAL REGISTRATIONS ChiCTR-IPR-15006209; NCT02660411.
Collapse
Affiliation(s)
- Shuang-Jie Cao
- Department of Anesthesiology, Peking University First Hospital, Beijing, China
| | - Yue Zhang
- Department of Anesthesiology, Peking University First Hospital, Beijing, China; Clinical Research Institute, Shenzhen Peking University-The Hong Kong University of Science & Technology Medical Center, Shenzhen, China
| | - Yu-Xiu Zhang
- Department of Anesthesiology, Peking University First Hospital, Beijing, China
| | - Wei Zhao
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ling-Hui Pan
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Xu-De Sun
- Department of Anesthesiology, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi'an, Shaanxi, China
| | - Zhen Jia
- Department of Anesthesiology, Affiliated Hospital of Qinghai University, Xining, Qinghai, China
| | - Wen Ouyang
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qing-Shan Ye
- Department of Anesthesiology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Fang-Xiang Zhang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Yong-Qing Guo
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Yan-Qiu Ai
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bin-Jiang Zhao
- Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jian-Bo Yu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Zhi-Heng Liu
- Department of Anesthesiology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen, Guangdong, China
| | - Ning Yin
- Department of Anesthesiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu, China; Department of Anesthesiology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xue-Ying Li
- Department of Biostatistics, Peking University First Hospital, Beijing, China
| | - Jia-Hui Ma
- Department of Anesthesiology, Peking University First Hospital, Beijing, China
| | - Hui-Juan Li
- Peking University Clinical Research Institute, Peking University Health Science Center, Beijing, China
| | - Mei-Rong Wang
- Peking University Clinical Research Institute, Peking University Health Science Center, Beijing, China
| | - Daniel I Sessler
- Outcomes Research Consortium, Cleveland Clinic, Cleveland, OH, USA; Department of Outcomes Research, Anesthesiology Institute, OH, USA
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK; National Clinical Research Center for Child Health, Hangzhou, China
| | - Dong-Xin Wang
- Department of Anesthesiology, Peking University First Hospital, Beijing, China; Outcomes Research Consortium, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
106
|
Liu X, Zhang Y, Li X, Xu J, Zhao C, Yang J. Raman Spectroscopy Combined with Malaria Protein for Early Capture and Recognition of Broad-Spectrum Circulating Tumor Cells. Int J Mol Sci 2023; 24:12072. [PMID: 37569448 PMCID: PMC10419290 DOI: 10.3390/ijms241512072] [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: 06/26/2023] [Revised: 07/22/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Early identification of tumors can significantly reduce the mortality rate. Circulating tumor cells (CTCs) are a type of tumor cell that detaches from the primary tumor and circulates through the bloodstream. Monitoring CTCs may allow the early identification of tumor progression. However, due to their rarity and heterogeneity, the enrichment and identification of CTCs is still challenging. Studies have shown that Raman spectroscopy could distinguish CTCs from metastatic cancer patients. VAR2CSA, a class of malaria proteins, has a strong broad-spectrum binding effect on various tumor cells and is a promising candidate biomarker for cancer detection. Here, recombinant malaria VAR2CSA proteins were synthesized, expressed, and purified. After confirming that various types of tumor cells can be isolated from blood by recombinant malaria VAR2CSA proteins, we further proved that the VAR2CSA combined with Raman spectroscopy could be used efficiently for tumor capture and type recognition using A549 cell lines spiked into the blood. This would allow the early screening and detection of a broad spectrum of CTCs. Finally, we synthesized and purified the malaria protein fusion antibody and confirmed its in vitro tumor-killing activity. Herein, this paper exploits the theoretical basis of a novel strategy to capture, recognize, and kill broad-spectrum types of CTCs from the peripheral blood.
Collapse
Affiliation(s)
- Xinning Liu
- Key Laboratory of Marine Drugs, Ministry of Education, Qingdao Marine Biomedical Research Institute, Ocean University of China, Qingdao 266071, China; (X.L.)
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Marine Science and Technology Center, Qingdao 266100, China
| | - Yidan Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, Qingdao Marine Biomedical Research Institute, Ocean University of China, Qingdao 266071, China; (X.L.)
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Marine Science and Technology Center, Qingdao 266100, China
| | - Xunrong Li
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics and Shandong Energy Institute, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266071, China
- University of Chinese Academy of Sciences, Beijing 100000, China
| | - Jian Xu
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics and Shandong Energy Institute, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266071, China
- University of Chinese Academy of Sciences, Beijing 100000, China
| | - Chenyang Zhao
- Key Laboratory of Marine Drugs, Ministry of Education, Qingdao Marine Biomedical Research Institute, Ocean University of China, Qingdao 266071, China; (X.L.)
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Marine Science and Technology Center, Qingdao 266100, China
| | - Jinbo Yang
- Key Laboratory of Marine Drugs, Ministry of Education, Qingdao Marine Biomedical Research Institute, Ocean University of China, Qingdao 266071, China; (X.L.)
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Marine Science and Technology Center, Qingdao 266100, China
| |
Collapse
|
107
|
Sanchez-Sandoval AL, Hernández-Plata E, Gomora JC. Voltage-gated sodium channels: from roles and mechanisms in the metastatic cell behavior to clinical potential as therapeutic targets. Front Pharmacol 2023; 14:1206136. [PMID: 37456756 PMCID: PMC10348687 DOI: 10.3389/fphar.2023.1206136] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
During the second half of the last century, the prevalent knowledge recognized the voltage-gated sodium channels (VGSCs) as the proteins responsible for the generation and propagation of action potentials in excitable cells. However, over the last 25 years, new non-canonical roles of VGSCs in cancer hallmarks have been uncovered. Their dysregulated expression and activity have been associated with aggressive features and cancer progression towards metastatic stages, suggesting the potential use of VGSCs as cancer markers and prognostic factors. Recent work has elicited essential information about the signalling pathways modulated by these channels: coupling membrane activity to transcriptional regulation pathways, intracellular and extracellular pH regulation, invadopodia maturation, and proteolytic activity. In a promising scenario, the inhibition of VGSCs with FDA-approved drugs as well as with new synthetic compounds, reduces cancer cell invasion in vitro and cancer progression in vivo. The purpose of this review is to present an update regarding recent advances and ongoing efforts to have a better understanding of molecular and cellular mechanisms on the involvement of both pore-forming α and auxiliary β subunits of VGSCs in the metastatic processes, with the aim at proposing VGSCs as new oncological markers and targets for anticancer treatments.
Collapse
Affiliation(s)
- Ana Laura Sanchez-Sandoval
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Medicina Genómica, Hospital General de México “Dr Eduardo Liceaga”, Mexico City, Mexico
| | - Everardo Hernández-Plata
- Consejo Nacional de Humanidades, Ciencias y Tecnologías and Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Juan Carlos Gomora
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
108
|
Subramani A, Cui W, Zhang Y, Friman T, Zhao Z, Huang W, Fonseca P, Lui WO, Narayanan V, Bobrowska J, Lekka M, Yan J, Conway DE, Holmgren L. Modulation of E-Cadherin Function through the AmotL2 Isoforms Promotes Ameboid Cell Invasion. Cells 2023; 12:1682. [PMID: 37443716 PMCID: PMC10340588 DOI: 10.3390/cells12131682] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 07/15/2023] Open
Abstract
The spread of tumor cells and the formation of distant metastasis remain the main causes of mortality in cancer patients. However, the mechanisms governing the release of cells from micro-environmental constraints remain unclear. E-cadherin negatively controls the invasion of epithelial cells by maintaining cell-cell contacts. Furthermore, the inactivation of E-cadherin triggers invasion in vitro. However, the role of E-cadherin is complex, as metastasizing cells maintain E-cadherin expression, which appears to have a positive role in the survival of tumor cells. In this report, we present a novel mechanism delineating how E-cadherin function is modulated to promote invasion. We have previously shown that E-cadherin is associated with p100AmotL2, which is required for radial actin formation and the transmission of mechanical force. Here, we present evidence that p60AmotL2, which is expressed in invading tumor cells, binds to the p100AmotL2 isoform and uncouples the mechanical constraint of radial actin filaments. We show for the first time that the coupling of E-cadherin to the actin cytoskeleton via p100AmotL2 is directly connected to the nuclear membrane. The expression of p60AmotL2 inactivates this connection and alters the properties of the nuclear lamina, potentiating the invasion of cells into micropores of the extracellular matrix. In summary, we propose that the balance of the two AmotL2 isoforms is important in the modulation of E-cadherin function and that an imbalance of this axis promotes ameboid cell invasion.
Collapse
Affiliation(s)
- Aravindh Subramani
- Department of Oncology and Pathology, U2, Bioclinicum J6:20, Solnavägen 30 Karolinska Institutet, Solna, 171 64 Stockholm, Sweden; (A.S.); (W.C.); (Y.Z.); (T.F.); (P.F.); (W.-O.L.)
| | - Weiyingqi Cui
- Department of Oncology and Pathology, U2, Bioclinicum J6:20, Solnavägen 30 Karolinska Institutet, Solna, 171 64 Stockholm, Sweden; (A.S.); (W.C.); (Y.Z.); (T.F.); (P.F.); (W.-O.L.)
| | - Yuanyuan Zhang
- Department of Oncology and Pathology, U2, Bioclinicum J6:20, Solnavägen 30 Karolinska Institutet, Solna, 171 64 Stockholm, Sweden; (A.S.); (W.C.); (Y.Z.); (T.F.); (P.F.); (W.-O.L.)
| | - Tomas Friman
- Department of Oncology and Pathology, U2, Bioclinicum J6:20, Solnavägen 30 Karolinska Institutet, Solna, 171 64 Stockholm, Sweden; (A.S.); (W.C.); (Y.Z.); (T.F.); (P.F.); (W.-O.L.)
| | - Zhihai Zhao
- Department of Physics, Faculty of Science: 2 Science Drive 3, S7-01-10, Lower Kent Ridge Road, Singapore 117542, Singapore; (Z.Z.); (W.H.); (J.Y.)
- Mechanobiology Institute (MBI): T-Lab, #10-02, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
| | - Wenmao Huang
- Department of Physics, Faculty of Science: 2 Science Drive 3, S7-01-10, Lower Kent Ridge Road, Singapore 117542, Singapore; (Z.Z.); (W.H.); (J.Y.)
- Mechanobiology Institute (MBI): T-Lab, #10-02, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
| | - Pedro Fonseca
- Department of Oncology and Pathology, U2, Bioclinicum J6:20, Solnavägen 30 Karolinska Institutet, Solna, 171 64 Stockholm, Sweden; (A.S.); (W.C.); (Y.Z.); (T.F.); (P.F.); (W.-O.L.)
| | - Weng-Onn Lui
- Department of Oncology and Pathology, U2, Bioclinicum J6:20, Solnavägen 30 Karolinska Institutet, Solna, 171 64 Stockholm, Sweden; (A.S.); (W.C.); (Y.Z.); (T.F.); (P.F.); (W.-O.L.)
| | - Vani Narayanan
- Department of Biomedical Engineering, Virginia Commonwealth University, 401 West Main Street, Richmond, VA 23284, USA; (V.N.); (D.E.C.)
| | - Justyna Bobrowska
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342 Krakow, Poland; (J.B.); (M.L.)
| | - Małgorzata Lekka
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342 Krakow, Poland; (J.B.); (M.L.)
| | - Jie Yan
- Department of Physics, Faculty of Science: 2 Science Drive 3, S7-01-10, Lower Kent Ridge Road, Singapore 117542, Singapore; (Z.Z.); (W.H.); (J.Y.)
- Mechanobiology Institute (MBI): T-Lab, #10-02, 5A Engineering Drive 1, National University of Singapore, Singapore 117411, Singapore
| | - Daniel E. Conway
- Department of Biomedical Engineering, Virginia Commonwealth University, 401 West Main Street, Richmond, VA 23284, USA; (V.N.); (D.E.C.)
| | - Lars Holmgren
- Department of Oncology and Pathology, U2, Bioclinicum J6:20, Solnavägen 30 Karolinska Institutet, Solna, 171 64 Stockholm, Sweden; (A.S.); (W.C.); (Y.Z.); (T.F.); (P.F.); (W.-O.L.)
| |
Collapse
|
109
|
Mun SK, Sim HB, Han JY, Kim H, Park DH, Chang DJ, Yee ST, Chang YT, Kim JJ. Visualization of Metastatic Lung Cancer with TiNIR. Tomography 2023; 9:1187-1195. [PMID: 37489464 PMCID: PMC10366764 DOI: 10.3390/tomography9040096] [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: 05/03/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 07/26/2023] Open
Abstract
The development of efficient biomarkers and probes for monitoring and treating cancer, specifically metastatic cancer, is a critical research area that can have a significant impact on both patient outcomes and drug discovery. In this context, TiNIR has been developed to detect tumor-initiating cells (TICs), with heme oxygenase 2 (HO2) as a promising therapeutic biomarker for tumor-initiating cells. In this study, TiNIR has demonstrated its effectiveness as an in vivo metastatic lung cancer tracker, highlighting its potential as a valuable tool in cancer research and therapy. The development of innovative approaches that selectively target metastatic cancers represents a promising avenue for improving survival rates and enhancing the quality of life of cancer patients.
Collapse
Affiliation(s)
- Seul-Ki Mun
- Department of Biomedical Science, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Republic of Korea
- Department of Pharmacy, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Republic of Korea
| | - Hyun Bo Sim
- Department of Biomedical Science, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Republic of Korea
| | - Ji Yeon Han
- Department of Biomedical Science, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Republic of Korea
| | - Hyeongyeong Kim
- Department of Biomedical Science, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Republic of Korea
| | - Dae-Han Park
- Department of Biomedical Science, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Republic of Korea
| | - Dong-Jo Chang
- Department of Pharmacy, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Republic of Korea
| | - Sung-Tae Yee
- Department of Pharmacy, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Republic of Korea
| | - Young-Tae Chang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Jong-Jin Kim
- Department of Biomedical Science, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Republic of Korea
| |
Collapse
|
110
|
Groenewoud A, Yin J, Gelmi MC, Alsafadi S, Nemati F, Decaudin D, Roman-Roman S, Kalirai H, Coupland SE, Jochemsen AG, Jager MJ, Engel FB, Snaar-Jagalska BE. Patient-derived zebrafish xenografts of uveal melanoma reveal ferroptosis as a drug target. Cell Death Discov 2023; 9:183. [PMID: 37321991 DOI: 10.1038/s41420-023-01446-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 03/24/2023] [Accepted: 04/24/2023] [Indexed: 06/17/2023] Open
Abstract
Uveal melanoma (UM) has a high risk to progress to metastatic disease with a median survival of 3.9 months after metastases detection, as metastatic UM responds poorly to conventional and targeted chemotherapy and is largely refractory to immunotherapy. Here, we present a patient-derived zebrafish UM xenograft model mimicking metastatic UM. Cells isolated from Xmm66 spheroids derived from metastatic UM patient material were injected into 2 days-old zebrafish larvae resulting in micro-metastases in the liver and caudal hematopoietic tissue. Metastasis formation could be reduced by navitoclax and more efficiently by the combinations navitoclax/everolimus and flavopiridol/quisinostat. We obtained spheroid cultures from 14 metastatic and 10 primary UM tissues, which were used for xenografts with a success rate of 100%. Importantly, the ferroptosis-related genes GPX4 and SLC7A11 are negatively correlated with the survival of UM patients (TCGA: n = 80; Leiden University Medical Centre cohort: n = 64), ferroptosis susceptibility is correlated with loss of BAP1, one of the key prognosticators for metastatic UM, and ferroptosis induction greatly reduced metastasis formation in the UM xenograft model. Collectively, we have established a patient-derived animal model for metastatic UM and identified ferroptosis induction as a possible therapeutic strategy for the treatment of UM patients.
Collapse
Affiliation(s)
- Arwin Groenewoud
- Institute of Biology, Leiden University, Leiden, The Netherlands.
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
- Bavarian Cancer Research Center (BZKF), 91054, Erlangen, Germany.
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany.
| | - Jie Yin
- Institute of Biology, Leiden University, Leiden, The Netherlands
| | - Maria Chiara Gelmi
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Samar Alsafadi
- Uveal Melanoma Translational Group, Department of Translational Research, Institut Curie, PSL Research University, 75248 Paris, France
| | - Fariba Nemati
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL Research University, 75248 Paris, France
| | - Didier Decaudin
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL Research University, 75248 Paris, France
| | - Sergio Roman-Roman
- Uveal Melanoma Translational Group, Department of Translational Research, Institut Curie, PSL Research University, 75248 Paris, France
| | - Helen Kalirai
- Liverpool Ocular Oncology Research Centre, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Sarah E Coupland
- Liverpool Ocular Oncology Research Centre, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Aart G Jochemsen
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Martine J Jager
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | | |
Collapse
|
111
|
Qian Z, Tian X, Miao Y, Xu X, Cheng X, Wu M, Yu Y. Bufalin inhibits the proliferation of lung cancer cells by suppressing Hippo-YAP pathway. Cell Signal 2023:110746. [PMID: 37286119 DOI: 10.1016/j.cellsig.2023.110746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 04/20/2023] [Accepted: 05/31/2023] [Indexed: 06/09/2023]
Abstract
Lung cancer has high morbidity and mortality. This study demonstrated that Bufalin inhibits the proliferation of lung cancer cells in vivo / in vitro by suppressing Hippo-YAP pathway. Here, we found that Bufalin promoted the binding of LATS and YAP to elevate the level of YAP phosphorylation. Phosphorylated YAP could not successfully enter the nucleus to activate the expression of downstream proliferation-related target genes Cyr61 and CTGF, whereas the YAP retained in the cytoplasm further bound to β-TrCP and underwent ubiquitination and degradation. This study verified the key role of YAP in stimulating the proliferation of lung cancer and revealed the anticancer target of Bufalin. Therefore, this study provides a theoretical basis for the anticancer effect of Bufalin, and suggests that Bufalin can be a potential anticancer drug.
Collapse
Affiliation(s)
- Zijun Qian
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Xiaoting Tian
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Yayou Miao
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xin Xu
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xuehua Cheng
- Department of TCM Geriatrics, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Mengyi Wu
- Shanghai University of Traditional Chinese Medicine, 201203, China
| | - Yongchun Yu
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| |
Collapse
|
112
|
Xing MW, Li CJ, Guo C, Wang BJ, Mu DL, Wang DX. Effect of intraoperative dexmedetomidine on long-term survival in older patients after major noncardiac surgery: 3-year follow-up of a randomized trial. J Clin Anesth 2023; 86:111068. [PMID: 36736209 DOI: 10.1016/j.jclinane.2023.111068] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/14/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023]
Abstract
STUDY OBJECTIVE To assess the impact of intraoperative dexmedetomidine on long-term outcomes of older patients following major noncardiac surgery mainly for cancer. DESIGN A long-term follow-up of patients enrolled in a randomized trial. SETTING The initial trial was performed in a tertiary care hospital in Beijing, China. PARTICIPANTS Patients aged 60 years or older who were scheduled for major noncardiac surgery. INTERVENTION Participants were randomized to receive either dexmedetomidine (a loading dose of 0.6 μg/kg over 10 min, followed by a continuous infusion of 0.5 μg/kg/h until 1 h before end of surgery) or placebo during anesthesia. MEASUREMENTS The primary endpoint was overall survival. Secondary endpoints included recurrence-free survival and event-free survival. Cox proportional hazard models were used to adjust for predefined confounding factors. Propensity score matching was employed for sensitive analysis. RESULTS Among 620 patients who were randomized in the initial trial, 619 were included in the long-term analysis (mean age 69 years, 40% female, 77% oncological surgery). The median follow-up duration was 42 months (interquartile range 41 to 45). Overall survival did not differ between the two groups: there were 49/309 (15.9%) deaths with dexmedetomidine versus 63/310 (20.3%) with placebo (adjusted hazard ratio [HR] 0.78, 95% CI 0.53-1.13, P = 0.187). Recurrence-free survival was improved with dexmedetomidine (68/309 [22.0%] events with dexmedetomidine versus 98/310 [31.6%] with placebo; adjusted HR 0.67, 95% CI 0.49-0.92, P = 0.012). Event-free survival was also improved with dexmedetomidine (120/309 [38.8%] events with dexmedetomidine versus 145/310 [46.8%] with placebo; adjusted HR 0.78, 95% CI 0.61-1.00, P = 0.047). Results were similar after propensity-score matching and in the subgroup of cancer patients. CONCLUSIONS In older patients having major noncardiac surgery mainly for cancer, intraoperative dexmedetomidine did not improve overall survival but was associated with improved recurrence-free and event-free survivals.
Collapse
Affiliation(s)
- Mao-Wei Xing
- Department of Anesthesiology, Peking University First Hospital, Beijing, China.
| | - Chun-Jing Li
- Department of Anesthesiology, Peking University First Hospital, Beijing, China.
| | - Chao Guo
- Department of Anesthesiology, Peking University First Hospital, Beijing, China.
| | - Bo-Jie Wang
- Department of Anesthesiology, Peking University First Hospital, Beijing, China; Department of Anesthesiology, the University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China.
| | - Dong-Liang Mu
- Department of Anesthesiology, Peking University First Hospital, Beijing, China.
| | - Dong-Xin Wang
- Department of Anesthesiology, Peking University First Hospital, Beijing, China; Outcomes Research Consortium, Cleveland, OH, USA.
| |
Collapse
|
113
|
Carneiro CS, Hapeman JD, Nedelcu AM. Synergistic inter-clonal cooperation involving crosstalk, co-option and co-dependency can enhance the invasiveness of genetically distant cancer clones. BMC Ecol Evol 2023; 23:20. [PMID: 37226092 DOI: 10.1186/s12862-023-02129-7] [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: 12/19/2022] [Accepted: 05/12/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Despite intensive research, cancer remains a major health problem. The difficulties in treating cancer reflect the complex nature of this disease, including high levels of heterogeneity within tumours. Intra-tumour heterogeneity creates the conditions for inter-clonal competition and selection, which could result in selective sweeps and a reduction in levels of heterogeneity. However, in addition to competing, cancer clones can also cooperate with each other, and the positive effects of these interactions on the fitness of clones could actually contribute to maintaining the heterogeneity of tumours. Consequently, understanding the evolutionary mechanisms and pathways involved in such activities is of great significance for cancer treatment. This is particularly relevant for metastasis (i.e., tumor cell migration, invasion, dispersal and dissemination), which is the most lethal phase during cancer progression. To explore if and how genetically distant clones can cooperate during migration and invasion, this study used three distinct cancer cell lines with different metastatic potentials. RESULTS We found that (i) the conditioned media from two invasive lines (breast and lung) increased the migration and invasion potential of a poorly metastatic line (breast), and (ii) this inter-clonal cooperative interaction involved the TGF-β1 signalling pathway. Furthermore, when the less aggressive line was co-cultured with the highly metastatic breast line, the invasive potential of both lines was enhanced, and this outcome was dependent on the co-option (through TGF-β1 autocrine-paracrine signalling) of the weakly metastatic clone into expressing an enhanced malignant phenotype that benefited both clones (i.e., a "help me help you" strategy). CONCLUSIONS Based on our findings, we propose a model in which crosstalk, co-option, and co-dependency can facilitate the evolution of synergistic cooperative interactions between genetically distant clones. Specifically, we suggest that synergistic cooperative interactions can easily emerge, regardless of the degree of overall genetic/genealogical relatedness, via crosstalk involving metastatic clones able to constitutively secrete molecules that induce and maintain their own malignant state (producer-responder clones) and clones that have the ability to respond to those signals (responder clones) and express a synergistic metastatic behaviour. Taking into account the lack of therapies that directly affect the metastatic process, interfering with such cooperative interactions during the early steps in the metastatic cascade could provide additional strategies to increase patient survival.
Collapse
Affiliation(s)
- Caroline S Carneiro
- Department of Biology, University of New Brunswick, Fredericton, NB, E3B 5A3, Canada
| | - Jorian D Hapeman
- Department of Biology, University of New Brunswick, Fredericton, NB, E3B 5A3, Canada
| | - Aurora M Nedelcu
- Department of Biology, University of New Brunswick, Fredericton, NB, E3B 5A3, Canada.
| |
Collapse
|
114
|
Kim JH, Park S, Jung E, Shin J, Kim YJ, Kim JY, Sessler JL, Seo JH, Kim JS. A dual-action niclosamide-based prodrug that targets cancer stem cells and inhibits TNBC metastasis. Proc Natl Acad Sci U S A 2023; 120:e2304081120. [PMID: 37186828 PMCID: PMC10214212 DOI: 10.1073/pnas.2304081120] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Chemotherapy typically destroys the tumor mass but rarely eradicates the cancer stem cells (CSCs) that can drive metastatic recurrence. A key current challenge is finding ways to eradicate CSCs and suppress their characteristics. Here, we report a prodrug, Nic-A, created by combining a carbonic anhydrase IX (CAIX) inhibitor, acetazolamide, with a signal transducer and transcriptional activator 3 (STAT3) inhibitor, niclosamide. Nic-A was designed to target triple-negative breast cancer (TNBC) CSCs and was found to inhibit both proliferating TNBC cells and CSCs via STAT3 dysregulation and suppression of CSC-like properties. Its use leads to a decrease in aldehyde dehydrogenase 1 activity, CD44high/CD24low stem-like subpopulations, and tumor spheroid-forming ability. TNBC xenograft tumors treated with Nic-A exhibited decreased angiogenesis and tumor growth, as well as decreased Ki-67 expression and increased apoptosis. In addition, distant metastases were suppressed in TNBC allografts derived from a CSC-enriched population. This study thus highlights a potential strategy for addressing CSC-based cancer recurrence.
Collapse
Affiliation(s)
- Ji Hyeon Kim
- Department of Chemistry, Korea University, Seoul02841, Korea
| | - Soeun Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul08308, Korea
| | - Eunsun Jung
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul08308, Korea
| | - Jinwoo Shin
- Department of Chemistry, Korea University, Seoul02841, Korea
| | - Yoon-Jae Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul08308, Korea
| | - Ji Young Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul08308, Korea
| | - Jonathan L. Sessler
- Department of Chemistry, The University of Texas at Austin, Austin, TX78712-1224
| | - Jae Hong Seo
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul02841, Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, Seoul08308, Korea
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul02841, Korea
| |
Collapse
|
115
|
Chen M, Li H, Xu X, Bao X, Xue L, Ai X, Xu J, Xu M, Shi Y, Zhen T, Li J, Yang Y, Ji Y, Fu Z, Xing K, Qing T, Wang Q, Zhong P, Zhu S. Identification of RAC1 in promoting brain metastasis of lung adenocarcinoma using single-cell transcriptome sequencing. Cell Death Dis 2023; 14:330. [PMID: 37202394 DOI: 10.1038/s41419-023-05823-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/20/2023]
Abstract
This study aims to give a new perspective to the biomarkers in the lung adenocarcinoma (LUAD) brain metastasis, pathways involved and potential therapeutics. We performed a comprehensive single-cell level transcriptomic analysis on one LUAD patient with circulating tumor cells (CTCs), primary tumor tissue and metastatic tumor tissue using scRNA-seq approach to identify metastasis related biomarkers. Further scRNA-seq were performed on 7 patients to validate the cancer metastatic hallmark. with single cells collected from either metastatic or primary LUAD tissues. Pathological and functional studies were also performed to evidence the critical role of RAC1 in the LUAD metastasis. Hallmark gene was verified based on immunohistochemistry staining, cytological experiment, survival information from The Cancer Genome Atlas (TCGA), and staining results from Human Protein Atlas (HPA) databases. PCA analysis revealed that CTCs were in the intermediate place between the metastatic group and primary group. In the unsupervised clustering analysis CTCs were closer to one of the metastatic tumor cells, implying heterogeneity of the metastatic tumor and origin of the CTCs were from metastatic site. Transitional phase related gene analysis identified RAC1 was enriched in metastatic tumor tissue (MTT) preferred gene set functioning as regulated cell death and apoptosis as well as promoted macromolecule organization. Compared with normal tissue, expression levels of RAC1 increased significantly in LUAD tissue based on HPA database. High expression of RAC1 predicts worse prognosis and higher-risk. EMT analysis identified the propensity of mesenchymal state in primary cells while epithelial signals were higher in the metastatic site. Functional clustering and pathway analyses suggested genes in RAC1 highly expressed cells played critical roles in adhesion, ECM and VEGF signaling pathways. Inhibition of RAC1 attenuates the proliferation, invasiveness and migration ability of lung cancer cells. Besides, through MRI T2WI results, we proved that RAC1 can promote brain metastasis in the RAC1-overexpressed H1975 cell burden nude mouse model. RAC1 and its mechanisms might promote drug design against LUAD brain metastasis.
Collapse
Affiliation(s)
- Mingyu Chen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, 200040, Shanghai, China
- School of Life Sciences, Fudan University, 200438, Shanghai, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences, Beijing, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Hanyue Li
- Department of Lung Tumor Clinical Center, Shanghai Chest Hospital, Shanghai Jiaotong University, 200030, Shanghai, China
| | - Xiaolin Xu
- Department of Cardiothoracic Surgery, Third Affiliated Hospital of Naval Military Medical University, 200003, Shanghai, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, 507 Zhengmin Road, Shanghai, PR China
| | - Xunxia Bao
- School of Life Science, Anhui Medical University, 230032, Hefei, China
| | - Lei Xue
- Department of Thoracic Surgery, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Military Medical University, 200003, Shanghai, China
| | - Xinghao Ai
- Department of Lung Tumor Clinical Center, Shanghai Chest Hospital, Shanghai Jiaotong University, 200030, Shanghai, China
| | - Jian Xu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, 200040, Shanghai, China
- School of Life Sciences, Fudan University, 200438, Shanghai, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences, Beijing, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Ming Xu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, 200040, Shanghai, China
- School of Life Sciences, Fudan University, 200438, Shanghai, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences, Beijing, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China
| | - Yong Shi
- Cinoasia Institute, 200438, Shanghai, China
| | | | - Jie Li
- Cinoasia Institute, 200438, Shanghai, China
| | - Yi Yang
- Cinoasia Institute, 200438, Shanghai, China
| | - Yang Ji
- Cinoasia Institute, 200438, Shanghai, China
| | | | | | - Tao Qing
- Cinoasia Institute, 200438, Shanghai, China
| | - Qiubo Wang
- Department of Clinical Laboratory, Wuxi 9th People's Hospital Affiliated to Soochow University, 214000, Wuxi, Jiangsu, China.
| | - Ping Zhong
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, 200040, Shanghai, China.
- School of Life Sciences, Fudan University, 200438, Shanghai, China.
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences, Beijing, China.
- Neurosurgical Institute of Fudan University, Shanghai, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China.
| | - Sibo Zhu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, 200040, Shanghai, China.
- School of Life Sciences, Fudan University, 200438, Shanghai, China.
| |
Collapse
|
116
|
Nohara LL, Ellis SLS, Dreier C, Dada S, Saranchova I, Munro L, Pfeifer CG, Coyle KM, Morrice JR, Shim DJS, Ahn P, De Voogd N, Williams DE, Cheng P, Garrovillas E, Andersen RJ, Jefferies WA. A novel cell-based screen identifies chemical entities that reverse the immune-escape phenotype of metastatic tumours. Front Pharmacol 2023; 14:1119607. [PMID: 37256225 PMCID: PMC10225555 DOI: 10.3389/fphar.2023.1119607] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/21/2023] [Indexed: 06/01/2023] Open
Abstract
Genetic and epigenetic events have been implicated in the downregulation of the cellular antigen processing and presentation machinery (APM), which in turn, has been associated with cancer evasion of the immune system. When these essential components are lacking, cancers develop the ability to subvert host immune surveillance allowing cancer cells to become invisible to the immune system and, in turn, promote cancer metastasis. Here we describe and validate the first high-throughput cell-based screening assay to identify chemical extracts and unique chemical entities that reverse the downregulation of APM components in cell lines derived from metastatic tumours. Through the screening of a library of 480 marine invertebrate extracts followed by bioassay-guided fractionation, curcuphenol, a common sesquiterpene phenol derived from turmeric, was identified as the active compound of one of the extracts. We demonstrate that curcuphenol induces the expression of the APM components, TAP-1 and MHC-I molecules, in cell lines derived from both metastatic prostate and lung carcinomas. Turmeric and curcumins that contain curcuphenol have long been utilized not only as a spice in the preparation of food, but also in traditional medicines for treating cancers. The remarkable discovery that a common component of spices can increase the expression of APM components in metastatic tumour cells and, therefore reverse immune-escape mechanisms, provides a rationale for the development of foods and advanced nutraceuticals as therapeutic candidates for harnessing the power of the immune system to recognize and destroy metastatic cancers.
Collapse
Affiliation(s)
- Lilian L. Nohara
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Samantha L. S. Ellis
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Carola Dreier
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Sarah Dada
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
- Departments of Medical Genetics, Zoology, and Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Iryna Saranchova
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
- Departments of Medical Genetics, Zoology, and Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Lonna Munro
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Cheryl G. Pfeifer
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Krysta M. Coyle
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Jessica R. Morrice
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Daniel Joo Sung Shim
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Paul Ahn
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Nicole De Voogd
- Netherlands Centre for Biodiversity Naturalis, Leiden, Netherlands
| | - David E. Williams
- Departments of Chemistry and Earth Ocean and Atmospheric Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ping Cheng
- Departments of Chemistry and Earth Ocean and Atmospheric Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Emmanuel Garrovillas
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
| | - Raymond J. Andersen
- Departments of Chemistry and Earth Ocean and Atmospheric Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Wilfred A. Jefferies
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
- The Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
- Departments of Medical Genetics, Zoology, and Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
117
|
Wang W, Liang X, Kong H, Yang Y, Xia Y, Wang Q, Xia A, Geng J. Correlation analysis of lung mucosa-colonizing bacteria with clinical features reveals metastasis-associated bacterial community structure in non-small cell lung cancer patients. Respir Res 2023; 24:129. [PMID: 37170267 PMCID: PMC10176848 DOI: 10.1186/s12931-023-02420-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 04/12/2023] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND Microbes colonizing lower airways can regulate the host immune profile and consequently participate in lung disease. Increasing evidence indicate that individual microbes promote lung cancer progression and are involved in metastasis incidence. To date, however, no study has revealed the community structure of lung bacteria in metastatic non-small cell lung cancer (NSCLC) patients. METHODS We prospectively enrolled 50 healthy subjects and 57 NSCLC patients. All healthy subjects and NSCLC patients underwent bronchoscope procedures for brush specimen collection. The 16 S ribosomal RNA gene was sequenced to characterize the community structure of lung mucosa-colonizing bacteria. The peripheral blood of NSCLC patients was also measured for leukocytes and cancer markers. RESULTS The lung bacteria of healthy subjects and NSCLC patients were divided into four communities. All community 2 members showed increased abundance in NSCLC patients compared with healthy subjects, and most community 2 members showed increased abundance in the metastatic NSCLC patients compared with the non-metastatic group. These bacteria were significantly and positively correlated with eosinophils, neutrophils and monocytes in the metastatic NSCLC group. In addition, the correlation between lung bacteria and cancer markers differed between the metastatic and non-metastatic NSCLC patients. Furthermore, bronchoalveolar lavage fluid from lung adenocarcinoma patients directly promoted NSCLC cell migration. CONCLUSIONS The community structure of lung mucosa-colonizing bacteria was relatively stable, but changed from the healthy population to NSCLC patients, especially the metastatic group. This distinct community structure and specific correlation with immune cells and cancer markers could help to distinguish NSCLC patients with or without metastasis.
Collapse
Affiliation(s)
- Wenxue Wang
- Department of Infectious Disease and Hepatic Disease, First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Jinbi Road #157, Kunming, Yunnan, 650032, China.
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China.
| | - Xiao Liang
- Department of Infectious Disease and Hepatic Disease, First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Jinbi Road #157, Kunming, Yunnan, 650032, China
| | - Hui Kong
- Department of Infectious Disease and Hepatic Disease, First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Jinbi Road #157, Kunming, Yunnan, 650032, China
| | - Yun Yang
- Department of Infectious Disease and Hepatic Disease, First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Jinbi Road #157, Kunming, Yunnan, 650032, China
| | - Yilan Xia
- Department of Infectious Disease and Hepatic Disease, First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Jinbi Road #157, Kunming, Yunnan, 650032, China
| | - Qiongjiao Wang
- Department of Infectious Disease and Hepatic Disease, First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Jinbi Road #157, Kunming, Yunnan, 650032, China
| | - Andong Xia
- Department of Infectious Disease and Hepatic Disease, First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Jinbi Road #157, Kunming, Yunnan, 650032, China
| | - Jiawei Geng
- Department of Infectious Disease and Hepatic Disease, First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Jinbi Road #157, Kunming, Yunnan, 650032, China.
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China.
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China.
| |
Collapse
|
118
|
Battistoni A, Lantier L, di Tommaso A, Ducournau C, Lajoie L, Samimi M, Coënon L, Rivière C, Epardaud M, Hertereau L, Poupée-Beaugé A, Rieu J, Mévélec MN, Lee GS, Moiré N, Germon S, Dimier-Poisson I. Nasal administration of recombinant Neospora caninum secreting IL-15/IL-15Rα inhibits metastatic melanoma development in lung. J Immunother Cancer 2023; 11:jitc-2023-006683. [PMID: 37192784 DOI: 10.1136/jitc-2023-006683] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND Metastases are the leading cause of mortality in many cancer types and lungs are one of the most common sites of metastasis alongside the liver, brain, and bones. In melanoma, 85% of late-stage patients harbor lung metastases. A local administration could enhance the targeting of metastases while limiting the systemic cytotoxicity. Therefore, intranasal administration of immunotherapeutic agents seems to be a promising approach to preferentially target lung metastases and decrease their burden on cancer mortality. From observations that certain microorganisms induce an acute infection of the tumor microenvironment leading to a local reactivating immune response, microbial-mediated immunotherapy is a next-generation field of investigation in which immunotherapies are engineered to overcome immune surveillance and escape from microenvironmental cancer defenses. METHODS The goal of our study is to evaluate the potential of the intranasal administration of Neospora caninum in a syngeneic C57BL6 mouse model of B16F10 melanoma lung metastases. It also compares the antitumoral properties of a wild-type N. caninum versus N. caninum secreting human interleukin (IL)-15 fused to the sushi domain of the IL-15 receptor α chain, a potent activator of cellular immune responses. RESULTS The treatment of murine lung metastases by intranasal administration of an N. caninum engineered to secrete human IL-15 impairs lung metastases from further progression with only 0,08% of lung surface harboring metastases versus 4,4% in wild-type N. caninum treated mice and 36% in untreated mice. The control of tumor development is associated with a strong increase in numbers, within the lung, of natural killer cells, CD8+ T cells and macrophages, up to twofold, fivefold and sixfold, respectively. Analysis of expression levels of CD86 and CD206 on macrophages surface revealed a polarization of these macrophages towards an antitumoral M1 phenotype. CONCLUSION Administration of IL-15/IL-15Rα-secreting N. caninum through intranasal administration, a non-invasive route, lend further support to N. caninum-demonstrated clear potential as an effective and safe immunotherapeutic approach for the treatment of metastatic solid cancers, whose existing therapeutic options are scarce. Combination of this armed protozoa with an intranasal route could reinforce the existing therapeutic arsenal against cancer and narrow the spectrum of incurable cancers.
Collapse
Affiliation(s)
- Arthur Battistoni
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
| | - Louis Lantier
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
- Kymeris Santé SA, Tours, France
| | - Anne di Tommaso
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
| | - Céline Ducournau
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
| | - Laurie Lajoie
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
| | - Mahtab Samimi
- Department de Dermatologie, CHRU de Tours, Tours, France
| | - Loïs Coënon
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
| | - Clément Rivière
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
| | | | - Leslie Hertereau
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
| | | | - Juliette Rieu
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
| | | | | | - Nathalie Moiré
- INRAE, Université de Tours, ISP, F-37380, Nouzilly, France
| | - Stephanie Germon
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
| | | |
Collapse
|
119
|
Yu H, Liang X, Zhang M, Fan Y, Wang G, Wang S, Sun J, Zhang J. LN-Net: Perfusion Pattern-Guided Deep Learning for Lymph Node Metastasis Diagnosis Based on Contrast-Enhanced Ultrasound Videos. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1248-1258. [PMID: 36803610 DOI: 10.1016/j.ultrasmedbio.2023.01.010] [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: 09/17/2022] [Revised: 01/10/2023] [Accepted: 01/14/2023] [Indexed: 05/11/2023]
Abstract
OBJECTIVE The blood flow in lymph nodes reflects important pathological features. However, most intelligent diagnosis based on contrast-enhanced ultrasound (CEUS) video focuses only on CEUS images, ignoring the process of extracting blood flow information. In the work described here, a parametric imaging method for describing blood perfusion pattern was proposed and a multimodal network (LN-Net) to predict lymph node metastasis was designed. METHODS First, the commercially available artificial intelligence object detection model YOLOv5 was improved to detect the lymph node region. Then the correlation and inflection point matching algorithms were combined to calculate the parameters of the perfusion pattern. Finally, the Inception-V3 architecture was used to extract the image features of each modality, with the blood perfusion pattern taken as the guiding factor in fusing the features with CEUS by sub-network weighting. DISCUSSION The average precision of the improved YOLOv5s algorithm compared with baseline was improved by 5.8%. LN-Net predicted lymph node metastasis with 84.9% accuracy, 83.7% precision and 80.3% recall. Compared with the model without blood flow feature guidance, accuracy was improved by 2.6%. The intelligent diagnosis method has good clinical interpretability. CONCLUSION A static parametric imaging map could describe a dynamic blood flow perfusion pattern, and as a guiding factor, it could improve the classification ability of the model with respect to lymph node metastasis.
Collapse
Affiliation(s)
- Hui Yu
- Department of Biomedical Engineering, College of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin, China; Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Xiaoyun Liang
- Department of Biomedical Engineering, College of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin, China
| | - Mengrui Zhang
- Department of General Surgery, General Hospital of Tianjin Medical University, Tianjin, China
| | - Yinuo Fan
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Guangpu Wang
- Department of Biomedical Engineering, College of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin, China
| | - Shuo Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Jinglai Sun
- Department of Biomedical Engineering, College of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin, China
| | - Jie Zhang
- Department of General Surgery, General Hospital of Tianjin Medical University, Tianjin, China.
| |
Collapse
|
120
|
Yang MH, Ha IJ, Ahn J, Kim CK, Lee M, Ahn KS. Potential function of loliolide as a novel blocker of epithelial-mesenchymal transition in colorectal and breast cancer cells. Cell Signal 2023; 105:110610. [PMID: 36707041 DOI: 10.1016/j.cellsig.2023.110610] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023]
Abstract
Loliolide (LL), a naturally occurring monoterpenoid lactone isolated from Vicia tenuifolia Roth, can exhibit numerous pharmacological effects such as those related to anti-Parkinson, anti-oxidant, anti-cholinesterase, and anti-depressant. Epithelial-mesenchymal transition (EMT) plays a pivotal role in regulating tumor metastasis. CXCR4 and CXCR7 are G-protein-coupled receptors (GPRs), which can be stimulated by CXCL12. CXCL12/CXCR4/CXCXR7 axis can cause activation of multiple pathways including MAPKs, JAK/STAT pathway, and manganese superoxide dismutase (MnSOD) signaling. These events can initiate EMT process and induce cell invasion and migration. Here, we investigated whether LL can modulate the CXCR4 and CXCR7 and EMT process in colon cancer and breast cancer cells. We found that LL suppressed levels of CXCR4 and CXCR7, and exerted an inhibitory effect on these chemokines even after stimulation by CXCL12. LL suppressed expression of MnSOD and mesenchymal markers, whereas induced epithelial markers. In addition, LL significantly attenuated cellular invasion, migration, and metastasis. We noted that LL inhibited CXCR4/7 and EMT process even after stimulation of CXCL12 and MnSOD overexpression. Therefore, in this study, we provide evidences that targeting CXCR4/7 and MnSOD could inhibit the invasion, migration, and metastasis of cancer cells as well as negatively regulate the EMT process. Overall, our study suggested that LL might act as a potent suppressor of EMT process against colon and breast cancer cells.
Collapse
Affiliation(s)
- Min Hee Yang
- KHU-KIST Department of Converging Science and Technology and Department of Science in Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - In Jin Ha
- Korean Medicine Clinical Trial Center (K-CTC), Korean Medicine Hospital, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jeongjun Ahn
- College of Pharmacy, Sunchon National University, 255 Jungangno, Suncheon-si, Jeonnam 57922, Republic of Korea.
| | - Chang-Kwon Kim
- College of Pharmacy, Sunchon National University, 255 Jungangno, Suncheon-si, Jeonnam 57922, Republic of Korea.
| | - Mina Lee
- College of Pharmacy, Sunchon National University, 255 Jungangno, Suncheon-si, Jeonnam 57922, Republic of Korea.
| | - Kwang Seok Ahn
- KHU-KIST Department of Converging Science and Technology and Department of Science in Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
121
|
Li T, Hu Z, Song F, Wu C, Miao Q, Wang Z, Feng W, Guo J, Chen Y. Photonic Hyperthermia Synergizes with Immune-Activators to Augment Tumor-Localized Immunotherapy. SMALL METHODS 2023; 7:e2300116. [PMID: 37075769 DOI: 10.1002/smtd.202300116] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/13/2023] [Indexed: 05/03/2023]
Abstract
Photothermal immunotherapy, the combination of photothermal hyperthermia and immunotherapy, is a noninvasive and desirable therapeutic strategy to address the deficiency of traditional photothermal ablation for tumor treatment. However, insufficient T-cell activation following photothermal treatment is a bottleneck to achieve satisfactory therapeutic effectiveness. In this work, a multifunctional nanoplatform is rationally designed and engineered on the basis of polypyrrole-based magnetic nanomedicine modified by T-cell activators of anti-CD3 and anti-CD28 monoclonal antibodies, which have achieved robust near infrared laser-triggered photothermal ablation and long-lasting T-cell activation, realizing diagnostic imaging-guided immunosuppressive tumor microenvironment regulation following photothermal hyperthermia by reinvigorating tumor-infiltrating lymphocytes. By virtue of high-efficient immunogenic cell death and dendritic cell maturation combined with T-cell activation, this nanosystem markedly restrains primary and abscopal tumors as well as metastatic tumors with negligible side effects in vivo, exerting the specific function for suppressing tumor recurrence and metastasis by establishing a long-term memory immune response.
Collapse
Affiliation(s)
- Tiankuan Li
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology and Ultrasound, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, P. R. China
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Zhongqian Hu
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology and Ultrasound, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, P. R. China
| | - Feifei Song
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, 200072, P. R. China
| | - Chenyao Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Qizeng Miao
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, 200072, P. R. China
| | - Zhongmin Wang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Jinhe Guo
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology and Ultrasound, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
122
|
Lima LTFD, Ganzella FADO, Cardoso GC, Pires VDS, Chequin A, Santos GL, Braun-Prado K, Galindo CM, Braz Junior O, Molento MB, Acco A, Adami ER, Costa ET, Cavichiolo Franco CR, Klassen G, Ramos EADS. l-carvone decreases breast cancer cells adhesion, migration, and invasion by suppressing FAK activation. Chem Biol Interact 2023; 378:110480. [PMID: 37059214 DOI: 10.1016/j.cbi.2023.110480] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/24/2023] [Accepted: 04/06/2023] [Indexed: 04/16/2023]
Abstract
Breast cancer is one of the most common types of cancer in the world and current therapeutic strategies present severe drawbacks. l-carvone (CRV), a monoterpene found in Mentha spicata (spearmint), has been reported to have potent anti-inflammatory activity. Here, we examined the role of CRV in breast cancer cell adhesion, migration and invasion in vitro and how this component could suppress the growth of Ehrlich carcinoma-bearing mice. In vivo, treatment with CRV significantly decreased tumor growth, increased tumor necrosis area, and reduced the expression of VEGF and HIF-1α in Ehrlich carcinoma-bearing mice. Furthermore, the anticancer efficacy of CRV was similar to currently used chemotherapy (Methotrexate), and the combination of CRV with MTX potentiated the chemotherapy effects. Further mechanistic investigation in vitro revealed that CRV modulates the interaction of breast cancer cells with the extracellular matrix (ECM) by disrupting focal adhesion, which was shown by scanning electron microscopy (SEM) and immunofluorescence. Moreover, CRV caused a decrease in β1-integrin expression and inhibited focal adhesion kinase (FAK) activation. FAK is one of the most important downstream activators of several metastatic processes, including MMP-2 mediated invasion and HIF-1α/VEGF angiogenesis stimulus, both of which were found to be reduced in MDA-MB-231 cells exposed to CRV. Our results provide new insight about targeting β1-integrin/FAK signaling pathway with CRV, which could be a new potential agent in the treatment of breast cancer.
Collapse
Affiliation(s)
- Lucas Trevisan França de Lima
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | | | - Gabriela Casani Cardoso
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | - Verônica Dos Santos Pires
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | - Andressa Chequin
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | - Giulia Luiza Santos
- Molecular Oncology Center, Research and Education Institute, Hospital Sirio-Libanes, São Paulo, SP, Brazil
| | - Karin Braun-Prado
- Department of Basic Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | | | - Odair Braz Junior
- Pos-graduate Program of Cellular and Molecular Biology, Federal University of Parana, Curitiba, PR, Brazil
| | - Marcelo Beltrão Molento
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil; Department of Veterinary Medicine, Federal University of Parana, Curitiba, PR, Brazil
| | - Alexandra Acco
- Pos-graduate Program of Pharmacology, Federal University of Parana, Curitiba, PR, Brazil
| | - Eliana Rezende Adami
- Pos-graduate Program of Pharmacology, Federal University of Parana, Curitiba, PR, Brazil
| | - Erico Tosoni Costa
- Molecular Oncology Center, Research and Education Institute, Hospital Sirio-Libanes, São Paulo, SP, Brazil
| | | | - Giseli Klassen
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil; Department of Basic Pathology, Federal University of Parana, Curitiba, PR, Brazil
| | - Edneia Amancio de Souza Ramos
- Pos-graduate Program of Microbiology, Parasitology and Pathology, Federal University of Parana, Curitiba, PR, Brazil; Department of Basic Pathology, Federal University of Parana, Curitiba, PR, Brazil.
| |
Collapse
|
123
|
Suvilesh KN, Manjunath Y, Pantel K, Kaifi JT. Preclinical models to study patient-derived circulating tumor cells and metastasis. Trends Cancer 2023; 9:355-371. [PMID: 36759267 DOI: 10.1016/j.trecan.2023.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 02/10/2023]
Abstract
Circulating tumor cells (CTCs) that are detached from the tumor can be precursors of metastasis. The majority of studies focus on enumeration of CTCs from patient blood to predict recurrence and therapy outcomes. Very few studies have managed to expand CTCs to investigate their functional dynamics with respect to genetic changes, tumorigenic potential, and response to drug treatment. A growing amount of evidence based on successful CTC expansion has revealed novel therapeutic targets that are associated with the process of metastasis. In this review, we summarize the successes, challenges, and limitations that collectively contribute to the better understanding of metastasis using patient-derived CTCs as blood-borne seeds of metastasis. The roadblocks and future avenues to move CTC-based scientific discoveries forward are also discussed.
Collapse
Affiliation(s)
- Kanve N Suvilesh
- Hugh E. Stephenson Jr., MD, Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO, USA.
| | - Yariswamy Manjunath
- Hugh E. Stephenson Jr., MD, Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Klaus Pantel
- Institute for Tumor Biology, University of Hamburg, Hamburg, Germany
| | - Jussuf T Kaifi
- Hugh E. Stephenson Jr., MD, Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA; Siteman Cancer Center, St. Louis, MO, USA.
| |
Collapse
|
124
|
Zhang W, Hu J, Liu R, Dai J, Yuan L, Liu Y, Chen B, Gong M, Xia F, Lou X. A Peptide-Conjugated Probe with Cleavage-Induced Morphological Change for Treatment on Tumor Cell Membrane. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207228. [PMID: 36793151 PMCID: PMC10104630 DOI: 10.1002/advs.202207228] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/01/2023] [Indexed: 06/18/2023]
Abstract
Despite the promising advancements of in situ forming nanoassembly for the inhibition of tumor growth and metastasis, the lack of sufficient triggering sites and hardly controlling the forming position restrict their further developments. Herein, a smart transformable peptide-conjugated probe (DMFA) with enzyme cleavage-induced morphological change is designed for treatment on the tumor cell membrane. Specifically, after self-assembling into nanoparticles and anchoring on the cell membrane with sufficient interaction sites rapidly and stably, DMFA will be efficiently cleaved into α-helix forming part (DP) and β-sheet forming part (LFA) by overexpressed matrix metalloproteinase-2. Thus, the promoted Ca2+ influx by DP-induced cell membrane breakage and decreased Na+ /K+ -ATPase activity by LFA-assembled nanofibers wrapping the cells can inhibit PI3K-Akt signaling pathway, leading to the inhibition of tumor cell growth and metastasis. This peptide-conjugated probe undergoes in situ morphological transformation on the cell membrane, exhibiting great potential in tumor therapy.
Collapse
Affiliation(s)
- Wei Zhang
- State Key Laboratory of Biogeology and Environmental GeologyEngineering Research Center of Nano‐Geomaterials of Ministry of EducationFaculty of Materials Science and ChemistryChina University of GeosciencesWuhan430074China
| | - Jing‐Jing Hu
- State Key Laboratory of Biogeology and Environmental GeologyEngineering Research Center of Nano‐Geomaterials of Ministry of EducationFaculty of Materials Science and ChemistryChina University of GeosciencesWuhan430074China
| | - Rui Liu
- State Key Laboratory of Biogeology and Environmental GeologyEngineering Research Center of Nano‐Geomaterials of Ministry of EducationFaculty of Materials Science and ChemistryChina University of GeosciencesWuhan430074China
| | - Jun Dai
- Department of Obstetrics and GynecologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Lizhen Yuan
- State Key Laboratory of Biogeology and Environmental GeologyEngineering Research Center of Nano‐Geomaterials of Ministry of EducationFaculty of Materials Science and ChemistryChina University of GeosciencesWuhan430074China
| | - Yiheng Liu
- State Key Laboratory of Biogeology and Environmental GeologyEngineering Research Center of Nano‐Geomaterials of Ministry of EducationFaculty of Materials Science and ChemistryChina University of GeosciencesWuhan430074China
| | - Bochao Chen
- State Key Laboratory of Biogeology and Environmental GeologyEngineering Research Center of Nano‐Geomaterials of Ministry of EducationFaculty of Materials Science and ChemistryChina University of GeosciencesWuhan430074China
| | - Mingxing Gong
- State Key Laboratory of Biogeology and Environmental GeologyEngineering Research Center of Nano‐Geomaterials of Ministry of EducationFaculty of Materials Science and ChemistryChina University of GeosciencesWuhan430074China
| | - Fan Xia
- State Key Laboratory of Biogeology and Environmental GeologyEngineering Research Center of Nano‐Geomaterials of Ministry of EducationFaculty of Materials Science and ChemistryChina University of GeosciencesWuhan430074China
| | - Xiaoding Lou
- State Key Laboratory of Biogeology and Environmental GeologyEngineering Research Center of Nano‐Geomaterials of Ministry of EducationFaculty of Materials Science and ChemistryChina University of GeosciencesWuhan430074China
| |
Collapse
|
125
|
Quah HS, Cao EY, Suteja L, Li CH, Leong HS, Chong FT, Gupta S, Arcinas C, Ouyang JF, Ang V, Celhar T, Zhao Y, Tay HC, Chan J, Takahashi T, Tan DSW, Biswas SK, Rackham OJL, Iyer NG. Single cell analysis in head and neck cancer reveals potential immune evasion mechanisms during early metastasis. Nat Commun 2023; 14:1680. [PMID: 36973261 PMCID: PMC10042873 DOI: 10.1038/s41467-023-37379-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
Profiling tumors at single-cell resolution provides an opportunity to understand complexities underpinning lymph-node metastases in head and neck squamous-cell carcinoma. Single-cell RNAseq (scRNAseq) analysis of cancer-cell trajectories identifies a subpopulation of pre-metastatic cells, driven by actionable pathways including AXL and AURK. Blocking these two proteins blunts tumor invasion in patient-derived cultures. Furthermore, scRNAseq analyses of tumor-infiltrating CD8 + T-lymphocytes show two distinct trajectories to T-cell dysfunction, corroborated by their clonal architecture based on single-cell T-cell receptor sequencing. By determining key modulators of these trajectories, followed by validation using external datasets and functional experiments, we uncover a role for SOX4 in mediating T-cell exhaustion. Finally, interactome analyses between pre-metastatic tumor cells and CD8 + T-lymphocytes uncover a putative role for the Midkine pathway in immune-modulation and this is confirmed by scRNAseq of tumors from humanized mice. Aside from specific findings, this study demonstrates the importance of tumor heterogeneity analyses in identifying key vulnerabilities during early metastasis.
Collapse
Affiliation(s)
- Hong Sheng Quah
- Cancer Therapeutics Research Laboratory, National Cancer Centre, Singapore, Singapore
- Academic Clinical Program in Oncology, Duke-NUS Medical School, Singapore, Singapore
| | - Elaine Yiqun Cao
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Lisda Suteja
- Cancer Therapeutics Research Laboratory, National Cancer Centre, Singapore, Singapore
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
| | - Constance H Li
- Cancer Therapeutics Research Laboratory, National Cancer Centre, Singapore, Singapore
- Academic Clinical Program in Oncology, Duke-NUS Medical School, Singapore, Singapore
| | - Hui Sun Leong
- Cancer Therapeutics Research Laboratory, National Cancer Centre, Singapore, Singapore
| | - Fui Teen Chong
- Cancer Therapeutics Research Laboratory, National Cancer Centre, Singapore, Singapore
| | - Shilpi Gupta
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Camille Arcinas
- Cancer Therapeutics Research Laboratory, National Cancer Centre, Singapore, Singapore
- Academic Clinical Program in Oncology, Duke-NUS Medical School, Singapore, Singapore
| | - John F Ouyang
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Vivian Ang
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Teja Celhar
- HuNIT platform, Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yunqian Zhao
- HuNIT platform, Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Hui Chen Tay
- HuNIT platform, Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jerry Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
| | - Takeshi Takahashi
- Laboratory Animal Research Department, Central Institute for Experimental Animals (CIEA), Kawasaki, Japan
| | - Daniel S W Tan
- Cancer Therapeutics Research Laboratory, National Cancer Centre, Singapore, Singapore
- Academic Clinical Program in Oncology, Duke-NUS Medical School, Singapore, Singapore
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Subhra K Biswas
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Owen J L Rackham
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
- The Alan Turing Institute, The British Library, London, United Kingdom
| | - N Gopalakrishna Iyer
- Cancer Therapeutics Research Laboratory, National Cancer Centre, Singapore, Singapore.
- Academic Clinical Program in Oncology, Duke-NUS Medical School, Singapore, Singapore.
- Department of Head and Neck Surgery, National Cancer Centre, Singapore, Singapore.
- Division of Medical Sciences, National Cancer Centre, Singapore, Singapore.
| |
Collapse
|
126
|
Lin CC, Yang CY, Hung TC, Wang CH, Wei SW, Schiro P, Tseng JY, Lin CH, Jiang JK. MiSelect R System: the validation of a new detection system of CTCs and their correlation with prognosis in non-metastatic CRC patients. Sci Rep 2023; 13:4773. [PMID: 36959311 PMCID: PMC10036555 DOI: 10.1038/s41598-023-31346-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 03/10/2023] [Indexed: 03/25/2023] Open
Abstract
Circulating tumor cells (CTCs) in blood are accepted as a prognostic marker for patients with metastatic colorectal cancer (CRC). However, there is limited data on the use of CTCs as a prognostic marker for non-metastatic patients. In the current study, we used a rare cell automated analysis platform, the MiSelect R System, to enumerate CTCs from blood in non-metastatic CRC patients, and corelated the number of CTCs with the clinical staging and survival. The presence of CTCs in mesenteric vein blood (MVB) samples from 101 CRC patients was significantly associated with T stage. Patients with 1 or more CTCs per 8 mL of MVB exhibited significantly worse disease-free survival (DFS) and cancer-specific survival (CSS) compared to patient without CTCs. The presence of CTCs before surgery is an independent marker for both DFS and CSS. CTC presence after surgical resection is also a prognostic marker. CTCs are a potentially useful prognostic and predictive biomarker in non-metastatic CRC patients that may further stratify patient's risk status within different stages of disease.
Collapse
Affiliation(s)
- Chun-Chi Lin
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, No.201, Shipai Rd. Sec. 2, Beitou Dist., Taipei City, 112, Taiwan
| | - Chih-Yung Yang
- Department of Teaching and Research, Taipei City Hospital, Taipei City, 104, Taiwan
| | - Tzu-Chao Hung
- MiCareo Taiwan Co., Ltd., 5F., No. 69, Ln. 77, Xing Ai Rd., Neihu Dist., Taipei City, 114, Taiwan
| | - Chun-Hung Wang
- MiCareo Taiwan Co., Ltd., 5F., No. 69, Ln. 77, Xing Ai Rd., Neihu Dist., Taipei City, 114, Taiwan
| | - Sheng-Wen Wei
- MiCareo Taiwan Co., Ltd., 5F., No. 69, Ln. 77, Xing Ai Rd., Neihu Dist., Taipei City, 114, Taiwan
| | - Perry Schiro
- MiCareo Taiwan Co., Ltd., 5F., No. 69, Ln. 77, Xing Ai Rd., Neihu Dist., Taipei City, 114, Taiwan
| | - Ju-Yu Tseng
- MiCareo Taiwan Co., Ltd., 5F., No. 69, Ln. 77, Xing Ai Rd., Neihu Dist., Taipei City, 114, Taiwan.
| | - Chi-Hung Lin
- Department of Biological Science and Technology, National Yang-Ming Chiao-Tung University, Hsinchu, 300, Taiwan
- Institute of Microbiology and Immunology, National Yang-Ming Chiao-Tung University, Taipei City, 112, Taiwan
- Cancer Progression Research Center, National Yang Ming Chiao-Tung University, Taipei City, 112, Taiwan
| | - Jeng-Kai Jiang
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, No.201, Shipai Rd. Sec. 2, Beitou Dist., Taipei City, 112, Taiwan.
- School of Medicine, National Yang-Ming Chiao-Tung University, Taipei City, 112, Taiwan.
| |
Collapse
|
127
|
Klosowski M, Haines L, Alfino L, McMellen A, Leibowitz M, Regan D. Naturally occurring canine sarcomas: Bridging the gap from mouse models to human patients through cross-disciplinary research partnerships. Front Oncol 2023; 13:1130215. [PMID: 37035209 PMCID: PMC10076632 DOI: 10.3389/fonc.2023.1130215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/20/2023] [Indexed: 04/11/2023] Open
Abstract
Fueled by support from the National Cancer Institute's "Cancer Moonshot" program, the past few years have witnessed a renewed interest in the canine spontaneous cancer model as an invaluable resource in translational oncology research. Increasingly, there is awareness that pet dogs with cancer provide an accessible bridge to improving the efficiency of cancer drug discovery and clinical therapeutic development. Canine tumors share many biological, genetic, and histologic features with their human tumor counterparts, and most importantly, retain the complexities of naturally occurring drug resistance, metastasis, and tumor-host immune interactions, all of which are difficult to recapitulate in induced or genetically engineered murine tumor models. The utility of canine models has been particularly apparent in sarcoma research, where the increased incidence of sarcomas in dogs as compared to people has facilitated comparative research resulting in treatment advances benefitting both species. Although there is an increasing awareness of the advantages in using spontaneous canine sarcoma models for research, these models remain underutilized, in part due to a lack of more permanent institutional and cross-institutional infrastructure to support partnerships between veterinary and human clinician-scientists. In this review, we provide an updated overview of historical and current applications of spontaneously occurring canine tumor models in sarcoma research, with particular attention to knowledge gaps, limitations, and growth opportunities within these applications. Furthermore, we propose considerations for working within existing veterinary translational and comparative oncology research infrastructures to maximize the benefit of partnerships between veterinary and human biomedical researchers within and across institutions to improve the utility and application of spontaneous canine sarcomas in translational oncology research.
Collapse
Affiliation(s)
- Marika Klosowski
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Laurel Haines
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Lauren Alfino
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Alexandra McMellen
- Center for Cancer and Blood Disorders, Children’s Hospital Colorado, Aurora, CO, United States
| | - Michael Leibowitz
- Center for Cancer and Blood Disorders, Children’s Hospital Colorado, Aurora, CO, United States
| | - Daniel Regan
- Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
128
|
Rastad H, Mozafary Bazargany MH, Samimisedeh P, Farahani M, Hashemnejad M, Moghadam S, Khodaparast Z, Shams R, Seifi-Alan M. Clinicopathological and prognostic value of lncRNA TPT1-AS1 in cancer: a systematic review study and meta-analysis. Pathol Res Pract 2023; 245:154403. [PMID: 37004278 DOI: 10.1016/j.prp.2023.154403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/26/2023] [Accepted: 03/03/2023] [Indexed: 03/07/2023]
Abstract
INTRODUCTION Aberrant expression of lncRNAs in cancer cells can impact their key phenotypes. We aimed to summarize available evidence on clinicopathological and prognostic value of lncRNA TPT1-AS1 in cancer. METHODS A systematic search was performed on Medline and Embase databases using relevant key terms covering lncRNA TPT1-AS1, cancer, and clinical outcomes. The effect size estimates and their 95 % confidence interval (CI) were pooled using random-effects models. Meta- analyses were conducted using STATA 16.0 software. RESULTS Seventeen articles met our eligibility criteria. Tumor tissue compared to normal tissue showed increased level of lncRNA TPT1-AS1 expression (pooled standardized mean difference (95 % CI): 0.65 (0.52-0.79)). Overexpression of this lncRNA was a significant predictor for poor prognosis (Pooled log-rank test P-value < 0.001); in patients with high-level of lncRNA TPT1-AS1, the risk of death at five years was 1.40 times greater than their counterparts. The pooled Odds ratios for association lncRNA TPT1-AS1 with tumor stage, tumor size, and lymph node metastasis were 1.94 (95 % CI: 0.90-4.19, 8 studies, I2 = 79.6 %), 2.33 (95 % CI: 1.31-4.14, 5 studies, I2 = 40.0 %), and 1.89 (95 % CI: 1.08-3.36, 5 studies, I2 = 61.7 %), respectively. Regarding the identified potential mechanisms, lncRNA TPT1-AS1 plays a role in cancer growth mainly by sponging miRNAs and regulating their downstream targets or controlling the expression of key cell cycle regulators. CONCLUSION In cancer patients, elevated expression of lncRNA TPT1-AS1 might be associated with a shorter Overall Survival, advanced stages, larger tumor size, and lymph node metastasis.
Collapse
Affiliation(s)
- Hadith Rastad
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Parham Samimisedeh
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Masoumeh Farahani
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Maryam Hashemnejad
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Somaye Moghadam
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Khodaparast
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Roshanak Shams
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mahnaz Seifi-Alan
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
129
|
Chen L, Jiang P, Shen X, Lyu J, Liu C, Li L, Huang Y. Cascade Delivery to Golgi Apparatus and On-Site Formation of Subcellular Drug Reservoir for Cancer Metastasis Suppression. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2204747. [PMID: 36585358 DOI: 10.1002/smll.202204747] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/07/2022] [Indexed: 06/17/2023]
Abstract
As the foremost cause of cancer-related death, metastasis consists of three steps: invasion, circulation, and colonization. Only targeting one single phase of the metastasis cascade may be insufficient since there are many alternative routes for tumor cells to disseminate. Here, to target the whole cascade of metastasis, hybrid erythrocyte and tumor cell membrane-coated nanoparticle (Hyb-NP) is designed with dual functions of increasing circulation time and recognizing primary, circulating, and colonized tumors. After loading with monensin, a recently reported metastasis inhibitor, the delivery system profoundly reduces spontaneous metastasis in an orthotopic breast cancer model. Underlying mechanism studies reveal that Hyb-NP can deliver monensin to its action site in the Golgi apparatus, and in return, monensin can block the exocytosis of Hyb-NP from the Golgi apparatus, forming a reservoir-like subcellular structure. Notably, the Golgi apparatus reservoir displays three vital functions for suppressing metastasis initialization, including enhanced subcellular drug retention, metastasis-related cytokine release inhibition, and directional migration inhibition. Collectively, based on metastasis cascade targeting at the tissue level, further formation of the Golgi apparatus drug reservoir at the subcellular level provides a potential therapeutic strategy for cancer metastasis suppression.
Collapse
Affiliation(s)
- Liqiang Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Peihang Jiang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Xinran Shen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Jiayan Lyu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Chendong Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
130
|
Park WY, Gray JM, Holewinski RJ, Andresson T, So JY, Carmona-Rivera C, Hollander MC, Yang HH, Lee M, Kaplan MJ, Cappell SD, Yang L. Apoptosis-induced nuclear expulsion in tumor cells drives S100a4-mediated metastatic outgrowth through the RAGE pathway. NATURE CANCER 2023; 4:419-435. [PMID: 36973439 PMCID: PMC10042736 DOI: 10.1038/s43018-023-00524-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/07/2023] [Indexed: 03/29/2023]
Abstract
Most tumor cells undergo apoptosis in circulation and at the metastatic organ sites due to host immune surveillance and a hostile microenvironment. It remains to be elucidated whether dying tumor cells have a direct effect on live tumor cells during the metastatic process and what the underlying mechanisms are. Here we report that apoptotic cancer cells enhance the metastatic outgrowth of surviving cells through Padi4-mediated nuclear expulsion. Tumor cell nuclear expulsion results in an extracellular DNA-protein complex that is enriched with receptor for advanced glycation endproducts (RAGE) ligands. The chromatin-bound RAGE ligand S100a4 activates RAGE receptors in neighboring surviving tumor cells, leading to Erk activation. In addition, we identified nuclear expulsion products in human patients with breast, bladder and lung cancer and a nuclear expulsion signature correlated with poor prognosis. Collectively, our study demonstrates how apoptotic cell death can enhance the metastatic outgrowth of neighboring live tumor cells.
Collapse
Affiliation(s)
- Woo-Yong Park
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Justin M Gray
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Ronald J Holewinski
- Protein Mass Spectrometry Group, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Thorkell Andresson
- Protein Mass Spectrometry Group, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Jae Young So
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Carmelo Carmona-Rivera
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - M Christine Hollander
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Howard H Yang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Maxwell Lee
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Steven D Cappell
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Li Yang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
131
|
Amgalan B, Day CP, Przytycka TM. Exploring tumor-normal cross-talk with TranNet: role of the environment in tumor progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.24.529899. [PMID: 36945455 PMCID: PMC10028821 DOI: 10.1101/2023.02.24.529899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
There is a growing awareness that tumor-adjacent normal tissues used as control samples in cancer studies do not represent fully healthy tissues. Instead, they are intermediates between healthy tissues and tumors. The factors that contribute to the deviation of such control samples from healthy state include exposure to the tumor-promoting factors, tumor-related immune response, and other aspects of tumor microenvironment. Characterizing the relation between gene expression of tumor-adjacent control samples and tumors is fundamental for understanding roles of microenvironment in tumor initiation and progression, as well as for identification of diagnostic and prognostic biomarkers for cancers. To address the demand, we developed and validated TranNet, a computational approach that utilizes gene expression in matched control and tumor samples to study the relation between their gene expression profiles. TranNet infers a sparse weighted bipartite graph from gene expression profiles of matched control samples to tumors. The results allow us to identify predictors (potential regulators) of this transition. To our knowledge, TranNet is the first computational method to infer such regulation. We applied TranNet to the data of several cancer types and their matched control samples from The Cancer Genome Atlas (TCGA). Many predictors identified by TranNet are genes associated with regulation by the tumor microenvironment as they are enriched in G-protein coupled receptor signaling, cell-to-cell communication, immune processes, and cell adhesion. Correspondingly, targets of inferred predictors are enriched in pathways related to tissue remodelling (including the epithelial-mesenchymal Transition (EMT)), immune response, and cell proliferation. This implies that the predictors are markers and potential stromal facilitators of tumor progression. Our results provide new insights for the relationships between tumor adjacent control sample, tumor and the tumor environment. Moreover, the set of predictors identified by TranNet will provide a valuable resource for future investigations. The TranNet method was implemented in python, source codes and the data sets used for and generated during this study are available at the Github site https://github.com/ncbi/TranNet .
Collapse
Affiliation(s)
- Bayarbaatar Amgalan
- National Center for Biotechnology Information, National Library of Medicine, NIH, Bethesda, Maryland, USA
| | - Chi-Ping Day
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Teresa M. Przytycka
- National Center for Biotechnology Information, National Library of Medicine, NIH, Bethesda, Maryland, USA
| |
Collapse
|
132
|
Pavlov VN, Urmantsev MF, Bakeev MR. Opportunities of ICG-fluorescent imaging of lymph nodes during radical cystectomy in patients with bladder cancer: A review. JOURNAL OF MODERN ONCOLOGY 2023. [DOI: 10.26442/18151434.2022.4.201874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Today, the "golden standard" for the treatment of aggressive forms of bladder cancer is radical cystectomy. An important stage is the pelvic lymphadenectomy. This procedure is crucial for staging the tumor process, determining the subsequent treatment tactics and improving the results of surgery. Currently, the concept of a signaling lymph node (LN) is actively developing, which allows reducing the necessary level of dissection of the pelvic LN. Reducing the number of resected LN increases the level of surgical safety of the patient. The leading method of intraoperative visualization of signal LN is the fluorescence of indocyanine green in the near infrared range of the spectrum. The prospects of this diagnostic method for radical cystectomy in patients with bladder cancer determine the vector of future scientific research in this area.
Collapse
|
133
|
Arpacı AH, Özkoçer SE, Güneş E, Elmas Ç, Işık B. Effects of recurrent ketamine exposure on brain histopathology in juvenile rats. Turk J Med Sci 2023; 53:19-28. [PMID: 36945933 PMCID: PMC10388022 DOI: 10.55730/1300-0144.5554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/12/2022] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Ketamine (KET) is a commonly used anesthetic agent. However, several previous studies reported that KET leads to neuronal damage in neurodevelopmental stages and has neuroprotective effects. The present experimental study aimed to determine the undesirable histopathological effects of KET in the cerebral cortex, striatum, and hippocampus after recurrent KET administration in juvenile rats. METHODS After ethical approval was obtained, 32 juvenile male Wistar Albino rats were randomized into four groups: 1 mg/kg serum saline intraperitoneally (i.p.), 5 mg/kg KET i.p., 20 mg/kg KET i.p., and 50 mg/kg KET i.p. KET was administered for three consecutive days at three-h intervals in three doses. Ten days after the last KET dose, the rats were sacrificed. Cerebral hemispheres were fixed. Hematoxylin and eosin stain was used for morphometric analysis. Hippocampi were evaluated by immunohistochemistry with anticleaved caspase-3 antibodies. Statistical analysis was conducted with SPSS 21 software using the ANOVA test and Bonferroni post hoc analysis method. RESULTS The experimental study findings revealed no difference between the groups' cell counts or sizes in cortical morphometry. No degenerative changes were observed in pyramidal and granular cells in the striatum. Mild gliosis was observed in the 20 mg/kg and 50 mg/kg KET administration groups. Immuno-histo-chemical analysis was conducted to determine apoptosis in the CA1 region of the hippocampus and revealed that caspase-3 positivity increased with the KET dose. However, there was no statistical difference between the groups. While it was lower than the control group in the 5 mg/kg KET group, it was similar to the control group in the 20 mg/kg KET group and higher in the 50 mg/kg KET group (p > 0.05). DISCUSSION : Repetitive KET exposure did not significantly affect juvenile cerebral morphology and apoptosis in hippocampal cells.
Collapse
Affiliation(s)
- Ayşe Hande Arpacı
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Ankara University, Ankara, Turkey
| | - Süheyla Esra Özkoçer
- Department of Histology and Embryology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Emel Güneş
- Department of Physiology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Çiğdem Elmas
- Department of Histology and Embryology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Berrin Işık
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
134
|
Ring A, Nguyen-Sträuli BD, Wicki A, Aceto N. Biology, vulnerabilities and clinical applications of circulating tumour cells. Nat Rev Cancer 2023; 23:95-111. [PMID: 36494603 PMCID: PMC9734934 DOI: 10.1038/s41568-022-00536-4] [Citation(s) in RCA: 140] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/07/2022] [Indexed: 12/13/2022]
Abstract
In recent years, exceptional technological advances have enabled the identification and interrogation of rare circulating tumour cells (CTCs) from blood samples of patients, leading to new fields of research and fostering the promise for paradigm-changing, liquid biopsy-based clinical applications. Analysis of CTCs has revealed distinct biological phenotypes, including the presence of CTC clusters and the interaction between CTCs and immune or stromal cells, impacting metastasis formation and providing new insights into cancer vulnerabilities. Here we review the progress made in understanding biological features of CTCs and provide insight into exploiting these developments to design future clinical tools for improving the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Alexander Ring
- Department of Biology, Institute for Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Bich Doan Nguyen-Sträuli
- Department of Biology, Institute for Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
- Department of Gynecology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Andreas Wicki
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Nicola Aceto
- Department of Biology, Institute for Molecular Health Sciences, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
135
|
Zhou X, Xu X, Hu Q, Wu Y, Yu F, He C, Qian Y, Han Y, Tang J, Hu H. Novel manganese and polyester dendrimer-based theranostic nanoparticles for MRI and breast cancer therapy. J Mater Chem B 2023; 11:648-656. [PMID: 36541124 DOI: 10.1039/d2tb01855a] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Therapeutic nanoplatforms are widely used in the diagnosis and treatment of breast cancer due to the merits of enabling high soft-tissue resolution and the availability of numerous therapeutic nanoparticles. It is thus vital to develop multifunctional theranostic nanoparticles for the visualization and dynamic monitoring of tumor therapy. In this study, we designed a manganese-based and hypericin-loaded polyester dendrimer nanoparticle (MHD) for magnetic resonance imaging (MRI) and hypericin-based photodynamic therapy (PDT) enhancement. We found that MHD could greatly enhance MRI contrast with a longitudinal relaxivity of 5.8 mM-1 s-1 due to the Mn-based paramagnetic dendrimer carrier. Meanwhile, the MRI-guided PDT inhibition of breast tumors could be achieved by the hypericin-carrying MHD and further improved by Mn2+-mediated alleviation of the hypoxic microenvironment and the enhancement of cellular ROS. Besides, MHD showed excellent biocompatibility and biosafety with liver and kidney clearance mechanisms. Thus, the high efficiency in MRI contrast enhancement and excellent tumor-inhibiting effects indicate MHD's potential as a novel, stable, and multifunctional nanotheranostic agent for breast cancer.
Collapse
Affiliation(s)
- Xiaoxuan Zhou
- Department of Radiology, Sir Run Run Shaw Hospital of School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310027, China.
| | - Xiaodan Xu
- Key Laboratory of Smart Biomaterials of Zhejiang Province, ZJU-Hangzhou Global Scientific and Technological Innovation Center, and College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China.
| | - Qiuhui Hu
- Department of Radiology, Sir Run Run Shaw Hospital of School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310027, China.
| | - Yan Wu
- Department of Radiology, Sir Run Run Shaw Hospital of School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310027, China.
| | - Feidan Yu
- Department of Radiology, Sir Run Run Shaw Hospital of School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310027, China.
| | - Chengbin He
- Department of Radiology, Sir Run Run Shaw Hospital of School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310027, China.
| | - Yue Qian
- Department of Radiology, Sir Run Run Shaw Hospital of School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310027, China.
| | - Yuxin Han
- Department of Radiology, Sir Run Run Shaw Hospital of School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310027, China.
| | - Jianbin Tang
- Key Laboratory of Smart Biomaterials of Zhejiang Province, ZJU-Hangzhou Global Scientific and Technological Innovation Center, and College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China.
| | - Hongjie Hu
- Department of Radiology, Sir Run Run Shaw Hospital of School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310027, China.
| |
Collapse
|
136
|
Najafi V, Yoosefian M, Hassani Z. Development of venetoclax performance using its new derivatives on BCL-2 protein inhibition. Cell Biochem Funct 2023; 41:58-66. [PMID: 36259104 DOI: 10.1002/cbf.3760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/26/2022] [Accepted: 10/04/2022] [Indexed: 01/11/2023]
Abstract
Cancer cells are resistant to apoptosis and this is one of the most obvious symptoms of cancer in humans. One of the most exciting strategies for treating cancer is to design regulators that increase cell death and stop cell growth. Members of the BCL-2 family of proteins play an important role in the regulation of apoptosis. In this study, an attempt was made to improve the performance of one of the anticancer drugs by designing new analogs of venetoclax (VNT). For this purpose, molecular docking studies were performed to determine the best binding state of VNT and its newly designed derivatives at the protein-binding site to estimate the binding energy. The best analog in terms of free energy was VNT-12 with the lowest energy (-12.15 kcal/mol). Finally, to investigate the inhibitory effect of the compounds on BCL-2 protein, molecular dynamics simulation was used, and by performing the relevant analyses during the simulation, it was observed that the newly designed ligand had better performance in inhibiting BCL-2 protein compared to VNT.
Collapse
Affiliation(s)
- Vahideh Najafi
- Department of Chemistry, Graduate University of Advanced Technology, Kerman, Iran
| | - Mehdi Yoosefian
- Department of Chemistry, Graduate University of Advanced Technology, Kerman, Iran
| | - Zahra Hassani
- Department of New Materials, Institute of Science, High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| |
Collapse
|
137
|
Brancato V. 3D Bioprinting for Cancer Models. Cancer Nanotechnol 2023. [DOI: 10.1007/978-3-031-17831-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
138
|
Wei X, Yang M. Cell- and subcellular organelle-targeting nanoparticle-mediated breast cancer therapy. Front Pharmacol 2023; 14:1180794. [PMID: 37089933 PMCID: PMC10117787 DOI: 10.3389/fphar.2023.1180794] [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: 03/06/2023] [Accepted: 03/27/2023] [Indexed: 04/25/2023] Open
Abstract
Breast cancer (BC) is the most prevalent malignant tumor, surpassing lung cancer as the most frequent malignancy in women. Drug resistance, metastasis, and immune escape are the major factors affecting patient survival and represent a huge challenge in BC treatment in clinic. The cell- and subcellular organelle-targeting nanoparticles-mediated targeted BC therapy may be an effective modality for immune evasion, metastasis, and drug resistance. Nanocarriers, efficiently delivering small molecules and macromolecules, are used to target subcellular apparatuses with excellent targeting, controlled delivery, and fewer side effects. This study summarizes and critically analyzes the latest organic nanoparticle-mediated subcellular targeted therapeutic based on chemotherapy, gene therapy, immunotherapy, and combination therapy in detail, and discusses the challenges and opportunities of nanoparticle therapy.
Collapse
Affiliation(s)
- Xue Wei
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Ming Yang
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Ming Yang,
| |
Collapse
|
139
|
Kundu T, Giri R, Hota SK, Sahu SK, Senapati U. Tumour budding in invasive breast carcinoma and its correlation with histopathological prognostic parameters and hormone receptor status. J Cancer Res Ther 2023; 19:S664-S668. [PMID: 38384036 DOI: 10.4103/jcrt.jcrt_656_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/25/2022] [Indexed: 02/23/2024]
Abstract
INTRODUCTION Tumor Budding (TB) is emerging as an important sensitive indicator of aggressiveness over traditional histopathological variables such as lymph node metastasis and lymphovascular invasion because TB at the invasive front was postulated as the first step of invasion and metastasis. But the role of TB in breast carcinoma is not yet standardized. AIM This study aims to assess the prognostic significance of TB in breast carcinoma. OBJECTIVES To study and grade TB in invasive breast carcinoma and correlate it with known histopathological prognostic markers and hormone receptor status. MATERIALS AND METHODS In this ambispective study, 51 cases of invasive breast carcinoma undergoing radical surgery were studied from September 2017 to September 2021. Tumor buds were defined as a single or cluster of up to four tumor cells at the invasive front of the tumor and were counted by using IHC Pancytokeratin in 200X and graded into a three-tier grading system. The correlation between TB with established histopathological parameters and hormone receptor status was studied. A Chi-square test was used and a P value < 0.05 was considered significant. RESULTS TB was seen in all cases of invasive breast carcinoma. Grade 1 TB was seen in 37.26% (19/51) of cases and Grade 2 TB and Grade 3 TB in 31.37% (16/51) of cases each. There was a lack of significant correlation between TB with histologic grade, lymph node status, and HER2 neu. Correlation between TB with Estrogen receptor (ER), Progesterone receptor (PR), and American Joint Committee on Cancer (AJCC) Prognostic stage group was noted but it was not statistically significant. CONCLUSION Although several studies have confirmed the prognostic value of TB in breast carcinoma, they had used several different methods of assessment. So, there is a need for a standardized method for the assessment of TB in breast carcinoma. Further standardization of TB may add its value as a prognostic factor.
Collapse
Affiliation(s)
- Tamanna Kundu
- Department of Pathology, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, Odisha, India
| | - Ranjana Giri
- Department of Pathology, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, Odisha, India
| | - Subhransu K Hota
- Department of Pathology, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, Odisha, India
| | - Subrat K Sahu
- Department of Oncosurgery, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, Odisha, India
| | - Urmila Senapati
- Department of Pathology, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, Odisha, India
| |
Collapse
|
140
|
Lu DY, Lu TR. Drug Sensitivity Testing for Cancer Therapy, Technique Analysis and Trends. Curr Rev Clin Exp Pharmacol 2023; 18:3-11. [PMID: 34515020 DOI: 10.2174/2772432816666210910104649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 02/08/2023]
Abstract
The techniques and qualities of drug sensitivity testing (DST) for anticancer treatment have grown rapidly in the past two decades worldwide. Much of DST progress came from advanced systems of technical versatility (faster, highly-throughput, highly-sensitive, and smaller in tumor quantity). As the earliest drug selective system, biomedical knowledge and technical advances for DST are mutually supported. More importantly, many pharmacological controversies are resolved by these technical advances. With this technical stride, the clinical landscape of DST entered into a new phase (>500 samples per testing and extremely low quantity of tumor cells). As a forerunner of the drug selection system, DST awaits a new version that can adapt to complicated therapeutic situations and diverse tumor categories in the clinic. By upholding this goal of pathogenic and therapeutic diversity, DST could eventually cure more cancer patients by establishing high-quality drug selection systems. To smoothen DST development, there is a need to increase the understanding of cancer biology, pathology and pharmacology (cancer heterogeneity, plasticity, metastasis and drug resistance) with well-informative parameters before chemotherapy. In this article, medicinal and technical insights into DST are especially highlighted.
Collapse
Affiliation(s)
- Da-Yong Lu
- School of Life Sciences, Shanghai University, Shanghai 200444, P.R. China
| | - Ting-Ren Lu
- College of Science, Shanghai University, Shanghai 200444, P.R. China
| |
Collapse
|
141
|
Sun M, Huang D, Liu Y, Chen H, Yu H, Zhang G, Chen Q, Chen H, Zhang J. Effects of Cinobufagin on the Proliferation, Migration, and Invasion of H1299 Lung Cancer Cells. Chem Biodivers 2023; 20:e202200961. [PMID: 36522286 DOI: 10.1002/cbdv.202200961] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/30/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Cinobufagin (CB), with its steroidal nucleus structure, is one of the major, biologically active components of Chan Su. Recent studies have shown that CB exerts inhibitory effects against numerous cancer cells. However, the effects of CB regarding the metastasis of non-small cell lung cancer (NSCLC) and the involved mechanisms need to be further studied. The purpose of the present study aimed to report the inhibitory function of CB against proliferation and metastasis of H1299 cells. CB inhibited proliferation of H1299 lung cancer cells with an IC50 value of 0.035±0.008 μM according to the results of MTT assays. Antiproliferative activity was also observed in colony forming cell assays. In addition, 5-ethynyl-2'-deoxyuridine (EdU) retention assays revealed that CB significantly inhibited the rate of DNA synthesis in H1299 cells. Moreover, results of the scratch wound healing assays and transwell migration assays displayed that CB exhibited significant inhibition against migration and invasion of H1299 cells. Furthermore, CB could concentration-dependently reduce the expression of integrin α2, β-catenin, FAK, Src, c-Myc, and STAT3 in H1299 cells. These western blotting results indicated that CB might target integrin α2, β-catenin, FAK and Src to suppress invasion and migration of NSCLC, which was consistent with the network pharmacology analysis results. Collectively, findings of the current study suggest that CB possesses promising activity against NSCLC growth and metastasis.
Collapse
Affiliation(s)
- Mingna Sun
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Dongyu Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yun Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.,Guangdong Provincial Institute of Biological Products and Materia Medica, Guangzhou, China
| | - Haifang Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Hua Yu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Guobin Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qilei Chen
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region, China
| | - Hubiao Chen
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong Special Administrative Region, China
| | - Jianye Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| |
Collapse
|
142
|
Chen CW, Zeng XY, Cheng CC, Wang CF, Chen JK. LSPR Sensing of Epithelial Cell Adhesion Molecules through Sphere and Cavity Plasmons of a Composite Ring Array of Poly[2-(dimethylamino)ethyl methacrylate]/Gold Nanoparticles. Anal Chem 2022; 94:17779-17786. [PMID: 36519823 DOI: 10.1021/acs.analchem.2c03149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Self-organization facilitates the formation of specific structures as a result of constituent interactions. In this study, the bottom of a 600 nm hole array photoresist template, which was deposited with a hydrophobic atom transfer radical polymerization (ATRP) initiator, was wetted by treatment with oxygen plasma. After the removal of the photoresist template, ring patterns of the ATRP initiator were formed at the interface between the hydrophobic and wetting regions. Poly[2-(dimethylamino)ethyl methacrylate] (PDMAEMA) was grafted from the ring array of the initiator to immobilize gold nanoparticles (AuNPs) as a uniform ring array on a silicon substrate via repeated swelling/shrinking cycles. The localized surface plasmon resonance (LSPR) peak of the PDMAEMA-AuNP hybrid ring (PAHR) red-shifted after 12 swelling/shrinking cycles. In comparison to gold nanoparticles, scalable gold nanorings can effectively develop a variety of nanostructures to design LSPR-based sensors and optimize the sensing accuracy and stability. To detect epithelial cell adhesion molecules (EpCAM) during the structural change from a ring to a disk, antiEpCAM was anchored onto the PAHR as a biosensor during swelling/shrinking. The coupling of antiEpCAM and EpCAM led to asymptotical convergence from rings to disks as well as blue shifts of the LSPR peaks. Linear correlation between the blue shift and EpCAM concentration showed a limit of detection of ∼27 pg mL-1 and a linear range of 25-200 pg mL-1 for the detection of EpCAM within 30 min. The simple method of combining lithography and plasma technology provides a versatile platform for developing the scalable ring structure of AuNPs for highly sensitive and selective biosensing.
Collapse
Affiliation(s)
- Chih-Wei Chen
- Division of Neurosurgery, Department of Surgery, Chi Mei Medical Center, Tainan710, Taiwan, ROC.,Department of Occupational Safety and Health/Institute of Industrial Safety and Disaster Prevention, College of Sustainable Environment, Chia Nan University of Pharmacy and Science, Tainan717, Taiwan, ROC.,Department of Materials and Science Engineering, National Taiwan University of Science and Technology, 43, Section 4, Keelung Road, Taipei106, Taiwan, ROC
| | - Xiang-Yun Zeng
- Department of Materials and Science Engineering, National Taiwan University of Science and Technology, 43, Section 4, Keelung Road, Taipei106, Taiwan, ROC
| | - Chih-Chia Cheng
- Graduate Institute of Applied Science and Technology, Advanced Membrane Materials Research Center, National Taiwan University of Science and Technology, Taipei106, Taiwan, ROC
| | - Chih-Feng Wang
- Graduate Institute of Applied Science and Technology, Advanced Membrane Materials Research Center, National Taiwan University of Science and Technology, Taipei106, Taiwan, ROC
| | - Jem-Kun Chen
- Department of Materials and Science Engineering, National Taiwan University of Science and Technology, 43, Section 4, Keelung Road, Taipei106, Taiwan, ROC
| |
Collapse
|
143
|
Onal S, Alkaisi MM, Nock V. Microdevice-based mechanical compression on living cells. iScience 2022; 25:105518. [PMID: 36444299 PMCID: PMC9699986 DOI: 10.1016/j.isci.2022.105518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Compressive stress enables the investigation of a range of cellular processes in which forces play an important role, such as cell growth, differentiation, migration, and invasion. Such solid stress can be introduced externally to study cell response and to mechanically induce changes in cell morphology and behavior by static or dynamic compression. Microfluidics is a useful tool for this, allowing one to mimic in vivo microenvironments in on-chip culture systems where force application can be controlled spatially and temporally. Here, we review the mechanical compression applications on cells with a broad focus on studies using microtechnologies and microdevices to apply cell compression, in comparison to off-chip bulk systems. Due to their unique features, microfluidic systems developed to apply compressive forces on single cells, in 2D and 3D culture models, and compression in cancer microenvironments are emphasized. Research efforts in this field can help the development of mechanoceuticals in the future.
Collapse
Affiliation(s)
- Sevgi Onal
- Electrical and Computer Engineering, University of Canterbury, Christchurch 8041, New Zealand
- MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
| | - Maan M. Alkaisi
- Electrical and Computer Engineering, University of Canterbury, Christchurch 8041, New Zealand
- MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
| | - Volker Nock
- Electrical and Computer Engineering, University of Canterbury, Christchurch 8041, New Zealand
- MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
- Biomolecular Interaction Centre, University of Canterbury, Christchurch 8041, New Zealand
| |
Collapse
|
144
|
Tian B, Bian Y, Bian DJ, Gao Y, Zhang X, Zhou SW, Zhang YH, Pang YN, Li ZS, Wang LW. Knowledge mapping of alternative splicing of cancer from 2012 to 2021: A bibliometric analysis. Front Oncol 2022; 12:1068805. [PMID: 36591484 PMCID: PMC9795218 DOI: 10.3389/fonc.2022.1068805] [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: 10/13/2022] [Accepted: 11/24/2022] [Indexed: 12/15/2022] Open
Abstract
Background As a processing method of RNA precursors, alternative splicing (AS) is critical to normal cellular activities. Aberrant AS events are associated with cancer development and can be promising targets to treat cancer. However, no detailed and unbiased study describes the current state of AS of cancer research. We aim to measure and recognize the current state and trends of AS cancer research in this study. Methods The Web of Science Core Collection was used to acquire the articles. Utilizing three bibliometric tools (CiteSpace, VOSviewer, R-bibliometrix), we were able to measure and recognize the influence and collaboration data of individual articles, journals, and co-citations. Analysis of co-occurrence and burst information helped us identify the trending research areas related to AS of cancer. Results From 2012 to 2021, the total number of papers on AS of cancer published in 766 academic journals was 3,507, authored by 20,406 researchers in 405 institutions from 80 countries/regions. Research involving AS of cancer genes was primarily conducted in the United States and China; simultaneously, the Chinese Academy of Sciences, Fudan University, and National Cancer Institute were the institutions with strong research capabilities. Scorilas Andreas is the scholar with the most publications, while the most co-citations were generated by Wang, Eric T. Plos One published the most papers on AS of cancer, while J Biol Chem was the most co-cited academic journal in this field. The results of keyword co-occurrence analysis can be divided into three types: molecular (P53, CD44, androgen receptor, srsf3, esrp1), pathological process (apoptosis, EMT, metastasis, angiogenesis, proliferation), and disease (breast cancer, colorectal cancer, prostate cancer, hepatocellular carcinoma, gastric cancer). Conclusion Research on AS of cancer has been increasing in intensity over the past decade. Current AS of cancer studies focused on the hallmarks of AS in cancer and AS signatures including diagnostic and therapeutic targets. Among them, the current trends are splicing factors regulating epithelial-mesenchymal transition and other hallmarks, aberrant splicing events in tumors, and further mechanisms. These might give researchers interested in this field a forward-looking perspective and inform further research.
Collapse
Affiliation(s)
- Bo Tian
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yan Bian
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - De-Jian Bian
- Department of Emergency, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Ye Gao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xun Zhang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Si-Wei Zhou
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yan-Hui Zhang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Ya-Nan Pang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China,Shanghai Institute of Pancreatic Diseases, Shanghai, China,*Correspondence: Ya-Nan Pang, ; Zhao-Shen Li, ; Luo-Wei Wang,
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China,*Correspondence: Ya-Nan Pang, ; Zhao-Shen Li, ; Luo-Wei Wang,
| | - Luo-Wei Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China,*Correspondence: Ya-Nan Pang, ; Zhao-Shen Li, ; Luo-Wei Wang,
| |
Collapse
|
145
|
Heightened Procoagulation after Post-Operative Thromboprophylaxis Completion in Patients with Metastatic Bone Disease from Primary Colorectal Cancer. J Clin Med 2022; 11:jcm11247397. [PMID: 36556013 PMCID: PMC9784178 DOI: 10.3390/jcm11247397] [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: 11/10/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Platelets play a role in venous thromboembolism (VTE) and in mediating colorectal cancer (CRC) progression. Still, platelets' role in hypercoagulability after surgical intervention for metastatic bone disease (MBD) is ill-defined. METHODS In this quantitative observational study, we utilized a high-resolution imaging approach to temporally examine platelet procoagulant membrane dynamics (PMD) in four patients with MBD from primary CRC (CRC/MBD), before and after surgical intervention, over a 6-month period. We coupled this investigation with thrombelastography, quantitative plasma shotgun proteomics, and biochemical analysis. RESULTS The plasma of CRC/MBD patients was enriched in ADAM1a, ADAMTS7, and physiological ligands for platelet glycoprotein-VI/spleen tyrosine kinase (GPVI/Syk) activation. Thromboprophylaxis attenuated procoagulation upon its initial prescription (post-operative day one, POD1); however, all patients experienced rebound procoagulation between POD3 and POD14, which was associated with Syk activation (Y525/Y526) in all patients, and a VTE event in two patients. Plasma levels of DNA-histone complexes increased steadily after surgery and remained elevated throughout the study period. Additionally, we increasingly sighted both homotypic and heterotypic platelet microaggregates after surgery in CRC/MBD patients, but not in healthy control participants' plasma. CONCLUSIONS Our data elucidates the cell biology of a prothrombo-inflammatory state caused by disease and vascular injury, and recalcitrant to thromboprophylaxis. New mechanistic insights into hypercoagulability in CRC/MBD patients may identify novel drug targets for effective thromboprophylaxis type and duration after orthopaedic surgery.
Collapse
|
146
|
Chen H, Li Q, Hu Q, Jiao X, Ren W, Wang S, Peng G. Double spiral chip-embedded micro-trapezoid filters (SMT filters) for the sensitive isolation of CTCs of prostate cancer by spectral detection. NANOSCALE ADVANCES 2022; 4:5392-5403. [PMID: 36540122 PMCID: PMC9724689 DOI: 10.1039/d2na00503d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 11/01/2022] [Indexed: 06/17/2023]
Abstract
Circulating tumor cells (CTCs) are cancer cells that are released from the original tumor and circulate in the blood vessels, carrying greatly similar constituents as the original tumor. Therefore, CTCs have a significant value in cancer prognosis, early diagnosis, and anti-cancer therapy. However, their rarity and heterogeneity make the isolation of CTCs an arduous task. In the present research, we propose a double spiral chip-embedded micro-trapezoid filter (SMT filter) for the sensitive isolation of the CTCs of prostate cancer by spectral detection. SMT filters were elongated to effectively capture CTCs and this distinctive design was conducive to their isolation and enrichment. The SMT filters were verified with tumor cells and artificial patient blood with a capture efficiency as high as 94% at a flow rate of 1.5 mL h-1. As a further validation, the SMT filters were validated in isolating CTCs from 10 prostate cancers and other cancers in 4 mL blood samples. Also, the CTCs tested positive for each patient blood sample, ranging from 83-114 CTCs. Significantly, we advanced hyperspectral imaging to detect the characteristic spectrum of CTCs both captured in situ on SMT filters and enriched after isolation. The CTCs could be positively identified by hyperspectral imaging with complete integrity of the cell morphology and an improved characteristic spectrum. This represents a breakthrough in the conventional surface-enhanced Raman scattering (SERS) spectroscopy of nanoparticles. Also, the characteristic spectrum of the CTCs would be highly beneficial for distinguishing the cancer type and accurate for enumerating tumor cells with varied intensities. Furthermore, a novel integrated flower-shaped microfilter was presented with all these aforementioned merits. The success of both the SMT filters and characteristic spectral detection indicated their feasibility for further clinical analysis, the evaluation of cancer therapy, and for potential application.
Collapse
Affiliation(s)
- Hongmei Chen
- School of Microelectronics and Data Science, Anhui University of Technology Maanshan 243002 P. R. China
- Shanghai Key Laboratory of Multidimensional Information Processing, East China Normal University Shanghai 200241 China
| | - Qingli Li
- Shanghai Key Laboratory of Multidimensional Information Processing, East China Normal University Shanghai 200241 China
| | - Qinghai Hu
- School of Chemistry and Chemical Engineering, Anhui University of Technology Maanshan 243002 P. R. China
| | - Xiaodong Jiao
- Department of Medical Oncology, Changzheng Hospital Shanghai 200070 P.R. China
| | - Wenjie Ren
- Shanghai Key Laboratory of Multidimensional Information Processing, East China Normal University Shanghai 200241 China
| | - Shuangshou Wang
- School of Chemistry and Chemical Engineering, Anhui University of Technology Maanshan 243002 P. R. China
| | - Guosheng Peng
- School of Microelectronics and Data Science, Anhui University of Technology Maanshan 243002 P. R. China
| |
Collapse
|
147
|
The Dual Role of PDCD10 in Cancers: A Promising Therapeutic Target. Cancers (Basel) 2022; 14:cancers14235986. [PMID: 36497468 PMCID: PMC9740655 DOI: 10.3390/cancers14235986] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/11/2022] Open
Abstract
Programmed cell death 10 (PDCD10) was initially considered as a protein associated with apoptosis. However, recent studies showed that PDCD10 is actually an adaptor protein. By interacting with multiple molecules, PDCD10 participates in various physiological processes, such as cell survival, migration, cell differentiation, vesicle trafficking, cellular senescence, neurovascular development, and gonadogenesis. Moreover, over the past few decades, accumulating evidence has demonstrated that the aberrant expression or mutation of PDCD10 is extremely common in various pathological processes, especially in cancers. The dysfunction of PDCD10 has been strongly implicated in oncogenesis and tumor progression. However, the updated data seem to indicate that PDCD10 has a dual role (either pro- or anti-tumor effects) in various cancer types, depending on cell/tissue specificity with different cellular interactors. In this review, we aimed to summarize the knowledge of the dual role of PDCD10 in cancers with a special focus on its cellular function and potential molecular mechanism. With these efforts, we hoped to provide new insight into the future development and application of PDCD10 as a clinical therapeutic target in cancers.
Collapse
|
148
|
Zhai BT, Sun J, Shi YJ, Zhang XF, Zou JB, Cheng JX, Fan Y, Guo DY, Tian H. Review targeted drug delivery systems for norcantharidin in cancer therapy. J Nanobiotechnology 2022; 20:509. [DOI: 10.1186/s12951-022-01703-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/11/2022] [Indexed: 12/04/2022] Open
Abstract
AbstractNorcantharidin (NCTD) is a demethylated derivative of cantharidin (CTD), the main anticancer active ingredient isolated from traditional Chinese medicine Mylabris. NCTD has been approved by the State Food and Drug Administration for the treatment of various solid tumors, especially liver cancer. Although NCTD greatly reduces the toxicity of CTD, there is still a certain degree of urinary toxicity and organ toxicity, and the poor solubility, short half-life, fast metabolism, as well as high venous irritation and weak tumor targeting ability limit its widespread application in the clinic. To reduce its toxicity and improve its efficacy, design of targeted drug delivery systems based on biomaterials and nanomaterials is one of the most feasible strategies. Therefore, this review focused on the studies of targeted drug delivery systems combined with NCTD in recent years, including passive and active targeted drug delivery systems, and physicochemical targeted drug delivery systems for improving drug bioavailability and enhancing its efficacy, as well as increasing drug targeting ability and reducing its adverse effects.
Graphical Abstract
Collapse
|
149
|
Wang Y, Li S, Weng L, Du H, Wang J, Xu X. LASS2 overexpression enhances early apoptosis of lung cancer cells through the caspase‑dependent pathway. Oncol Rep 2022; 48:220. [PMID: 36300249 DOI: 10.3892/or.2022.8435] [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: 06/10/2021] [Accepted: 06/27/2022] [Indexed: 11/06/2022] Open
Abstract
In a previous study by the authors, the longevity assurance homolog 2 (LASS2) gene was determined to inhibit activity of vacuolar H+‑ATPase (V‑ATPase) by combining with the C subunit (ATP6L) of V‑ATPase. However, the influence of LASS2 overexpression and silencing on apoptosis of human lung cancer cells 95D or 95C remains unclear. Thus, the effect of LASS2 on apoptosis and its potential mechanisms were investigated in 95D and 95C cells. Using the lentiviral transfection method, lentiviral vectors of LASS2 overexpression and silencing were transfected into 95D and 95C cells, respectively. The apoptotic ability of tumor cells was observed by flow cytometry. The expression levels of LASS2, Bcl‑2, Bax, cytochrome c, caspase‑9, and caspase‑3 were detected by western blotting. CCK‑8 assay was used to detect the growth ability of tumor cells in vitro. Flow cytometric analysis revealed that LASS2 overexpression could promote the early apoptosis of lung cancer cells 95D. CCK‑8 assay demonstrated that LASS2 overexpression inhibited the proliferation of 95D cells. Additionally, LASS2 overexpression decreased the expression of Bcl‑2, induced the release of cytochrome c from mitochondria, and promoted the activation of caspase‑9 and caspase‑3. There was a significant difference in the expression of Bcl‑2, cytochrome c, caspase‑9 and caspase‑3 in the LASS2‑overexpression group compared with the normal and negative control groups. Alternatively, the aforementioned experiments in lung cancer cells 95C following LASS2 silencing produced the opposite effects. LASS2 may induce early apoptosis of lung cancer cells by influencing the caspase‑dependent mitochondrial pathway.
Collapse
Affiliation(s)
- Yamei Wang
- Department of Pathology, College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010059, P.R. China
| | - Shirong Li
- Department of Pathology, College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010059, P.R. China
| | - Lixin Weng
- Department of Pathology, College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010059, P.R. China
| | - Hua Du
- Department of Pathology, College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010059, P.R. China
| | - Jingyuan Wang
- Department of Pathology, College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010059, P.R. China
| | - Xiaoyan Xu
- Department of Pathology, College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010059, P.R. China
| |
Collapse
|
150
|
Sun Y, Yang J, Li Y, Luo J, Sun J, Li D, Wang Y, Wang K, Yang L, Wu L, Sun X. Single low-dose INC280-loaded theranostic nanoparticles achieve multirooted delivery for MET-targeted primary and liver metastatic NSCLC. Mol Cancer 2022; 21:212. [PMID: 36457016 PMCID: PMC9717478 DOI: 10.1186/s12943-022-01681-y] [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: 08/04/2022] [Accepted: 11/11/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) patients with primary tumors and liver metastases have substantially reduced survival. Since mesenchymal-epithelial transition factor (MET) plays a significant role in the molecular mechanisms of advanced NSCLC, small molecule MET inhibitor capmatinib (INC280) hold promise for clinically NSCLC treatment. However, the major obstacles of MET-targeted therapy are poor drug solubility and off-tumor effects, even oral high-dosing regimens cannot significantly increase the therapeutic drug concentration in primary and metastatic NSCLC. METHODS We developed a multirooted delivery system INC280-PFCE nanoparticles (NPs) by loading INC280 into perfluoro-15-crown-5-ether for improving MET-targeted therapy. Biodistribution and anti-MET/antimetastatic effects of NPs were validated in orthotopic NSCLC and NSCLC liver metastasis models in a single low-dose. The efficacy of INC280-PFCE NPs was also explored in human NSCLC specimens. RESULTS INC280-PFCE NPs exhibited excellent antitumor ability in vitro. In orthotopic NSCLC models, sustained release and prolonged retention behaviors of INC280-PFCE NPs within tumors could be visualized in real-time by 19F magnetic resonance imaging (19F-MRI), and single pulmonary administration of NPs showed more significant tumor growth inhibition than oral administration of free INC280 at a tenfold higher dose. Furthermore, a single low-dose INC280-PFCE NPs administered intravenously suppressed widespread dissemination of liver metastasis without systemic toxicity. Finally, we verified the clinical translation potential of INC280-PFCE NPs in human NSCLC specimens. CONCLUSIONS These results demonstrated high anti-MET/antimetastatic efficacies, real-time MRI visualization and high biocompatibility of NPs after a single low-dose.
Collapse
Affiliation(s)
- Yige Sun
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Jie Yang
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Yingbo Li
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Jing Luo
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Jiemei Sun
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Daoshuang Li
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Yuchen Wang
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Kai Wang
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Lili Yang
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Lina Wu
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Xilin Sun
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| |
Collapse
|