1
|
Hasan S, Awasthi P, Malik S, Dwivedi M. Immunotherapeutic strategies to induce inflection in the immune response: therapy for cancer and COVID-19. Biotechnol Genet Eng Rev 2024; 40:3571-3610. [PMID: 36411974 DOI: 10.1080/02648725.2022.2147661] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 10/11/2022] [Indexed: 11/23/2022]
Abstract
Cancer has agonized the human race for millions of years. The present decade witnesses biological therapeutics to combat cancer effectively. Cancer Immunotherapy involves the use of therapeutics for manipulation of the immune system by immune agents like cytokines, vaccines, and transfection agents. Recently, this therapeutic approach has got vast attention due to the current pandemic COVID-19 and has been very effective. Concerning cancer, immunotherapy is based on the activation of the host's antitumor response by enhancing effector cell number and the production of soluble mediators, thereby reducing the host's suppressor mechanisms by induction of a tumour killing environment and by modulating immune checkpoints. In the present era, immunotherapies have gained traction and momentum as a pedestal of cancer treatment, improving the prognosis of many patients with a wide variety of haematological and solid malignancies. Food supplements, natural immunomodulatory drugs, and phytochemicals, with recent developments, have shown positive trends in cancer treatment by improving the immune system. The current review presents the systematic studies on major immunotherapeutics and their development for the effective treatment of cancers as well as in COVID-19. The focus of the review is to highlight comparative analytics of existing and novel immunotherapies in cancers, concerning immunomodulatory drugs and natural immunosuppressants, including immunotherapy in COVID-19 patients.
Collapse
Affiliation(s)
- Saba Hasan
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - Prankur Awasthi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University, Ranchi, Jharkhand, India
| | - Manish Dwivedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| |
Collapse
|
2
|
Baumann Z, Wiethe C, Vecchi CM, Richina V, Lopes T, Bentires-Alj M. Optimized full-spectrum flow cytometry panel for deep immunophenotyping of murine lungs. CELL REPORTS METHODS 2024:100885. [PMID: 39481389 DOI: 10.1016/j.crmeth.2024.100885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 07/22/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024]
Abstract
The lung immune system consists of both resident and circulating immune cells that communicate intricately. The immune system is activated by exposure to bacteria and viruses, when cancer initiates in the lung (primary lung cancer), or when metastases of other cancer types, including breast cancer, spread to and develop in the lung (secondary lung cancer). Thus, in these pathological situations, a comprehensive and quantitative assessment of changes in the lung immune system is of paramount importance for understanding mechanisms of infectious diseases, lung cancer, and metastasis but also for developing efficacious treatments. Unfortunately, lung tissue exhibits high autofluorescence, and this high background signal makes high-parameter flow cytometry analysis complicated. Here, we provide an optimized 30-parameter antibody panel for the analysis of all major immune cell types and states in normal and metastatic murine lungs using spectral flow cytometry.
Collapse
Affiliation(s)
- Zora Baumann
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; Department of Surgery, University Hospital Basel, 4031 Basel, Switzerland
| | | | - Cinja M Vecchi
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; Department of Surgery, University Hospital Basel, 4031 Basel, Switzerland
| | - Veronica Richina
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; Department of Surgery, University Hospital Basel, 4031 Basel, Switzerland
| | - Telma Lopes
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Mohamed Bentires-Alj
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; Department of Surgery, University Hospital Basel, 4031 Basel, Switzerland.
| |
Collapse
|
3
|
Wang H, Li J, Yu K, Lu Y, Ma M, Li Y. The cellular localization and oncogenic or tumor suppressive effects of angiomiotin-like protein 2 in tumor and normal cells. IUBMB Life 2024; 76:764-779. [PMID: 38717123 DOI: 10.1002/iub.2830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/10/2024] [Indexed: 10/19/2024]
Abstract
Angiomiotin (AMOT) family comprises three members: AMOT, AMOT-like protein 1 (AMOTL1), and AMOT-like protein 2 (AMOTL2). AMOTL2 is widely expressed in endothelial cells, epithelial cells, and various cancer cells. Specifically, AMOTL2 predominantly localizes in the cytoplasm and nucleus in human normal cells, whereas associates with cell-cell junctions and actin cytoskeleton in non-human cells, and locates at cell junctions or within the recycling endosomes in cancer cells. AMOTL2 is implicated in regulation of tube formation, cell polarity, and shape, although the specific impact on tumorigenesis remains to be conclusively determined. It has been shown that AMOTL2 enhances tumor growth and metastasis in pancreatic, breast, and colon cancer, however inhibits cell proliferation and migration in lung, hepatocellular cancer, and glioblastoma. In addition to its role in cell shape and cytoskeletal dynamics through co-localization with F-actin, AMOTL2 modulates the transcription of Yes-associated protein (YAP) by binding to it, thereby affecting its phosphorylation and cellular sequestration. Furthermore, the stability and cellular localization of AMOTL2, influenced by its phosphorylation and ubiquitination mediated by specific proteins, affects its cellular function. Additionally, we observe that AMOTL2 is predominantly downregulated in some tumors, but significantly elevated in colorectal adenocarcinoma (COAD). Moreover, overall analysis, GSEA and ROC curve analysis indicate that AMOTL2 exerts as an oncogenic protein in COAD by modulating Wnt pathway, participating in synthesis of collagen formation, and interacting with extracellular matrix receptor. In addition, AMOTL2 potentially regulates the distribution of immune cells infiltration in COAD. In summary, AMOTL2 probably functions as an oncogene in COAD. Consequently, further in-depth mechanistic research is required to elucidate the precise roles of AMOTL2 in various cancers.
Collapse
Affiliation(s)
- Huizhen Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jing Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Kexun Yu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yida Lu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mengdi Ma
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yongxiang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
4
|
Zhang C, Li J, Xu D, Wang D, Qi M, Chen F, Wu B, Deng K, Huang S. Surface Molecularly Engineered Mitochondria Conduct Immunophenotype Repolarization of Tumor-Associated Macrophages to Potentiate Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403044. [PMID: 39119940 PMCID: PMC11481252 DOI: 10.1002/advs.202403044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/25/2024] [Indexed: 08/10/2024]
Abstract
Reprogramming tumor-associated macrophages (TAMs) to an inflammatory phenotype effectively increases the potential of immune checkpoint blockade (ICB) therapy. Artificial mitochondrial transplantation, an emerging and safe strategy, has made brilliant achievements in regulating the function of recipient cells in preclinic and clinic, but its performance in reprogramming the immunophenotype of TAMs has not been reported. Here, the metabolism of M2 TAMs is proposed resetting from oxidative phosphorylation (OXPHOS) to glycolysis for polarizing M1 TAMs through targeted transplantation of mannosylated mitochondria (mPEI/M1mt). Mitochondria isolated from M1 macrophages are coated with mannosylated polyethyleneimine (mPEI) through electrostatic interaction to form mPEI/M1mt, which can be targeted uptake by M2 macrophages expressed a high level of mannose receptors. Mechanistically, mPEI/M1mt accelerates phosphorylation of NF-κB p65, MAPK p38 and JNK by glycolysis-mediated elevation of intracellular ROS, thus prompting M1 macrophage polarization. In vivo, the transplantation of mPEI/M1mt excellently potentiates therapeutic effects of anti-PD-L1 by resetting an antitumor proinflammatory tumor microenvironment and stimulating CD8 and CD4 T cells dependent immune response. Altogether, this work provides a novel platform for improving cancer immunotherapy, meanwhile, broadens the scope of mitochondrial transplantation technology in clinics in the future.
Collapse
Affiliation(s)
- Cai‐Ju Zhang
- Department of Radiology, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhan430071China
- Department of RadiologyHainan Hospital Affiliated to Hainan Medical UniversityHainan570311China
| | - Jia‐Mi Li
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of ChemistryWuhan UniversityWuhan430072China
- Department of RadiologyRenmin Hospital of Wuhan UniversityJiefang Road 238,Wuchang DistrictWuhanHubei430060China
| | - Dan Xu
- Department of Nuclear Medicine, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhan430071China
| | - Dan‐Dan Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of ChemistryWuhan UniversityWuhan430072China
| | - Ming‐Hui Qi
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of ChemistryWuhan UniversityWuhan430072China
| | - Feng Chen
- Department of RadiologyHainan Hospital Affiliated to Hainan Medical UniversityHainan570311China
| | - Bo Wu
- Department of Radiology, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhan430071China
| | - Kai Deng
- Department of Radiology, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhan430071China
| | - Shi‐Wen Huang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of ChemistryWuhan UniversityWuhan430072China
- Department of Orthopedic Trauma and MicrosurgeryZhongnan Hospital of Wuhan UniversityWuhan430071China
| |
Collapse
|
5
|
Li JB, Guo SS, Liu T, Lin ZC, Gong WJ, Tang LQ, Guo L, Mo HY, Mai HQ, Chen QY. Joint modeling of longitudinal health-related quality of life during concurrent chemoradiotherapy period and long-term survival among patients with advanced nasopharyngeal carcinoma. Radiat Oncol 2024; 19:125. [PMID: 39304905 DOI: 10.1186/s13014-024-02473-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 06/16/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND To investigate the prognosis of longitudinal health-related quality of life (HRQOL) during concurrent chemoradiotherapy (CCRT) on survival outcomes in patients with advanced nasopharyngeal carcinoma (NPC). METHODS During 2012-2014, 145 adult NPC patients with stage II-IVb NPC were investigated weekly using the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire core 30 (EORCT QLQ-C30) during their CCRT period. The effects of longitudinal trends of HRQOL on survival outcomes were estimated using joint modeling, and hazard ratios (HRs) with 95% confidence intervals (95% CIs) were reported as a 10-point increase in HRQOL scores. RESULTS After a median follow-up of 83.4 months, the multivariable models showed significant associations of longitudinal increasing scores in fatigue and appetite loss during the CCRT period with distant metastasis-free survival: 10-point increases in scores of fatigue and appetite loss domains during CCRT period were significantly associated with 75% (HR: 1.75, 95% CI: 1.01, 3.02; p = 0.047) and 59% (HR: 1.59, 95% CI: 1.09, 2.59; p = 0.018) increase in the risk of distant metastasis, respectively. The prognostic effects of the longitudinal HRQOL trend on overall survival and progress-free survival were statistically non-significant. CONCLUSION Increases in fatigue and appetite loss of HRQOL during the CCRT period are significantly associated with high risks of distant metastasis in advanced NPC patients. Nutritional support and psychological intervention are warranted for NPC patients during the treatment period.
Collapse
Grants
- No.81803105, No.81425018, No.81672868, No.82002852 and No.81802775 National Natural Science Foundation of China
- No.81803105, No.81425018, No.81672868, No.82002852 and No.81802775 National Natural Science Foundation of China
- No.81803105, No.81425018, No.81672868, No.82002852 and No.81802775 National Natural Science Foundation of China
- No.81803105, No.81425018, No.81672868, No.82002852 and No.81802775 National Natural Science Foundation of China
- No.81803105, No.81425018, No.81672868, No.82002852 and No.81802775 National Natural Science Foundation of China
- No.81803105, No.81425018, No.81672868, No.82002852 and No.81802775 National Natural Science Foundation of China
- No. 2018A030310238, No.2017A030312003 Natural Science Foundation of Guangdong Province
- No. A2018201 Medical Science and Technology Research Fund of Guangdong Province
- 2022YFC2705005 National Key Research and Development Program of China
Collapse
Affiliation(s)
- Ji-Bin Li
- Department of Clinical Research, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Shan-Shan Guo
- Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, 651 Dong Feng East Road, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Ting Liu
- Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, 651 Dong Feng East Road, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Zhuo-Chen Lin
- Department of Medical Records, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Wei-Jie Gong
- Department of General Practice, Health Science Center, Shenzhen University, Shenzhen, 518037, People's Republic of China
| | - Lin-Quan Tang
- Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, 651 Dong Feng East Road, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Ling Guo
- Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, 651 Dong Feng East Road, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Hao-Yuan Mo
- Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, 651 Dong Feng East Road, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Hai-Qiang Mai
- Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, 651 Dong Feng East Road, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Qiu-Yan Chen
- Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, 651 Dong Feng East Road, Guangzhou, 510060, People's Republic of China.
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
6
|
Li X, Hu Y, Zhang X, Shi X, Parak WJ, Pich A. Transvascular transport of nanocarriers for tumor delivery. Nat Commun 2024; 15:8172. [PMID: 39289401 PMCID: PMC11408679 DOI: 10.1038/s41467-024-52416-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 09/05/2024] [Indexed: 09/19/2024] Open
Abstract
Nanocarriers (NCs) play a crucial role in delivering theranostic agents to tumors, making them a pivotal focus of research. However, the persistently low delivery efficiency of engineered NCs has been a significant challenge in the advancement of nanomedicine, stirring considerable debate. Transvascular transport is a critical pathway for NC delivery from vessels to tumors, yet a comprehensive understanding of the interactions between NCs and vascular systems remains elusive. In recent years, considerable efforts have been invested in elucidating the transvascular transport mechanisms of NCs, leading to promising advancements in tumor delivery and theranostics. In this context, we highlight various delivery mechanisms, including the enhanced permeability and retention effect, cooperative immune-driven effect, active transcytosis, and cell/bacteria-mediated delivery. Furthermore, we explore corresponding strategies aimed at enhancing transvascular transport of NCs for efficient tumor delivery. These approaches offer intriguing solutions spanning physicochemical, biological, and pharmacological domains to improve delivery and therapeutic outcomes. Additionally, we propose a forward-looking delivery framework that relies on advanced tumor/vessel models, high-throughput NC libraries, nano-bio interaction datasets, and artificial intelligence, which aims to guide the design of next-generation carriers and implementation strategies for optimized delivery.
Collapse
Affiliation(s)
- Xin Li
- DWI-Leibniz-Institute for Interactive Materials, Aachen, 52056, Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen, 52074, Germany
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, 201804, China
| | - Yong Hu
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, 201804, China
| | - Xingcai Zhang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA.
| | - Xiangyang Shi
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Wolfgang J Parak
- Center for Hybrid Nanostructures (CHyN), University of Hamburg, Hamburg, 20607, Germany.
| | - Andrij Pich
- DWI-Leibniz-Institute for Interactive Materials, Aachen, 52056, Germany.
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen, 52074, Germany.
- Aachen Maastricht Institute for Biobased Materials, Maastricht University, RD Geleen, 6167, The Netherlands.
| |
Collapse
|
7
|
Shao M, Gao Y, Xu X, Chan DW, Du J. Exosomes: Key Factors in Ovarian Cancer Peritoneal Metastasis and Drug Resistance. Biomolecules 2024; 14:1099. [PMID: 39334866 PMCID: PMC11430201 DOI: 10.3390/biom14091099] [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: 08/03/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Ovarian cancer remains a leading cause of death among gynecological cancers, largely due to its propensity for peritoneal metastasis and the development of drug resistance. This review concentrates on the molecular underpinnings of these two critical challenges. We delve into the role of exosomes, the nano-sized vesicles integral to cellular communication, in orchestrating the complex interactions within the tumor microenvironment that facilitate metastatic spread and thwart therapeutic efforts. Specifically, we explore how exosomes drive peritoneal metastasis by promoting epithelial-mesenchymal transition in peritoneal mesothelial cells, altering the extracellular matrix, and supporting angiogenesis, which collectively enable the dissemination of cancer cells across the peritoneal cavity. Furthermore, we dissect the mechanisms by which exosomes contribute to the emergence of drug resistance, including the sequestration and expulsion of chemotherapeutic agents, the horizontal transfer of drug resistance genes, and the modulation of critical DNA repair and apoptotic pathways. By shedding light on these exosome-mediated processes, we underscore the potential of exosomal pathways as novel therapeutic targets, offering hope for more effective interventions against ovarian cancer's relentless progression.
Collapse
Affiliation(s)
- Ming Shao
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
- Department of Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen 518172, China
| | - Yunran Gao
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, China
| | - Xiling Xu
- School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, China
| | - David Wai Chan
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Juan Du
- Department of Central Laboratory, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen 518172, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| |
Collapse
|
8
|
Zhang Y, Ding X, Zhang X, Li Y, Xu R, Li HJ, Zuo D, Chen G. Unveiling the contribution of tumor-associated macrophages in driving epithelial-mesenchymal transition: a review of mechanisms and therapeutic Strategies. Front Pharmacol 2024; 15:1404687. [PMID: 39286635 PMCID: PMC11402718 DOI: 10.3389/fphar.2024.1404687] [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: 03/21/2024] [Accepted: 08/15/2024] [Indexed: 09/19/2024] Open
Abstract
Tumor-associated macrophages (TAMs), fundamental constituents of the tumor microenvironment (TME), significantly influence cancer development, primarily by promoting epithelial-mesenchymal transition (EMT). EMT endows cancer cells with increased motility, invasiveness, and resistance to therapies, marking a pivotal juncture in cancer progression. The review begins with a detailed exposition on the origins of TAMs and their functional heterogeneity, providing a foundational understanding of TAM characteristics. Next, it delves into the specific molecular mechanisms through which TAMs induce EMT, including cytokines, chemokines and stromal cross-talking. Following this, the review explores TAM-induced EMT features in select cancer types with notable EMT characteristics, highlighting recent insights and the impact of TAMs on cancer progression. Finally, the review concludes with a discussion of potential therapeutic targets and strategies aimed at mitigating TAM infiltration and disrupting the EMT signaling network, thereby underscoring the potential of emerging treatments to combat TAM-mediated EMT in cancer. This comprehensive analysis reaffirms the necessity for continued exploration into TAMs' regulatory roles within cancer biology to refine therapeutic approaches and improve patient outcomes.
Collapse
Affiliation(s)
- Yijia Zhang
- Department of Pharmacy, Taizhou Second People's Hospital (Mental Health Center affiliated to Taizhou University School of Medicine), Taizhou University, Taizhou, Zhejiang, China
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaofei Ding
- Department of Pharmacology, Taizhou University, Taizhou, Zhejiang, China
| | - Xue Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Ye Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Rui Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Hai-Jun Li
- Department of Pharmacy, Taizhou Second People's Hospital (Mental Health Center affiliated to Taizhou University School of Medicine), Taizhou University, Taizhou, Zhejiang, China
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Guang Chen
- Department of Pharmacy, Taizhou Second People's Hospital (Mental Health Center affiliated to Taizhou University School of Medicine), Taizhou University, Taizhou, Zhejiang, China
- Department of Pharmacology, Taizhou University, Taizhou, Zhejiang, China
| |
Collapse
|
9
|
Kang K, Lin X, Chen P, Liu H, Liu F, Xiong W, Li G, Yi M, Li X, Wang H, Xiang B. T cell exhaustion in human cancers. Biochim Biophys Acta Rev Cancer 2024; 1879:189162. [PMID: 39089484 DOI: 10.1016/j.bbcan.2024.189162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
T cell exhaustion refers to a progressive state in which T cells become functionally impaired due to sustained antigenic stimulation, which is characterized by increased expression of immune inhibitory receptors, but weakened effector functions, reduced self-renewal capacity, altered epigenetics, transcriptional programme and metabolism. T cell exhaustion is one of the major causes leading to immune escape of cancer, creating an environment that supports tumor development and metastatic spread. In addition, T cell exhaustion plays a pivotal role to the efficacy of current immunotherapies for cancer. This review aims to provide a comprehensive view of roles of T cell exhaustion in cancer development and progression. We summerized the regulatory mechanisms that involved in T cell exhaustion, including transcription factors, epigenetic and metabolic reprogramming events, and various microenvironmental factors such as cytokines, microorganisms, and tumor autocrine substances. The paper also discussed the challenges posed by T cell exhaustion to cancer immunotherapies, including immune checkpoint blockade (ICB) therapies and chimeric antigen receptor T cell (CAR-T) therapy, highlightsing the obstacles encountered in ICB therapies and CAR-T therapies due to T cell exhaustion. Finally, the article provides an overview of current therapeutic options aimed to reversing or alleviating T cell exhaustion in ICB and CAR-T therapies. These therapeutic approaches seek to overcome T cell exhaustion and enhance the effectiveness of immunotherapies in treating tumors.
Collapse
Affiliation(s)
- Kuan Kang
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China
| | - Xin Lin
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China
| | - Pan Chen
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China
| | - Huai Liu
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Feng Liu
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Wei Xiong
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China
| | - Guiyuan Li
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China
| | - Mei Yi
- Department of Dermatology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Infammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China.
| | - Hui Wang
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China.
| | - Bo Xiang
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China; FuRong Laboratory, Changsha 410078, Hunan, China.
| |
Collapse
|
10
|
Lim RMH, Lee JY, Kannan B, Ko TK, Chan JY. Molecular and immune pathobiology of human angiosarcoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189159. [PMID: 39032539 DOI: 10.1016/j.bbcan.2024.189159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Angiosarcoma is a rare endothelial-derived malignancy that is extremely diverse in anatomy, aetiology, molecular and immune characteristics. While novel therapeutic approaches incorporating targeted agents and immunotherapy have yielded significant improvements in patient outcomes across several cancers, their impact on angiosarcoma remains modest. Contributed by its heterogeneous nature, there is currently a lack of novel drug targets in this disease entity and no reliable biomarkers that predict response to conventional treatment. This review aims to examine the molecular and immune landscape of angiosarcoma in association with its aetiology, anatomical sites, prognosis and therapeutic options. We summarise current efforts to characterise angiosarcoma subtypes based on molecular and immune profiling. Finally, we highlight promising technologies such as single-cell spatial "omics" that may further our understanding of angiosarcoma and propose strategies that can be similarly applied for the study of other rare cancers.
Collapse
Affiliation(s)
| | - Jing Yi Lee
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, Singapore
| | - Bavani Kannan
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore
| | - Tun Kiat Ko
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore
| | - Jason Yongsheng Chan
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, Singapore; Division of Medical Oncology, National Cancer Centre Singapore, Singapore.
| |
Collapse
|
11
|
Cao Y, Zhou Z, He S, Liu W. TTYH3 promotes the malignant progression of oral squamous cell carcinoma SCC-9 cells by regulating tumor-associated macrophage polarization. Arch Oral Biol 2024; 165:106028. [PMID: 38908074 DOI: 10.1016/j.archoralbio.2024.106028] [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: 03/05/2024] [Revised: 06/04/2024] [Accepted: 06/08/2024] [Indexed: 06/24/2024]
Abstract
OBJECTIVE This study was designed to investigate the biological role and the reaction mechanism of Tweety family member 3 (TTYH3) in oral squamous cell carcinoma (OSCC). DESIGN The mRNA and protein expressions of TTYH3 were assessed with RT-qPCR and western blot. After silencing TTYH3 expression, the proliferation of OSCC cells were detected using cell counting kit-8 (CCK-8) assay, 5-ethynyl-2'-deoxyuridine (EdU) staining and colony formation assay. Cell migration and invasion were detected using wound healing and transwell. Gelatin zymography protease assay was used to detect matrix metalloproteinase-2 (MMP2) and matrix metalloproteinase-2 (MMP9) activity and western blot was used to detect the expressions of proteins associated with proliferation and epithelial-mesenchymal transition (EMT). The mRNA expression of TTYH3 in THP-1-derived macrophage was detected using real-time reverse transcriptase-polymerase chain reaction (RT-qPCR). The number of CD86-positive cells and CD206-positive cells was detected using immunofluorescence assay. RT-qPCR was used to detect the expressions of M2 markers arginase 1 (ARG1), chitinase-like 3 (YM1) and mannose receptor C-type 1 (MRC1). RESULTS In this study, it was found that TTYH3 expression was upregulated in OSCC cell lines and TTYH3 knockdown could inhibit the proliferation, migration, invasion and EMT process in OSCC via suppressing M2 polarization of tumor-associated macrophages. CONCLUSIONS Collectively, TTYH3 facilitated the progression of OSCC through the regulation of tumor-associated macrophages polarization.
Collapse
Affiliation(s)
- Yuhui Cao
- Department of Stomatology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325088, China
| | - Zhihui Zhou
- Department of Stomatology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325088, China
| | - Shuai He
- Department of Stomatology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325088, China
| | - Wenhui Liu
- Department of Stomatology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325088, China.
| |
Collapse
|
12
|
Liu Q, Xu J, Dai B, Guo D, Sun C, Du X. Single-cell resolution profiling of the immune microenvironment in primary and metastatic nasopharyngeal carcinoma. J Cancer Res Clin Oncol 2024; 150:391. [PMID: 39158776 PMCID: PMC11333513 DOI: 10.1007/s00432-024-05900-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/17/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is an assertive malignancy with partially understood underlying mechanisms, urging further study into its diverse and dynamic tumor microenvironment (TME) to bolster diagnosis, treatment, and prognostic accuracy. AIMS To track the evolutionary route of metastasis, here we perform a yielding scRNA-seq data from 24 primary carcinoma, 7 peripheral blood mononuclear cell (PBMC) nasopharyngeal carcinoma, and 7 metastatic carcinoma patients' sample. MATERIALS AND METHODS Following high quality control and filtration, a total of 292,298 cells from these tumors were classified into 10 clusters: T cells, B cells, Macrophages/Monocytes, Natural Killer (NK) cells, Plasma cells, plasmacytoid Dendritic Cells, Migratory Dendritic Cells, Mast cells, Cancer-Associated Fibroblasts, and Epithelial cells. RESULTS By comparing and analyzing the different functional capacities of cellular entities within primary and metastatic nasopharyngeal carcinoma, coupled with a detailed investigation into the heterogeneity and differential fate trajectories of T cells, B cells, and myeloid cells, as well as assessing the interactions of cell-cell communicative heterogeneity between these carcinogenic states, we established single-cell atlases for primary and metastatic tumors and identified a large number of potential therapeutic targets. CONCLUSION This comprehensive analysis significantly advances our understanding of nasopharyngeal carcinoma (NPC) metastasis by detailing the evolutionary dynamics and the impact of the tumor microenvironment at a single-cell resolution, thereby laying a crucial foundation for future metastatic tumor research and providing new insights into immune heterogeneity, molecular interactions, and potential therapeutic strategies for NPC.
Collapse
Affiliation(s)
- Qiuping Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, People's Republic of China
| | - Jingping Xu
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, People's Republic of China
| | - Bingyi Dai
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, People's Republic of China
| | - Danni Guo
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, People's Republic of China
| | - Changling Sun
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, People's Republic of China
| | - Xiaodong Du
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu Province, People's Republic of China.
| |
Collapse
|
13
|
Zhao J, Zhang K, Sui D, Wang S, Li Y, Tang X, Liu X, Song Y, Deng Y. Recent advances in sialic acid-based active targeting chemoimmunotherapy promoting tumor shedding: a systematic review. NANOSCALE 2024; 16:14621-14639. [PMID: 39023195 DOI: 10.1039/d4nr01740d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Tumors have always been a major public health concern worldwide, and attempts to look for effective treatments have never ceased. Sialic acid is known to be a crucial element for tumor development and its receptors are highly expressed on tumor-associated immune cells, which perform significant roles in establishing the immunosuppressive tumor microenvironment and further boosting tumorigenesis, progression, and metastasis. Obviously, it is essential to consider sophisticated crosstalk between tumors, the immune system, and preparations, and understand the links between pharmaceutics and immunology. Sialic acid-based chemoimmunotherapy enables active targeting drug delivery via mediating the recognition between the sialic acid-modified nano-drug delivery system represented by liposomes and sialic acid-binding receptors on tumor-associated immune cells, which inhibit their activity and utilize their homing ability to deliver drugs. Such a "Trojan horse" strategy has remarkably improved the shortcomings of traditional passive targeting treatments, unexpectedly promoted tumor shedding, and persistently induced robust immunological memory, thus highlighting its prospective application potential for targeting various tumors. Herein, we review recent advances in sialic acid-based active targeting chemoimmunotherapy to promote tumor shedding, summarize the current viewpoints on the tumor shedding mechanism, especially the formation of durable immunological memory, and analyze the challenges and opportunities of this attractive approach.
Collapse
Affiliation(s)
- Jingyi Zhao
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Kunfeng Zhang
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Dezhi Sui
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Shuo Wang
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Yantong Li
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Xueying Tang
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Xinrong Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| |
Collapse
|
14
|
Santos RLD, Martins MR, Tavares VL, Neto JPD, Torres LC. Analysis of the expression of cytokines and chemokines, platelet-leukocyte aggregates, sCD40L and sCD62P in cutaneous melanoma. J Surg Oncol 2024. [PMID: 39129330 DOI: 10.1002/jso.27748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/14/2024] [Accepted: 05/22/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Cutaneous melanoma (CM) is a malignancy with a variable incidence worldwide and a poor advanced-stage prognosis. Melanoma growth is closely associated with the immune system. METHODS A cross-sectional study was performed on CM patients admitted at the Hospital de Cancer de Pernambuco (HCP) between 2015 and 2018. Fifty-one CM patients were included, and 30 healthy individuals. The study aimed to evaluate the association of platelet activation mechanisms and inflammatory response in patients with cutaneous melanoma. RESULTS Elevated serum IL10 and low serum TNF levels in CM patients compared to controls (p < 0.05). High IL6 levels in patients with negative lymph nodes LN (-) compared to positive lymph nodes group (LN +, p = 0.0005). Low RANTES levels in patients compared to controls (p < 0.05). Elevated levels of platelet-lymphocyte (PLA), platelet-monocytes (PMA), and platelet-neutrophils (PNA) aggregates were observed in patients compared to controls (p < 0.05). CM patients with stage II had lower PMA levels than stages I and III (p < 0.05). High PMA levels were observed in patients with LN (+) compared to the LN (-) group (p < 0.0001). Patients with SSM had high levels of sCD40L and sCD62P compared to controls (p < 0.05)). High sCD40L levels in stage II compared to the stage III group, and sCD62P in stages I and II compared to the stage III group (p < 0.05). High sCD62P levels in patients with LN (-) compared to the group LN (+) (p < 0.05). CONCLUSION It was observed the immunosuppressive profile in CM may favor tumor progression. High levels of platelet-leukocyte aggregates, sCD40L, and sCD62P may be associated with the worst prognosis.
Collapse
Affiliation(s)
- Rogerio Luiz Dos Santos
- Translational Research Laboratory Prof. CA Hart (IMIP), Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Hospital de Câncer de Pernambuco, Recife, Brazil
- Skin Cancer Department, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Mário R Martins
- Translational Research Laboratory Prof. CA Hart (IMIP), Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Hospital de Câncer de Pernambuco, Recife, Brazil
- Skin Cancer Department, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Valéria Lobo Tavares
- Translational Research Laboratory Prof. CA Hart (IMIP), Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Hospital de Câncer de Pernambuco, Recife, Brazil
| | | | - Leuridan Cavalcante Torres
- Translational Research Laboratory Prof. CA Hart (IMIP), Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, Brazil
- Hospital de Câncer de Pernambuco, Recife, Brazil
- Postgraduate program in Translational Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| |
Collapse
|
15
|
Zhang H, Zhang Y, Zhang Y, Li H, Ou M, Yu Y, Zhang F, Yin H, Mao Z, Mei L. Catalytic activity of violet phosphorus-based nanosystems and the role of metabolites in tumor therapy. Nat Commun 2024; 15:6783. [PMID: 39117634 PMCID: PMC11310355 DOI: 10.1038/s41467-024-50769-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 07/22/2024] [Indexed: 08/10/2024] Open
Abstract
Although nanocatalytic medicine has demonstrated its advantages in tumor therapy, the outcomes heavily relie on substrate concentration and the metabolic pathways are still indistinct. We discover that violet phosphorus quantum dots (VPQDs) can catalyze the production of reactive oxygen species (ROS) without requiring external stimuli and the catalytic substrates are confirmed to be oxygen (O2) and hydrogen peroxide (H2O2) through the computational simulation and experiments. Considering the short of O2 and H2O2 at the tumor site, we utilize calcium peroxide (CaO2) to supply catalytic substrates for VPQDs and construct nanoparticles together with them, named VPCaNPs. VPCaNPs can induce oxidative stress in tumor cells, particularly characterized by a significant increase in hydroxyl radicals and superoxide radicals, which cause substantial damage to the structure and function of cells, ultimately leading to cell apoptosis. Intriguingly, O2 provided by CaO2 can degrade VPQDs slowly, and the degradation product, phosphate, as well as CaO2-generated calcium ions, can promote tumor calcification. Antitumor immune activation and less metastasis are also observed in VPCaNPs administrated animals. In conclusion, our study unveils the anti-tumor activity of VPQDs as catalysts for generating cytotoxic ROS and the degradation products can promote tumor calcification, providing a promising strategy for treating tumors.
Collapse
Affiliation(s)
- Hanjie Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, PR China
| | - Yitong Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, PR China
| | - Yushi Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, PR China
| | - Hanyue Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, PR China
| | - Meitong Ou
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, PR China
| | - Yongkang Yu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Fan Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, PR China
| | - Huijuan Yin
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, PR China
- Integrative regeneration laboratory, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, PR China
| | - Zhuo Mao
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, PR China.
| | - Lin Mei
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, PR China.
| |
Collapse
|
16
|
Deng M, Chen X, Qiu J, Liu G, Huang C. A Neural Network-Based Scoring System for Predicting Prognosis and Therapy in Breast Cancer. Curr Protoc 2024; 4:e1122. [PMID: 39166828 DOI: 10.1002/cpz1.1122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Breast cancer is a prevalent malignancy affecting women worldwide. Currently, there are no precise molecular biomarkers with immense potential for accurately predicting breast cancer development, which limits clinical management options. Recent evidence has highlighted the importance of metastatic and tumor-infiltrating immune cells in modulating the antitumor therapy response. However, the prognostic value of using these features in combination, and their potential for guiding individualized treatment for breast cancer, remains vague. To address this challenge, we recently developed the metastatic and immunogenomic risk score (MIRS), a comprehensive and user-friendly scoring system that leverages advanced bioinformatics methods to facilitate transcriptomics data analysis. To help users become familiar with the MIRS tool and apply it effectively in analyzing new breast cancer datasets, we describe detailed protocols that require no advanced programming skills. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Calculating a MIRS score from transcriptomics data Basic Protocol 2: Predicting clinical outcomes from MIRS scores Basic Protocol 3: Evaluating treatment responses and guiding therapeutic strategies in breast cancer patients Basic Protocol 4: Guidelines for utilizing the MIRS webtool.
Collapse
Affiliation(s)
- Min Deng
- MOE Frontier Science Center for Precision Oncology, Cancer Center, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Xinyu Chen
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, China
| | - Jiayue Qiu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, China
| | - Guiyou Liu
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Chen Huang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, China
| |
Collapse
|
17
|
Yang Y, Ge J, Zhong X, Liu L, Chen L, Lu S, Ren J, Chen Y, Sun S, Song Z, Cheng Y, Cheng L. Turning Waste into Wealth: A Potent Sono-Immune Strategy Based on Microcystis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2401974. [PMID: 38889229 DOI: 10.1002/adma.202401974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/16/2024] [Indexed: 06/20/2024]
Abstract
Currently, sonodynamic therapy (SDT) has limited therapeutic outcomes and immune responses, highlighting the urgent need for enhanced strategies that can stimulate robust and long-lasting antitumor effects. Microcystis, a notorious microalga, reveals the possibility of mediating SDT owing to the presence of gas vesicles (GVs) and phycocyanin (PC). Herein, a nontoxic strain of Microcystis elabens (labeled Me) is developed as a novel agent for SDT because it generates O2 under red light (RL) illumination, while GVs and PC act as cavitation nuclei and sonosensitizers, respectively. Moreover, algal debris is released after ultrasound (US) irradiation, which primes the Toll-like receptor pathway to initiate a cascade of immune responses. This sono-immune strategy inhibits CT26 colon tumor growth largely by promoting dendritic cell (DC) maturation and cytotoxic T-cell activation. After combination with the immune checkpoint blockade (ICB), the therapeutic outcome is further amplified, accompanied by satisfactory abscopal and immune memory effects; the similar potency is proven in the "cold" 4T1 triple-negative breast tumor. In addition, Me exhibits good biosafety without significant acute or chronic toxicity. Briefly, this study turns waste into wealth by introducing sono-immunotherapy based on Microcystis that achieved encouraging therapeutic effects on cancer, which is expected to be translated into the clinic.
Collapse
Affiliation(s)
- Yuqi Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
- Monash Suzhou Research Institute, Monash University, Suzhou, 215000, China
- Department of Materials Science and Engineering, Monash University, Clayton, VIC3800, Australia
| | - Jun Ge
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Xiaoyan Zhong
- Department of Toxicology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Luyao Liu
- Department of Toxicology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Linfu Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Shunyi Lu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Jiacheng Ren
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Youdong Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Shumin Sun
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Zhuorun Song
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Yuan Cheng
- Monash Suzhou Research Institute, Monash University, Suzhou, 215000, China
- Department of Materials Science and Engineering, Monash University, Clayton, VIC3800, Australia
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| |
Collapse
|
18
|
Liu H, Huang M, Xin D, Wang H, Yu H, Pu W. Natural products with anti-tumorigenesis potential targeting macrophage. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 131:155794. [PMID: 38875811 DOI: 10.1016/j.phymed.2024.155794] [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: 04/07/2024] [Revised: 05/06/2024] [Accepted: 05/30/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND Inflammation is a risk factor for tumorigenesis. Macrophage, a subset of immune cells with high plasticity, plays a multifaceted role in this process. Natural products, which are bioactive compounds derived from traditional herbs or foods, have exhibited diverse effects on macrophages and tumorigenesis making them a valuable resource of drug discovery or optimization in tumor prevention. PURPOSE Provide a comprehensive overview of the various roles of macrophages in tumorigenesis, as well as the effects of natural products on tumorigenesis by modulating macrophage function. METHODS A thorough literature search spanning the past two decades was carried out using PubMed, Web of Science, Elsevier, and CNKI following the PRISMA guidelines. The search terms employed included "macrophage and tumorigenesis", "natural products, macrophages and tumorigenesis", "traditional Chinese medicine and tumorigenesis", "natural products and macrophage polarization", "macrophage and tumor related microenvironment", "macrophage and tumor signal pathway", "toxicity of natural products" and combinations thereof. Furthermore, certain articles are identified through the tracking of citations from other publications or by accessing the websites of relevant journals. Studies that meet the following criteria are excluded: (1) Articles not written in English or Chinese; (2) Full texts were not available; (3) Duplicate articles and irrelevant studies. The data collected was organized and summarized based on molecular mechanisms or compound structure. RESULTS This review elucidates the multifaceted effect of macrophages on tumorigenesis, encompassing process such as inflammation, angiogenesis, and tumor cell invasion by regulating metabolism, non-coding RNA, signal transduction and intercellular crosstalk. Natural products, including vitexin, ovatodiolide, ligustilide, and emodin, as well as herbal remedies, have demonstrated efficacy in modulating macrophage function, thereby attenuating tumorigenesis. These interventions mainly focus on mitigating the initial inflammatory response or modifying the inflammatory environment within the precancerous niche. CONCLUSIONS These mechanistic insights of macrophages in tumorigenesis offer valuable ideas for researchers. The identified natural products facilitate the selection of promising candidates for future cancer drug development.
Collapse
Affiliation(s)
- Hao Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Manru Huang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Dandan Xin
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Hong Wang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| | - Haiyang Yu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China.
| | - Weiling Pu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
19
|
Omidvar S, Vahedian V, Sourani Z, Yari D, Asadi M, Jafari N, Khodavirdilou L, Bagherieh M, Shirzad M, Hosseini V. The molecular crosstalk between innate immunity and DNA damage repair/response: Interactions and effects in cancers. Pathol Res Pract 2024; 260:155405. [PMID: 38981346 DOI: 10.1016/j.prp.2024.155405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 06/08/2024] [Accepted: 06/12/2024] [Indexed: 07/11/2024]
Abstract
DNA damage can lead to erroneous alterations and mutations which in turn can result into wide range of disease condition including aging, severe inflammation, and, most importantly, cancer. Due to the constant exposure to high-risk factors such as exogenous and endogenous DNA-damaging agents, cells may experience DNA damage impairing stability and integrity of the genome. These perturbations in DNA structure can arise from several mutations in the genome. Therefore, DNA Damage Repair/Response (DDR) detects and then corrects these potentially tumorigenic problems by inducing processes such as DNA repair, cell cycle arrest, apoptosis, etc. Additionally, DDR can activate signaling pathways related to immune system as a protective mechanism against genome damage. These protective machineries are ignited and spread through a network of molecules including DNA damage sensors, transducers, kinases and downstream effectors. In this review, we are going to discuss the molecular crosstalk between innate immune system and DDR, as well as their potential effects on cancer pathophysiology.
Collapse
Affiliation(s)
- Sahar Omidvar
- Cancer Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| | - Vahid Vahedian
- Department of Hematology, Transfusion Medicine and Cellular Therapy, Division of Hematology/Oncology, Clinical Hospital, Faculty of Medicine, University of Sao Paulo (FMUSP-HC), Sao Paulo, Brazil; Department of Clinical Medicine, Division of Medical Investigation Laboratory (LIM-31), Clinical Hospital, Faculty of Medicine, University of Sao Paulo (FMUSP-HC), Sao Paulo, Brazil; Comprehensive Center for Translational and Precision Oncology (CTO), SP State Cancer Institute (ICESP), Sao Paulo, Brazil.
| | - Zahra Sourani
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| | - Davood Yari
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| | - Mehrdad Asadi
- Department of Medical Laboratory Sciences and Microbiology, Faculty of Medical Sciences, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran.
| | - Negin Jafari
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Lida Khodavirdilou
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA.
| | - Molood Bagherieh
- Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran.
| | - Moein Shirzad
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| | - Vahid Hosseini
- Department of Medical Laboratory Sciences and Microbiology, Faculty of Medical Sciences, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran; Infectious Diseases Research Center, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran.
| |
Collapse
|
20
|
Ota G, Inoue R, Saito A, Kono Y, Kitayama J, Sata N, Horie H. Reduced Abundance of Phocaeicola in Mucosa-associated Microbiota Is Associated with Distal Colorectal Cancer Metastases Possibly through an Altered Local Immune Environment. J Anus Rectum Colon 2024; 8:235-245. [PMID: 39086872 PMCID: PMC11286368 DOI: 10.23922/jarc.2024-014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/17/2024] [Indexed: 08/02/2024] Open
Abstract
Objectives The aim of this study was to identify the microbiota whose decrease in tumor area was associated with the metastatic process of distal colorectal cancer (CRC). Methods Twenty-eight consecutive patients with distal CRC undergoing surgical resection in our hospital were enrolled. Microbiota in 28 specimens from surgically resected colorectal cancers were analyzed using 16S ribosomal ribonucleic acid gene amplicon sequencing and the relative abundance (RA) of microbiota was evaluated. The densities of tumor-infiltrating lymphocytes (TIL) and tumor associated macrophages (TAM) in the colorectal cancers were immunohistochemically evaluated. Results Phocaeicola was the most abundant microbiota in normal mucosa. The RA of Phocaeicola in tumor tissues tended to be lower than that in normal mucosa although the difference was not significant (p=0.0732). The RA of Phocaeicola at tumor sites did not correlate either with depth of tumor invasion (pT-stage) or tumor size, however they were significantly reduced in patients with nodal metastases (p<0.05) and those with distant metastases (p<0.001). The RA of Phocaeicola at tumor sites showed positive correlation with the densities of CD3(+) or CD8(+) TIL. Since P. vulgatus was the most dominant species (47%) of the Phocaeicola, the RA of P. vulgatus and CRC metastasis and its association with TIL and TAM were also investigated. P. vulgatus showed a similar trend to genus Phocaeicola but was not statistically significant. Conclusions A relative reduction of Phocaeicola attenuates the local anti-tumor immune response in distal CRC, which may facilitate metastatic spread.
Collapse
Affiliation(s)
- Gaku Ota
- Department of Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Ryo Inoue
- Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University, Hirakata, Japan
| | - Akira Saito
- Department of Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Yoshihiko Kono
- Department of Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Joji Kitayama
- Department of Surgery, Jichi Medical University, Shimotsuke, Japan
- Clinical Research Center, Division of Translational Research, Jichi Medical University, Shimotsuke, Japan
| | - Naohiro Sata
- Department of Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Hisanaga Horie
- Department of Surgery, Jichi Medical University, Shimotsuke, Japan
- Department of Operating Room Management, Jichi Medical University Hospital, Shimotsuke, Japan
| |
Collapse
|
21
|
Pratticò F, Garajová I. Focus on Pancreatic Cancer Microenvironment. Curr Oncol 2024; 31:4241-4260. [PMID: 39195299 DOI: 10.3390/curroncol31080316] [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: 06/16/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
Pancreatic ductal adenocarcinoma remains one of the most lethal solid tumors due to its local aggressiveness and metastatic potential, with a 5-year survival rate of only 13%. A robust connection between pancreatic cancer microenvironment and tumor progression exists, as well as resistance to current anticancer treatments. Pancreatic cancer has a complex tumor microenvironment, characterized by an intricate crosstalk between cancer cells, cancer-associated fibroblasts and immune cells. The complex composition of the tumor microenvironment is also reflected in the diversity of its acellular components, such as the extracellular matrix, cytokines, growth factors and secreted ligands involved in signaling pathways. Desmoplasia, the hallmark of the pancreatic cancer microenvironment, contributes by creating a dense and hypoxic environment that promotes further tumorigenesis, provides innate systemic resistance and suppresses anti-tumor immune invasion. We discuss the complex crosstalk among tumor microenvironment components and explore therapeutic strategies and opportunities in pancreatic cancer research. Better understanding of the tumor microenvironment and its influence on pancreatic cancer progression could lead to potential novel therapeutic options, such as integration of immunotherapy and cytokine-targeted treatments.
Collapse
Affiliation(s)
- Fabiana Pratticò
- Medical Oncology Unit, University Hospital of Parma, 43100 Parma, Italy
| | - Ingrid Garajová
- Medical Oncology Unit, University Hospital of Parma, 43100 Parma, Italy
| |
Collapse
|
22
|
Li M, Zhao X. LILRB4 in acute myeloid leukemia: From prognostic biomarker to immunotherapeutic target. Chin Med J (Engl) 2024:00029330-990000000-01138. [PMID: 38973293 DOI: 10.1097/cm9.0000000000003195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Indexed: 07/09/2024] Open
Abstract
ABSTRACT Leukocyte immunoglobulin-like receptor (LILR) B4 (also known as ILT3/CD85k) is an immune checkpoint protein that is highly expressed in solid tumors and hematological malignancies and plays a significant role in the pathophysiology of cancer. LILRB4 is highly expressed in acute myeloid leukemia (AML), and this phenotype is associated with adverse patient outcomes. Its differential expression in tumors compared to normal tissues, its presence in tumor stem cells, and its multifaceted roles in tumorigenesis position it as a promising therapeutic target in AML. Currently, several immunotherapies targeting LILRB4 are undergoing clinical trials. This review summarizes advancements made in the study of LILRB4 in AML, focusing on its structure, ligands, expression, and significance in normal tissues and AML; its protumorigenic effects and mechanisms in AML; and the application of LILRB4-targeted therapies in AML. These insights highlight the potential advantages of LILRB4 as an immunotherapeutic target in the context of AML.
Collapse
Affiliation(s)
- Muzi Li
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing 100044, China
| | | |
Collapse
|
23
|
Pan JJ, Xie SZ, Zheng X, Xu JF, Xu H, Yin RQ, Luo YL, Shen L, Chen ZR, Chen YR, Yu SZ, Lu L, Zhu WW, Lu M, Qin LX. Acetyl-CoA metabolic accumulation promotes hepatocellular carcinoma metastasis via enhancing CXCL1-dependent infiltration of tumor-associated neutrophils. Cancer Lett 2024; 592:216903. [PMID: 38670307 DOI: 10.1016/j.canlet.2024.216903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024]
Abstract
High levels of acetyl-CoA are considered a key metabolic feature of metastatic cancers. However, the impacts of acetyl-CoA metabolic accumulation on cancer microenvironment remodeling are poorly understood. In this study, using human hepatocellular carcinoma (HCC) tissues and orthotopic xenograft models, we found a close association between high acetyl-CoA levels in HCCs, increased infiltration of tumor-associated neutrophils (TANs) in the cancer microenvironment and HCC metastasis. Cytokine microarray and enzyme-linked immunosorbent assays (ELISA) revealed the crucial role of the chemokine (C-X-C motif) ligand 1(CXCL1). Mechanistically, acetyl-CoA accumulation induces H3 acetylation-dependent upregulation of CXCL1 gene expression. CXCL1 recruits TANs, leads to neutrophil extracellular traps (NETs) formation and promotes HCC metastasis. Collectively, our work linked the accumulation of acetyl-CoA in HCC cells and TANs infiltration, and revealed that the CXCL1-CXC receptor 2 (CXCR2)-TANs-NETs axis is a potential target for HCCs with high acetyl-CoA levels.
Collapse
Affiliation(s)
- Jun-Jie Pan
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Sun-Zhe Xie
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Xin Zheng
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Jian-Feng Xu
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Hao Xu
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Rui-Qi Yin
- Department of Infectious Diseases, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Yun-Ling Luo
- Department of Infectious Diseases, Rui'an People's Hospital, Wenzhou Medical University, 168 Ruifeng Avenue, Zhejiang 325200, China
| | - Li Shen
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Zheng-Ru Chen
- Department of Infectious Diseases, Rui'an People's Hospital, Wenzhou Medical University, 168 Ruifeng Avenue, Zhejiang 325200, China
| | - Yi-Ran Chen
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Shi-Zhe Yu
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Lu Lu
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China
| | - Wen-Wei Zhu
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China.
| | - Ming Lu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Lun-Xiu Qin
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, 12 Urumqi Road, Shanghai 200040, China; Institutes of Biomedical Sciences, Fudan University, 130 Dongan Road, Shanghai 200032, China.
| |
Collapse
|
24
|
Pan Y, Cheng J, Zhu Y, Zhang J, Fan W, Chen X. Immunological nanomaterials to combat cancer metastasis. Chem Soc Rev 2024; 53:6399-6444. [PMID: 38745455 DOI: 10.1039/d2cs00968d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Metastasis causes greater than 90% of cancer-associated deaths, presenting huge challenges for detection and efficient treatment of cancer due to its high heterogeneity and widespread dissemination to various organs. Therefore, it is imperative to combat cancer metastasis, which is the key to achieving complete cancer eradication. Immunotherapy as a systemic approach has shown promising potential to combat metastasis. However, current clinical immunotherapies are not effective for all patients or all types of cancer metastases owing to insufficient immune responses. In recent years, immunological nanomaterials with intrinsic immunogenicity or immunomodulatory agents with efficient loading have been shown to enhance immune responses to eliminate metastasis. In this review, we would like to summarize various types of immunological nanomaterials against metastasis. Moreover, this review will summarize a series of immunological nanomaterial-mediated immunotherapy strategies to combat metastasis, including immunogenic cell death, regulation of chemokines and cytokines, improving the immunosuppressive tumour microenvironment, activation of the STING pathway, enhancing cytotoxic natural killer cell activity, enhancing antigen presentation of dendritic cells, and enhancing chimeric antigen receptor T cell therapy. Furthermore, the synergistic anti-metastasis strategies based on the combinational use of immunotherapy and other therapeutic modalities will also be introduced. In addition, the nanomaterial-mediated imaging techniques (e.g., optical imaging, magnetic resonance imaging, computed tomography, photoacoustic imaging, surface-enhanced Raman scattering, radionuclide imaging, etc.) for detecting metastasis and monitoring anti-metastasis efficacy are also summarized. Finally, the current challenges and future prospects of immunological nanomaterial-based anti-metastasis are also elucidated with the intention to accelerate its clinical translation.
Collapse
Affiliation(s)
- Yuanbo Pan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Junjie Cheng
- Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Yang Zhu
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, Fujian, China.
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 211198, China.
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
| |
Collapse
|
25
|
Chen W, Zhao Z, Zhou H, Dong S, Li X, Hu S, Zhong S, Chen K. Development of prognostic signatures and risk index related to lipid metabolism in ccRCC. Front Oncol 2024; 14:1378095. [PMID: 38939337 PMCID: PMC11208495 DOI: 10.3389/fonc.2024.1378095] [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: 03/07/2024] [Accepted: 05/31/2024] [Indexed: 06/29/2024] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) is a metabolic disorder characterized by abnormal lipid accumulation in the cytoplasm. Lipid metabolism-related genes may have important clinical significance for prognosis prediction and individualized treatment. Methods We collected bulk and single-cell transcriptomic data of ccRCC and normal samples to identify key lipid metabolism-related prognostic signatures. qPCR was used to confirm the expression of signatures in cancer cell lines. Based on the identified signatures, we developed a lipid metabolism risk score (LMRS) as a risk index. We explored the potential application value of prognostic signatures and LMRS in precise treatment from multiple perspectives. Results Through comprehensive analysis, we identified five lipid metabolism-related prognostic signatures (ACADM, ACAT1, ECHS1, HPGD, DGKZ). We developed a risk index LMRS, which was significantly associated with poor prognosis in patients. There was a significant correlation between LMRS and the infiltration levels of multiple immune cells. Patients with high LMRS may be more likely to respond to immunotherapy. The different LMRS groups were suitable for different anticancer drug treatment regimens. Conclusion Prognostic signatures and LMRS we developed may be applied to the risk assessment of ccRCC patients, which may have potential guiding significance in the diagnosis and precise treatment of ccRCC patients.
Collapse
Affiliation(s)
- Wenbo Chen
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Zhenyu Zhao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Zhou
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Dong
- Department of Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoyu Li
- Department of Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Sheng Hu
- Department of Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shan Zhong
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Ke Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
26
|
McGinnis CS, Miao Z, Superville D, Yao W, Goga A, Reticker-Flynn NE, Winkler J, Satpathy AT. The temporal progression of lung immune remodeling during breast cancer metastasis. Cancer Cell 2024; 42:1018-1031.e6. [PMID: 38821060 PMCID: PMC11255555 DOI: 10.1016/j.ccell.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 03/23/2024] [Accepted: 05/06/2024] [Indexed: 06/02/2024]
Abstract
Tumor metastasis requires systemic remodeling of distant organ microenvironments that impacts immune cell phenotypes, population structure, and intercellular communication. However, our understanding of immune phenotypic dynamics in the metastatic niche remains incomplete. Here, we longitudinally assayed lung immune transcriptional profiles in the polyomavirus middle T antigen (PyMT) and 4T1 metastatic breast cancer models from primary tumorigenesis, through pre-metastatic niche formation, to the final stages of metastatic outgrowth at single-cell resolution. Computational analyses of these data revealed a TLR-NFκB inflammatory program enacted by both peripherally derived and tissue-resident myeloid cells that correlated with pre-metastatic niche formation and mirrored CD14+ "activated" myeloid cells in the primary tumor. Moreover, we observed that primary tumor and metastatic niche natural killer (NK) cells are differentially regulated in mice and human patient samples, with the metastatic niche featuring elevated cytotoxic NK cell proportions. Finally, we identified cell-type-specific dynamic regulation of IGF1 and CCL6 signaling during metastatic progression that represents anti-metastatic immunotherapy candidate pathways.
Collapse
Affiliation(s)
- Christopher S McGinnis
- Department of Pathology, Stanford University, Stanford, CA 94305, USA; Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| | - Zhuang Miao
- Department of Pathology, Stanford University, Stanford, CA 94305, USA; Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA
| | - Daphne Superville
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA 94158, USA; Department of Cell and Tissue Biology, UCSF, San Francisco, CA 94143, USA; Department of Medicine, UCSF, San Francisco, CA 94143, USA
| | - Winnie Yao
- Department of Pathology, Stanford University, Stanford, CA 94305, USA; Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA
| | - Andrei Goga
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA 94158, USA; Department of Cell and Tissue Biology, UCSF, San Francisco, CA 94143, USA; Department of Medicine, UCSF, San Francisco, CA 94143, USA
| | | | - Juliane Winkler
- Center for Cancer Research, Medical University of Vienna, Vienna 1090, Austria.
| | - Ansuman T Satpathy
- Department of Pathology, Stanford University, Stanford, CA 94305, USA; Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA.
| |
Collapse
|
27
|
Yi Y, Qin G, Yang H, Jia H, Zeng Q, Zheng D, Ye S, Zhang Z, Liu TM, Luo KQ, Deng CX, Xu RH. Mesenchymal Stromal Cells Increase the Natural Killer Resistance of Circulating Tumor Cells via Intercellular Signaling of cGAS-STING-IFNβ-HLA. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400888. [PMID: 38638003 PMCID: PMC11151078 DOI: 10.1002/advs.202400888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/17/2024] [Indexed: 04/20/2024]
Abstract
Circulating tumor cells (CTCs) shed from primary tumors must overcome the cytotoxicity of immune cells, particularly natural killer (NK) cells, to cause metastasis. The tumor microenvironment (TME) protects tumor cells from the cytotoxicity of immune cells, which is partially executed by cancer-associated mesenchymal stromal cells (MSCs). However, the mechanisms by which MSCs influence the NK resistance of CTCs remain poorly understood. This study demonstrates that MSCs enhance the NK resistance of cancer cells in a gap junction-dependent manner, thereby promoting the survival and metastatic seeding of CTCs in immunocompromised mice. Tumor cells crosstalk with MSCs through an intercellular cGAS-cGAMP-STING signaling loop, leading to increased production of interferon-β (IFNβ) by MSCs. IFNβ reversely enhances the type I IFN (IFN-I) signaling in tumor cells and hence the expression of human leukocyte antigen class I (HLA-I) on the cell surface, protecting the tumor cells from NK cytotoxicity. Disruption of this loop reverses NK sensitivity in tumor cells and decreases tumor metastasis. Moreover, there are positive correlations between IFN-I signaling, HLA-I expression, and NK tolerance in human tumor samples. Thus, the NK-resistant signaling loop between tumor cells and MSCs may serve as a novel therapeutic target.
Collapse
Affiliation(s)
- Ye Yi
- Center of Reproduction, Development and Aging, Cancer Center, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Guihui Qin
- Center of Reproduction, Development and Aging, Cancer Center, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Hongmei Yang
- Center of Reproduction, Development and Aging, Cancer Center, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Hao Jia
- Center of Reproduction, Development and Aging, Cancer Center, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Qibing Zeng
- Center of Reproduction, Development and Aging, Cancer Center, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Dejin Zheng
- Center of Reproduction, Development and Aging, Cancer Center, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Sen Ye
- Center of Reproduction, Development and Aging, Cancer Center, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Zhiming Zhang
- Center of Reproduction, Development and Aging, Cancer Center, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, 999078, China
| | - Tzu-Ming Liu
- Center of Reproduction, Development and Aging, Cancer Center, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, 999078, China
- Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macao SAR, 999078, China
| | - Kathy Qian Luo
- Center of Reproduction, Development and Aging, Cancer Center, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, 999078, China
- Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macao SAR, 999078, China
| | - Chu-Xia Deng
- Center of Reproduction, Development and Aging, Cancer Center, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, 999078, China
- Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macao SAR, 999078, China
| | - Ren-He Xu
- Center of Reproduction, Development and Aging, Cancer Center, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, 999078, China
- Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macao SAR, 999078, China
| |
Collapse
|
28
|
Fan J, Zhu J, Zhu H, Xu H. Potential therapeutic targets in myeloid cell therapy for overcoming chemoresistance and immune suppression in gastrointestinal tumors. Crit Rev Oncol Hematol 2024; 198:104362. [PMID: 38614267 DOI: 10.1016/j.critrevonc.2024.104362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 03/26/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024] Open
Abstract
In the tumor microenvironment (TME), myeloid cells play a pivotal role. Myeloid-derived immunosuppressive cells, including tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), are central components in shaping the immunosuppressive milieu of the tumor. Within the TME, a majority of TAMs assume an M2 phenotype, characterized by their pro-tumoral activity. These cells promote tumor cell growth, angiogenesis, invasion, and migration. In contrast, M1 macrophages, under appropriate activation conditions, exhibit cytotoxic capabilities against cancer cells. However, an excessive M1 response may lead to pro-tumoral inflammation. As a result, myeloid cells have emerged as crucial targets in cancer therapy. This review concentrates on gastrointestinal tumors, detailing methods for targeting macrophages to enhance tumor radiotherapy and immunotherapy sensitivity. We specifically delve into monocytes and tumor-associated macrophages' various functions, establishing an immunosuppressive microenvironment, promoting tumorigenic inflammation, and fostering neovascularization and stromal remodeling. Additionally, we examine combination therapeutic strategies.
Collapse
Affiliation(s)
- Jiawei Fan
- Department of Gastroenterology, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR China
| | - Jianshu Zhu
- Department of Spine Surgery, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR China
| | - He Zhu
- Department of Gastroenterology, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR China
| | - Hong Xu
- Department of Gastroenterology, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR China.
| |
Collapse
|
29
|
Kalashgrani MY, Mousavi SM, Akmal MH, Gholami A, Omidifar N, Chiang WH, Lai CW, Ripaj Uddin M, Althomali RH, Rahman MM. Biosensors for metastatic cancer cell detection. Clin Chim Acta 2024; 559:119685. [PMID: 38663472 DOI: 10.1016/j.cca.2024.119685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 05/04/2024]
Abstract
Early detection and effective cancer treatment are critical to improving metastatic cancer cell diagnosis and management today. In particular, accurate qualitative diagnosis of metastatic cancer cell represents an important step in the diagnosis of cancer. Today, biosensors have been widely developed due to the daily need to measure different chemical and biological species. Biosensors are utilized to quantify chemical and biological phenomena by generating signals that are directly proportional to the quantity of the analyte present in the reaction. Biosensors are widely used in disease control, drug delivery, infection detection, detection of pathogenic microorganisms, and markers that indicate a specific disease in the body. These devices have been especially popular in the field of metastatic cancer cell diagnosis and treatment due to their portability, high sensitivity, high specificity, ease of use and short response time. This article examines biosensors for metastatic cancer cells. It also studies metastatic cancer cells and the mechanism of metastasis. Finally, the function of biosensors and biomarkers in metastatic cancer cells is investigated.
Collapse
Affiliation(s)
| | - Seyyed Mojtaba Mousavi
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taiwan
| | - Muhammad Hussnain Akmal
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taiwan
| | - Ahmad Gholami
- Biotechnology Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Navid Omidifar
- Department of Pathology, Shiraz University of Medical Sciences, Shiraz 71468-64685, Iran
| | - Wei-Hung Chiang
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taiwan.
| | - Chin Wei Lai
- Nanotechnology and Catalysis Research Centre (NANOCAT), Level 3, Block A, Institute for Advanced Studies (IAS), Universiti Malaya (UM), 50603 Kuala Lumpur, Malaysia
| | - Md Ripaj Uddin
- Institute of National Analytical Research and Service (INARS), Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhanmondi, Dhaka, Bangladesh
| | - Raed H Althomali
- Department of Chemistry, College of Art and Science, Prince Sattam bin Abdulaziz University, Wadi Al-Dawasir 11991, Al Kharj, Saudi Arabia
| | - Mohammed M Rahman
- Center of Excellence for Advanced Materials Research (CEAMR) & Department of Chemistry, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
30
|
Bloomquist R, Mondal AK, Vashisht A, Sahajpal N, Jones K, Vashisht V, Singh H, Farmaha J, Kolhe R. Gene Regulatory Network Analysis of Post-Mortem Lungs Unveils Novel Insights into COVID-19 Pathogenesis. Viruses 2024; 16:853. [PMID: 38932146 PMCID: PMC11209433 DOI: 10.3390/v16060853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
The novel coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has emerged as one of the most significant global health crises in recent history. The clinical characteristics of COVID-19 patients have revealed the possibility of immune activity changes contributing to disease severity. Nevertheless, limited information is available regarding the immune response in human lung tissue, which is the primary site of infection. In this study, we conducted an extensive analysis of lung tissue to screen for differentially expressed miRNAs and mRNAs in five individuals who died due to COVID-19 and underwent a rapid autopsy, as well as seven control individuals who died of other causes unrelated to COVID-19. To analyze the host response gene expression, miRNA microarray and Nanostring's nCounter XT gene expression assay were performed. Our study identified 37 downregulated and 77 upregulated miRNAs in COVID-19 lung biopsy samples compared to the controls. A total of 653 mRNA transcripts were differentially expressed between the two sample types, with most transcripts (472) being downregulated in COVID-19-positive specimens. Hierarchical and PCA K-means clustering analysis showed distinct clustering between COVID-19 and control samples. Enrichment and network analyses revealed differentially expressed genes important for innate immunity and inflammatory response in COVID-19 lung biopsies. The interferon-signaling pathway was highly upregulated in COVID-19 specimens while genes involved in interleukin-17 signaling were downregulated. These findings shed light on the mechanisms of host cellular responses to COVID-19 infection in lung tissues and could help identify new targets for the prevention and treatment of COVID-19 infection.
Collapse
Affiliation(s)
- Ryan Bloomquist
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30909, USA; (R.B.); (A.K.M.); (A.V.); (K.J.); (V.V.); (H.S.); (J.F.)
- School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Ashis K. Mondal
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30909, USA; (R.B.); (A.K.M.); (A.V.); (K.J.); (V.V.); (H.S.); (J.F.)
| | - Ashutosh Vashisht
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30909, USA; (R.B.); (A.K.M.); (A.V.); (K.J.); (V.V.); (H.S.); (J.F.)
| | | | - Kimya Jones
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30909, USA; (R.B.); (A.K.M.); (A.V.); (K.J.); (V.V.); (H.S.); (J.F.)
| | - Vishakha Vashisht
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30909, USA; (R.B.); (A.K.M.); (A.V.); (K.J.); (V.V.); (H.S.); (J.F.)
| | - Harmanpreet Singh
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30909, USA; (R.B.); (A.K.M.); (A.V.); (K.J.); (V.V.); (H.S.); (J.F.)
| | - Jaspreet Farmaha
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30909, USA; (R.B.); (A.K.M.); (A.V.); (K.J.); (V.V.); (H.S.); (J.F.)
| | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA 30909, USA; (R.B.); (A.K.M.); (A.V.); (K.J.); (V.V.); (H.S.); (J.F.)
| |
Collapse
|
31
|
Bayatli E, Ozgural O, Dogan I, Ozpiskin OM, Hasimoglu S, Kuzukiran YC, Zaimoglu M, Eroglu U, Kahilogullari G, Ugur HC, Caglar YS. Prediction of Meningioma Grade Using Hematological Parameters. World Neurosurg 2024; 185:e893-e899. [PMID: 38453007 DOI: 10.1016/j.wneu.2024.02.148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/09/2024]
Abstract
OBJECTIVE Predicting the aggressiveness of meningiomas may influence the surgical strategy timing. Because of the paucity of robust markers, the systemic immune-inflammation (SII) index is a novel biomarker to be an independent predictor of poor prognosis in various cancers including gliomas. We aimed to investigate the value of SII as well as neutrophil-lymphocyte ratio (NLR) and platelet-lymphocyte ratio (PLR) indices in predicting prognosis. METHODS Records including demographic, clinical, and laboratory data of patients operated on due to intracranial meningioma in 2017-2023 were retrospectively reviewed. RESULTS A total of 234 patients were included in this study. All of SII index, NLR, and PLR values at presentation were significantly higher in grade ≥2 meningiomas. A positive correlation was observed between SII index and Ki67 index (r=0.313; P<0.001); between NLR and Ki67 index (r=0.330; P<0.001); and between PLR and Ki67 index (r=0.223; P<0.01). SII index (optimal cutoff level >618), NLR (optimal cutoff level >3.53), and PLR (optimal cutoff level >121.2) showed significant predictive values. CONCLUSIONS This is the first study to assess the prognostic value of the SII index in patients with intracranial meningiomas. Increased SII index, NLR and PLR were correlated with higher grade and higher Ki-67 index. They also harbor the potential to screen patients that may need more aggressive treatments or more frequent follow-up examinations.
Collapse
Affiliation(s)
- Eyup Bayatli
- Ankara University, School of Medicine, Department of Neurosurgery, Ankara, Turkey
| | - Onur Ozgural
- Ankara University, School of Medicine, Department of Neurosurgery, Ankara, Turkey
| | - Ihsan Dogan
- Ankara University, School of Medicine, Department of Neurosurgery, Ankara, Turkey.
| | - Omer Mert Ozpiskin
- Ankara University, School of Medicine, Department of Neurosurgery, Ankara, Turkey
| | - Siavash Hasimoglu
- Ankara University, School of Medicine, Department of Neurosurgery, Ankara, Turkey
| | - Yusuf Cem Kuzukiran
- Ankara University, School of Medicine, Department of Neurosurgery, Ankara, Turkey
| | - Murat Zaimoglu
- Ankara University, School of Medicine, Department of Neurosurgery, Ankara, Turkey
| | - Umit Eroglu
- Ankara University, School of Medicine, Department of Neurosurgery, Ankara, Turkey
| | - Gokmen Kahilogullari
- Ankara University, School of Medicine, Department of Neurosurgery, Ankara, Turkey
| | - Hasan Caglar Ugur
- Ankara University, School of Medicine, Department of Neurosurgery, Ankara, Turkey
| | - Y Sukru Caglar
- Ankara University, School of Medicine, Department of Neurosurgery, Ankara, Turkey
| |
Collapse
|
32
|
Astuti Y, Raymant M, Quaranta V, Clarke K, Abudula M, Smith O, Bellomo G, Chandran-Gorner V, Nourse C, Halloran C, Ghaneh P, Palmer D, Jones RP, Campbell F, Pollard JW, Morton JP, Mielgo A, Schmid MC. Efferocytosis reprograms the tumor microenvironment to promote pancreatic cancer liver metastasis. NATURE CANCER 2024; 5:774-790. [PMID: 38355776 PMCID: PMC11136665 DOI: 10.1038/s43018-024-00731-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024]
Abstract
Pancreatic ductal adenocarcinoma is a highly metastatic disease and macrophages support liver metastases. Efferocytosis, or engulfment of apoptotic cells by macrophages, is an essential process in tissue homeostasis and wound healing, but its role in metastasis is less well understood. Here, we found that the colonization of the hepatic metastatic site is accompanied by low-grade tissue injury and that efferocytosis-mediated clearance of parenchymal dead cells promotes macrophage reprogramming and liver metastasis. Mechanistically, progranulin expression in macrophages is necessary for efficient efferocytosis by controlling lysosomal acidification via cystic fibrosis transmembrane conductance regulator and the degradation of lysosomal cargo, resulting in LXRα/RXRα-mediated macrophage conversion and upregulation of arginase 1. Pharmacological blockade of efferocytosis or macrophage-specific genetic depletion of progranulin impairs macrophage conversion, improves CD8+ T cell functions, and reduces liver metastasis. Our findings reveal how hard-wired functions of macrophages in tissue repair contribute to liver metastasis and identify potential targets for prevention of pancreatic ductal adenocarcinoma liver metastasis.
Collapse
Affiliation(s)
- Yuliana Astuti
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Meirion Raymant
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Valeria Quaranta
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Kim Clarke
- Computational Biology Facility, University of Liverpool, Liverpool, UK
| | - Maidinaimu Abudula
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Olivia Smith
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Gaia Bellomo
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | | | - Craig Nourse
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Christopher Halloran
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Paula Ghaneh
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Daniel Palmer
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Robert P Jones
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Fiona Campbell
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | | | - Jennifer P Morton
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Ainhoa Mielgo
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Michael C Schmid
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK.
| |
Collapse
|
33
|
Zannikou M, Fish EN, Platanias LC. Signaling by Type I Interferons in Immune Cells: Disease Consequences. Cancers (Basel) 2024; 16:1600. [PMID: 38672681 PMCID: PMC11049350 DOI: 10.3390/cancers16081600] [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: 03/11/2024] [Revised: 04/08/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
This review addresses interferon (IFN) signaling in immune cells and the tumor microenvironment (TME) and examines how this affects cancer progression. The data reveal that IFNs exert dual roles in cancers, dependent on the TME, exhibiting both anti-tumor activity and promoting cancer progression. We discuss the abnormal IFN signaling induced by cancerous cells that alters immune responses to permit their survival and proliferation.
Collapse
Affiliation(s)
- Markella Zannikou
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, 303 East Superior Ave., Chicago, IL 60611, USA
| | - Eleanor N. Fish
- Toronto General Hospital Research Institute, University Health Network, 67 College Street, Toronto, ON M5G 2M1, Canada;
- Department of Immunology, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Leonidas C. Platanias
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology-Oncology, Feinberg School of Medicine, Northwestern University, 303 East Superior Ave., Chicago, IL 60611, USA
- Department of Medicine, Jesse Brown Veterans Affairs Medical Center, 820 S. Damen Ave., Chicago, IL 60612, USA
| |
Collapse
|
34
|
Wang L, Ma L, Song Z, Zhou L, Chen K, Wang X, Liu Z, Wang B, Shen C, Guo X, Jia X. Single-cell transcriptome analysis profiling lymphatic invasion-related TME in colorectal cancer. Sci Rep 2024; 14:8911. [PMID: 38632387 PMCID: PMC11024122 DOI: 10.1038/s41598-024-59656-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/12/2024] [Indexed: 04/19/2024] Open
Abstract
Lymphatic invasion (LI) is extremely aggressive and induces worse prognosis among patients with colorectal cancer (CRC). Thus, it is critical to characterize the cellular and molecular mechanisms underlying LI in order to establish novel and efficacious therapeutic targets that enhance the prognosis of CRC patients. RNA-seq data, clinical and survival information of colon adenocarcinoma (COAD) patients were obtained from the TCGA database. In addition, three scRNA-seq datasets of CRC patients were acquired from the GEO database. Data analyses were conducted with the R packages. We assessed the tumor microenvironment (TME) differences between LI+ and LI- based scRNA-seq data, LI+ cells exhibited augmented abundance of immunosuppression and invasive subset. Marked extracellular matrix network activation was also observed in LI+ cells within SPP1+ macrophages. We revealed that an immunosuppressive and pro-angiogenic TME strongly enhanced LI, as was evidenced by the CD4+ Tregs, CD8+ GZMK+, SPP1+ macrophages, e-myCAFs, and w-myCAFs subcluster infiltrations. Furthermore, we identified potential LI targets that influenced tumor development, metastasis, and immunotherapeutic response. Finally, a novel LIRS model was established based on the expression of 14 LI-related signatures, and in the two testing cohorts, LIRS was also proved to have accurate prognostic predictive ability. In this report, we provided a valuable resource and extensive insights into the LI of CRC. Our conclusions can potentially benefit the establishment of highly efficacious therapeutic targets as well as diagnostic biomarkers that improve patient outcomes.
Collapse
Affiliation(s)
- Liping Wang
- Department of Geriatrics, Liaocheng People's Hospital, Liaocheng, 252000, Shandong, China
| | - Liming Ma
- Harbin Inji Technology Co., Ltd., Harbin, 150060, Heilongjiang, China
| | - Zhaona Song
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, 252000, Shandong, China
| | - Li Zhou
- Beijing Easyresearch Technology Limited, Beijing, 100049, China
| | - Kexin Chen
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, 252000, Shandong, China
| | - Xizi Wang
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, 252000, Shandong, China
| | - Zhen Liu
- Harbin Inji Technology Co., Ltd., Harbin, 150060, Heilongjiang, China
| | - Baozhong Wang
- Department of Oncology, Liaocheng People's Hospital, Liaocheng, 252000, Shandong, China
| | - Chen Shen
- Department of Data and Information, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, Zhejiang, China
| | - Xianchao Guo
- Harbin Inji Technology Co., Ltd., Harbin, 150060, Heilongjiang, China.
| | - Xiaodong Jia
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, 252000, Shandong, China.
| |
Collapse
|
35
|
Rojas-Quintero J, Díaz MP, Palmar J, Galan-Freyle NJ, Morillo V, Escalona D, González-Torres HJ, Torres W, Navarro-Quiroz E, Rivera-Porras D, Bermúdez V. Car T Cells in Solid Tumors: Overcoming Obstacles. Int J Mol Sci 2024; 25:4170. [PMID: 38673757 PMCID: PMC11050550 DOI: 10.3390/ijms25084170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 04/28/2024] Open
Abstract
Chimeric antigen receptor T cell (CAR T cell) therapy has emerged as a prominent adoptive cell therapy and a therapeutic approach of great interest in the fight against cancer. This approach has shown notorious efficacy in refractory hematological neoplasm, which has bolstered its exploration in the field of solid cancers. However, successfully managing solid tumors presents considerable intrinsic challenges, which include the necessity of guiding the modified cells toward the tumoral region, assuring their penetration and survival in adverse microenvironments, and addressing the complexity of identifying the specific antigens for each type of cancer. This review focuses on outlining the challenges faced by CAR T cell therapy when used in the treatment of solid tumors, as well as presenting optimizations and emergent approaches directed at improving its efficacy in this particular context. From precise localization to the modulation of the tumoral microenvironment and the adaptation of antigen recognition strategies, diverse pathways will be examined to overcome the current limitations and buttress the therapeutic potential of CAR T cells in the fight against solid tumors.
Collapse
Affiliation(s)
- Joselyn Rojas-Quintero
- Medicine, Pulmonary, Critical Care, and Sleep Medicine Department, Baylor College of Medicine, Houston, TX 77030, USA;
| | - María P. Díaz
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | - Jim Palmar
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | - Nataly J. Galan-Freyle
- Centro de Investigaciones en Ciencias de la Vida, Universidad Simón Bolívar, Barranquilla 080002, Colombia; (N.J.G.-F.); (E.N.-Q.)
| | - Valery Morillo
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | - Daniel Escalona
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | | | - Wheeler Torres
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | - Elkin Navarro-Quiroz
- Centro de Investigaciones en Ciencias de la Vida, Universidad Simón Bolívar, Barranquilla 080002, Colombia; (N.J.G.-F.); (E.N.-Q.)
- Facultad de Ciencias Básicas y Biomédicas, Barranquilla 080002, Colombia
| | - Diego Rivera-Porras
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Cúcuta 540001, Colombia;
| | - Valmore Bermúdez
- Centro de Investigaciones en Ciencias de la Vida, Universidad Simón Bolívar, Barranquilla 080002, Colombia; (N.J.G.-F.); (E.N.-Q.)
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla 080002, Colombia;
| |
Collapse
|
36
|
Zhou T, Qian H, Zhang D, Fang W, Yao M, Shi H, Chen T, Chai C, Guo B. PGRN inhibits CD8 +T cell recruitment and promotes breast cancer progression by up-regulating ICAM-1 on TAM. Cancer Immunol Immunother 2024; 73:76. [PMID: 38554213 PMCID: PMC10981592 DOI: 10.1007/s00262-024-03655-z] [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: 08/16/2023] [Accepted: 02/11/2024] [Indexed: 04/01/2024]
Abstract
BACKGROUND Tumor microenvironment actually reduces antitumor effect against the immune attack by exclusion of CD8+T cells. Progranulin (PGRN) is a multifunctional growth factor with significant pathological effects in multiple tumors; however, its role in immunity evasion of breast cancer (BCa) is not completely understood. METHODS We depleted GRN (PGRN gene) genetically in mice or specifically in PY8119 murine BCa cell line, and mouse models of orthotopic or subcutaneous transplantation were used. Chimeric mice-deficient of PGRN (Grn-/-) in bone marrow (BM) compartment was also generated. Association of PGRN expression with chemokine production or BCa development was investigated by histological and immunological assays. RESULTS We found PGRN was involved in exhaustion of cytotoxic CD8+T cell in BCa with the increasing expressions of M2 markers and intercellular cell adhesion molecule-1 (ICAM-1) on macrophages. Specifically, ablation of PGRN in PY8119 cells reduced tumor burden, accompanied by the infiltrating of cytotoxic CD8+T cells into tumor nests. Moreover, our result revealed that blockade of PD-1 in PGRN-depleted tumors exhibited better antitumor effect in vivo and significantly decreased tumor burden. CONCLUSION These findings suggest that inhibition of PGRN may act as a potential immune-therapeutic strategy by recovering infiltration of CD8+T cell in BCa tissue and thereby enhancing the response to anti-PD-1 therapy.
Collapse
Affiliation(s)
- Ting Zhou
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Husun Qian
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Dian Zhang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Wenli Fang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - MengLi Yao
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - He Shi
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Tingmei Chen
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Chengsen Chai
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Bianqin Guo
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, People's Republic of China.
| |
Collapse
|
37
|
Cui C, Tang X, Xing J, Sheng X, Chi H, Zhan W. Single-cell RNA-seq revealed heterogeneous responses and functional differentiation of hemocytes against white spot syndrome virus infection in Litopenaeus vannamei. J Virol 2024; 98:e0180523. [PMID: 38323810 PMCID: PMC10949519 DOI: 10.1128/jvi.01805-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/12/2024] [Indexed: 02/08/2024] Open
Abstract
Shrimp hemocytes are the vital immune cells participating in innate immune response to defend against viruses. However, the lack of specific molecular markers for shrimp hemocyte hindered the insightful understanding of their functional clusters and differential roles in combating microbial infections. In this study, we used single-cell RNA sequencing to map the transcriptomic landscape of hemocytes from the white spot syndrome virus (WSSV)-infected Litopenaeus vannamei and conjointly analyzed with our previous published single-cell RNA sequencing technology data from the healthy hemocytes. A total of 16 transcriptionally distinct cell clusters were identified, which occupied different proportions in healthy and WSSV-infected hemocytes and exerted differential roles in antiviral immune response. Following mapping of the sequencing data to the WSSV genome, we found that all types of hemocytes could be invaded by WSSV virions, especially the cluster 8, which showed the highest transcriptional levels of WSSV genes and exhibited a cell type-specific antiviral response to the viral infection. Further evaluation of the cell clusters revealed the delicate dynamic balance between hemocyte immune response and viral infestation. Unsupervised pseudo-time analysis of hemocytes showed that the hemocytes in immune-resting state could be significantly activated upon WSSV infection and then functionally differentiated to different hemocyte subsets. Collectively, our results revealed the differential responses of shrimp hemocytes and the process of immune-functional differentiation post-WSSV infection, providing essential resource for the systematic insight into the synergistic immune response mechanism against viral infection among hemocyte subtypes. IMPORTANCE Current knowledge of shrimp hemocyte classification mainly comes from morphology, which hinder in-depth characterization of cell lineage development, functional differentiation, and different immune response of hemocyte types during pathogenic infections. Here, single-cell RNA sequencing was used for mapping hemocytes during white spot syndrome virus (WSSV) infection in Litopenaeus vannamei, identifying 16 cell clusters and evaluating their potential antiviral functional characteristics. We have described the dynamic balance between viral infestation and hemocyte immunity. And the functional differentiation of hemocytes under WSSV stimulation was further characterized. Our results provided a comprehensive transcriptional landscape and revealed the heterogeneous immune response in shrimp hemocytes during WSSV infection.
Collapse
Affiliation(s)
- Chuang Cui
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Heng Chi
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
38
|
Wang M, Xue W, Yuan H, Wang Z, Yu L. Nano-Drug Delivery Systems Targeting CAFs: A Promising Treatment for Pancreatic Cancer. Int J Nanomedicine 2024; 19:2823-2849. [PMID: 38525013 PMCID: PMC10959015 DOI: 10.2147/ijn.s451151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/06/2024] [Indexed: 03/26/2024] Open
Abstract
Currently, pancreatic cancer (PC) is one of the most lethal malignant tumors. PC is typically diagnosed at a late stage, exhibits a poor response to conventional treatment, and has a bleak prognosis. Unfortunately, PC's survival rate has not significantly improved since the 1960s. Cancer-associated fibroblasts (CAFs) are a key component of the pancreatic tumor microenvironment (TME). They play a vital role in maintaining the extracellular matrix and facilitating the intricate communication between cancer cells and infiltrated immune cells. Exploring therapeutic approaches targeting CAFs may reverse the current landscape of PC therapy. In recent years, nano-drug delivery systems have evolved rapidly and have been able to accurately target and precisely release drugs with little or no toxicity to the whole body. In this review, we will comprehensively discuss the origin, heterogeneity, potential targets, and recent advances in the nano-drug delivery system of CAFs in PC. We will also propose a novel integrated treatment regimen that utilizes a nano-drug delivery system to target CAFs in PC, combined with radiotherapy and immunotherapy. Additionally, we will address the challenges that this regimen currently faces.
Collapse
Affiliation(s)
- Mingjie Wang
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Wenxiang Xue
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Hanghang Yuan
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Zhicheng Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Lei Yu
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| |
Collapse
|
39
|
Jiang Q, Xiao D, Wang A, Yu Q, Yin Y, Wu J, Zhang Y, Jin T, Kuang B, Jia Y. CLEC4E upregulation in gastric cancer: A potential therapeutic target correlating with tumor-associated macrophages. Heliyon 2024; 10:e27172. [PMID: 38463883 PMCID: PMC10920739 DOI: 10.1016/j.heliyon.2024.e27172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/12/2024] Open
Abstract
Background CLEC4E has been reported to promote lung cancer progression. Tumor-associated macrophages (TAMs) play an important role in tumorigenesis. Whether the expression of CLEC4E in TAMs is associated with gastric carcinogenesis remains unclear. Methods The TIMER, UALCAN, UCSC Xena, and KM plotter databases are used to examine the expression of CLEC4E and its prognostic significance in gastric cancer (GC). Additionally, GO, KEGG, and GSEA analysis were conducted, and single-cell RNA-seq (scRNA-seq) datasets were utilized. The Coremine medical database was used to predict therapeutic drugs, and molecular docking was performed. Human GC samples were obtained, and co-culture models were constructed to evaluate the effects of CLEC4E in TAMs on tumor growth, migration, and invasion in vitro. Results CLEC4E was significantly upregulated in GC, and high CLEC4E expression was associated with poor prognosis. Western blotting and immunostaining showed increased protein levels of CLEC4E in GC. GO, KEGG, and GSEA results indicated that CLEC4E is involved in immune response. Immune infiltration analysis demonstrated that CLEC4E expression positively correlated with multiple immune cell types. scRNA-seq analyses revealed that CLEC4E was predominantly expressed in myeloid cells specifically TAMs, in GC. In vitro experiments confirmed that MFC induced CLEC4E expression in TAMs to mediate tumor progression. Specifically targeting CLEC4E by si-CLEC4E or stigmasterol inhibited cancer cell migration and invasion. Conclusion CLEC4E is a potential prognostic biomarker and new therapeutic target for GC that can be specifically targeted by stigmasterol.
Collapse
Affiliation(s)
- Qin Jiang
- Department of Gastroenterology, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, 430000, China
| | - Dan Xiao
- Department of Gastroenterology, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, 430000, China
| | - Ao Wang
- Department of Gastroenterology, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, 430000, China
| | - Qiong Yu
- Department of Gastroenterology, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, 430000, China
| | - Ying Yin
- Department of Gastroenterology, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, 430000, China
| | - Jingchong Wu
- Department of Gastroenterology, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, 430000, China
| | - Yan Zhang
- Department of Gastroenterology, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, 430000, China
| | - Tian Jin
- Department of Pathology, Hanchuan People's Hospital, Hanchuan, 431600, China
| | - Baicheng Kuang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yegui Jia
- Department of Gastroenterology, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, 430000, China
| |
Collapse
|
40
|
Ravi K, Manoharan TJM, Wang KC, Pockaj B, Nikkhah M. Engineered 3D ex vivo models to recapitulate the complex stromal and immune interactions within the tumor microenvironment. Biomaterials 2024; 305:122428. [PMID: 38147743 PMCID: PMC11098715 DOI: 10.1016/j.biomaterials.2023.122428] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/28/2023]
Abstract
Cancer thrives in a complex environment where interactions between cellular and acellular components, surrounding the tumor, play a crucial role in disease development and progression. Despite significant progress in cancer research, the mechanism driving tumor growth and therapeutic outcomes remains elusive. Two-dimensional (2D) cell culture assays and in vivo animal models are commonly used in cancer research and therapeutic testing. However, these models suffer from numerous shortcomings including lack of key features of the tumor microenvironment (TME) & cellular composition, cost, and ethical clearance. To that end, there is an increased interest in incorporating and elucidating the influence of TME on cancer progression. Advancements in 3D-engineered ex vivo models, leveraging biomaterials and microengineering technologies, have provided an unprecedented ability to reconstruct native-like bioengineered cancer models to study the heterotypic interactions of TME with a spatiotemporal organization. These bioengineered cancer models have shown excellent capabilities to bridge the gap between oversimplified 2D systems and animal models. In this review article, we primarily provide an overview of the immune and stromal cellular components of the TME and then discuss the latest state-of-the-art 3D-engineered ex vivo platforms aiming to recapitulate the complex TME features. The engineered TME model, discussed herein, are categorized into three main sections according to the cellular interactions within TME: (i) Tumor-Stromal interactions, (ii) Tumor-Immune interactions, and (iii) Complex TME interactions. Finally, we will conclude the article with a perspective on how these models can be instrumental for cancer translational studies and therapeutic testing.
Collapse
Affiliation(s)
- Kalpana Ravi
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA
| | | | - Kuei-Chun Wang
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA
| | | | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA; Biodesign Virginia G. Piper Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
41
|
Asadi M, Zarredar H, Zafari V, Soleimani Z, Saeedi H, Caner A, Shanehbandi D. Immune Features of Tumor Microenvironment: A Genetic Spotlight. Cell Biochem Biophys 2024; 82:107-118. [PMID: 37870699 DOI: 10.1007/s12013-023-01192-7] [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: 06/03/2023] [Accepted: 10/10/2023] [Indexed: 10/24/2023]
Abstract
A tumor represents a highly intricate tissue entity, characterized by an exceptionally complex microenvironment that starkly contrasts with the typical physiological surroundings of healthy tissues. Within this tumor microenvironment (TME), every component and factor assume paramount importance in the progression of malignancy and exerts a pivotal influence on a patient's clinical outcome. One of the remarkable aspects of the TME is its remarkable heterogeneity, not only across different types of cancers but even within the same histological category of tumors. In-depth research has illuminated the intricate interplay between specific immune cells and molecules and the dynamic characteristics of the TME. Recent investigations have yielded compelling evidence that several mutations harbored by tumor cells possess the capacity to instigate substantial alterations in the TME. These mutations, often acting as drivers of tumorigenesis, can orchestrate a cascade of events that remodel the TME, thereby influencing crucial aspects of cancer behavior, including its invasiveness, immune evasion, and response to therapies. It is within this nuanced context that the present study endeavors to provide a concise yet comprehensive summary of how specific mutations, within the genetic landscape of cancer cells, can instigate profound changes in TME features. By elucidating the intricate relationship between genetic mutations and the TME, this research aims to contribute to a deeper understanding of cancer biology. Ultimately, the knowledge gained from this study holds the potential to inform the development of more targeted and effective treatments, thereby offering new hope to patients grappling with the complexities of cancer.
Collapse
Affiliation(s)
- Milad Asadi
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Turkey
| | - Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Venus Zafari
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Turkey
| | - Zahra Soleimani
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Saeedi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayse Caner
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Turkey.
- The University of Texas, MD Anderson Cancer Center, Houston, USA.
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
42
|
Seow-En I, Koh YX, Zhao Y, Ang BH, Tan IEH, Chok AY, Tan EJKW, Au MKH. Predictive modeling algorithms for liver metastasis in colorectal cancer: A systematic review of the current literature. Ann Hepatobiliary Pancreat Surg 2024; 28:14-24. [PMID: 38129965 PMCID: PMC10896689 DOI: 10.14701/ahbps.23-078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/16/2023] [Indexed: 12/23/2023] Open
Abstract
This study aims to assess the quality and performance of predictive models for colorectal cancer liver metastasis (CRCLM). A systematic review was performed to identify relevant studies from various databases. Studies that described or validated predictive models for CRCLM were included. The methodological quality of the predictive models was assessed. Model performance was evaluated by the reported area under the receiver operating characteristic curve (AUC). Of the 117 articles screened, seven studies comprising 14 predictive models were included. The distribution of included predictive models was as follows: radiomics (n = 3), logistic regression (n = 3), Cox regression (n = 2), nomogram (n = 3), support vector machine (SVM, n = 2), random forest (n = 2), and convolutional neural network (CNN, n = 2). Age, sex, carcinoembryonic antigen, and tumor staging (T and N stage) were the most frequently used clinicopathological predictors for CRCLM. The mean AUCs ranged from 0.697 to 0.870, with 86% of the models demonstrating clear discriminative ability (AUC > 0.70). A hybrid approach combining clinical and radiomic features with SVM provided the best performance, achieving an AUC of 0.870. The overall risk of bias was identified as high in 71% of the included studies. This review highlights the potential of predictive modeling to accurately predict the occurrence of CRCLM. Integrating clinicopathological and radiomic features with machine learning algorithms demonstrates superior predictive capabilities.
Collapse
Affiliation(s)
- Isaac Seow-En
- Department of Colorectal Surgery, Singapore General Hospital and National Cancer Centre Singapore, Singapore
| | - Ye Xin Koh
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Centre Singapore, Singapore
- Duke-National University of Singapore Medical School, Singapore
- Liver Transplant Service, SingHealth Duke-National University of Singapore Transplant Centre, Singapore
| | - Yun Zhao
- Department of Colorectal Surgery, Singapore General Hospital and National Cancer Centre Singapore, Singapore
- Group Finance Analytics, Singapore Health Services, Singapore
| | - Boon Hwee Ang
- Group Finance Analytics, Singapore Health Services, Singapore
| | | | - Aik Yong Chok
- Department of Colorectal Surgery, Singapore General Hospital and National Cancer Centre Singapore, Singapore
| | - Emile John Kwong Wei Tan
- Department of Colorectal Surgery, Singapore General Hospital and National Cancer Centre Singapore, Singapore
| | - Marianne Kit Har Au
- Group Finance Analytics, Singapore Health Services, Singapore
- Finance, SingHealth Community Hospitals, Singapore
| |
Collapse
|
43
|
Yang K, Yi T. Tumor cell stemness in gastrointestinal cancer: regulation and targeted therapy. Front Mol Biosci 2024; 10:1297611. [PMID: 38455361 PMCID: PMC10918437 DOI: 10.3389/fmolb.2023.1297611] [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: 09/20/2023] [Accepted: 11/14/2023] [Indexed: 03/09/2024] Open
Abstract
The cancer stem cells are a rare group of self-renewable cancer cells capable of the initiation, progression, metastasis and recurrence of tumors, and also a key contributor to the therapeutic resistance. Thus, understanding the molecular mechanism of tumor stemness regulation, especially in the gastrointestinal (GI) cancers, is of great importance for targeting CSC and designing novel therapeutic strategies. This review aims to elucidate current advancements in the understanding of CSC regulation, including CSC biomarkers, signaling pathways, and non-coding RNAs. We will also provide a comprehensive view on how the tumor microenvironment (TME) display an overall tumor-promoting effect, including the recruitment and impact of cancer-associated fibroblasts (CAFs), the establishment of an immunosuppressive milieu, and the induction of angiogenesis and hypoxia. Lastly, this review consolidates mainstream novel therapeutic interventions targeting CSC stemness regulation.
Collapse
Affiliation(s)
- Kangqi Yang
- School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Tuo Yi
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
44
|
Nair R, Paul P, Maji I, Gupta U, Mahajan S, Aalhate M, Guru SK, Singh PK. Exploring the current landscape of chitosan-based hybrid nanoplatforms as cancer theragnostic. Carbohydr Polym 2024; 326:121644. [PMID: 38142105 DOI: 10.1016/j.carbpol.2023.121644] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 11/24/2023] [Indexed: 12/25/2023]
Abstract
In the last decade, investigators have put significant efforts to develop several diagnostic and therapeutic strategies against cancer. Many novel nanoplatforms, including lipidic, metallic, and inorganic nanocarriers, have shown massive potential at preclinical and clinical stages for cancer diagnosis and treatment. Each of these nano-systems is distinct with its own benefits and limitations. The need to overcome the limitations of single-component nano-systems, improve their morphological and biological features, and achieve multiple functionalities has resulted in the emergence of hybrid nanoparticles (HNPs). These HNPs integrate multicomponent nano-systems with diagnostic and therapeutic functions into a single nano-system serving as promising nanotools for cancer theragnostic applications. Chitosan (CS) being a mucoadhesive, biodegradable, and biocompatible biopolymer, has emerged as an essential element for the development of HNPs offering several advantages over conventional nanoparticles including pH-dependent drug delivery, sustained drug release, and enhanced nanoparticle stability. In addition, the free protonable amino groups in the CS backbone offer flexibility to its structure, making it easy for the modification and functionalization of CS, resulting in better drug targetability and cell uptake. This review discusses in detail the existing different oncology-directed CS-based HNPs including their morphological characteristics, in-vitro/in-vivo outcomes, toxicity concerns, hurdles in clinical translation, and future prospects.
Collapse
Affiliation(s)
- Rahul Nair
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Priti Paul
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Indrani Maji
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Ujala Gupta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Mayur Aalhate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Santosh Kumar Guru
- Department of Biological Science, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India.
| |
Collapse
|
45
|
Wang M, Liu C, Wang Y, Jamil M, Alhomrani M, Alamri AS, Alsanie WF, Alsharif A, Ali M, Jabeen N. Bone morphogenetic protein 1: a prognostic indicator and potential biomarker in three cancer types. Am J Transl Res 2024; 16:400-414. [PMID: 38463598 PMCID: PMC10918118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/10/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND Bone morphogenetic protein 1 (BMP1) is a metalloprotease that plays a role in activating both transforming growth factor-β (TGF-β) and BMP signaling pathways. TGF-β has been identified as a factor initiating and facilitating cancer development. Consequently, we propose the hypothesis that dysregulation of BMP1 could potentially contribute to the onset and advancement of human cancers. METHODS In this research, we aimed to analyze BMP1 expression level and the associated clinical outcomes across various cancers using online cancer OMICS databases, advanced Bioinformatics tools, and molecular analyses. RESULTS The outcomes of our web server-based expression analysis indicated an up-regulation of BMP1 in a majority of the human cancers examined. External validation using clinical samples also showed higher expression of BMP1. Moreover, heightened BMP1 expression exhibited a noteworthy correlation with reduced overall survival (OS) duration in Bladder Cancer (BLCA), Kidney Renal Clear Cell Carcinoma (KIRC), and Lung Adenocarcinoma (LUAD) patients. This suggests a substantial involvement of the BMP1 gene in the development and progression of these three types of cancers. The major signaling pathways related with BMP1 enriched genes were "ECM-receptor interaction, Amoebiasis, Focal adhesion, Protein digestion and absorption, progesterone-mediated, PI3K-Akt signaling pathway, and platelet activation". Moreover, we also explored some interesting correlations among BMP1 expression and its DNA promoter methylation level, CD8+ T immune cells level, and genetic variations. CONCLUSION In conclusion, our study has provided some solid basis for BMP1 to be used as a reliable common biomarker for BLCA, KIRC, and LUAD patients.
Collapse
Affiliation(s)
- Ming Wang
- Department of Oncology, Hebei Yanda HospitalLangfang 065200, Hebei, China
| | - Chunmei Liu
- Department of Oncology, Hebei Yanda HospitalLangfang 065200, Hebei, China
| | - Yingjie Wang
- Department of Oncology, Hebei Yanda HospitalLangfang 065200, Hebei, China
| | - Muhammad Jamil
- PARC Arid Zone Research CentreDera Ismail Khan 29050, Pakistan
| | - Majid Alhomrani
- College of Applied Medical Sciences, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif UniversityP.O. Box 11099, Taif 21944, Saudi Arabia
- Research Centre for Health Sciences, Taif UniversityTaif 26571, Saudi Arabia
| | - Abdulhakeem S Alamri
- College of Applied Medical Sciences, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif UniversityP.O. Box 11099, Taif 21944, Saudi Arabia
- Research Centre for Health Sciences, Taif UniversityTaif 26571, Saudi Arabia
| | - Walaa F Alsanie
- College of Applied Medical Sciences, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif UniversityP.O. Box 11099, Taif 21944, Saudi Arabia
- Research Centre for Health Sciences, Taif UniversityTaif 26571, Saudi Arabia
| | - Abdulaziz Alsharif
- College of Applied Medical Sciences, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif UniversityP.O. Box 11099, Taif 21944, Saudi Arabia
- Research Centre for Health Sciences, Taif UniversityTaif 26571, Saudi Arabia
| | - Mubarik Ali
- Animal Science Institute, National Agricultural Research CenterIslamabad 54000, Pakistan
| | - Norina Jabeen
- Department of Rural Sociology, University of Agriculture FaisalabadFaisalabad 38040, Punjab, Pakistan
| |
Collapse
|
46
|
Kuang T, Qiu Z, Wang K, Zhang L, Dong K, Wang W. Pan-immune inflammation value as a prognostic biomarker for cancer patients treated with immune checkpoint inhibitors. Front Immunol 2024; 15:1326083. [PMID: 38410508 PMCID: PMC10895004 DOI: 10.3389/fimmu.2024.1326083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 01/24/2024] [Indexed: 02/28/2024] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) represent a paradigm shift in the development of cancer therapy. However, the improved efficacy of ICIs remains to be further investigated. We conducted a systematic review and meta-analysis to evaluate the pan-immunoinflammatory value (PIV) and PILE score used to predict response to ICI therapy. Methods We searched selected databases for studies on pan-immune inflammation values and their association with outcomes of treatment with immune checkpoint inhibitors. We used hazard ratios (HRS) and 95% confidence intervals (CI) to summarize survival outcomes. All data analyses were performed using STATA 15.0. Results 7 studies comprising 982 patients were included in the meta-analysis. The pooled results showed that higher PIV was significantly associated with shorter overall survival OS (HR = 1.895, 95%CI: 1.548-2.318) and progression-free survival (PFS) (HR = 1.582, 95%CI: 1.324-1.890). Subgroup analyses also confirmed the reliability of the results. Conclusions High PIV and PILE metrics are associated with lower survival in cancer patients receiving ICIs.
Collapse
Affiliation(s)
- Tianrui Kuang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease, Wuhan, China
| | - Zhendong Qiu
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease, Wuhan, China
| | - Kunpeng Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease, Wuhan, China
| | - Lilong Zhang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease, Wuhan, China
| | - Keshuai Dong
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease, Wuhan, China
| | - Weixing Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease, Wuhan, China
| |
Collapse
|
47
|
Jiang Y, Lu L. New insight into the agonism of protease-activated receptors as an immunotherapeutic strategy. J Biol Chem 2024; 300:105614. [PMID: 38159863 PMCID: PMC10810747 DOI: 10.1016/j.jbc.2023.105614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 01/03/2024] Open
Abstract
The activation and mobilization of immune cells play a crucial role in immunotherapy. Existing therapeutic interventions, such as cytokines administration, aim to enhance immune cell activity. However, these approaches usually result in modest effectiveness and toxic side effects, thereby restricting their clinical application. Protease-activated receptors (PARs), a subfamily of G protein-coupled receptors, actively participate in the immune system by directly activating immune cells. The activation of PARs by proteases or synthetic ligands can modulate immune cell behavior, signaling, and responses to treat immune-related diseases, suggesting the significance of PARs agonism in immunotherapy. However, the agonism of PARs in therapeutical applications remains rarely discussed, since it has been traditionally considered that PARs activation facilitates disease progressions. This review aims to comprehensively summarize the activation, rather than inhibition, of PARs in immune-related physiological responses and diseases. Additionally, we will discuss the emerging immunotherapeutic potential of PARs agonism, providing a new strategic direction for PARs-mediated immunotherapy.
Collapse
Affiliation(s)
- Yuhong Jiang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China.
| | - Lei Lu
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
48
|
Gorji L, Brown ZJ, Pawlik TM. Advances and considerations in the use of immunotherapies for primary hepato-biliary malignancies. Surg Oncol 2024; 52:102031. [PMID: 38128340 DOI: 10.1016/j.suronc.2023.102031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 11/28/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
Hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA) comprise the two most common primary liver malignancies. Curative treatment options often include hepatectomy or liver transplantation; however, many patients present with advanced disease that is not amenable to surgical management. In turn, many patients are treated with systemic or targeted therapy. The tumor microenvironment (TME) is a complex network of immune cells and somatic cells, which can foster an environment for disease development and progression, as well as susceptibility and resistance to systemic therapeutic agents. In particular, the TME is comprised of both immune and non-immune cells. Immune cells such as T lymphocytes, natural killer (NK) cells, macrophages, and neutrophils reside in the TME and can affect tumorigenesis, disease progression, as well as response to therapy. Given the importance of the immune system, there are many emerging approaches for cancer immunotherapy. We herein provide a review the latest data on immunotherapy for primary HCC and BTC relative to the TME.
Collapse
Affiliation(s)
- Leva Gorji
- Department of Surgery, Kettering Health Dayton, Dayton, OH, USA.
| | - Zachary J Brown
- Department of Surgery, Division of Surgical Oncology, New York University - Long Island, Mineola, NY, 11501, USA.
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Cancer Hospital, Columbus, OH, USA.
| |
Collapse
|
49
|
Liu S, Li Y, Shi L, Liu J, Ren Y, Laman JD, van der Mei HC, Busscher HJ. Maintaining sidedness and fluidity in cell membrane coatings supported on nano-particulate and planar surfaces. Bioact Mater 2024; 32:344-355. [PMID: 37927898 PMCID: PMC10622627 DOI: 10.1016/j.bioactmat.2023.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/14/2023] [Accepted: 10/10/2023] [Indexed: 11/07/2023] Open
Abstract
Supported cell membrane coatings meet many requirements set to bioactive nanocarriers and materials, provided sidedness and fluidity of the natural membrane are maintained upon coating. However, the properties of a support-surface responsible for maintaining correct sidedness and fluidity are unknown. Here, we briefly review the properties of natural membranes and membrane-isolation methods, with focus on the asymmetric distribution of functional groups in natural membranes (sidedness) and the ability of molecules to float across a membrane to form functional domains (fluidity). This review concludes that hydrophilic sugar-residues of glycoproteins in the outer-leaflet of cell membranes direct the more hydrophobic inner-leaflet towards a support-surface to create a correctly-sided membrane coating, regardless of electrostatic double-layer interactions. On positively-charged support-surfaces however, strong, electrostatic double-layer attraction of negatively-charged membranes can impede homogeneous coating. In correctly-sided membrane coatings, fluidity is maintained regardless of whether the surface carries a positive or negative charge. However, membranes are frozen on positively-charged, highly-curved, small nanoparticles and localized nanoscopic structures on a support-surface. This leaves an unsupported membrane coating in between nanostructures on planar support-surfaces that is in dual-sided contact with its aqueous environment, yielding enhanced fluidity in membrane coatings on nanostructured, planar support-surfaces as compared with smooth ones.
Collapse
Affiliation(s)
- Sidi Liu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, 199 Ren'ai Road, Suzhou, 215123, PR China
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Yuanfeng Li
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University Wenzhou, Zhejiang, 325035, PR China
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, PR China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, PR China
| | - Jian Liu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, 199 Ren'ai Road, Suzhou, 215123, PR China
| | - Yijin Ren
- University of Groningen and University Medical Center Groningen, Department of Orthodontics, Hanzeplein 1, 9700 RB, Groningen, the Netherlands
| | - Jon D. Laman
- University of Groningen and University Medical Center Groningen, Department of Pathology and Medical Biology, Hanzeplein 1, 9700 RB, Groningen, the Netherlands
| | - Henny C. van der Mei
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| | - Henk J. Busscher
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
| |
Collapse
|
50
|
Wu J, Hu W, Yang W, Long Y, Chen K, Li F, Ma X, Li X. Knockdown of SQLE promotes CD8+ T cell infiltration in the tumor microenvironment. Cell Signal 2024; 114:110983. [PMID: 37993027 DOI: 10.1016/j.cellsig.2023.110983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/24/2023]
Abstract
Cholesterol biosynthesis and metabolism are critical aspects that shape the process of tumor development and associated microenvironmental conditions owing to the ability of cholesterol to drive tumor growth and invasion. Squalene Epoxidase (SQLE) is the second rate-limiting enzyme involved in the synthesis of cholesterol. The functional role of SQLE within the tumor microenvironment, however, has yet to be established. Here we show that SQLE is distinctively expressed across most types of cancer, and the expression level is highly correlated with tumor mutation burden and microsatellite instability. Accordingly, SQLE was identified as a prognostic risk factor in cancer patients. In addition, we observed a negative correlation between SQLE expression and immune cell infiltration across multiple cancers, and murine xenograft model further confirmed that SQLE knockdown was associated with enhanced intratumoral CD8+ T cell infiltration. Using next-generation sequencing, we identified 410 genes distinctively expressed in tumors exhibiting SQLE inhibition. KEGG and GO analysis further verified that SQLE altered the immune response in the tumor microenvironment. Furthermore, we found that the metabolism and translation of proteins is the main binding factor with SQLE. Our findings ascertain that SQLE is a potential target in multiple cancers and suppressing SQLE establishes an essential mechanism for shaping tumor microenvironment.
Collapse
Affiliation(s)
- Jun Wu
- Key Laboratory of Brain, Cognition and Education Sciences, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, South China Normal University, Guangzhou 510631, China
| | - Weibin Hu
- Key Laboratory of Brain, Cognition and Education Sciences, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, South China Normal University, Guangzhou 510631, China
| | - Wenhui Yang
- Key Laboratory of Brain, Cognition and Education Sciences, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, South China Normal University, Guangzhou 510631, China
| | - Yihao Long
- Key Laboratory of Brain, Cognition and Education Sciences, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, South China Normal University, Guangzhou 510631, China
| | - Kaizhao Chen
- Key Laboratory of Brain, Cognition and Education Sciences, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, South China Normal University, Guangzhou 510631, China
| | - Fugui Li
- Cancer Research Institute of Zhongshan City, Zhongshan City People's Hospital, Zhongshan 528403, China
| | - Xiaodong Ma
- Key Laboratory of Brain, Cognition and Education Sciences, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, South China Normal University, Guangzhou 510631, China.
| | - Xun Li
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China.
| |
Collapse
|