1
|
Jeng LB, Shih FY, Liao YW, Shyu WC, Teng CF. Hypoxic tumor cell line lysate-pulsed dendritic cell vaccine exhibits better therapeutic effects on hepatocellular carcinoma. Br J Cancer 2025:10.1038/s41416-025-02975-w. [PMID: 40050434 DOI: 10.1038/s41416-025-02975-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 02/03/2025] [Accepted: 02/26/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Dendritic cell (DC) vaccine is a promising immunotherapy for hepatocellular carcinoma (HCC) via triggering antigen-specific anti-tumor immunity. Hypoxia contributes to higher level and broader spectrum of antigen expression in tumor cells. METHODS This study aims to compare immunological activity and therapeutic efficacy between hypoxic and normoxic HCC cell line lysate-pulsed DC vaccines. RESULTS The results showed that hypoxic HCC cell line lysate-pulsed DC vaccines exhibited a stronger activity in producing interleukin-12 and promoting T cell proliferation and cytotoxicity in vitro. In HCC mice, hypoxic HCC cell line lysate-pulsed DC vaccines displayed a better efficacy in improving survival time and tumor volume and inducing intratumoral cytotoxic T cell infiltration and activation as well as tumor cell apoptosis. Adenylate kinase 4-derived antigens were important for hypoxic HCC cell line lysate-pulsed DC vaccine-elicited T cell killing. CONCLUSIONS In conclusion, this study demonstrated hypoxic HCC cell line lysate-pulsed DC vaccine as a potential therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Long-Bin Jeng
- Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan, ROC
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan, ROC
- Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan, ROC
- School of Medicine, China Medical University, Taichung, Taiwan, ROC
| | - Fu-Ying Shih
- Ph.D. Program for Biotech Pharmaceutical Industry, China Medical University, Taichung, Taiwan, ROC
| | - Yu-Wen Liao
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, ROC
| | - Woei-Cherng Shyu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, ROC
- Translational Medicine Research Center, China Medical University Hospital, Taichung, 404, Taiwan, ROC
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan, ROC
- Department of Occupational Therapy, Asia University, Taichung, Taiwan, ROC
| | - Chiao-Fang Teng
- Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan, ROC.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, ROC.
- Master Program for Cancer Biology and Drug Discovery, China Medical University, Taichung, Taiwan, ROC.
| |
Collapse
|
2
|
Zhang B, Zhu Y, Tang Y, Liu L, Liu Y, Li Y, Yu W, Lu L. The mediator subunit complex protein MED15 promotes lipid deposition and cancer progression during hypoxia. J Biol Chem 2025; 301:108296. [PMID: 39947475 PMCID: PMC11930138 DOI: 10.1016/j.jbc.2025.108296] [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: 11/28/2024] [Revised: 01/29/2025] [Accepted: 02/06/2025] [Indexed: 03/09/2025] Open
Abstract
Hypoxia, a hallmark of solid tumors, is associated with increased lipid droplet (LD) accumulation. However, the mechanisms underlying this remain elusive. Here, we identify Mediator complex subunit 15 (MED15) as a critical regulator of hypoxia-inducible factor (HIF) signaling, potentially impacting LD accumulation. In mammalian cells, we elucidated that MED15, as a HIF target gene, participates in promoting HIF transcriptional activity without affecting HIFα protein levels, creating a positive feedback loop. Furthermore, zebrafish deficiency in med15 displayed decreased HIF activity and impaired tolerance to hypoxic stress. Functionally, MED15 deficiency attenuated the proliferation of colon and renal cancer cells in vitro and tumor growth in vivo. Mechanistically, MED15 acts upstream of carnitine palmitoyltransferase 1A (CPT1A), a key enzyme in fatty acid oxidation, ultimately promoting HIF-mediated LD accumulation. Disrupting the MED15-CPT1A axis impairs this process. These findings reveal a novel MED15-HIF-CPT1A axis that promotes LD formation, potentially contributing to hypoxic tumor progression.
Collapse
Affiliation(s)
- Boqi Zhang
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Yu Zhu
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Yanfei Tang
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Lu Liu
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Yunzhang Liu
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Yun Li
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Wengong Yu
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Ling Lu
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, China.
| |
Collapse
|
3
|
Abreu MM, Chocron AF, Smadja DM. From cold to hot: mechanisms of hyperthermia in modulating tumor immunology for enhanced immunotherapy. Front Immunol 2025; 16:1487296. [PMID: 40092992 PMCID: PMC11906415 DOI: 10.3389/fimmu.2025.1487296] [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: 08/27/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
The emergence of immunotherapies has revolutionized cancer treatment by leveraging the immune system to target malignancies, offering new hope where traditional therapies often fall short. Within this context, hyperthermia (HT) has re-emerged as a promising adjunctive treatment, capable of enhancing the effectiveness of radiotherapy, chemotherapy, and immunotherapy. HT influences both the innate and adaptive immune systems, enhancing the activity of immune cells such as neutrophils, NK cells, and dendritic cells, while also modulating the tumor microenvironment (TME) to promote immunogenic cell death (ICD) and reduce immunosuppressive conditions. These effects contribute to the transformation of immunologically "cold" tumors into "hot" tumors, making them more susceptible to immune-mediated destruction. Furthermore, HT can amplify the efficacy of immune checkpoint inhibitors (ICIs) by improving immune cell infiltration, inducing damage-associated molecular pattern (DAMP) release, and enhancing antigen presentation. Preclinical and clinical studies support the combination of HT with ICIs, demonstrating improved outcomes in otherwise resistant tumors. However, the full therapeutic potential of the different technologies allowing to apply HT remains to be fully understood, and further research is needed to optimize treatment protocols, explore the differential impacts of local versus whole-body hyperthermia, and identify biomarkers for patient stratification. This review underscores the multifaceted role of HT in immunity and its potential to significantly enhance the efficacy of immunotherapy.
Collapse
Affiliation(s)
- M Marc Abreu
- Medicine Department, BTT Medical Institute, Aventura, FL, United States
- BTT Engineering Department, BTT Medical Institute, Aventura, FL, United States
| | - Alberto F Chocron
- Medicine Department, BTT Medical Institute, Aventura, FL, United States
- Research Service, Miami Veteran Administration Medical Center, Miami, FL, United States
| | - David M Smadja
- Department of Hematology, AP-HP, Georges Pompidou European Hospital, Paris, France
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
4
|
Branco H, Xavier CPR, Riganti C, Vasconcelos MH. Hypoxia as a critical player in extracellular vesicles-mediated intercellular communication between tumor cells and their surrounding microenvironment. Biochim Biophys Acta Rev Cancer 2025; 1880:189244. [PMID: 39672279 DOI: 10.1016/j.bbcan.2024.189244] [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: 06/18/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
In the past years, increasing attention has been paid to the role of extracellular vesicles (EVs) as mediators of intercellular communication in cancer. These small size particles mediate the intercellular transfer of important bioactive molecules involved in malignant initiation and progression. Hypoxia, or low partial pressure of oxygen, is recognized as a remarkable feature of solid tumors and has been demonstrated to exert a profound impact on tumor prognosis and therapeutic efficacy. Indeed, the high-pitched growth rate and chaotic neovascular architecture that embodies solid tumors results in a profound reduction in oxygen pressure within the tumor microenvironment (TME). In response to oxygen-deprived conditions, tumor cells and their surrounding milieu develop homeostatic adaptation mechanisms that contribute to the establishment of a pro-tumoral phenotype. Latest evidence suggests that the hypoxic microenvironment that surrounds the tumor bulk may be a clincher for the observed elevated levels of circulating EVs in cancer patients. Thus, it is proposed that EVs may play a role in mediating intercellular communication in response to hypoxic conditions. This review focuses on the EVs-mediated crosstalk that is established between tumor cells and their surrounding immune, endothelial, and stromal cell populations, within the hypoxic TME.
Collapse
Affiliation(s)
- Helena Branco
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Cristina P R Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, University Institute of Health Sciences - CESPU, 4585-116 Gandra, Portugal.
| | - Chiara Riganti
- Department of Oncology, University of Torino, 10126 Torino, Italy; Interdepartmental Research Center for Molecular Biotechnology "G. Tarone", University of Torino, 10126 Torino, Italy
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
5
|
Li W, Chen J, Guo Z. Targeting metabolic pathway enhance CAR-T potency for solid tumor. Int Immunopharmacol 2024; 143:113412. [PMID: 39454410 DOI: 10.1016/j.intimp.2024.113412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/01/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024]
Abstract
Chimeric antigen receptor (CAR) T cells have great potential in cancer therapy, particularly in treating hematologic malignancies. However, their efficacy in solid tumors remains limited, with a significant proportion of patients failing to achieve long-term complete remission. One major challenge is the premature exhaustion of CAR-T cells, often due to insufficient metabolic energy. The survival, function and metabolic adaptation of CAR-T cells are key determinants of their therapeutic efficacy. We explore how targeting metabolic pathways in the tumor microenvironment can enhance CAR-T cell therapy by addressing metabolic competition and immunosuppression that impair CAR-T cell function. Tumors undergo metabolically reprogrammed to meet their rapid proliferation, thereby modulating metabolic pathways in immune cells to promote immunosuppression. The distinct metabolic requirements of tumors and T cells create a competitive environment, affecting the efficacy of CAR-T cell therapy. Recent research on glucose, lipid and amino acid metabolism, along with the interactions between tumor and immune cell metabolism, has revealed that targeting these metabolic processes can enhance antitumor immune responses. Combining metabolic interventions with existing antitumor therapies can fulfill the metabolic demands of immune cells, providing new ideas for tumor immunometabolic therapies. This review discusses the latest advances in the immunometabolic mechanisms underlying tumor immunosuppression, their implications for immunotherapy, and summarizes potential metabolic targets to improve the efficacy of CAR-T therapy.
Collapse
Affiliation(s)
- Wenying Li
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Jiannan Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
6
|
Wu L, Bai R, Zhang Y, Chen H, Wu J, Chen Z, Wang H, Zhao L. METTL3-VISTA axis-based combination immunotherapy for APC truncation colorectal cancer. J Immunother Cancer 2024; 12:e009865. [PMID: 39653551 PMCID: PMC11629097 DOI: 10.1136/jitc-2024-009865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 11/17/2024] [Indexed: 12/12/2024] Open
Abstract
OBJECTIVE Although immune checkpoint blockade (ICB) therapy represents a bright spot in antitumor immunotherapy, its clinical benefits in colorectal cancer (CRC) are limited. Therefore, a new target for mediating CRC immunosuppression is urgently needed. Adenomatous polyposis coli (APC) mutations have been reported as early-stage characteristic events in CRC, but the role of truncated APC in the CRC immune microenvironment remains unclear and its clinical significance has yet to be explored. DESIGN Adenocarcinoma formation in the colon of the APCMin/+ mouse model, which displays features associated with the translation of truncated APC proteins, was induced by azoxymethane/dextran sodium sulfate. Multiplexed immunohistochemical consecutive staining on single slides and flow cytometry were used to explore the activation of immune cells and the expression of the immune checkpoint V-domain immunoglobulin suppressor of T-cell activation (VISTA) in the CRC tissues of APCWT and APCMin/+ mice. The construction of truncated APC vectors and an initial subserosal graft tumor mouse model was employed to mimic the tumor microenvironment (TME) during APC mutation. Methylated RNA immunoprecipitation-quantitative PCR assays were performed to investigate the N6-methyladenosine (m6A)-dependent transcriptional regulation of hypoxia-inducible factor-1 alpha (HIF1α) by methyltransferase-like protein 3 (METTL3). Mettl3fl/fl vil1-cre+/- mice were used to demonstrate that targeting METTL3 is a mediator that mitigates the deleterious effects of the APC978∆-HIF1α axis on antitumor immunity. A chimeric VISTA humanized mouse model was used to evaluate the drug efficacy of the VISTA-targeted compound onvatilimab. RESULTS We showed that APC978∆, a truncated APC protein, mediated overexpression of METTL3, resulting in m6A methylation of HIF1α messenger RNA and high expression of HIF1α. Furthermore, HIF1α promotes the migration of myeloid-derived suppressor cells to the TME by binding to the promoters of MCP-1 and MIF. In addition, HIF1α enhances the expression of the immune checkpoint VISTA on CRC cells, weakening tumor immune monitoring. CONCLUSIONS We elucidate that an underappreciated function of truncated APC in CRC is its ability to drive an immunosuppressive program that boosts tumor progression. Our work could provide a new perspective for the clinical application of immunotherapy in patients with CRC resistant to ICB therapy.
Collapse
Affiliation(s)
- Ling Wu
- Department of Pathology, Shunde Hospital of Southern Medical University, Foshan, Guangdong, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Rui Bai
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yujie Zhang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Hao Chen
- Department of Pathology, Shunde Hospital of Southern Medical University, Foshan, Guangdong, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianghua Wu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zetao Chen
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hui Wang
- Department of Medical Oncology, Guangzhou Medical University Affiliated Cancer Hospital, Guangzhou, Guangdong, China
| | - Liang Zhao
- Department of Pathology, Shunde Hospital of Southern Medical University, Foshan, Guangdong, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Wang Y, Wang X, Liu Y, Xu J, Zhu J, Zheng Y, Qi Q. A novel hypoxia- and lactate metabolism-related prognostic signature to characterize the immune landscape and predict immunotherapy response in osteosarcoma. Front Immunol 2024; 15:1467052. [PMID: 39569192 PMCID: PMC11576178 DOI: 10.3389/fimmu.2024.1467052] [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: 07/19/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
Background Immunotherapy has shown considerable promise in cancer treatment, yet only a minority of osteosarcoma patients derive benefits from this approach. Hypoxia and lactate metabolism are two predominant characteristics of the tumor microenvironment. These features are crucial for molding the immune landscape and thus have the potential to act as predictive indicators for immunotherapy response. Methods Prognostic modeled genes were identified through univariate and multivariate Cox regression as well as LASSO regression analyses. The tumor microenvironment was evaluated using ESTIMATE, CIBERSORT, and ImmuCellAI analyses. Tide prediction and expression of immune checkpoints, MHC molecules, chemokines, interleukins, interferons, receptors, and other cytokines were utilized to estimate immunotherapy efficacy. Single-cell analysis was performed to demonstrate the expression of modeled genes among various immune cell types. Experimental validation was carried out to verify the expression and functions of SFXN4 and SQOR. Results A potent signature was constructed with 8 genes related to hypoxia and lactate metabolism, including MAFF, COL5A2, FAM162A, SQOR, UQCRB, SFXN4, PFKFB2 and COX6A2. A nomogram incorporating risk scores and other clinical features demonstrated excellent predictive capacity. Osteosarcoma patients with high-risk scores exhibited poor prognosis and more "cold" tumor characteristics. According to the ESTIMATE algorithm, these patients displayed lower immune, stromal, and ESTIMATE scores, partially attributed to inadequate infiltration of key immunocytes. The Ciborsort analysis similarly indicated that high-risk individuals had diminished infiltration of critical anti-tumor immune cells such as Cytotoxic T cells, CD4+ T cells, and NK cells. The low expression levels of certain immune checkpoints, MHC molecules, chemokines, interleukins, interferons, receptors, and other cytokines in high-risk cases suggested their unsatisfactory responses to immune treatment. Tide prediction further demonstrated that fewer individuals classified as high risk may exhibit sensitivity to immune checkpoint inhibitor therapy. Notably, SFXN4 was found to be highly expressed in osteosarcoma tissues and cells; it promoted the growth, migration, and invasion of osteosarcoma cells, while SQOR had the opposite effect. Conclusion Our research has developed a robust hypoxia- and lactate metabolism-related gene signature, providing a solid theoretical foundation for prognosis prediction, classification of "cold" and "hot" tumors, accessing immunotherapy response, and directing personalized treatment for osteosarcoma.
Collapse
Affiliation(s)
- Yizhuo Wang
- The Second Department of Orthopedic Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Wang
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yang Liu
- The Second Department of Orthopedic Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiayuan Xu
- The Second Department of Orthopedic Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiyuan Zhu
- Department of pathology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yufu Zheng
- The Second Department of Orthopedic Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Quan Qi
- The Second Department of Orthopedic Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
8
|
Ghosh MK, Kumar S, Begam S, Ghosh S, Basu M. GBM immunotherapy: Exploring molecular and clinical frontiers. Life Sci 2024; 356:123018. [PMID: 39214286 DOI: 10.1016/j.lfs.2024.123018] [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: 05/31/2024] [Revised: 08/21/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
GBM is the most common, aggressive, and intracranial primary brain tumor; it originates from the glial progenitor cells, has poor overall survival (OS), and has limited treatment options. In this decade, GBM immunotherapy is in trend and preferred over several conventional therapies, due to their better patient survival outcome. This review explores the clinical trials of several immunotherapeutic approaches (immune checkpoint blockers (ICBs), CAR T-cell therapy, cancer vaccines, and adoptive cell therapy) with their efficacy and safety. Despite significant progress, several challenges (viz., immunosuppressive microenvironment, heterogeneity, and blood-brain barrier (BBB)) were experienced that hamper their immunotherapeutic potential. Furthermore, these challenges were clinically studied to be resolved by multiple combinatorial approaches, discussed in the later part of the review. Thus, this review suggests the clinical use and potential of immunotherapy in GBM and provides the holistic recent knowledge and future perspectives.
Collapse
Affiliation(s)
- Mrinal K Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata 700091, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India.
| | - Sunny Kumar
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata 700091, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Sabana Begam
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata 700091, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Sayani Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata 700091, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Malini Basu
- Department of Microbiology, Dhruba Chand Halder College, Dakshin Barasat, South 24 Parganas, PIN-743372, India
| |
Collapse
|
9
|
Li X, Xu J, Li X, Shi J, Wei C, Liang Q. Profiling hypoxia signaling reveals a lncRNA signature contributing to immunosuppression in high-grade glioma. Front Immunol 2024; 15:1471388. [PMID: 39416790 PMCID: PMC11479907 DOI: 10.3389/fimmu.2024.1471388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
Background Hypoxic conditions in glioma are linked to tumor aggressiveness, poor prognosis, and treatment resistance. Long non-coding RNAs (lncRNAs) play key roles in the hypoxic and immune microenvironment of cancers, but their link to hypoxia-induced immunosuppression in high-grade glioma (HGG) is not well-studied. Methods Gene expression profiles from TCGA and CGGA, along with clinical and genomic data, were analyzed. Bioinformatics methods including Consensus Clustering, Pearson correlation, and Cox regression analyses were used. Cell proliferation was assessed using cell counting kit-8 and colony formation assays. Glioma-macrophage interactions were evaluated using a co-culture model. Results Hypoxia subtype clustering showed hypoxic stress correlates with worse HGG prognosis. Eight hypoxia-related lncRNAs (AP000695.4, OSMR-AS1, AC078883.3, RP11-545E17.3, LINC01057, LINC01503, TP73-AS1, and LINC00672) with prognostic value were identified, forming a risk signature that separated patients into distinct prognostic groups. Multivariate Cox regression confirmed the signature as an independent prognostic factor. High-risk patients had greater hypoxia, leading to an immunosuppressive environment and immunotherapy resistance via tumor-associated macrophages (TAMs). TP73-AS1 significantly influenced hypoxia-induced TAM infiltration and M2 polarization. Conclusions We profiled hypoxic stress in HGG and developed an 8-lncRNA hypoxia-related signature predicting patient survival and immunotherapy response, emphasizing its role in hypoxia-induced immunosuppression.
Collapse
Affiliation(s)
- Xinqiao Li
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jingcheng Xu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xue Li
- International Department, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jianghua Shi
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chunmi Wei
- Department of Radiotherapy, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qingyu Liang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
10
|
Zhang Y, Liu Y, Li T, Yang X, Lang S, Pei P, Pei H, Chang L, Hu L, Liu T, Yang K. Engineered bacteria breach tumor physical barriers to enhance radio-immunotherapy. J Control Release 2024; 373:867-878. [PMID: 39097194 DOI: 10.1016/j.jconrel.2024.07.076] [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: 05/06/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024]
Abstract
Radiotherapy widely applied for local tumor therapy in clinic has been recently reinvigorated by the discovery that radiotherapy could activate systematic antitumor immune response. Nonetheless, the endogenous radio-immune effect is still incapable of radical tumor elimination due to the prevention of immune cell infiltration by the physical barrier in tumor microenvironment (TME). Herein, an engineered Salmonella secreting nattokinase (VNPNKase) is developed to synergistically modulate the physical and immune characteristics of TME to enhance radio-immunotherapy of colon tumors. The facultative anaerobic VNPNKase enriches at the tumor site after systemic administration, continuously secreting abundant NKase to degrade fibronectin, dredge the extracellular matrix (ECM), and inactivate cancer-associated fibroblasts (CAFs). The VNPNKase- dredged TME facilitates the infiltration of CD103+ dendritic cells (DCs) and thus the presentation of tumor-associated antigens (TAAs) after radiotherapy, recruiting sufficient CD8+ T lymphocytes to specifically eradicate localized tumors. Moreover, the pre-treatment of VNPNKase before radiotherapy amplifies the abscopal effect and achieves a long-term immune memory effect, preventing the metastasis and recurrence of tumors. Our research suggests that this strategy using engineered bacteria to breach tumor physical barrier for promoting immune cell infiltration possesses great promise as a translational strategy to enhance the effectiveness of radio-immunotherapy in treating solid tumors.
Collapse
Affiliation(s)
- Yanxiang Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yue Liu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Tingting Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xulu Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Shanshan Lang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Pei Pei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Hailong Pei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Lei Chang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Lin Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Teng Liu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China; Department of Pathology, the First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu 215000, China.
| |
Collapse
|
11
|
Dell’Anno F, Giugliano R, Listorti V, Razzuoli E. A Review on Canine and Human Soft Tissue Sarcomas: New Insights on Prognosis Factors and Treatment Measures. Vet Sci 2024; 11:362. [PMID: 39195816 PMCID: PMC11358912 DOI: 10.3390/vetsci11080362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/26/2024] [Accepted: 08/03/2024] [Indexed: 08/29/2024] Open
Abstract
Soft tissue sarcomas (STSs) represent a diverse group of tumors arising from mesenchymal cells, affecting both humans and animals, including dogs. Although STSs represent a class of rare tumors, especially in humans, they pose significant clinical challenges due to their potential for local recurrence and distant metastasis. Dogs, as a model for human STSs, offer several advantages, including exposure to similar environmental risk factors, genetic diversity among breeds, and the spontaneous development of tumors. Furthermore, canine tumors closely mimic the heterogeneity and complexity of human tumors, making them valuable for research into disease progression and treatment effectiveness. Current treatment approaches for STSs in both dogs and humans primarily involve surgery, radiation therapy, and chemotherapy, with treatment decisions based on tumor characteristics and patient factors. However, the development of novel therapeutic strategies is essential, given the high failure rate of new drugs in clinical trials. To better design new tailored treatments, comprehension of the tumor microenvironment (TME) is fundamental, since it plays a crucial role in STS initiation and progression by modulating tumor behavior, promoting angiogenesis, and suppressing immune responses. Notably, TME features include cancer-associated fibroblasts (CAFs), extracellular matrix (ECM) alterations, and tumor-associated macrophages (TAMs) that, depending on their polarization state, can affect immune responses and thus the patient's prognosis. In this review, new therapeutical approaches based on immunotherapy will be deeply explored as potential treatment options for both dogs and humans with STSs. In conclusion, this review provides an overview of the current understanding of STSs in dogs and humans, emphasizing the importance of the TME and potential treatment strategies.
Collapse
Affiliation(s)
- Filippo Dell’Anno
- National Reference Center of Veterinary and comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, 16129 Genova, Italy; (F.D.); (V.L.); (E.R.)
- Department of Public Health, Experimental and Forensic Medicine, Section of Biostatistics and Clinical Epidemiology, University of Pavia, 27100 Pavia, Italy
| | - Roberta Giugliano
- National Reference Center of Veterinary and comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, 16129 Genova, Italy; (F.D.); (V.L.); (E.R.)
| | - Valeria Listorti
- National Reference Center of Veterinary and comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, 16129 Genova, Italy; (F.D.); (V.L.); (E.R.)
| | - Elisabetta Razzuoli
- National Reference Center of Veterinary and comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, 16129 Genova, Italy; (F.D.); (V.L.); (E.R.)
| |
Collapse
|
12
|
Zhu G, Cao L, Wu J, Xu M, Zhang Y, Wu M, Li J. Co-morbid intersections of cancer and cardiovascular disease and targets for natural drug action: Reprogramming of lipid metabolism. Biomed Pharmacother 2024; 176:116875. [PMID: 38850662 DOI: 10.1016/j.biopha.2024.116875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/24/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
Cancer and cardiovascular diseases are major contributors to global morbidity and mortality, and their seemingly separate pathologies are intricately intertwined. In the context of cancer, the cardiovascular disease encompasses not only the side effects arising from anti-tumor treatments but also the metabolic shifts induced by oncological conditions. A growing body of research indicates that lipid metabolic reprogramming serves as a distinctive hallmark of tumors. Furthermore, anomalies in lipid metabolism play a significant role in the development of cardiovascular disease. This study delves into the cardiac implications of lipid metabolic reprogramming within the cancer context, closely examining abnormalities in lipid metabolism present in tumors, cardiac tissue, and immune cells within the microenvironment. Additionally, we examined risk factors such as obesity and anti-tumor therapy. Despite progress, a gap remains in the availability of drugs targeting lipid metabolism modulation for treating tumors and mitigating cardiac risk, with limited advancement seen in prior studies. Here, we present a review of previous research on natural drugs that exhibit both shared and distinct therapeutic effects on tumors and cardiac health by modulating lipid metabolism. Our aim is to provide insights for potential drug development.
Collapse
Affiliation(s)
- Guanghui Zhu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Luchang Cao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Jingyuan Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Manman Xu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ying Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Min Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Jie Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
13
|
Pu W, Ma C, Wang B, Zhu W, Chen H. The "Heater" of "Cold" Tumors-Blocking IL-6. Adv Biol (Weinh) 2024; 8:e2300587. [PMID: 38773937 DOI: 10.1002/adbi.202300587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/13/2024] [Indexed: 05/24/2024]
Abstract
The resolution of inflammation is not simply the end of the inflammatory response but rather a complex process that involves various cells, inflammatory factors, and specialized proresolving mediators following the occurrence of inflammation. Once inflammation cannot be cleared by the body, malignant tumors may be induced. Among them, IL-6, as an immunosuppressive factor, activates a variety of signal transduction pathways and induces tumorigenesis. Monitoring IL-6 can be used for the diagnosis, efficacy evaluation and prognosis of tumor patients. In terms of treatment, improving the efficacy of targeted and immunotherapy remains a major challenge. Blocking IL-6 and its mediated signaling pathways can regulate the tumor immune microenvironment and enhance immunotherapy responses by activating immune cells. Even transform "cold" tumors that are difficult to respond to immunotherapy into immunogenic "hot" tumors, acting as a "heater" for "cold" tumors, restarting the tumor immune cycle, and reducing immunotherapy-related toxic reactions and drug resistance. In clinical practice, the combined application of IL-6 inhibition with targeted therapy and immunotherapy may produce synergistic results. Nevertheless, additional clinical trials are imperative to further validate the safety and efficacy of this therapeutic approach.
Collapse
Affiliation(s)
- Weigao Pu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
- Department of Tumour Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Chenhui Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
- Department of Tumour Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Bofang Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
- Department of Tumour Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Weidong Zhu
- General Surgery Department of Lintao County People's Hospital in Gansu Province, Lanzhou, Gansu, 730030, China
| | - Hao Chen
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
- Department of Tumour Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou, Gansu, 730030, China
| |
Collapse
|
14
|
Zhu K, Wang L, Xiao Y, Zhang X, You G, Chen Y, Wang Q, Zhao L, Zhou H, Chen G. Nanomaterial-related hemoglobin-based oxygen carriers, with emphasis on liposome and nano-capsules, for biomedical applications: current status and future perspectives. J Nanobiotechnology 2024; 22:336. [PMID: 38880905 PMCID: PMC11180412 DOI: 10.1186/s12951-024-02606-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/30/2024] [Indexed: 06/18/2024] Open
Abstract
Oxygen is necessary for life and plays a key pivotal in maintaining normal physiological functions and treat of diseases. Hemoglobin-based oxygen carriers (HBOCs) have been studied and developed as a replacement for red blood cells (RBCs) in oxygen transport due to their similar oxygen-carrying capacities. However, applications of HBOCs are hindered by vasoactivity, oxidative toxicity, and a relatively short circulatory half-life. With advancements in nanotechnology, Hb encapsulation, absorption, bioconjugation, entrapment, and attachment to nanomaterials have been used to prepare nanomaterial-related HBOCs to address these challenges and pend their application in several biomedical and therapeutic contexts. This review focuses on the progress of this class of nanomaterial-related HBOCs in the fields of hemorrhagic shock, ischemic stroke, cancer, and wound healing, and speculates on future research directions. The advancements in nanomaterial-related HBOCs are expected to lead significant breakthroughs in blood substitutes, enabling their widespread use in the treatment of clinical diseases.
Collapse
Affiliation(s)
- Kai Zhu
- Academy of Military Medical Sciences, Beijing, 100850, China
| | - Lijun Wang
- Academy of Military Medical Sciences, Beijing, 100850, China
- Department of Morphology Laboratory, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China
| | - Yao Xiao
- Academy of Military Medical Sciences, Beijing, 100850, China
| | - Xiaoyong Zhang
- Academy of Military Medical Sciences, Beijing, 100850, China
| | - Guoxing You
- Academy of Military Medical Sciences, Beijing, 100850, China
| | - Yuzhi Chen
- Academy of Military Medical Sciences, Beijing, 100850, China
| | - Quan Wang
- Academy of Military Medical Sciences, Beijing, 100850, China
| | - Lian Zhao
- Academy of Military Medical Sciences, Beijing, 100850, China.
| | - Hong Zhou
- Academy of Military Medical Sciences, Beijing, 100850, China.
| | - Gan Chen
- Academy of Military Medical Sciences, Beijing, 100850, China.
| |
Collapse
|
15
|
Alqarni A, Jasim SA, Altalbawy FMA, Kaur H, Kaur I, Rodriguez-Benites C, Deorari M, Alwaily ER, Al-Ani AM, Redhee AH. Challenges and opportunities for cancer stem cell-targeted immunotherapies include immune checkpoint inhibitor, cancer stem cell-dendritic cell vaccine, chimeric antigen receptor immune cells, and modified exosomes. J Biochem Mol Toxicol 2024; 38:e23719. [PMID: 38764138 DOI: 10.1002/jbt.23719] [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: 02/10/2024] [Revised: 04/15/2024] [Accepted: 05/06/2024] [Indexed: 05/21/2024]
Abstract
Cancer stem cells (CSCs) are associated with the tumor microenvironment (TME). CSCs induce tumorigenesis, tumor recurrence and progression, and resistance to standard therapies. Indeed, CSCs pose an increasing challenge to current cancer therapy due to their stemness or self-renewal properties. The molecular and cellular interactions between heterogeneous CSCs and surrounding TME components and tumor-supporting immune cells show synergistic effects toward treatment failure. In the immunosuppressive TME, CSCs express various immunoregulatory proteins, growth factors, metabolites and cytokines, and also produce exosomes, a type of extracellular vesicles, to protect themselves from host immune surveillance. Among these, the identification and application of CSC-derived exosomes could be considered for the development of therapeutic approaches to eliminate CSCs or cancer, in addition to targeting the modulators that remodel the composition of the TME, as reviewed in this study. Here, we introduce the role of CSCs and how their interaction with TME complicates immunotherapies, and then present the CSC-based immunotherapy and the limitation of these therapies. We describe the biology and role of tumor/CSC-derived exosomes that induce immune suppression in the TME, and finally, introduce their potentials for the development of CSC-based targeted immunotherapy in the future.
Collapse
Affiliation(s)
- Abdullah Alqarni
- Department of Diagnostics Dental Sciences and Oral Biology, College of Dentistry, King Khalid University, Abha, Saudi Arabia
| | | | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | - Harpreet Kaur
- School of Basic and Applied Sciences, Shobhit University, Gangoh, India
- Department of Health and Allied Sciences, Arka Jain University, Jamshedpur, India
| | - Irwanjot Kaur
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, India
| | - Carlos Rodriguez-Benites
- Departamento Académico de Física, Facultad de Ciencias Físicas y Matemáticas, Universidad Nacional de Trujillo, Trujillo, Perú
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Enas R Alwaily
- Microbiology Research Group, College of Pharmacy, Al-Ayen University, Thi Qar, Iraq
| | - Ahmed M Al-Ani
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq
| | - Ahmed H Redhee
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
16
|
Ghiyabi E, Arabameri A, Charmi M. Mathematical modeling of hypoxia and adenosine to explore tumor escape mechanisms in DC-based immunotherapy. Sci Rep 2024; 14:11387. [PMID: 38762567 PMCID: PMC11102449 DOI: 10.1038/s41598-024-62209-6] [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: 03/22/2024] [Accepted: 05/14/2024] [Indexed: 05/20/2024] Open
Abstract
Identifying and controlling tumor escape mechanisms is crucial for improving cancer treatment effectiveness. Experimental studies reveal tumor hypoxia and adenosine as significant contributors to such mechanisms. Hypoxia exacerbates adenosine levels in the tumor microenvironment. Combining inhibition of these factors with dendritic cell (DC)-based immunotherapy promises improved clinical outcomes. However, challenges include understanding dynamics, optimal vaccine dosages, and timing. Mathematical models, including agent-based, diffusion, and ordinary differential equations, address these challenges. Here, we employ these models for the first time to elucidate how hypoxia and adenosine facilitate tumor escape in DC-based immunotherapy. After parameter estimation using experimental data, we optimize vaccination protocols to minimize tumor growth. Sensitivity analysis highlights adenosine's significant impact on immunotherapy efficacy. Its suppressive role impedes treatment success, but inhibiting adenosine could enhance therapy, as suggested by the model. Our findings shed light on hypoxia and adenosine-mediated tumor escape mechanisms, informing future treatment strategies. Additionally, identifiability analysis confirms accurate parameter determination using experimental data.
Collapse
Affiliation(s)
- Elahe Ghiyabi
- Department of Electrical Engineering, University of Zanjan, Zanjan, Iran
| | - Abazar Arabameri
- Department of Electrical Engineering, University of Zanjan, Zanjan, Iran.
| | - Mostafa Charmi
- Department of Electrical Engineering, University of Zanjan, Zanjan, Iran
| |
Collapse
|
17
|
Song LN, Wang B, Cai JL, Zhang PL, Chen SP, Zhou ZJ, Dai Z. Stratifying ICIs-responsive tumor microenvironment in HCC: from parsing out immune-hypoxic crosstalk to clinically applicable MRI-radiomics models. Br J Cancer 2024; 130:1356-1364. [PMID: 38355839 PMCID: PMC11014931 DOI: 10.1038/s41416-023-02463-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: 04/02/2023] [Accepted: 10/04/2023] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND We aimed to redefine Immune checkpoint inhibitors (ICIs)-responsive "hot" TME and develop a corresponding stratification model to maximize ICIs-efficacy in Hepatocellular Carcinoma (HCC). METHODS Hypoxic scores were designed, and the relevance to immunotherapy responses were validated in pan-cancers through single cell analysis. Multi-omics analysis using the hypoxic scores and immune infiltrate abundance was performed to redefine the ICIs-responsive TME subtype in HCC patients from TCGA (n = 363) and HCCDB database (n = 228). The immune hypoxic stress index (IHSI) was constructed to stratify the ICIs-responsive TME subtype, with exploring biological mechanism in vitro and in vivo. MRI-radiomics models were built for clinical applicability. RESULTS The hypoxic scores were lower in the dominant cell-subclusters of responders in pan-cancers. The higher immune infiltrate-normoxic (HIN) subtype was redefined as the ICIs-responsive TME. Stratification of the HIN subtype using IHSI effectively identified ICIs-responders in Melanoma (n = 122) and urological cancer (n = 22). TRAF3IP3, the constituent gene of IHSI, was implicated in ICIs-relevant "immune-hypoxic" crosstalk by stimulating MAVS/IFN-I pathway under normoxic condition. MRI-radiomics models assessing TRAF3IP3 with HIF1A expression (AUC > 0.80) screened ICIs-Responders in HCC cohort (n = 75). CONCLUSION The hypoxic-immune stratification redefined ICIs-responsive TME and provided MRI-Radiomics models for initial ICIs-responders screening, with IHSI facilitating further identification.
Collapse
Affiliation(s)
- Li-Na Song
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Biao Wang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Jia-Liang Cai
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Pei-Ling Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Shi-Ping Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zheng-Jun Zhou
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Zhi Dai
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China.
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
18
|
Chen G, Xu J, Ma S, Ji X, Carney JB, Wang C, Gao X, Chen P, Fan B, Chen J, Yue Y, James TD. Visual monitoring of biocatalytic processes using small molecular fluorescent probes: strategies-mechanisms-applications. Chem Commun (Camb) 2024; 60:2716-2731. [PMID: 38353179 DOI: 10.1039/d3cc05626k] [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: 03/06/2024]
Abstract
Real-time monitoring of biocatalytic-based processes is significantly improved and simplified when they can be visualized. Visual monitoring can be achieved by integrating a fluorescent unit with the biocatalyst. Herein, we outline the design strategies of fluorescent probes for monitoring biocatalysis: (1) probes for monitoring biocatalytic transfer: γ-glutamine is linked to the fluorophore as both a recognition group and for intramolecular charge transfer (ICT) inhibition; the probe is initially in an off state and is activated via the transfer of the γ-glutamine group and the release of the free amino group, which results in restoration of the "Donor-π-Acceptor" (D-π-A) system and fluorescence recovery. (2) Probes for monitoring biocatalytic oxidation: a propylamine is connected to the fluorophore as a recognition group, which cages the hydroxyl group, leading to the inhibition of ICT; propylamine is oxidized and subsequently β-elimination occurs, resulting in exposure of the hydroxyl group and fluorescence recovery. (3) Probes for monitoring biocatalytic reduction: a nitro group attached to a fluorophore as a fluorescence quenching group, this is converted to an amino group by catalytic reduction, resulting in fluorescence recovery. (4) Probes for monitoring biocatalytic hydrolysis: β-D-galactopyranoside or phosphate acts as a recognition group attached to hydroxyl groups of the fluorophore; the subsequent biocatalytic hydrolysis reaction releases the hydroxyl group resulting in fluorescence recovery. Following these 4 mechanisms, fluorophores including cyanine, coumarin, rhodamine, and Nile-red, have been used to develop systems for monitoring biocatalytic reactions. We anticipate that these strategies will result in systems able to rapidly diagnose and facilitate the treatment of serious diseases.
Collapse
Affiliation(s)
- Guang Chen
- The Youth Innovation Team of Shaanxi Universities, Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Jie Xu
- The Youth Innovation Team of Shaanxi Universities, Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Siyue Ma
- The Youth Innovation Team of Shaanxi Universities, Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Xinrui Ji
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada.
| | - Jared B Carney
- Department of Chemistry, Delaware State University, Dover, Delaware 19901, USA.
| | - Chao Wang
- The Youth Innovation Team of Shaanxi Universities, Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Xiaoyong Gao
- Jiangsu Simba Biological Medicine Co., Ltd. Gaogang Distrct Qidizhihui Park, Taizhou City, China
| | - Pu Chen
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada.
| | - Baolei Fan
- Hubei University of Science and Technology, No. 88, Xianning Avenue, Xianan District, Xianning 437000, China.
| | - Ji Chen
- Jiangsu Simba Biological Medicine Co., Ltd. Gaogang Distrct Qidizhihui Park, Taizhou City, China
| | - Yanfeng Yue
- Department of Chemistry, Delaware State University, Dover, Delaware 19901, USA.
| | - Tony D James
- Department of Chemistry, University of Bath, Bath BA2 7AY, UK.
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| |
Collapse
|
19
|
Zhao C, Zhu X, Tan J, Mei C, Cai X, Kong F. Lipid-based nanoparticles to address the limitations of GBM therapy by overcoming the blood-brain barrier, targeting glioblastoma stem cells, and counteracting the immunosuppressive tumor microenvironment. Biomed Pharmacother 2024; 171:116113. [PMID: 38181717 DOI: 10.1016/j.biopha.2023.116113] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/24/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most common primary malignant brain tumor, characterized by high heterogeneity, strong invasiveness, poor prognosis, and a low survival rate. A broad range of nanoparticles have been recently developed as drug delivery systems for GBM therapy owing to their inherent size effect and ability to cross the blood-brain barrier (BBB). Lipid-based nanoparticles (LBNPs), such as liposomes, solid lipid NPs (SLNs), and nano-structured lipid carriers (NLCs), have emerged as the most promising drug delivery system for the treatment of GBM because of their unique size, surface modification possibilities, and proven bio-safety. In this review, the main challenges of the current clinical treatment of GBM and the strategies on how novel LBNPs overcome them were explored. The application and progress of LBNP-based drug delivery systems in GBM chemotherapy, immunotherapy, and gene therapy in recent years were systematically reviewed, and the prospect of LBNPs for GBM treatment was discussed.
Collapse
Affiliation(s)
- Changhong Zhao
- School of Medicine, Hubei Polytechnic University, Huangshi 435003, China; Lantian Pharmaceuticals Co., Ltd, Hubei, China.
| | - Xinshu Zhu
- School of Medical Science and Laboratory Medicine, Jiangsu College of Nursing, Huai'an 223005, China
| | - Jianmei Tan
- School of Medicine, Hubei Polytechnic University, Huangshi 435003, China
| | - Chao Mei
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
| | - Xiang Cai
- Lantian Pharmaceuticals Co., Ltd, Hubei, China; School of Business, Hubei University of Science and Technology, China
| | - Fei Kong
- School of Medicine, Hubei Polytechnic University, Huangshi 435003, China; School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China.
| |
Collapse
|
20
|
Li A, Fang J. Anti‐angiogenic therapy enhances cancer immunotherapy: Mechanism and clinical application. INTERDISCIPLINARY MEDICINE 2024; 2. [DOI: 10.1002/inmd.20230025] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/18/2023] [Indexed: 01/04/2025]
Abstract
AbstractImmunotherapy, specifically immune checkpoint inhibitors, is revolutionizing cancer treatment, achieving durable control of previously incurable or advanced tumors. However, only a certain group of patients exhibit effective responses to immunotherapy. Anti‐angiogenic therapy aims to block blood vessel growth in tumors by depriving them of essential nutrients and effectively impeding their growth. Emerging evidence shows that tumor vessels exhibit structural and functional abnormalities, resulting in an immunosuppressive microenvironment and poor response to immunotherapy. Both preclinical and clinical studies have used anti‐angiogenic agents to enhance the effectiveness of immunotherapy against cancer. In this review, we concentrate on the synergistic effect of anti‐angiogenic and immune therapies in cancer management, dissect the direct effects and underlying mechanisms of tumor vessels on recruiting and activating immune cells, and discuss the potential of anti‐angiogenic agents to improve the effectiveness of immunotherapy. Lastly, we outline challenges and opportunities for the anti‐angiogenic strategy to enhance immunotherapy. Considering the increasing approval of the combination of anti‐angiogenic and immune therapies in treating cancers, this comprehensive review would be timely and important.
Collapse
Affiliation(s)
- An‐Qi Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism Guangdong Provincial Key Laboratory of New Drug Screening School of Pharmaceutical Sciences Southern Medical University Guangzhou China
| | - Jian‐Hong Fang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism Guangdong Provincial Key Laboratory of New Drug Screening School of Pharmaceutical Sciences Southern Medical University Guangzhou China
- Department of Hepatobiliary Surgery I General Surgery Center Zhujiang Hospital Southern Medical University Guangzhou China
| |
Collapse
|
21
|
Meng X, Ma F, Yu D. The diverse effects of cisplatin on tumor microenvironment: Insights and challenges for the delivery of cisplatin by nanoparticles. ENVIRONMENTAL RESEARCH 2024; 240:117362. [PMID: 37827371 DOI: 10.1016/j.envres.2023.117362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/11/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
Cisplatin is a well-known platinum-based chemotherapy medication that is widely utilized for some malignancies. Despite the direct cytotoxic consequences of cisplatin on tumor cells, studies in the recent decade have revealed that cisplatin can also affect different cells and their secretions in the tumor microenvironment (TME). Cisplatin has complex impacts on the TME, which may contribute to its anti-tumor activity or drug resistance mechanisms. These regulatory effects of cisplatin play a paramount function in tumor growth, invasion, and metastasis. This paper aims to review the diverse impacts of cisplatin and nanoparticles loaded with cisplatin on cancer cells and also non-cancerous cells in TME. The impacts of cisplatin on immune cells, tumor stroma, cancer cells, and also hypoxia will be discussed in the current review. Furthermore, we emphasize the challenges and prospects of using cisplatin in combination with other adjuvants and therapeutic modalities that target TME. We also discuss the potential synergistic effects of cisplatin with immune checkpoint inhibitors (ICIs) and other agents with anticancer potentials such as polyphenols and photosensitizers. Furthermore, the potential of nanoparticles for targeting TME and better delivery of cisplatin into tumors will be discussed.
Collapse
Affiliation(s)
- Xinxin Meng
- Zhuji Sixth People's Hospital of Zhejiang Province, Zhuji, Zhejiang, 311801, China
| | - Fengyun Ma
- Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Shaoxing University, Zhuji, Zhejiang, 311800, China.
| | - Dingli Yu
- Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Shaoxing University, Zhuji, Zhejiang, 311800, China
| |
Collapse
|
22
|
Zhao Y, Lv HJ, Deng XY, Chen P, Garstka MA, Shi BY, Fu J. Translocated HMGB3 is involved in papillary thyroid cancer progression by activating cytoplasmic TLR3 and transmembrane TREM1. Cell Cycle 2023; 22:2584-2601. [PMID: 38197217 PMCID: PMC10936681 DOI: 10.1080/15384101.2024.2302244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 01/11/2024] Open
Abstract
The family of high mobility group box (HMGB) proteins participates in various biological processes including immunity, inflammation, as well as cancer formation and progression. However, its role in thyroid cancer remains to be clarified. We performed quantitative RT-PCR (qRT-PCR), western blot, enzyme-linked immunosorbent, immunohistochemistry, and immunofluorescence assays to evaluate the expression level and subcellular location of HMGB3. The effects of HMGB3 knockdown on malignant biological behaviors of thyroid cancer were determined by cell proliferation assays, cell cycle and apoptosis assays, and transwell chamber migration and invasion assays. Differential expression genes (DEGs) altered by HMGB3 were analyzed using the Ingenuity Pathway Analysis (IPA) and TRRUST v2 database. HMGB3 correlated pathways predicted by bioinformatic analysis were then confirmed using western blot, co-immunoprecipitation, dual-luciferase reporter assay, and flow cytometry. We found that HMGB3 is overexpressed and its downregulation inhibits cell viability, promotes cell apoptosis and cell cycle arrest, and suppresses cell migration and invasion in thyroid cancer. In PTC, both tissue and serum levels of HMGB3 are elevated and are correlated with lymph node metastasis and advanced tumor stage. Mechanistically, we observed the translocation of HMGB3 in PTC, induced at least partially by hypoxia. Cytoplasmic HMGB3 activates nucleic-acid-mediated TLR3/NF-κB signaling and extracellular HMGB3 interacts with the transmembrane TREM1 receptor in PTC. This study demonstrates the oncogenic role of HMGB3 cytoplasmic and extracellular translocation in papillary thyroid cancers; we recommend its future use as a potential circulating biomarker and therapeutic target for PTC.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
- Department of Endocrinology, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Hong-Jun Lv
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
| | - Xue-Yang Deng
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| | - Pu Chen
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
| | - Malgorzata A. Garstka
- Core Research Laboratory; Department of Endocrinology; National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Bing-Yin Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
| | - Jiao Fu
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
| |
Collapse
|
23
|
Ou Z, Lu Y, Xu D, Luo Z. Hypoxia mediates immune escape of pancreatic cancer cells by affecting miR-1275/AXIN2 in natural killer cells. Front Immunol 2023; 14:1271603. [PMID: 38035113 PMCID: PMC10684956 DOI: 10.3389/fimmu.2023.1271603] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023] Open
Abstract
Given the increasing incidence of pancreatic cancer and the low survival rate, the exploration of the complex tumor microenvironment and the development of novel treatment options is urgent. NK cells, known for their cytotoxic abilities and modulation of other immune cells, are vital in recognizing and killing cancer cells. However, hypoxic conditions in the tumor microenvironment have been found to impair NK cell functionality and contribute to tumor immune escape. Therefore, we aimed to uncover the mechanism through which hypoxia mediates the immune escape of pancreatic cancer cells, focusing on the influence of miR-1275/AXIN2 on NK cells. Using a combination of GEO dataset screening, Tumor Immune Estimation Resource 2.0 immunoscore screening, and the Cancer Genome Atlas data, we identified a correlation between miR-1275 and NK cells. The down-regulation of miR-1275 was associated with decreased NK cell activity and survival in patients with pancreatic cancer. Pathway analysis further linked miR-1275 expression with the hypoxic HIF1A pathway. In vitro experiments were conducted using the NK-92 cell, revealing that hypoxia significantly reduced miR-1275 expression and correspondingly decreased the cell-killing ability of NK cells. Upregulation of miR-1275 increased perforin, IFN-γ and TNF-α expression levels and enhanced NK cell cytotoxicity. Additionally, miR-1275 was found to bind to and inhibit AXIN2 expression, which when overexpressed, partially alleviated the promotive effect of upregulated miR-1275 on NK-92 cell killing ability. In conclusion, this research underscores the critical role of the miR-1275/AXIN2 axis in hypoxia-mediated immune escape in pancreatic cancer, thus opening new potential avenues for treatment strategies.
Collapse
Affiliation(s)
- Zhenglin Ou
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, China
| | - Yebin Lu
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, China
| | - Dayong Xu
- Department of General Surgery, The First Hospital of Changsha, Changsha, Hunan, China
| | - Zhen Luo
- Department of General Surgery, The First Hospital of Changsha, Changsha, Hunan, China
| |
Collapse
|
24
|
Abstract
Ischemia or hypoxia can lead to pathological changes in the metabolism and function of tissues and then lead to various diseases. Timely and effective blood resuscitation or improvement of hypoxia is very important for the treatment of diseases. However, there is a need to develop stable, nontoxic, and immunologically inert oxygen carriers due to limitations such as blood shortages, different blood types, and the risk of transmitting infections. With the development of various technologies, oxygen carriers based on hemoglobin and perfluorocarbon have been widely studied in recent years. This paper reviews the development and application of hemoglobin and perfluorocarbon oxygen carriers. The design of oxygen carriers was analyzed, and their application as blood substitutes or oxygen carriers in various hypoxic diseases was discussed. Finally, the characteristics and future research of ideal oxygen carriers were prospected to provide reference for follow-up research.
Collapse
Affiliation(s)
- Qingsong Ye
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Deyuan Zheng
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Kaiyuan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, 22 Hankou Road, Nanjing 210093, China
| |
Collapse
|
25
|
Kwantwi LB. Exosome-mediated crosstalk between tumor cells and innate immune cells: implications for cancer progression and therapeutic strategies. J Cancer Res Clin Oncol 2023; 149:9487-9503. [PMID: 37154928 DOI: 10.1007/s00432-023-04833-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 05/02/2023] [Indexed: 05/10/2023]
Abstract
The increasing number of cancer-associated deaths despite the substantial improvement in diagnosis and treatment has sparked discussions on the need for novel biomarkers and therapeutic strategies for cancer. Exosomes have become crucial players in tumor development and progression, largely due to the diverse nature of their cargo content released to recipient cells. Importantly, exosome-mediated crosstalk between tumor and stromal cells is essential in reprogramming the tumor microenvironment to facilitate tumor progression. As a result, exosomes have gradually become a marker for the early diagnosis of many diseases and an important tool in drug delivery systems. However, the precise mechanisms by which exosomes participate in tumor progression remain elusive, multifaceted, and a double-edged sword, thus requiring further clarification. The available evidence suggests that exosomes can facilitate communication between innate immune cells and tumor cells to either support or inhibit tumor progression. Herein, this review focused on exosome-mediated intercellular communication between tumor cells and macrophages, neutrophils, mast cells, monocytes, dendritic cells, and natural killer cells. Specifically, how such intercellular communication affects tumor progression has been described. It has also been discussed that, depending on their cargo, exosomes can suppress or promote tumor cell progression. In addition, the potential application of exosomes and strategies to target exosomes in cancer treatment has been comprehensively discussed.
Collapse
Affiliation(s)
- Louis Boafo Kwantwi
- Department of Medical Imaging Sciences, Klintaps College of Health and Allied Sciences, Accra, DTD. TDC, 30A Klagon, Com. 19, Tema, Ghana.
| |
Collapse
|
26
|
Stampone E, Bencivenga D, Capellupo MC, Roberti D, Tartaglione I, Perrotta S, Della Ragione F, Borriello A. Genome editing and cancer therapy: handling the hypoxia-responsive pathway as a promising strategy. Cell Mol Life Sci 2023; 80:220. [PMID: 37477829 PMCID: PMC10361942 DOI: 10.1007/s00018-023-04852-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/14/2023] [Accepted: 06/29/2023] [Indexed: 07/22/2023]
Abstract
The precise characterization of oxygen-sensing pathways and the identification of pO2-regulated gene expression are both issues of critical importance. The O2-sensing system plays crucial roles in almost all the pivotal human processes, including the stem cell specification, the growth and development of tissues (such as embryogenesis), the modulation of intermediate metabolism (including the shift of the glucose metabolism from oxidative to anaerobic ATP production and vice versa), and the control of blood pressure. The solid cancer microenvironment is characterized by low oxygen levels and by the consequent activation of the hypoxia response that, in turn, allows a complex adaptive response characterized mainly by neoangiogenesis and metabolic reprogramming. Recently, incredible advances in molecular genetic methodologies allowed the genome editing with high efficiency and, above all, the precise identification of target cells/tissues. These new possibilities and the knowledge of the mechanisms of adaptation to hypoxia suggest the effective development of new therapeutic approaches based on the manipulation, targeting, and exploitation of the oxygen-sensor system molecular mechanisms.
Collapse
Affiliation(s)
- Emanuela Stampone
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Via Luigi De Crecchio, 7, 80138, Naples, Italy
| | - Debora Bencivenga
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Via Luigi De Crecchio, 7, 80138, Naples, Italy
| | - Maria Chiara Capellupo
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Via Luigi De Crecchio, 7, 80138, Naples, Italy
| | - Domenico Roberti
- Department of the Woman, the Child and of the General and Specialty Surgery, University of Campania "L. Vanvitelli", Via Luigi De Crecchio, 2, 80138, Naples, Italy
| | - Immacolata Tartaglione
- Department of the Woman, the Child and of the General and Specialty Surgery, University of Campania "L. Vanvitelli", Via Luigi De Crecchio, 2, 80138, Naples, Italy
| | - Silverio Perrotta
- Department of the Woman, the Child and of the General and Specialty Surgery, University of Campania "L. Vanvitelli", Via Luigi De Crecchio, 2, 80138, Naples, Italy
| | - Fulvio Della Ragione
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Via Luigi De Crecchio, 7, 80138, Naples, Italy.
| | - Adriana Borriello
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Via Luigi De Crecchio, 7, 80138, Naples, Italy.
| |
Collapse
|
27
|
Ramachandramoorthy H, Dang T, Srinivasa A, Nguyen KT, Nguyen P. Development of a Smart Portable Hypoxic Chamber with Accurate Sensing, Control and Visualization of In Vitro Cell Culture for Replication of Cancer Microenvironment. Cancers (Basel) 2023; 15:3645. [PMID: 37509306 PMCID: PMC10378062 DOI: 10.3390/cancers15143645] [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: 02/15/2023] [Revised: 07/09/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Clinical resistance towards treatment is a major concern in cancer therapy. This is due to in vitro studies lacking essential microenvironmental aspects. Tumor-hypoxia is an important pathophysiological phenomenon in numerous malignant tumors. Various studies have shown the importance of a hypoxic microenvironment (HME) in cancer drug resistance and its effects on cellular signaling and metabolism pathways. Most drugs fail in transition from a laboratory to clinical trials because of the variability in the testing microenvironment conditions. It is, thus, very crucial that research work needs to replicate these conditions in vitro to test the drugs and/or drug carriers for cancer therapy. Previous works have used a portable hypoxia chamber to reduce the cell microenvironment to hypoxic conditions. These techniques lack reliability and consistency due to a lack of control and visualization. In this research, we developed a smart portable hypoxia chamber that could accurately control the oxygen inside the portable chamber and have a global visualization. The proposed hypoxia chamber provided ease of use with the ranges of 1% to 20% oxygen with increments of 0.5%, as well as reproducibility and accuracy. The chamber displayed great precision on reaching the set oxygen limit and a high stability in maintaining that set level of oxygen compared to the uncontrolled setup for extended durations (24 h). For instance, at a 2% oxygen level, our automated system maintained this level over 1400 min, whereas the oxygen level fluctuated up to 4.5% in the conventional hypoxic chamber. We have also demonstrated the pitfalls of uncontrolled and non-visualized hypoxia chamber setup and the dire need for our system. The hypoxia-induced factor (HIF-1α) expression in cancer cell lines was tested and compared between the conventional hypoxia setup and our automated hypoxia chamber. We observed that there was a twofold increase in HIF-1α expression in the automated controlled chamber compared to the conventional device. The device also provided real-time sensing, visualization and control of the chamber conditions, which could aid in complex in vitro studies.
Collapse
Affiliation(s)
- Harish Ramachandramoorthy
- Department of Bioengineering, University of Texas, Arlington, TX 76019, USA
- Joint Bioengineering Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tuan Dang
- Department of Computer Science, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Ankitha Srinivasa
- Department of Bioengineering, University of Texas, Arlington, TX 76019, USA
| | - Kytai Truong Nguyen
- Department of Bioengineering, University of Texas, Arlington, TX 76019, USA
- Joint Bioengineering Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Phuc Nguyen
- Department of Computer Science, University of Texas at Arlington, Arlington, TX 76019, USA
| |
Collapse
|
28
|
Mortezaee K, Majidpoor J, Kharazinejad E. The impact of hypoxia on tumor-mediated bypassing anti-PD-(L)1 therapy. Biomed Pharmacother 2023; 162:114646. [PMID: 37011483 DOI: 10.1016/j.biopha.2023.114646] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/04/2023] Open
Abstract
Extending the durability of response is the current focus in cancer immunotherapy with immune checkpoint inhibitors (ICIs). However, factors like non-immunogenic tumor microenvironment (TME) along with aberrant angiogenesis and dysregulated metabolic systems are negative contributors. Hypoxia is a key TME condition and a critical promoter of tumor hallmarks. It acts on immune and non-immune cells within TME in order for promoting immune evasion and therapy resistance. Extreme hypoxia is a major promoter of resistance to the programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) inhibitor therapy. Hypoxia inducible factor-1 (HIF-1) acts as a key mediator of hypoxia and a critical promoter of resistance to the anti-PD-(L)1. Targeting hypoxia or HIF-1 can thus be an effective strategy for reinvigoration of cellular immunity against cancer. Among various strategies presented so far, the key focus is over vascular normalization, which is an approach highly effective for reducing the rate of hypoxia, increasing drug delivery into the tumor area, and boosting the efficacy of anti-PD-(L)1.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Islamic Republic of Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Islamic Republic of Iran
| | - Ebrahim Kharazinejad
- Department of Anatomy, Faculty of Medicine, Abadan University of Medical Sciences, Abadan, Islamic Republic of Iran.
| |
Collapse
|
29
|
Wang L, Liu Y, Dai Y, Tang X, Yin T, Wang C, Wang T, Dong L, Shi M, Qin J, Xue M, Cao Y, Liu J, Liu P, Huang J, Wen C, Zhang J, Xu Z, Bai F, Deng X, Peng C, Chen H, Jiang L, Chen S, Shen B. Single-cell RNA-seq analysis reveals BHLHE40-driven pro-tumour neutrophils with hyperactivated glycolysis in pancreatic tumour microenvironment. Gut 2023; 72:958-971. [PMID: 35688610 PMCID: PMC10086491 DOI: 10.1136/gutjnl-2021-326070] [Citation(s) in RCA: 122] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 05/27/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Innate immunity plays important roles in pancreatic ductal adenocarcinoma (PDAC), as non-T-cell-enriched tumour. Neutrophils are major players in innate immune system. Here, we aimed to explore the heterogeneity and pro-tumour mechanisms of neutrophils in PDAC. DESIGN We analysed single-cell transcriptomes of peripheral blood polymorphonuclear leucocytes (PMNs) and tumour-infiltrating immune cells from five patients with PDAC, and performed immunofluorescence/immunohistochemistry staining, multi-omics analysis and in vitro experiments to validate the discoveries of bioinformatics analysis. RESULTS Exploration of the heterogeneity of tumour-associated neutrophils (TANs) revealed a terminally differentiated pro-tumour subpopulation (TAN-1) associated with poor prognosis, an inflammatory subpopulation (TAN-2), a population of transitional stage that have just migrated to tumour microenvironment (TAN-3) and a subpopulation preferentially expressing interferon-stimulated genes (TAN-4). Glycolysis signature was upregulated along neutrophil transition trajectory, and TAN-1 was featured with hyperactivated glycolytic activity. The glycolytic switch of TANs was validated by integrative multi-omics approach of transcriptomics, proteomics and metabolomics analysis. Activation of glycolytic activity by LDHA overexpression induced immunosuppression and pro-tumour functions in neutrophil-like differentiated HL-60 (dHL-60) cells. Mechanistic studies revealed BHLHE40, downstream to hypoxia and endoplasmic reticulum stress, was a key regulator in polarisation of neutrophils towards TAN-1 phenotype, and direct transcriptional regulation of BHLHE40 on TAN-1 marker genes was demonstrated by chromatin immunoprecipitation assay. Pro-tumour and immunosuppression functions were observed in dHL-60 cells overexpressing BHLHE40. Importantly, immunohistochemistry analysis of PDAC tissues revealed the unfavourable prognostic value of BHLHE40+ neutrophils. CONCLUSION The dynamic properties of TANs revealed by this study will be helpful in advancing PDAC therapy targeting innate immunity.
Collapse
Affiliation(s)
- Liwen Wang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Yihao Liu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Yuting Dai
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xiaomei Tang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Tong Yin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Chaofu Wang
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Ting Wang
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Lei Dong
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Minmin Shi
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Jiejie Qin
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Meilin Xue
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Yizhi Cao
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Jia Liu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Pengyi Liu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Jinyan Huang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Center for Biomedical Big Data, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Chenlei Wen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Jun Zhang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Zhiwei Xu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Fan Bai
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing, People's Republic of China
| | - Xiaxing Deng
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Chenghong Peng
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Hao Chen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Lingxi Jiang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Saijuan Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Baiyong Shen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| |
Collapse
|
30
|
Mortezaee K, Majidpoor J. The impact of hypoxia on extracellular vesicle secretome profile of cancer. Med Oncol 2023; 40:128. [PMID: 36964452 DOI: 10.1007/s12032-023-01995-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/09/2023] [Indexed: 03/26/2023]
Abstract
Extracellular vesicles (EVs) are emerging as key mediators of cell-to-cell communications and signal transporters between tumor and stroma, and hypoxia is a critical characteristic of tumor microenvironment (TME) in solid cancers. Hypoxia stimulates tumor cells to generate and secrete more EVs, and the EVs shed from cancer transfer biological information to boost hypoxia and hypoxia inducible factor (HIF) functionality. Hypoxia alters EV secretome profile to carry pro-tumorigenic factors for promoting numerous tumor-related processes including increased cancer cell proliferation and survival, immune escape, aberrant angiogenesis, and invasion and metastasis. Exosomal hypoxia inducible factor (HIF)-1α is an essential driver of epithelial-mesenchymal transition (EMT) and stemness profile in cancer. Hypoxic cancer-derived EVs are also contributed to therapy resistance. In fact, EVs are messengers of hypoxic tolerance in cancer, which enable adaptation of tumor cells to changes occurring within TME for their further resistance and metastasis. Tracing EVs shed from hypoxic tumor cells into plasma provide important information about the genomic signature of cancer. In this review, we aimed to discuss about key tumorigenic events promoted by inter-connections between hypoxia and EVs, mainly exosomes, secreted into tumor area focusing on key hallmarks of cancer.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
31
|
Zhang C, Fei Y, Wang H, Hu S, Liu C, Hu R, Du Q. CAFs orchestrates tumor immune microenvironment—A new target in cancer therapy? Front Pharmacol 2023; 14:1113378. [PMID: 37007004 PMCID: PMC10064291 DOI: 10.3389/fphar.2023.1113378] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/23/2023] [Indexed: 03/15/2023] Open
Abstract
Cancer immunotherapy has opened a new landscape in cancer treatment, however, the poor specificity and resistance of most targeted therapeutics have limited their therapeutic efficacy. In recent years, the role of CAFs in immune regulation has been increasingly noted as more evidence has been uncovered regarding the link between cancer-associated fibroblasts (CAFs) and the evolutionary process of tumor progression. CAFs interact with immune cells to shape the tumor immune microenvironment (TIME) that favors malignant tumor progression, a crosstalk process that leads to the failure of cancer immunotherapies. In this review, we outline recent advances in the immunosuppressive function of CAFs, highlight the mechanisms of CAFs-immune cell interactions, and discuss current CAF-targeted therapeutic strategies for future study.
Collapse
Affiliation(s)
- Chunxue Zhang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yuxiang Fei
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hui Wang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Sheng Hu
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Chao Liu
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Qianming Du, ; Rong Hu, ; Chao Liu,
| | - Rong Hu
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Jiangsu Nanjing, China
- *Correspondence: Qianming Du, ; Rong Hu, ; Chao Liu,
| | - Qianming Du
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Qianming Du, ; Rong Hu, ; Chao Liu,
| |
Collapse
|
32
|
Yuan Z, Li Y, Zhang S, Wang X, Dou H, Yu X, Zhang Z, Yang S, Xiao M. Extracellular matrix remodeling in tumor progression and immune escape: from mechanisms to treatments. Mol Cancer 2023; 22:48. [PMID: 36906534 PMCID: PMC10007858 DOI: 10.1186/s12943-023-01744-8] [Citation(s) in RCA: 273] [Impact Index Per Article: 136.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/11/2023] [Indexed: 03/13/2023] Open
Abstract
The malignant tumor is a multi-etiological, systemic and complex disease characterized by uncontrolled cell proliferation and distant metastasis. Anticancer treatments including adjuvant therapies and targeted therapies are effective in eliminating cancer cells but in a limited number of patients. Increasing evidence suggests that the extracellular matrix (ECM) plays an important role in tumor development through changes in macromolecule components, degradation enzymes and stiffness. These variations are under the control of cellular components in tumor tissue via the aberrant activation of signaling pathways, the interaction of the ECM components to multiple surface receptors, and mechanical impact. Additionally, the ECM shaped by cancer regulates immune cells which results in an immune suppressive microenvironment and hinders the efficacy of immunotherapies. Thus, the ECM acts as a barrier to protect cancer from treatments and supports tumor progression. Nevertheless, the profound regulatory network of the ECM remodeling hampers the design of individualized antitumor treatment. Here, we elaborate on the composition of the malignant ECM, and discuss the specific mechanisms of the ECM remodeling. Precisely, we highlight the impact of the ECM remodeling on tumor development, including proliferation, anoikis, metastasis, angiogenesis, lymphangiogenesis, and immune escape. Finally, we emphasize ECM "normalization" as a potential strategy for anti-malignant treatment.
Collapse
Affiliation(s)
- Zhennan Yuan
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yingpu Li
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Sifan Zhang
- Department of Neurobiology, Harbin Medical University, Harbin, 150081, China
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - He Dou
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xi Yu
- Department of Gynecological Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Zhiren Zhang
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.,Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin, 150001, China
| | - Shanshan Yang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150000, China.
| | - Min Xiao
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
33
|
Gao C, Kwong CHT, Wang Q, Kam H, Xie B, Lee SMY, Chen G, Wang R. Conjugation of Macrophage-Mimetic Microalgae and Liposome for Antitumor Sonodynamic Immunotherapy via Hypoxia Alleviation and Autophagy Inhibition. ACS NANO 2023; 17:4034-4049. [PMID: 36739531 DOI: 10.1021/acsnano.3c00041] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Sonodynamic therapy (SDT) is a noninvasive technique for local antitumor treatment; however, its clinical application is often limited by the low tumor accumulation of SDT agents, tumor's hypoxic microenvironment, and cytoprotective effects of autophagy. To address these issues, herein we developed surface-engineered chlorella (Chl, a green algae) as a targeted drug carrier and sustainable oxygen supplier (via photosynthesis) for significantly improved SDT via hypoxia alleviation as well as autophagy inhibition of chloroquine phosphate. In this design, the macrophage membrane was coated onto Chl to form macrophage-mimetic Chl (MChl) to increase its biocompatibility and targeted tumor accumulation driven by the inflammatory-homing effects of macrophage membranes. In addition, the membrane coating on Chl allowed lipid insertion to yield β-cyclodextrin (β-CD) modified MChl (CD-MChl). Subsequently, supramolecular conjugates of MChl-NP were constructed via host-guest interactions between CD-MChl and adamantane (ADA)-modified liposome (ADA-NP), and the anchored liposome went with CD-MChl hand-in-hand to the tumor tissues for co-delivery of Chl, hematoporphyrin, and chloroquine phosphate (loaded in ADA-NP). The synergistic therapy achieved via local oxygenation, SDT, and autophagy inhibition maximally improved the therapeutic efficacy of MChl-CQ-HP-NP against melanoma. Tumor rechallenging results revealed that the changes of tumor microenvironment including hypoxia alleviation, SDT induced immunogenic cell death, and autophagy inhibition collectively induced a strong antitumor immune response and memory.
Collapse
Affiliation(s)
- Cheng Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR 999078, China
| | - Cheryl H T Kwong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR 999078, China
| | - Qingfu Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR 999078, China
| | - Hiotong Kam
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR 999078, China
| | - Beibei Xie
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR 999078, China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR 999078, China
| | - Guosong Chen
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR 999078, China
| |
Collapse
|
34
|
Mortezaee K, Majidpoor J, Najafi S, Tasa D. Bypassing anti-PD-(L)1 therapy: Mechanisms and management strategies. Biomed Pharmacother 2023; 158:114150. [PMID: 36577330 DOI: 10.1016/j.biopha.2022.114150] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Resistance to immune checkpoint inhibitors (ICIs) is a major issue of the current era in cancer immunotherapy. Immune evasion is a multi-factorial event, which occurs generally at a base of cold immunity. Despite advances in the field, there are still unsolved challenges about how to combat checkpoint hijacked by tumor cells and what are complementary treatment strategies to render durable anti-tumor outcomes. A point is that anti-programed death-1 receptor (PD-1)/anti-programmed death-ligand 1 (PD-L1) is not the solo path of immune escape, and responses in many types of solid tumors to the PD-1/PD-L1 inhibitors are not satisfactory. Thus, seeking mechanisms inter-connecting tumor with its immune ecosystem nearby unravel more about resistance mechanisms so as to develop methods for sustained reinvigoration of immune activity against cancer. In this review, we aimed to discuss about common and specific paths taken by tumor cells to evade immune surveillance, describing novel detection strategies, as well as suggesting some approaches to recover tumor sensitivity to the anti-PD-(L)1 therapy based on the current knowledge.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Tasa
- Hepatopancreatobiliary Surgery Fellowship, Organ Transplantation Group, Massih Daneshvari Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Surgery, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
35
|
Brummel K, Eerkens AL, de Bruyn M, Nijman HW. Tumour-infiltrating lymphocytes: from prognosis to treatment selection. Br J Cancer 2023; 128:451-458. [PMID: 36564565 PMCID: PMC9938191 DOI: 10.1038/s41416-022-02119-4] [Citation(s) in RCA: 105] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Tumour-infiltrating lymphocytes (TILs) are considered crucial in anti-tumour immunity. Accordingly, the presence of TILs contains prognostic and predictive value. In 2011, we performed a systematic review and meta-analysis on the prognostic value of TILs across cancer types. Since then, the advent of immune checkpoint blockade (ICB) has renewed interest in the analysis of TILs. In this review, we first describe how our understanding of the prognostic value of TIL has changed over the last decade. New insights on novel TIL subsets are discussed and give a broader view on the prognostic effect of TILs in cancer. Apart from prognostic value, evidence on the predictive significance of TILs in the immune therapy era are discussed, as well as new techniques, such as machine learning that strive to incorporate these predictive capacities within clinical trials.
Collapse
Affiliation(s)
- Koen Brummel
- University of Groningen, University Medical Center Groningen, Department of Obstetrics and Gynecology, Groningen, The Netherlands
| | - Anneke L Eerkens
- University of Groningen, University Medical Center Groningen, Department of Obstetrics and Gynecology, Groningen, The Netherlands
| | - Marco de Bruyn
- University of Groningen, University Medical Center Groningen, Department of Obstetrics and Gynecology, Groningen, The Netherlands
| | - Hans W Nijman
- University of Groningen, University Medical Center Groningen, Department of Obstetrics and Gynecology, Groningen, The Netherlands.
| |
Collapse
|
36
|
Tang YY, Wang DC, Wang YQ, Huang AF, Xu WD. Emerging role of hypoxia-inducible factor-1α in inflammatory autoimmune diseases: A comprehensive review. Front Immunol 2023; 13:1073971. [PMID: 36761171 PMCID: PMC9905447 DOI: 10.3389/fimmu.2022.1073971] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/13/2022] [Indexed: 01/26/2023] Open
Abstract
Hypoxia-inducible factor-1α (HIF-1α) is a primary metabolic sensor, and is expressed in different immune cells, such as macrophage, dendritic cell, neutrophil, T cell, and non-immune cells, for instance, synovial fibroblast, and islet β cell. HIF-1α signaling regulates cellular metabolism, triggering the release of inflammatory cytokines and inflammatory cells proliferation. It is known that microenvironment hypoxia, vascular proliferation, and impaired immunological balance are present in autoimmune diseases. To date, HIF-1α is recognized to be overexpressed in several inflammatory autoimmune diseases, such as systemic lupus erythematosus (SLE), rheumatoid arthritis, and function of HIF-1α is dysregulated in these diseases. In this review, we narrate the signaling pathway of HIF-1α and the possible immunopathological roles of HIF-1α in autoimmune diseases. The collected information will provide a theoretical basis for the familiarization and development of new clinical trials and treatment based on HIF-1α and inflammatory autoimmune disorders in the future.
Collapse
Affiliation(s)
- Yang-Yang Tang
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Da-Cheng Wang
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - You-Qiang Wang
- Department of Laboratory Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - An-Fang Huang
- Department of Rheumatology and Immunology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wang-Dong Xu
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China,*Correspondence: Wang-Dong Xu,
| |
Collapse
|
37
|
Peng C, Ye H, li Z, Duan X, Yang W, Yi Z. Multi-omics characterization of a scoring system to quantify hypoxia patterns in patients with head and neck squamous cell carcinoma. J Transl Med 2023; 21:15. [PMID: 36627705 PMCID: PMC9830846 DOI: 10.1186/s12967-022-03869-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 12/29/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The 5-year survival rate of patients with head and neck squamous cell carcinoma (HNSCC) remains < 50%. Hypoxia patterns are a hallmark of HNSCC that are associated with its occurrence and progression. However, the precise role of hypoxia during HNSCC, such as the relationship between hypoxia, tumor immune landscape and cell communication orchestration remains largely unknown. The current study integrated data from bulk and single-cell RNA sequencing analyses to define the relationship between hypoxia and HNSCC. METHODS A scoring system named the hypoxia score (HS) was constructed based on hypoxia-related genes (HRGs) expression. The predictive value of HS response for patient outcomes and different treatments was evaluated. Single-cell datasets and cell communication were utilized to rule out cell populations which hypoxia targeted on. RESULTS The survival outcomes, immune/Estimate scores, responses to targeted inhibitors, and chemotherapeutic, and immunotherapy responses were distinct between a high HS group and a low HS group (all P < 0.05). Single-cell datasets showed different distributions of HS in immune cell populations (P < 0.05). Furthermore, HLA-DPA1/CD4 axis was identified as a unique interaction between CD4 + T Conv and pDC cells. CONCLUSIONS Altogether, the quantification for hypoxia patterns is a potential biomarker for prognosis, individualized chemotherapeutic and immunotherapy strategies. The portrait of cell communication characteristics over the HNSCC ecosystem enhances the understanding of hypoxia patterns in HNSCC.
Collapse
Affiliation(s)
- Cong Peng
- grid.459540.90000 0004 1791 4503Department of Otolaryngology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Huiping Ye
- grid.459540.90000 0004 1791 4503Department of Otolaryngology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Zhengyang li
- grid.459540.90000 0004 1791 4503Department of Otolaryngology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Xiaofeng Duan
- grid.459540.90000 0004 1791 4503Department of Oral and Maxillofacial Surgery, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Wen Yang
- grid.452244.1Department of Pathology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhuguang Yi
- grid.459540.90000 0004 1791 4503Department of Otolaryngology, Guizhou Provincial People’s Hospital, Guiyang, China
| |
Collapse
|
38
|
Mortezaee K, Majidpoor J. Transforming growth factor-β signalling in tumour resistance to the anti-PD-(L)1 therapy: Updated. J Cell Mol Med 2023; 27:311-321. [PMID: 36625080 PMCID: PMC9889687 DOI: 10.1111/jcmm.17666] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/19/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
Low frequency of durable responses in patients treated with immune checkpoint inhibitors (ICIs) demands for taking complementary strategies in order to boost immune responses against cancer. Transforming growth factor-β (TGF-β) is a multi-tasking cytokine that is frequently expressed in tumours and acts as a critical promoter of tumour hallmarks. TGF-β promotes an immunosuppressive tumour microenvironment (TME) and defines a bypass mechanism to the ICI therapy. A number of cells within the stroma of tumour are influenced from TGF-β activity. There is also evidence of a relation between TGF-β with programmed death-ligand 1 (PD-L1) expression within TME, and it influences the efficacy of anti-programmed death-1 receptor (PD-1) or anti-PD-L1 therapy. Combination of TGF-β inhibitors with anti-PD(L)1 has come to the promising outcomes, and clinical trials are under way in order to use agents with bifunctional capacity and fusion proteins for bonding TGF-β traps with anti-PD-L1 antibodies aiming at reinvigorating immune responses and promoting persistent responses against advanced stage cancers, especially tumours with immunologically cold ecosystem.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of MedicineKurdistan University of Medical SciencesSanandajIran
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Diseases Research CenterGonabad University of Medical SciencesGonabadIran
| |
Collapse
|
39
|
Xia Z, Quan Y. Effect of tumor microenvironment on ferroptosis: inhibition or promotion. Front Oncol 2023; 13:1155511. [PMID: 37213276 PMCID: PMC10196176 DOI: 10.3389/fonc.2023.1155511] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/24/2023] [Indexed: 05/23/2023] Open
Abstract
Ferroptosis is a type of lipid peroxidation-induced, iron-dependent programmed cell death. Emerging evidence suggests that ferroptosis is intimately connected to tumorigenesis, development, treatment and plays a major role in tumor immune regulation. This study focused on the connection between ferroptosis and immune regulation, which may offer a theoretical basis for targeting ferroptosis and tumor immunotherapy.
Collapse
Affiliation(s)
- Zhengzhen Xia
- The First Clinical Medical School, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yi Quan
- The First Clinical Medical School, Guangdong Medical University, Zhanjiang, Guangdong, China
- Department of Oncology Medical Center, The First People’s Hospital of Zhaoqing, Zhaoqing, Guangdong, China
- *Correspondence: Yi Quan,
| |
Collapse
|
40
|
Smith PJ, McKeown SR, Patterson LH. Targeting DNA topoisomerase IIα (TOP2A) in the hypoxic tumour microenvironment using unidirectional hypoxia-activated prodrugs (uHAPs). IUBMB Life 2023; 75:40-54. [PMID: 35499745 PMCID: PMC10084299 DOI: 10.1002/iub.2619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/24/2022] [Accepted: 04/03/2022] [Indexed: 12/29/2022]
Abstract
The hypoxic tumour microenvironment (hTME), arising from inadequate and chaotic vascularity, can present a major obstacle for the treatment of solid tumours. Hypoxic tumour cells compromise responses to treatment since they can generate resistance to radiotherapy, chemotherapy and immunotherapy. The hTME impairs the delivery of a range of anti-cancer drugs, creates routes for metastasis and exerts selection pressures for aggressive phenotypes; these changes potentially occur within an immunosuppressed environment. Therapeutic strategies aimed at the hTME include targeting the molecular changes associated with hypoxia. An alternative approach is to exploit the prevailing lack of oxygen as a principle for the selective activation of prodrugs to target cellular components within the hTME. This review focuses on the design concepts and rationale for the use of unidirectional Hypoxia-Activated Prodrugs (uHAPs) to target the hTME as exemplified by the uHAPs AQ4N and OCT1002. These agents undergo irreversible reduction in a hypoxic environment to active forms that target DNA topoisomerase IIα (TOP2A). This nuclear enzyme is essential for cell division and is a recognised chemotherapeutic target. An activated uHAP interacts with the enzyme-DNA complex to induce DNA damage, cell cycle arrest and tumour cell death. uHAPs are designed to overcome the shortcomings of conventional HAPs and offer unique pharmacodynamic properties for effective targeting of TOP2A in the hTME. uHAP therapy in combination with standard of care treatments has the potential to enhance outcomes by co-addressing the therapeutic challenge presented by the hTME.
Collapse
Affiliation(s)
- Paul J Smith
- Cancer and Genetics Division, School of Medicine, Cardiff University, Cardiff, UK
| | | | - Laurence H Patterson
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Bradford, UK
| |
Collapse
|
41
|
Najafi S, Majidpoor J, Mortezaee K. The impact of microbiota on PD-1/PD-L1 inhibitor therapy outcomes: A focus on solid tumors. Life Sci 2022; 310:121138. [DOI: 10.1016/j.lfs.2022.121138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 10/02/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
|
42
|
Mortezaee K, Majidpoor J, Najafi S. VISTA immune regulatory effects in bypassing cancer immunotherapy: Updated. Life Sci 2022; 310:121083. [DOI: 10.1016/j.lfs.2022.121083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/04/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022]
|
43
|
Li Y, Azmi AS, Mohammad RM. Deregulated transcription factors and poor clinical outcomes in cancer patients. Semin Cancer Biol 2022; 86:122-134. [PMID: 35940398 DOI: 10.1016/j.semcancer.2022.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/22/2022] [Accepted: 08/04/2022] [Indexed: 01/27/2023]
Abstract
Transcription factors are a group of proteins, which possess DNA-binding domains, bind to DNA strands of promoters or enhancers, and initiate transcription of genes with cooperation of RNA polymerase and other co-factors. They play crucial roles in regulating transcription during embryogenesis and development. Their physiological status in different cell types is also important to maintain cellular homeostasis. Therefore, any deregulation of transcription factors will lead to the development of cancer cells and tumor progression. Based on their functions in cancer cells, transcription factors could be either oncogenic or tumor suppressive. Furthermore, transcription factors have been shown to modulate cancer stem cells, epithelial-mesenchymal transition (EMT) and drug response; therefore, measuring deregulated transcription factors is hypothesized to predict treatment outcomes of patients with cancers and targeting deregulated transcription factors could be an encouraging strategy for cancer therapy. Here, we summarize the current knowledge of major deregulated transcription factors and their effects on causing poor clinical outcome of patients with cancer. The information presented here will help to predict the prognosis and drug response and to design novel drugs and therapeutic strategies for the treatment of cancers by targeting deregulated transcription factors.
Collapse
Affiliation(s)
- Yiwei Li
- Karmanos Cancer Institute and Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Asfar S Azmi
- Karmanos Cancer Institute and Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ramzi M Mohammad
- Karmanos Cancer Institute and Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
44
|
Extracellular vesicle isolation, purification and evaluation in cancer diagnosis. Expert Rev Mol Med 2022; 24:e41. [PMID: 36268744 DOI: 10.1017/erm.2022.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Strategies for non-invasive biomarker discovery in early detection of cancer are an urgent need. Extracellular vesicles (EVs) have generated increasing attention from the scientific community and are under intensive investigations due to their unique biological profiles and their non-invasive nature. EVs are membrane-enclosed vesicles with variable sizes and function. Such vesicles are actively secreted from multiple cell types and are considered as key vehicles for inter-cellular communications and signalling. The stability and potential to easily cross biological barriers enable EVs for exerting durable effects on target cells. These along with easy access to such vesicles, the consistent secretion from tumour during all stages of tumorigenesis and their content providing a reservoir of molecules as well as mirroring the identity of the cell of origin are virtues that have made EVs appealing to be assessed in liquid biopsy approaches and for using as a promising resource of biomarkers in cancer diagnosis and therapy and monitoring targeted cancer therapy. Early detection of EVs will guide time-scheduled personalised therapy. Surveying reliable and sensitive methods for rapid isolation of EVs from biofluids, the purity of isolated vesicles and their molecular profiling and marker specification for clinical translation in patients with cancer are issues in the area and the hot topics of many recent studies. Here, the focus is over methods for EV isolation and stratification for digging more information about liquid biopsy-based diagnosis. Extending knowledge regarding EV-based strategies is a key to validate independent patient follow-up for cancer diagnosis at early stages and inspecting the efficacy of therapeutics.
Collapse
|
45
|
Bai R, Li Y, Jian L, Yang Y, Zhao L, Wei M. The hypoxia-driven crosstalk between tumor and tumor-associated macrophages: mechanisms and clinical treatment strategies. Mol Cancer 2022; 21:177. [PMID: 36071472 PMCID: PMC9454207 DOI: 10.1186/s12943-022-01645-2] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/25/2022] [Indexed: 02/08/2023] Open
Abstract
Given that hypoxia is a persistent physiological feature of many different solid tumors and a key driver for cancer malignancy, it is thought to be a major target in cancer treatment recently. Tumor-associated macrophages (TAMs) are the most abundant immune cells in the tumor microenvironment (TME), which have a large impact on tumor development and immunotherapy. TAMs massively accumulate within hypoxic tumor regions. TAMs and hypoxia represent a deadly combination because hypoxia has been suggested to induce a pro-tumorigenic macrophage phenotype. Hypoxia not only directly affects macrophage polarization, but it also has an indirect effect by altering the communication between tumor cells and macrophages. For example, hypoxia can influence the expression of chemokines and exosomes, both of which have profound impacts on the recipient cells. Recently, it has been demonstrated that the intricate interaction between cancer cells and TAMs in the hypoxic TME is relevant to poor prognosis and increased tumor malignancy. However, there are no comprehensive literature reviews on the molecular mechanisms underlying the hypoxia-mediated communication between tumor cells and TAMs. Therefore, this review has the aim to collect all recently available data on this topic and provide insights for developing novel therapeutic strategies for reducing the effects of hypoxia.
Collapse
Affiliation(s)
- Ruixue Bai
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, 110122, People's Republic of China.,Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Yunong Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, 110122, People's Republic of China
| | - Lingyan Jian
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Yuehui Yang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China. .,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, 110122, People's Republic of China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China. .,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, 110122, People's Republic of China. .,Shenyang Kangwei Medical Laboratory Analysis Co. LTD, Shenyang, 110000, People's Republic of China.
| |
Collapse
|
46
|
Epithelial-mesenchymal transition in cancer stemness and heterogeneity: updated. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:193. [PMID: 36071302 DOI: 10.1007/s12032-022-01801-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 07/15/2022] [Indexed: 10/14/2022]
Abstract
Epithelial-mesenchymal transition (EMT) as a trans-differentiation program and a key process in tumor progression is linked positively with increased expansion of cancer stem cells and cells with stem-like properties. This is mediated through modulation of critical tumorigenic events and is positively correlated with hypoxic conditions in tumor microenvironment. The presence of cells eliciting diverse phenotypical states inside tumor is representative of heterogeneity and higher tumor resistance to therapy. In this review, we aimed to discuss about the current understanding toward EMT, stemness, and heterogeneity in tumors of solid organs, their contribution to the key tumorigenic events along with major signaling pathway involved, and, finally, to suggest some strategies to target these critical events.
Collapse
|
47
|
Zhang M, Wei T, Zhang X, Guo D. Targeting lipid metabolism reprogramming of immunocytes in response to the tumor microenvironment stressor: A potential approach for tumor therapy. Front Immunol 2022; 13:937406. [PMID: 36131916 PMCID: PMC9483093 DOI: 10.3389/fimmu.2022.937406] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/12/2022] [Indexed: 12/26/2022] Open
Abstract
The tumor microenvironment (TME) has become a major research focus in recent years. The TME differs from the normal extracellular environment in parameters such as nutrient supply, pH value, oxygen content, and metabolite abundance. Such changes may promote the initiation, growth, invasion, and metastasis of tumor cells, in addition to causing the malfunction of tumor-infiltrating immunocytes. As the neoplasm develops and nutrients become scarce, tumor cells transform their metabolic patterns by reprogramming glucose, lipid, and amino acid metabolism in response to various environmental stressors. Research on carcinoma metabolism reprogramming suggests that like tumor cells, immunocytes also switch their metabolic pathways, named “immunometabolism”, a phenomenon that has drawn increasing attention in the academic community. In this review, we focus on the recent progress in the study of lipid metabolism reprogramming in immunocytes within the TME and highlight the potential target molecules, pathways, and genes implicated. In addition, we discuss hypoxia, one of the vital altered components of the TME that partially contribute to the initiation of abnormal lipid metabolism in immune cells. Finally, we present the current immunotherapies that orchestrate a potent antitumor immune response by mediating the lipid metabolism of immunocytes, highlight the lipid metabolism reprogramming capacity of various immunocytes in the TME, and propose promising new strategies for use in cancer therapy.
Collapse
Affiliation(s)
- Ming Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Digestive Organ Transplantation, Zhengzhou, China
| | - Tingju Wei
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaodan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Digestive Organ Transplantation, Zhengzhou, China
| | - Danfeng Guo
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Digestive Organ Transplantation, Zhengzhou, China
- *Correspondence: Danfeng Guo,
| |
Collapse
|
48
|
Mortezaee K, Majidpoor J. Dysregulated metabolism: A friend-to-foe skewer of macrophages. Int Rev Immunol 2022:1-17. [DOI: 10.1080/08830185.2022.2095374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
49
|
Hu J, Li X, Yang L, Li H. Hypoxia, a key factor in the immune microenvironment. Biomed Pharmacother 2022; 151:113068. [PMID: 35676780 DOI: 10.1016/j.biopha.2022.113068] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/15/2022] [Accepted: 04/28/2022] [Indexed: 11/29/2022] Open
Abstract
The physical and chemical pressures in the tumor microenvironment (TME) play an important role in tumor development by regulating stromal elements, including immune cells. Hypoxia can induce a cascade of events in tumor initiation and development via immune regulation. As a dangerous factor, hypoxia activates multiple signaling pathways to reshape the immune microenvironment, leading to immunosuppression. Consequently, targeting hypoxia in the TME is a potential strategy to prevent immune escape and inhibit malignant tumor progression. In this review, we summarized the role of hypoxia-induced factors in the tumor immune escape process and provide a novel pathway to restrain tumor progression and development.
Collapse
Affiliation(s)
- Jingyao Hu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China.
| | - Xinyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China.
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China.
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China.
| |
Collapse
|
50
|
NK and cells with NK-like activities in cancer immunotherapy-clinical perspectives. Med Oncol 2022; 39:131. [PMID: 35716327 DOI: 10.1007/s12032-022-01735-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/13/2022] [Indexed: 01/10/2023]
Abstract
Natural killer (NK) cells are lymphoid cells of innate immunity that take important roles in immune surveillance. NK cells are considered as a bridge between innate and adaptive immunity, and their infiltration into tumor area is related positively with prolonged patient survival. They are defined as CD16+ CD56+ CD3- cells in clinic. NK cells promote cytolytic effects on target cells and induce their apoptosis. Loss of NK cell cytotoxic activity and reduction in the number of activating receptors are the current issues for application of such cells in cellular immunotherapy, which resulted in the diminished long-term effects. The focus of this review is to discuss about the activity of NK cells and cells with NK-like activity including natural killer T (NKT), cytokine-induced killer (CIK) and lymphokine-activated killer (LAK) cells in immunotherapy of human solid cancers.
Collapse
|