1
|
Gao H, Qu L, Li M, Guan X, Zhang S, Deng X, Wang J, Xing F. Unlocking the potential of chimeric antigen receptor T cell engineering immunotherapy: Long road to achieve precise targeted therapy for hepatobiliary pancreatic cancers. Int J Biol Macromol 2025; 297:139829. [PMID: 39814310 DOI: 10.1016/j.ijbiomac.2025.139829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 01/03/2025] [Accepted: 01/11/2025] [Indexed: 01/18/2025]
Abstract
Innovative therapeutic strategies are urgently needed to address the ongoing global health concern of hepatobiliary pancreatic malignancies. This review summarizes the latest and most comprehensive research of chimeric antigen receptor (CAR-T) cell engineering immunotherapy for treating hepatobiliary pancreatic cancers. Commencing with an exploration of the distinct anatomical location and the immunosuppressive, hypoxic tumor microenvironment (TME), this review critically assesses the limitations of current CAR-T therapy in hepatobiliary pancreatic cancers and proposes corresponding solutions. Various studies aim at enhancing CAR-T cell efficacy in these cancers through improving T cell persistence, enhancing antigen specificity and reducing tumor heterogeneity, also modulating the immunosuppressive and hypoxic TME. Additionally, the review examines the application of emerging nanoparticles and biotechnologies utilized in CAR-T therapy for these cancers. The results suggest that constructing optimized CAR-T cells to overcome physical barrier, manipulating the TME to relieve immunosuppression and hypoxia, designing CAR-T combination therapies, and selecting the most suitable delivery strategies, all together could collectively enhance the safety of CAR-T engineering and advance the effectiveness of adaptive cell therapy for hepatobiliary pancreatic cancers.
Collapse
Affiliation(s)
- Hongli Gao
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Lianyue Qu
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001, China
| | - Mu Li
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xin Guan
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Shuang Zhang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xin Deng
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Jin Wang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Fei Xing
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
2
|
Liu B, Tian H, Momeni MR. The interplay of exercise and green tea: a new road in cancer therapy. Cancer Cell Int 2025; 25:6. [PMID: 39773739 PMCID: PMC11705833 DOI: 10.1186/s12935-024-03632-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025] Open
Abstract
Exercise is one of the most important activities for every individual due to its proven health beneficials. Several investigations have highlighted the advantageous impacts of aerobic exercise, largely attributed to its capacity to enhance the body's capability to defend against threats against oxidative stress. The information currently accessible suggests that adding regular aerobic exercise to a daily routine greatly decreases the chances of developing serious cancer and passing away. An unevenness in the levels of free radicals and the body's antioxidant defenses, made up of enzyme and non-enzyme antioxidants, results in oxidative pressure. Generally, an imbalance in the levels of oxidative stress triggers the creation of harmful reactive oxygen or nitrogen compounds, causing the development or progression of numerous ailments, including cancer. The equilibrium between pro-oxidant and antioxidant substances is a direct indicator of this imbalance. Green tea and its derivatives are rich sources of bioactive substances such as flavonoids and polyphenols which possess antioxidant abilities. Moreover, modulation of epigenetic targets as well as inflammatory pathways including ERK1/2 and NF-κB are other proposed mechanisms for its antioxidant activity. Recent studies demonstrate the promise of green tea as an antioxidant, showing its ability to decrease the likelihood of developing cancer by impacting actions like cell growth, blood vessel formation, and spread of cancer cells. This summary will concentrate on the complex network of different pathways related to physical activity and consumption of green tea. In particular, the focus of this research will be on examining how oxidative stress contributes to health and investigating the potential antioxidant properties of green tea, and the interconnected relationship between exercise and green tea in the treatment of cancer. Elucidation of these different pathways would help scientists for development of better therapeutic targets and further increase of current anticancer agents efficiency.
Collapse
Affiliation(s)
- Bing Liu
- Henan University of Chinese Medicine, Zhengzhou, 450000, Henan, China
| | - Heyu Tian
- Henan University of Chinese Medicine, Zhengzhou, 450000, Henan, China.
| | | |
Collapse
|
3
|
Buzaglo GBB, Telles GD, Araújo RB, Junior GDS, Ruberti OM, Ferreira MLV, Derchain SFM, Vechin FC, Conceição MS. The Therapeutic Potential of Physical Exercise in Cancer: The Role of Chemokines. Int J Mol Sci 2024; 25:13740. [PMID: 39769501 PMCID: PMC11678861 DOI: 10.3390/ijms252413740] [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: 07/17/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 01/11/2025] Open
Abstract
The global increase in cancer cases and mortality has been associated with inflammatory processes, in which chemokines play crucial roles. These molecules, a subfamily of cytokines, are essential for the migration, adhesion, interaction, and positioning of immune cells throughout the body. Chemokines primarily originate in response to pathogenic stimuli and inflammatory cytokines. They are expressed by lymphocytes in the bloodstream and are divided into four classes (CC, CXC, XC, and CX3C), playing multifaceted roles in the tumor environment (TME). In the TME, chemokines regulate immune behavior by recruiting cells such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), which promote tumor survival. Additionally, they directly influence tumor behavior, promoting pathological angiogenesis, invasion, and metastasis. On the other hand, chemokines can also induce antitumor responses by mobilizing CD8+ T cells and natural killer (NK) cells to the tumor, reducing pro-inflammatory chemokines and enhancing essential antitumor responses. Given the complex interaction between chemokines, the immune system, angiogenic factors, and metastasis, it becomes evident how important it is to target these pathways in therapeutic interventions to counteract cancer progression. In this context, physical exercise emerges as a promising strategy due to its role modulating the expression of anti-inflammatory chemokines and enhancing the antitumor response. Aerobic and resistance exercises have been associated with a beneficial inflammatory profile in cancer, increased infiltration of CD8+ T cells in the TME, and improvement of intratumoral vasculature. This creates an environment less favorable to tumor growth and supports the circulation of antitumor immune cells and chemokines. Therefore, understanding the impact of exercise on the expression of chemokines can provide valuable insights for therapeutic interventions in cancer treatment and prevention.
Collapse
Affiliation(s)
- Glenda B. B. Buzaglo
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Guilherme D. Telles
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (G.D.T.); (F.C.V.)
| | - Rafaela B. Araújo
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Gilmar D. S. Junior
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Olivia M. Ruberti
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Marina L. V. Ferreira
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Sophie F. M. Derchain
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Campinas, Campinas, Sao Paulo 13083-881, Brazil;
| | - Felipe C. Vechin
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (G.D.T.); (F.C.V.)
| | - Miguel S. Conceição
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| |
Collapse
|
4
|
Brummer C, Singer K, Henrich F, Peter K, Strobl C, Neueder B, Bruss C, Renner K, Pukrop T, Herr W, Aigner M, Kreutz M. The Tumor Metabolite 5'-Deoxy-5'Methylthioadenosine (MTA) Inhibits Maturation and T Cell-Stimulating Capacity of Dendritic Cells. Cells 2024; 13:2114. [PMID: 39768204 PMCID: PMC11727219 DOI: 10.3390/cells13242114] [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: 10/29/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
Metabolite accumulation in the tumor microenvironment fosters immune evasion and limits the efficiency of immunotherapeutic approaches. Methylthioadenosine phosphorylase (MTAP), which catalyzes the degradation of 5'-deoxy-5'methylthioadenosine (MTA), is downregulated in many cancer entities. Consequently, MTA accumulates in the microenvironment of MTAP-deficient tumors, where it is known to inhibit tumor-infiltrating T cells and NK cells. However, the impact of MTA on other intra-tumoral immune cells has not yet been fully elucidated. To study the effects of MTA on dendritic cells (DCs), human monocytes were maturated into DCs with (MTA-DC) or without MTA (co-DC) and analyzed for activation, differentiation, and T cell-stimulating capacity. MTA altered the cytokine secretion profile of monocytes and impaired their maturation into dendritic cells. MTA-DCs produced less IL-12 and showed a more immature-like phenotype characterized by decreased expression of the co-stimulatory molecules CD80, CD83, and CD86 and increased expression of the monocyte markers CD14 and CD16. Consequently, MTA reduced the capability of DCs to stimulate T cells. Mechanistically, the MTA-induced effects on monocytes and DCs were mediated by a mechanism beyond adenosine receptor signaling. These results provide new insights into how altered polyamine metabolism impairs the maturation of monocyte-derived DCs and impacts the crosstalk between T and dendritic cells.
Collapse
Affiliation(s)
- Christina Brummer
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Katrin Singer
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Frederik Henrich
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
- Department of Internal Medicine 5, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Katrin Peter
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Carolin Strobl
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
- Department of Internal Medicine 5, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Bernadette Neueder
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Christina Bruss
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Kathrin Renner
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
- Department of Otorhinolaryngology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Tobias Pukrop
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
- Comprehensive Cancer Center Eastern Bavaria (CCCO), 93053 Regensburg, Germany
- Center of Translational Oncology (CTO), 93053 Regensburg, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Michael Aigner
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
- Department of Internal Medicine 5, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Marina Kreutz
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| |
Collapse
|
5
|
Sirini C, De Rossi L, Moresco MA, Casucci M. CAR T cells in solid tumors and metastasis: paving the way forward. Cancer Metastasis Rev 2024; 43:1279-1296. [PMID: 39316265 DOI: 10.1007/s10555-024-10213-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024]
Abstract
CAR T cell therapy, hailed as a breakthrough in cancer treatment due to its remarkable outcomes in hematological malignancies, encounters significant hurdles when applied to solid tumors. While notable responses to CAR T cells remain sporadic in these patients, challenges persist due to issues such as on-target off-tumor toxicity, difficulties in their trafficking and infiltration into the tumor, and the presence of a hostile and immunosuppressive microenvironment. This review aims to explore recent endeavors aimed at overcoming these obstacles in CAR T cell therapy for solid tumors. Specifically, we will delve into promising strategies for enhancing tumor specificity through antigen targeting, addressing tumor heterogeneity, overcoming physical barriers, and counteracting the immune-suppressive microenvironment.
Collapse
Affiliation(s)
- Camilla Sirini
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Laura De Rossi
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Marta Angiola Moresco
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Monica Casucci
- Innovative Immunotherapies Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy.
| |
Collapse
|
6
|
Hao J, Huang Z, Zhang S, Song K, Wang J, Gao C, Fang Z, Zhang N. Deciphering the multifaceted roles and clinical implications of 2-hydroxyglutarate in cancer. Pharmacol Res 2024; 209:107437. [PMID: 39349213 DOI: 10.1016/j.phrs.2024.107437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/13/2024] [Accepted: 09/24/2024] [Indexed: 10/02/2024]
Abstract
Increasing evidence indicates that 2-hydroxyglutarate (2HG) is an oncometabolite that drives tumour formation and progression. Due to mutations in isocitrate dehydrogenase (IDH) and the dysregulation of other enzymes, 2HG accumulates significantly in tumour cells. Due to its structural similarity to α-ketoglutarate (αKG), accumulated 2HG leads to the competitive inhibition of αKG-dependent dioxygenases (αKGDs), such as KDMs, TETs, and EGLNs. This inhibition results in epigenetic alterations in both tumour cells and the tumour microenvironment. This review comprehensively discusses the metabolic pathways of 2HG and the subsequent pathways influenced by elevated 2HG levels. We will delve into the molecular mechanisms by which 2HG exerts its oncogenic effects, particularly focusing on epigenetic modifications. This review will also explore the various methods available for the detection of 2HG, emphasising both current techniques and emerging technologies. Furthermore, 2HG shows promise as a biomarker for clinical diagnosis and treatment. By integrating these perspectives, this review aims to provide a comprehensive overview of the current understanding of 2HG in cancer biology, highlight the importance of ongoing research, and discuss future directions for translating these findings into clinical applications.
Collapse
Affiliation(s)
- Jie Hao
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Ziyi Huang
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Siyue Zhang
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Kefan Song
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Juncheng Wang
- Advanced Medical Research Institute, Shandong University, Jinan, China
| | - Chao Gao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Zhiqing Fang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Ning Zhang
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
7
|
Del Baldo G, Carai A, Mastronuzzi A. Chimeric antigen receptor adoptive immunotherapy in central nervous system tumors: state of the art on clinical trials, challenges, and emerging strategies to addressing them. Curr Opin Oncol 2024; 36:545-553. [PMID: 38989708 PMCID: PMC11460750 DOI: 10.1097/cco.0000000000001076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
PURPOSE OF REVIEW Central nervous system (CNS) tumors represent a significant unmet medical need due to their enduring burden of high mortality and morbidity. Chimeric antigen receptor (CAR) T-cell therapy emerges as a groundbreaking approach, offering hope for improved treatment outcomes. However, despite its successes in hematological malignancies, its efficacy in solid tumors, including CNS tumors, remains limited. Challenges such as the intricate tumor microenvironment (TME), antigenic heterogeneity, and CAR T-cell exhaustion hinder its effectiveness. This review aims to explore the current landscape of CAR T-cell therapy for CNS tumors, highlighting recent advancements and addressing challenges in achieving therapeutic efficacy. RECENT FINDINGS Innovative strategies aim to overcome the barriers posed by the TME and antigen diversity, prevent CAR T-cell exhaustion through engineering approaches and combination therapies with immune checkpoint inhibitors to improving treatment outcomes. SUMMARY Researchers have been actively working to address these challenges. Moreover, addressing the unique challenges associated with neurotoxicity in CNS tumors requires specialized management strategies. These may include the development of grading systems, monitoring devices, alternative cell platforms and incorporation of suicide genes. Continued research efforts and clinical advancements are paramount to overcoming the existing challenges and realizing the full potential of CAR T-cell therapy in treating CNS tumors.
Collapse
Affiliation(s)
- Giada Del Baldo
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy Bambino Gesù Children's Hospital, IRCCS
- Department of Experimental Medicine, Sapienza University of Rome
| | - Andrea Carai
- Department of Neurosciences, Neurosurgery Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Angela Mastronuzzi
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy Bambino Gesù Children's Hospital, IRCCS
| |
Collapse
|
8
|
Cui J, Chai S, Liu R, Shen G. Targeting PGK1: A New Frontier in Breast Cancer Therapy Under Hypoxic Conditions. Curr Issues Mol Biol 2024; 46:12214-12229. [PMID: 39590319 PMCID: PMC11593045 DOI: 10.3390/cimb46110725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/21/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Breast cancer represents one of the most prevalent malignant neoplasms affecting women, and its pathogenesis has garnered significant scholarly interest. Research indicates that the progression of breast cancer is intricately regulated by glucose metabolism. Under hypoxic conditions within the tumor microenvironment, breast cancer cells generate ATP and essential biosynthetic precursors for growth via the glycolytic pathway. Notably, phosphoglycerate kinase 1 (PGK1) is intimately associated with the regulation of hypoxia-inducible factors in breast cancer and plays a crucial role in modulating glycolytic processes. Further investigation into the role of PGK1 in breast cancer pathogenesis is anticipated to identify novel therapeutic targets and strategies. This review consolidates current research on the regulation of glucose metabolism and the function of PGK1 in breast cancer within hypoxic conditions. It aims to offer a significant theoretical foundation for elucidating the mechanisms underlying breast cancer progression and metastasis, thereby facilitating the development of innovative treatment approaches.
Collapse
Affiliation(s)
- Jiayong Cui
- Clinical Medicine College, Graduate School of Qinghai University, Xining 810000, China; (J.C.); (S.C.); (R.L.)
- Breast Disease Diagnosis and Treatment Cencer, Affiliated Hospital of Qinghai University, Xining 810000, China
| | - Shengjun Chai
- Clinical Medicine College, Graduate School of Qinghai University, Xining 810000, China; (J.C.); (S.C.); (R.L.)
| | - Rui Liu
- Clinical Medicine College, Graduate School of Qinghai University, Xining 810000, China; (J.C.); (S.C.); (R.L.)
- Breast Disease Diagnosis and Treatment Cencer, Affiliated Hospital of Qinghai University, Xining 810000, China
| | - Guoshuang Shen
- Breast Disease Diagnosis and Treatment Cencer, Affiliated Hospital of Qinghai University, Xining 810000, China
| |
Collapse
|
9
|
Kang X, Mita N, Zhou L, Wu S, Yue Z, Babu RJ, Chen P. Nanotechnology in Advancing Chimeric Antigen Receptor T Cell Therapy for Cancer Treatment. Pharmaceutics 2024; 16:1228. [PMID: 39339264 PMCID: PMC11435308 DOI: 10.3390/pharmaceutics16091228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has emerged as a groundbreaking treatment for hematological cancers, yet it faces significant hurdles, particularly regarding its efficacy in solid tumors and concerning associated adverse effects. This review provides a comprehensive analysis of the advancements and ongoing challenges in CAR-T therapy. We highlight the transformative potential of nanotechnology in enhancing CAR-T therapy by improving targeting precision, modulating the immune-suppressive tumor microenvironment, and overcoming physical barriers. Nanotechnology facilitates efficient CAR gene delivery into T cells, boosting transfection efficiency and potentially reducing therapy costs. Moreover, nanotechnology offers innovative solutions to mitigate cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Cutting-edge nanotechnology platforms for real-time monitoring of CAR-T cell activity and cytokine release are also discussed. By integrating these advancements, we aim to provide valuable insights and pave the way for the next generation of CAR-T cell therapies to overcome current limitations and enhance therapeutic outcomes.
Collapse
Affiliation(s)
- Xuejia Kang
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL 36849, USA; (L.Z.); (S.W.)
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA; (N.M.); (Z.Y.); (R.J.B.)
| | - Nur Mita
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA; (N.M.); (Z.Y.); (R.J.B.)
- Faculty of Pharmacy, Mulawarman University, Samarinda 75119, Kalimantan Timur, Indonesia
| | - Lang Zhou
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL 36849, USA; (L.Z.); (S.W.)
| | - Siqi Wu
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL 36849, USA; (L.Z.); (S.W.)
| | - Zongliang Yue
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA; (N.M.); (Z.Y.); (R.J.B.)
| | - R. Jayachandra Babu
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA; (N.M.); (Z.Y.); (R.J.B.)
| | - Pengyu Chen
- Materials Research and Education Center, Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL 36849, USA; (L.Z.); (S.W.)
| |
Collapse
|
10
|
Peng X, He Z, Yuan D, Liu Z, Rong P. Lactic acid: The culprit behind the immunosuppressive microenvironment in hepatocellular carcinoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189164. [PMID: 39096976 DOI: 10.1016/j.bbcan.2024.189164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/05/2024]
Abstract
As a solid tumor with high glycolytic activity, hepatocellular carcinoma (HCC) produces excess lactic acid and increases extracellular acidity, thus forming a unique immunosuppressive microenvironment. L-lactate dehydrogenase (LDH) and monocarboxylate transporters (MCTs) play a very important role in glycolysis. LDH is the key enzyme for lactic acid (LA) production, and MCT is responsible for the cellular import and export of LA. The synergistic effect of the two promotes the formation of an extracellular acidic microenvironment. In the acidic microenvironment of HCC, LA can not only promote the proliferation, survival, transport and angiogenesis of tumor cells but also have a strong impact on immune cells, ultimately leading to an inhibitory immune microenvironment. This article reviews the role of LA in HCC, especially its effect on immune cells, summarizes the progress of LDH and MCT-related drugs, and highlights the potential of immunotherapy targeting lactate combined with HCC.
Collapse
Affiliation(s)
- Xiaopei Peng
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Molecular Imaging Research Center, Central South University, Changsha, Hunan 410013, China
| | - Zhenhu He
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Molecular Imaging Research Center, Central South University, Changsha, Hunan 410013, China
| | - Dandan Yuan
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Molecular Imaging Research Center, Central South University, Changsha, Hunan 410013, China
| | - Zhenguo Liu
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Pengfei Rong
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Molecular Imaging Research Center, Central South University, Changsha, Hunan 410013, China.
| |
Collapse
|
11
|
Hu X, Ouyang W, Chen H, Liu Z, Lai Z, Yao H. Claudin-9 (CLDN9) promotes gastric cancer progression by enhancing the glycolysis pathway and facilitating PD-L1 lactylation to suppress CD8+ T cell anti-tumor immunity. CANCER PATHOGENESIS AND THERAPY 2024. [DOI: 10.1016/j.cpt.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/31/2024]
|
12
|
Song X, Hao C, Li Y, Li Y, Dong H, Wei Q, Wei M, Li H, Zhao L. Chiral inorganic nanomaterials in the tumor microenvironment: A new chapter in cancer therapy. Pharmacol Res 2024; 208:107386. [PMID: 39216840 DOI: 10.1016/j.phrs.2024.107386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/20/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Chirality plays a crucial function in the regulation of normal physiological processes and is widespread in organisms. Chirality can be imparted to nanomaterials, whether they are natural or manmade, through the process of asymmetric assembly and/or grafting of molecular chiral groups or linkers. Chiral inorganic nanomaterials possess unique physical and chemical features that set them apart from regular nanomaterials. They also have the ability to interact with cells and tissues in a specific manner, making them useful in various biomedical applications, particularly in the treatment of tumors. Despite the growing amount of research on chiral inorganic nanomaterials in the tumor microenvironment (TME) and their promising potential applications, there is a lack of literature that comprehensively summarizes the intricate interactions between chiral inorganic nanomaterials and TME. In this review, we introduce the fundamental concept, classification, synthesis methods, and physicochemical features of chiral inorganic nanomaterials. Next, we briefly outline the components of TME, such as T cells, macrophages, dendritic cells, and weak acids, and then discuss the anti-tumor effects of several chiral inorganic nanoparticles targeting these components and their potential for possible application during cancer therapy. Finally, the present challenges faced by chiral inorganic nanomaterials in cancer treatment and their future areas of investigation are disclosed.
Collapse
Affiliation(s)
- Xueyi Song
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Chenjing Hao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Yao Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Yunong Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Hongzhi Dong
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Qian Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Heran Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China.
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| |
Collapse
|
13
|
Nakagawara K, Ando M, Srirat T, Mise-Omata S, Hayakawa T, Ito M, Fukunaga K, Yoshimura A. NR4A ablation improves mitochondrial fitness for long persistence in human CAR-T cells against solid tumors. J Immunother Cancer 2024; 12:e008665. [PMID: 39151930 PMCID: PMC11331892 DOI: 10.1136/jitc-2023-008665] [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] [Accepted: 07/19/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND Antitumor effect of chimeric antigen receptor (CAR)-T cells against solid tumors is limited due to various factors, such as low infiltration rate, poor expansion capacity, and exhaustion of T cells within the tumor. NR4A transcription factors have been shown to play important roles in T-cell exhaustion in mice. However, the precise contribution of each NR4a factor to human T-cell differentiation remains to be clarified. METHODS In this study, we deleted NR4A family factors, NR4A1, NR4A2, and NR4A3, in human CAR-T cells recognizing human epidermal growth factor receptor type 2 (HER2) by using the CRISPR/Cas9 system. We induced T-cell exhaustion in these cells in vitro through repeated co-culturing of CAR-T cells with Her2+A549 lung adenocarcinoma cells and evaluated cell surface markers such as memory and exhaustion phenotypes, proliferative capacity, cytokine production and metabolic activity. We validated the antitumor toxicity of NR4A1/2/3 triple knockout (TKO) CAR-T cells in vivo by transferring CAR-T cells into A549 tumor-bearing immunodeficient mice. RESULTS Human NR4A-TKO CAR-T cells were resistant against exhaustion induced by repeated antigen stimulation in vitro, and maintained higher tumor-killing activity both in vitro and in vivo compared with control CAR-T cells. A comparison of the effectiveness of NR4A single, double, and TKOs demonstrated that triple KO was the most effective in avoiding exhaustion. Furthermore, a strong enhancement of antitumor effects by NR4A TKO was also observed in T cells from various donors including aged persons. Mechanistically, NR4A TKO CAR-T cells showed enhanced mitochondrial oxidative phosphorylation, therefore could persist for longer periods within the tumors. CONCLUSIONS NR4A factors regulate CAR-T cell persistence and stemness through mitochondrial gene expression, therefore NR4A is a highly promising target for the generation of superior CAR-T cells against solid tumors.
Collapse
MESH Headings
- Humans
- Animals
- Mice
- Mitochondria/metabolism
- Immunotherapy, Adoptive/methods
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/immunology
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 2/genetics
- Receptors, Thyroid Hormone/metabolism
- Receptors, Thyroid Hormone/genetics
- Neoplasms/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Xenograft Model Antitumor Assays
- Female
- DNA-Binding Proteins
- Receptors, Steroid
Collapse
Affiliation(s)
- Kensuke Nakagawara
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Ando
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Tanakorn Srirat
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Setsuko Mise-Omata
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Taeko Hayakawa
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Minako Ito
- Division of Allergy and Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| |
Collapse
|
14
|
Zhang T, Zhang X, Chen J, Zhang X, Zhang Y. Harnessing microbial antigens as cancer antigens: a promising avenue for cancer immunotherapy. Front Immunol 2024; 15:1411490. [PMID: 39139570 PMCID: PMC11319170 DOI: 10.3389/fimmu.2024.1411490] [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: 04/03/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024] Open
Abstract
Immunotherapy has revolutionized cancer treatment by leveraging the immune system's innate capabilities to combat malignancies. Despite the promise of tumor antigens in stimulating anti-tumor immune responses, their clinical utility is hampered by limitations in eliciting robust and durable immune reactions, exacerbated by tumor heterogeneity and immune evasion mechanisms. Recent insights into the immunogenic properties of host homologous microbial antigens have sparked interest in their potential for augmenting anti-tumor immunity while minimizing off-target effects. This review explores the therapeutic potential of microbial antigen peptides in tumor immunotherapy, beginning with an overview of tumor antigens and their challenges in clinical translation. We further explore the intricate relationship between microorganisms and tumor development, elucidating the concept of molecular mimicry and its implications for immune recognition of tumor-associated antigens. Finally, we discuss methodologies for identifying and characterizing microbial antigen peptides, highlighting their immunogenicity and prospects for therapeutic application.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Xilong Zhang
- Department of Burns and Plastic Surgery, First People’s Hospital of Xuzhou City, Xuzhou, China
| | - Jianquan Chen
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Xiuwei Zhang
- Department of Burns and Plastic Surgery, First People’s Hospital of Xuzhou City, Xuzhou, China
| | - Yunlei Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
15
|
Chen Q, Sun Y, Li H. Application of CAR-T cell therapy targeting mesothelin in solid tumor treatment. Discov Oncol 2024; 15:289. [PMID: 39023820 PMCID: PMC11258118 DOI: 10.1007/s12672-024-01159-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T-cell therapy is one of the most effective immunotherapies. CAR-T-cell therapy has achieved great success in the treatment of hematological malignancies. However, due to the characteristics of solid malignant tumors, such as on-target effects, off-tumor toxicity, an immunosuppressive tumor microenvironment (TME), and insufficient trafficking, CAR-T-cell therapy for solid tumors is still in the exploration stage. Mesothelin (MSLN) is a molecule expressed on the surface of various solid malignant tumor cells that is suitable as a target of tumor cells with high MSLN expression for CAR-T-cell therapy. This paper briefly described the development of CAR-T cell therapy and the structural features of MSLN, and especially summarized the strategies of structure optimization of MSLN-targeting CAR-T-cells and the enhancement methods of MSLN-targeting CAR-T cell anti-tumor efficacy by summarizing some preclinical experiment and clinical trials. When considering MSLN-targeting CAR-T-cell therapy as an example, this paper summarizes the efforts made by researchers in CAR-T-cell therapy for solid tumors and summarizes feasible treatment plans by integrating the existing research results.
Collapse
Affiliation(s)
- Qiuhong Chen
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan, 250014, People's Republic of China
| | - Yang Sun
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan, 250014, People's Republic of China
| | - Hua Li
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan, 250014, People's Republic of China.
| |
Collapse
|
16
|
Chen L, Yin G, Wang Z, Liu Z, Sui C, Chen K, Song T, Xu W, Qi L, Li X. A predictive radiotranscriptomics model based on DCE-MRI for tumor immune landscape and immunotherapy in cholangiocarcinoma. Biosci Trends 2024; 18:263-276. [PMID: 38853000 DOI: 10.5582/bst.2024.01121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
This study aims to determine the predictive role of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) derived radiomic model in tumor immune profiling and immunotherapy for cholangiocarcinoma. To perform radiomic analysis, immune related subgroup clustering was first performed by single sample gene set enrichment analysis (ssGSEA). Second, a total of 806 radiomic features for each phase of DCE-MRI were extracted by utilizing the Python package Pyradiomics. Then, a predictive radiomic signature model was constructed after a three-step features reduction and selection, and receiver operating characteristic (ROC) curve was employed to evaluate the performance of this model. In the end, an independent testing cohort involving cholangiocarcinoma patients with anti-PD-1 Sintilimab treatment after surgery was used to verify the potential application of the established radiomic model in immunotherapy for cholangiocarcinoma. Two distinct immune related subgroups were classified using ssGSEA based on transcriptome sequencing. For radiomic analysis, a total of 10 predictive radiomic features were finally identified to establish a radiomic signature model for immune landscape classification. Regarding to the predictive performance, the mean AUC of ROC curves was 0.80 in the training/validation cohort. For the independent testing cohort, the individual predictive probability by radiomic model and the corresponding immune score derived from ssGSEA was significantly correlated. In conclusion, radiomic signature model based on DCE-MRI was capable of predicting the immune landscape of chalangiocarcinoma. Consequently, a potentially clinical application of this developed radiomic model to guide immunotherapy for cholangiocarcinoma was suggested.
Collapse
Affiliation(s)
- Lu Chen
- Department of Hepatobiliary Cancer, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Guotao Yin
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ziyang Wang
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Nuclear Medicine, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zifan Liu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chunxiao Sui
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Kun Chen
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Tianqiang Song
- Department of Hepatobiliary Cancer, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wengui Xu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lisha Qi
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xiaofeng Li
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
17
|
Zhang Y, Wang C, Li JJ. Revisiting the role of mesenchymal stromal cells in cancer initiation, metastasis and immunosuppression. Exp Hematol Oncol 2024; 13:64. [PMID: 38951845 PMCID: PMC11218091 DOI: 10.1186/s40164-024-00532-4] [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: 01/26/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024] Open
Abstract
Immune checkpoint blockade (ICB) necessitates a thorough understanding of intricate cellular interactions within the tumor microenvironment (TME). Mesenchymal stromal cells (MSCs) play a pivotal role in cancer generation, progression, and immunosuppressive tumor microenvironment. Within the TME, MSCs encompass both resident and circulating counterparts that dynamically communicate and actively participate in TME immunosurveillance and response to ICB. This review aims to reevaluate various facets of MSCs, including their potential self-transformation to function as cancer-initiating cells and contributions to the creation of a conducive environment for tumor proliferation and metastasis. Additionally, we explore the immune regulatory functions of tumor-associated MSCs (TA-MSCs) and MSC-derived extracellular vesicles (MSC-EVs) with analysis of potential connections between circulating and tissue-resident MSCs. A comprehensive understanding of the dynamics of MSC-immune cell communication and the heterogeneous cargo of tumor-educated versus naïve MSCs may unveil a new MSC-mediated immunosuppressive pathway that can be targeted to enhance cancer control by ICB.
Collapse
Affiliation(s)
- Yanyan Zhang
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Radiation Oncology, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Charles Wang
- Department of Radiation Oncology, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Jian Jian Li
- Department of Radiation Oncology, School of Medicine, University of California Davis, Sacramento, CA, USA.
- NCI-Designated Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA.
| |
Collapse
|
18
|
Zha J, Zhang J, Lu J, Zhang G, Hua M, Guo W, Yang J, Fan G. A review of lactate-lactylation in malignancy: its potential in immunotherapy. Front Immunol 2024; 15:1384948. [PMID: 38779665 PMCID: PMC11109376 DOI: 10.3389/fimmu.2024.1384948] [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: 02/11/2024] [Accepted: 04/04/2024] [Indexed: 05/25/2024] Open
Abstract
Lactic acid was formerly regarded as a byproduct of metabolism. However, extensive investigations into the intricacies of cancer development have revealed its significant contributions to tumor growth, migration, and invasion. Post-translational modifications involving lactate have been widely observed in histone and non-histone proteins, and these modifications play a crucial role in regulating gene expression by covalently attaching lactoyl groups to lysine residues in proteins. This discovery has greatly enhanced our comprehension of lactic acid's involvement in disease pathogenesis. In this article, we provide a comprehensive review of the intricate relationship between lactate and tumor immunity, the occurrence of lactylation in malignant tumors, and the exploitation of targeted lactate-lactylation in tumor immunotherapy. Additionally, we discuss future research directions, aiming to offer novel insights that could inform the investigation, diagnosis, and treatment of related diseases.
Collapse
Affiliation(s)
- Jinhui Zha
- Department of Urology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
- Department of General Surgery, Shenzhen University General Hospital, Shenzhen, China
| | - Junan Zhang
- Department of Basic Medicine, Shenzhen University, Shenzhen, China
| | - Jingfen Lu
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guangcheng Zhang
- Department of Urology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
- Department of General Surgery, Shenzhen University General Hospital, Shenzhen, China
| | - Mengzhan Hua
- Department of Basic Medicine, Shenzhen University, Shenzhen, China
| | - Weiming Guo
- Department of Sports Medicine Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Jing Yang
- Endocrinology Department, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Gang Fan
- Department of Urology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
19
|
Shi C, Zhao S, Mi L, Niu D, Hu F, Han W, Li B. Fucoidan MF4 from Fucus vesiculosus inhibits Lewis lung cancer via STING-TBK1-IRF3 pathway. Int J Biol Macromol 2024; 267:131336. [PMID: 38583840 DOI: 10.1016/j.ijbiomac.2024.131336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 03/28/2024] [Accepted: 04/01/2024] [Indexed: 04/09/2024]
Abstract
Fucoidan, a sulfated polysaccharide of marine origin found in brown algae and sea cucumbers, has been identified as a neuroprotective compound. In this study, a novel fucoidan MF4 was extracted from Fucus vesiculosus and isolated using Q-Sepharose fast-flow ion-exchange chromatography. The physicochemical properties of MF4 were characterized. MF4 is primarily composed of fucose, xylose, galactose, glucose, and mannose in a molar ratio of 12.3: 4.9: 1.1: 1.0: 1.1, with an average molecular weight of 67.7 kDa. Notably, MF4 demonstrated suppression of LLC tumor growth in vivo. RNA-sequencing analysis revealed that MF4 enhanced the expression of type I interferon-associated downstream genes in macrophages. Furthermore, MF4 increased the levels of phosphorylated TBK1 and IRF3 proteins in vitro. By activating the STING-TBK1-IRF3 signaling pathway, MF4 may enhance the antitumor activity of macrophages. Taken together, MF4 has promising potential as an antitumor and immunomodulatory agent.
Collapse
Affiliation(s)
- Chuanqin Shi
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266003, China; Center of Translational Medicine, Zibo Central Hospital, Zibo 255020, China
| | - Shihua Zhao
- Department of Endocrinology, Zibo Central Hospital, Zibo 255020, China
| | - Liyan Mi
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 261400, China
| | - Deying Niu
- Department of Neurosurgery, Zibo Central Hospital, Zibo 255020, China
| | - Fanwen Hu
- Departmet of Pharmacy, Jinan Dermatosis Prevention and Contorl Hospital, Jinan 250000, China
| | - Wenwei Han
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao 266071, China.
| | - Bing Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266003, China; Department of Dermatology, The Affiliated Haici Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
20
|
Ji P, Jin XK, Deng XC, Zhang SM, Liang JL, Li QR, Chen WH, Zhang XZ. Metabolic Regulation-Mediated Reversion of the Tumor Immunosuppressive Microenvironment for Potentiating Cooperative Metabolic Therapy and Immunotherapy. NANO LETTERS 2024; 24:4691-4701. [PMID: 38588212 DOI: 10.1021/acs.nanolett.4c01307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Tumor cells exhibit heightened glucose (Glu) consumption and increased lactic acid (LA) production, resulting in the formation of an immunosuppressive tumor microenvironment (TME) that facilitates malignant proliferation and metastasis. In this study, we meticulously engineer an antitumor nanoplatform, denoted as ZLGCR, by incorporating glucose oxidase, LA oxidase, and CpG oligodeoxynucleotide into zeolitic imidazolate framework-8 that is camouflaged with a red blood cell membrane. Significantly, ZLGCR-mediated consumption of Glu and LA not only amplifies the effectiveness of metabolic therapy but also reverses the immunosuppressive TME, thereby enhancing the therapeutic outcomes of CpG-mediated antitumor immunotherapy. It is particularly important that the synergistic effect of metabolic therapy and immunotherapy is further augmented when combined with immune checkpoint blockade therapy. Consequently, this engineered antitumor nanoplatform will achieve a cooperative tumor-suppressive outcome through the modulation of metabolism and immune responses within the TME.
Collapse
Affiliation(s)
- Ping Ji
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
- Institute of Precision Medicine Peking University Shenzhen Hospital, Shenzhen 518036, P. R. China
| | - Xiao-Kang Jin
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xin-Chen Deng
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Shi-Man Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Jun-Long Liang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Qian-Ru Li
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Wei-Hai Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
- Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
- Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| |
Collapse
|
21
|
Chen C, Park AK, Monroy I, Ren Y, Kim SI, Chaurasiya S, Priceman SJ, Fong Y. Using Oncolytic Virus to Retask CD19-Chimeric Antigen Receptor T Cells for Treatment of Pancreatic Cancer: Toward a Universal Chimeric Antigen Receptor T-Cell Strategy for Solid Tumor. J Am Coll Surg 2024; 238:436-447. [PMID: 38214445 DOI: 10.1097/xcs.0000000000000964] [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: 01/13/2024]
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T cells targeting the B-cell antigen CD19 are standard therapy for relapsed or refractory B-cell lymphoma and leukemia. CAR T cell therapy in solid tumors is limited due to an immunosuppressive tumor microenvironment and a lack of tumor-restricted antigens. We recently engineered an oncolytic virus (CF33) with high solid tumor affinity and specificity to deliver a nonsignaling truncated CD19 antigen (CD19t), allowing targeting by CD19-CAR T cells. Here, we tested this combination against pancreatic cancer. STUDY DESIGN We engineered CF33 to express a CD19t (CF33-CD19t) target. Flow cytometry and ELISA were performed to quantify CD19t expression, immune activation, and killing by virus and CD19-CAR T cells against various pancreatic tumor cells. Subcutaneous pancreatic human xenograft tumor models were treated with virus, CAR T cells, or virus+CAR T cells. RESULTS In vitro, CF33-CD19t infection of tumor cells resulted in >90% CD19t cell-surface expression. Coculturing CD19-CAR T cells with infected cells resulted in interleukin-2 and interferon gamma secretion, upregulation of T-cell activation markers, and synergistic cell killing. Combination therapy of virus+CAR T cells caused significant tumor regression (day 13): control (n = 16, 485 ± 20 mm 3 ), virus alone (n = 20, 254 ± 23 mm 3 , p = 0.0001), CAR T cells alone (n = 18, 466 ± 25 mm 3 , p = NS), and virus+CAR T cells (n = 16, 128 ± 14 mm 3 , p < 0.0001 vs control; p = 0.0003 vs virus). CONCLUSIONS Engineered CF33-CD19t effectively infects and expresses CD19t in pancreatic tumors, triggering cell killing and increased immunogenic response by CD19-CAR T cells. Notably, CF33-CD19t can turn cold immunologic tumors hot, enabling solid tumors to be targetable by agents designed against liquid tumor antigens.
Collapse
Affiliation(s)
- Courtney Chen
- From the Departments of Surgery (Chen, Kim, Chaurasiya, Fong)
| | - Anthony K Park
- Hematology and Hematopoietic Cell Transplantation (Park, Monroy, Ren, Priceman)
- Irell and Manella Graduate School of Biological Sciences (Park), City of Hope, Duarte, CA
| | - Isabel Monroy
- Hematology and Hematopoietic Cell Transplantation (Park, Monroy, Ren, Priceman)
| | - Yuwei Ren
- Hematology and Hematopoietic Cell Transplantation (Park, Monroy, Ren, Priceman)
| | - Sang-In Kim
- From the Departments of Surgery (Chen, Kim, Chaurasiya, Fong)
| | | | - Saul J Priceman
- Hematology and Hematopoietic Cell Transplantation (Park, Monroy, Ren, Priceman)
- Immuno-Oncology, Beckman Research Institute (Priceman)
| | - Yuman Fong
- From the Departments of Surgery (Chen, Kim, Chaurasiya, Fong)
| |
Collapse
|
22
|
吴 朋, 杨 智, 李 青, 王 德. [Advances in Research on Cell Metabolic Interactions in the Tumor Microenvironment]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:482-489. [PMID: 38645846 PMCID: PMC11026886 DOI: 10.12182/20240360606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Indexed: 04/23/2024]
Abstract
Metabolic reprogramming plays a critical role in tumorigenesis and tumor progression. The metabolism and the proliferation of tumors are regulated by both intrinsic factors within the tumor and the availability of metabolites in the tumor microenvironment (TME). The metabolic niche within the TME is primarily orchestrated at 3 levels: 1) the regulation of tumor metabolism by factors intrinsic to the tumors, 2) the interaction between tumor cells and T cells, macrophages, and stromal cells, and 3) the metabolic heterogeneity of tumor cells within the tissue space. Herein, we provided a concise overview of the various metabolic regulatory modes observed in tumor cells. Additionally, we extensively analyzed the interaction between tumor cells and other cells within the TME, as well as the metabolic characteristics and functions of different types of cells. Ultimately, this review provides a theoretical basis and novel insights for the precision treatment of tumors.
Collapse
Affiliation(s)
- 朋飞 吴
- 四川大学华西医院 呼吸与共病研究院 精准医学研究中心/精准医学四川省重点实验室 (成都 610041)Precision Medicine Research Center, Precision Medicine Key Laboratory of Sichuan Province, Institute of Respiratory and Comorbidity, West China Hospital, Sichuan University, Chengdu 610041, China
- 四川大学华西医院 呼吸与共病研究院 呼吸健康研究所 (成都 610041)Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, Institute of Respiratory and Comorbidity, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 智 杨
- 四川大学华西医院 呼吸与共病研究院 精准医学研究中心/精准医学四川省重点实验室 (成都 610041)Precision Medicine Research Center, Precision Medicine Key Laboratory of Sichuan Province, Institute of Respiratory and Comorbidity, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 青晏 李
- 四川大学华西医院 呼吸与共病研究院 精准医学研究中心/精准医学四川省重点实验室 (成都 610041)Precision Medicine Research Center, Precision Medicine Key Laboratory of Sichuan Province, Institute of Respiratory and Comorbidity, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 德年 王
- 四川大学华西医院 呼吸与共病研究院 精准医学研究中心/精准医学四川省重点实验室 (成都 610041)Precision Medicine Research Center, Precision Medicine Key Laboratory of Sichuan Province, Institute of Respiratory and Comorbidity, West China Hospital, Sichuan University, Chengdu 610041, China
- 四川大学华西医院 呼吸与共病研究院 呼吸健康研究所 (成都 610041)Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, Institute of Respiratory and Comorbidity, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
23
|
Levstek L, Janžič L, Ihan A, Kopitar AN. Biomarkers for prediction of CAR T therapy outcomes: current and future perspectives. Front Immunol 2024; 15:1378944. [PMID: 38558801 PMCID: PMC10979304 DOI: 10.3389/fimmu.2024.1378944] [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: 01/30/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy holds enormous potential for the treatment of hematologic malignancies. Despite its benefits, it is still used as a second line of therapy, mainly because of its severe side effects and patient unresponsiveness. Numerous researchers worldwide have attempted to identify effective predictive biomarkers for early prediction of treatment outcomes and adverse effects in CAR T cell therapy, albeit so far only with limited success. This review provides a comprehensive overview of the current state of predictive biomarkers. Although existing predictive metrics correlate to some extent with treatment outcomes, they fail to encapsulate the complexity of the immune system dynamics. The aim of this review is to identify six major groups of predictive biomarkers and propose their use in developing improved and efficient prediction models. These groups include changes in mitochondrial dynamics, endothelial activation, central nervous system impairment, immune system markers, extracellular vesicles, and the inhibitory tumor microenvironment. A comprehensive understanding of the multiple factors that influence therapeutic efficacy has the potential to significantly improve the course of CAR T cell therapy and patient care, thereby making this advanced immunotherapy more appealing and the course of therapy more convenient and favorable for patients.
Collapse
Affiliation(s)
| | | | | | - Andreja Nataša Kopitar
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
24
|
Yousefi A, Sotoodehnejadnematalahi F, Nafissi N, Zeinali S, Azizi M. MicroRNA-561-3p indirectly regulates the PD-L1 expression by targeting ZEB1, HIF1A, and MYC genes in breast cancer. Sci Rep 2024; 14:5845. [PMID: 38462658 PMCID: PMC10925600 DOI: 10.1038/s41598-024-56511-6] [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: 05/23/2023] [Accepted: 03/07/2024] [Indexed: 03/12/2024] Open
Abstract
Globally, breast cancer is the second most common cause of cancer-related deaths among women. In breast cancer, microRNAs (miRNAs) are essential for both the initiation and development of tumors. It has been suggested that the tumor suppressor microRNA-561-3p (miR-561-3p) is crucial in arresting the growth of cancer cells. Further research is necessary to fully understand the role and molecular mechanism of miR-561 in human BC. The aim of this study was to investigate the inhibitory effect of miR-561-3p on ZEB1, HIF1A, and MYC expression as oncogenes that have the most impact on PD-L1 overexpression and cellular processes such as proliferation, apoptosis, and cell cycle in breast cancer (BC) cell lines. The expression of ZEB1, HIF1A, and MYC genes and miR-561-3p were measured in BC clinical samples and cell lines via qRT-PCR. The luciferase assay, MTT, Annexin-PI staining, and cell cycle experiments were used to assess the effect of miR-561-3p on candidate gene expression, proliferation, apoptosis, and cell cycle progression. Flow cytometry was used to investigate the effects of miR-561 on PD-L1 suppression in the BC cell line. The luciferase assay showed that miRNA-561-3p targets the 3'-UTRs of ZEB1, HIF1A and MYC genes significantly. In BC tissues, the qRT-PCR results demonstrated that miR-561-3p expression was downregulated and the expression of ZEB1, HIF1A and MYC genes was up-regulated. It was shown that overexpression of miR-561-3p decreased PD-L1 expression and BC cell proliferation, and induced apoptosis and cell cycle arrest through downregulation of candidate oncogenes. Furthermore, inhibition of candidate genes by miR-561-3p reduced PD-L1 at both mRNA and protein levels. Our research investigated the impact of miR-561-3p on the expression of ZEB1, HIF1A and MYC in breast cancer cells for the first time. Our findings may help clarify the role of miR-561-3p in PD-L1 regulation and point to this miR as a potential biomarker and novel therapeutic target for cancer immunotherapy.
Collapse
Affiliation(s)
- Atena Yousefi
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Nahid Nafissi
- Breast Surgery Department, Iran University of Medical Sciences, Tehran, Iran
| | - Sirous Zeinali
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, 69th Pasteur Street, Kargar Avenue, Tehran, Iran
| | - Masoumeh Azizi
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, 69th Pasteur Street, Kargar Avenue, Tehran, Iran.
| |
Collapse
|
25
|
Yu X, Yang J, Xu J, Pan H, Wang W, Yu X, Shi S. Histone lactylation: from tumor lactate metabolism to epigenetic regulation. Int J Biol Sci 2024; 20:1833-1854. [PMID: 38481814 PMCID: PMC10929197 DOI: 10.7150/ijbs.91492] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/19/2024] [Indexed: 01/25/2025] Open
Abstract
The Warburg Effect is one of the most well-known cancer hallmarks. This metabolic pattern centered on lactate has extremely complex effects on various aspects of tumor microenvironment, including metabolic remodeling, immune suppression, cancer cell migration, and drug resistance development. Based on accumulating evidence, metabolites are likely to participate in the regulation of biological processes in the microenvironment and to form a feedback loop. Therefore, further revealing the key mechanism of lactate-mediated oncological effects is a reasonable scientific idea. The discovery and refinement of histone lactylation in recent years has laid a firm foundation for the above idea. Histone lactylation is a post-translational modification that occurs at lysine sites on histones. Specific enzymes, known as "writers" and "erasers", catalyze the addition or removal, respectively, of lactacyl group at target lysine sites. An increasing number of investigations have reported this modification as key to multiple cellular procedures. In this review, we discuss the close connection between histone lactylation and a series of biological processes in the tumor microenvironment, including tumorigenesis, immune infiltration, and energy metabolism. Finally, this review provides insightful perspectives, identifying promising avenues for further exploration and potential clinical application in this field of research.
Collapse
Affiliation(s)
- Xiaoning Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Jing Yang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Haoqi Pan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No.270 Dong' An Road, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, No.270 Dong' An Road, 200032, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No.270 Dong' An Road, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, No.270 Dong' An Road, 200032, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
26
|
Shukla M, Bhowmick R, Ganguli P, Sarkar RR. Metabolic reprogramming and signalling cross-talks in tumour-immune interaction: a system-level exploration. ROYAL SOCIETY OPEN SCIENCE 2024; 11:231574. [PMID: 38481985 PMCID: PMC10933535 DOI: 10.1098/rsos.231574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/19/2023] [Accepted: 01/23/2024] [Indexed: 04/26/2024]
Abstract
Tumour-immune microenvironment (TIME) is pivotal in tumour progression and immunoediting. Within TIME, immune cells undergo metabolic adjustments impacting nutrient supply and the anti-tumour immune response. Metabolic reprogramming emerges as a promising approach to revert the immune response towards a pro-inflammatory state and conquer tumour dominance. This study proposes immunomodulatory mechanisms based on metabolic reprogramming and employs the regulatory flux balance analysis modelling approach, which integrates signalling, metabolism and regulatory processes. For the first time, a comprehensive system-level model is constructed to capture signalling and metabolic cross-talks during tumour-immune interaction and regulatory constraints are incorporated by considering the time lag between them. The model analysis identifies novel features to enhance the immune response while suppressing tumour activity. Particularly, altering the exchange of succinate and oxaloacetate between glioma and macrophage enhances the pro-inflammatory response of immune cells. Inhibition of glutamate uptake in T-cells disrupts the antioxidant mechanism of glioma and reprograms metabolism. Metabolic reprogramming through adenosine monophosphate (AMP)-activated protein kinase (AMPK), coupled with glutamate uptake inhibition, was identified as the most impactful combination to restore T-cell function. A comprehensive understanding of metabolism and gene regulation represents a favourable approach to promote immune cell recovery from tumour dominance.
Collapse
Affiliation(s)
- Mudita Shukla
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune, Maharashtra, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Rupa Bhowmick
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune, Maharashtra, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Piyali Ganguli
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune, Maharashtra, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Ram Rup Sarkar
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune, Maharashtra, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| |
Collapse
|
27
|
Xuekai L, Yan S, Jian C, Yifei S, Xinyue W, Wenyuan Z, Shuwen H, Xi Y. Advances in reprogramming of energy metabolism in tumor T cells. Front Immunol 2024; 15:1347181. [PMID: 38415258 PMCID: PMC10897011 DOI: 10.3389/fimmu.2024.1347181] [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: 11/30/2023] [Accepted: 01/29/2024] [Indexed: 02/29/2024] Open
Abstract
Cancer is a leading cause of human death worldwide, and the modulation of the metabolic properties of T cells employed in cancer immunotherapy holds great promise for combating cancer. As a crucial factor, energy metabolism influences the activation, proliferation, and function of T cells, and thus metabolic reprogramming of T cells is a unique research perspective in cancer immunology. Special conditions within the tumor microenvironment and high-energy demands lead to alterations in the energy metabolism of T cells. In-depth research on the reprogramming of energy metabolism in T cells can reveal the mechanisms underlying tumor immune tolerance and provide important clues for the development of new tumor immunotherapy strategies as well. Therefore, the study of T cell energy metabolism has important clinical significance and potential applications. In the study, the current achievements in the reprogramming of T cell energy metabolism were reviewed. Then, the influencing factors associated with T cell energy metabolism were introduced. In addition, T cell energy metabolism in cancer immunotherapy was summarized, which highlighted its potential significance in enhancing T cell function and therapeutic outcomes. In summary, energy exhaustion of T cells leads to functional exhaustion, thus resulting in immune evasion by cancer cells. A better understanding of reprogramming of T cell energy metabolism may enable immunotherapy to combat cancer and holds promise for optimizing and enhancing existing therapeutic approaches.
Collapse
Affiliation(s)
- Liu Xuekai
- Department of Clinical Laboratory, Aerospace Center Hospital, Beijing, China
| | - Song Yan
- Department of Clinical Laboratory, Aerospace Center Hospital, Beijing, China
| | - Chu Jian
- Department of Medical Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Department of Gastroenterology, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital), Huzhou, China
- Department of Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Song Yifei
- Department of Medical Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Department of Gastroenterology, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital), Huzhou, China
- Department of Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Wu Xinyue
- Department of Medical Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Department of Gastroenterology, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital), Huzhou, China
- Department of Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Zhang Wenyuan
- Department of Gynecology, Heyuan Hospital of Traditional Chinese Medicine, Heyuan, China
| | - Han Shuwen
- Department of Medical Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Department of Gastroenterology, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital), Huzhou, China
- Department of Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Yang Xi
- Department of Medical Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Department of Gastroenterology, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital), Huzhou, China
- Department of Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| |
Collapse
|
28
|
Francescone R, Crawford HC, Vendramini-Costa DB. Rethinking the Roles of Cancer-Associated Fibroblasts in Pancreatic Cancer. Cell Mol Gastroenterol Hepatol 2024; 17:737-743. [PMID: 38316215 PMCID: PMC10966284 DOI: 10.1016/j.jcmgh.2024.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/07/2024]
Abstract
Bearing a dismal 5-year survival rate, pancreatic ductal adenocarcinoma (PDAC) is a challenging disease that features a unique fibroinflammatory tumor microenvironment. As major components of the PDAC tumor microenvironment, cancer-associated fibroblasts are still poorly understood and their contribution to the several hallmarks of PDAC, such as resistance to therapies, immunosuppression, and high incidence of metastasis, is likely underestimated. There have been encouraging advances in the understanding of these fascinating cells, but many controversies remain, leaving the field still actively exploring the full scope of their contributions in PDAC progression. Here we pose several important considerations regarding PDAC cancer-associated fibroblast functions. We posit that transcriptomic analyses be interpreted with caution, when aiming to uncover the functional contributions of these cells. Moreover, we propose that normalizing these functions, rather than eliminating them, will provide the opportunity to enhance therapeutic response. Finally, we propose that cancer-associated fibroblasts should not be studied in isolation, but in conjunction with its extracellular matrix, because their respective functions are coordinated and concordant.
Collapse
Affiliation(s)
- Ralph Francescone
- Department of Surgery, Henry Ford Health, Detroit, Michigan; Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, Michigan
| | - Howard C Crawford
- Department of Surgery, Henry Ford Health, Detroit, Michigan; Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, Michigan
| | - Debora Barbosa Vendramini-Costa
- Department of Surgery, Henry Ford Health, Detroit, Michigan; Henry Ford Pancreatic Cancer Center, Henry Ford Health, Detroit, Michigan.
| |
Collapse
|
29
|
Poveda-Garavito N, Combita AL. Contribution of the TIME in BCP-ALL: the basis for novel approaches therapeutics. Front Immunol 2024; 14:1325255. [PMID: 38299154 PMCID: PMC10827891 DOI: 10.3389/fimmu.2023.1325255] [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: 10/20/2023] [Accepted: 12/28/2023] [Indexed: 02/02/2024] Open
Abstract
The bone marrow (BM) niche is a microenvironment where both immune and non-immune cells functionally interact with hematopoietic stem cells (HSC) and more differentiated progenitors, contributing to the regulation of hematopoiesis. It is regulated by various signaling molecules such as cytokines, chemokines, and adhesion molecules in its microenvironment. However, despite the strict regulation of BM signals to maintain their steady state, accumulating evidence in B-cell precursor acute lymphoblastic leukemia (BCP-ALL) indicates that leukemic cells can disrupt the physiological hematopoietic niche in the BM, creating a new leukemia-supportive microenvironment. This environment favors immunological evasion mechanisms and the interaction of these cells with the development and progression of BCP-ALL. With a growing understanding of the tumor immune microenvironment (TIME) in the development and progression of BCP-ALL, current strategies focused on "re-editing" TIME to promote antitumor immunity have been developed. In this review, we summarize how TIME cells are disrupted by the presence of leukemic cells, evading immunosurveillance mechanisms in the BCP-ALL model. We also explore the crosstalk between TIME and leukemic cells that leads to treatment resistance, along with the most promising immuno-therapy strategies. Understanding and further research into the role of the BM microenvironment in leukemia progression and relapse are crucial for developing more effective treatments and reducing patient mortality.
Collapse
Affiliation(s)
- Nathaly Poveda-Garavito
- Grupo de Investigación en Biología del Cáncer, Instituto Nacional de Cancerología (INC), Bogotá, Colombia
- Grupo de Investigación Traslacional en Oncología, Instituto Nacional de Cancerología (INC), Bogotá, Colombia
- Departamento de Microbiología, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Alba Lucía Combita
- Grupo de Investigación en Biología del Cáncer, Instituto Nacional de Cancerología (INC), Bogotá, Colombia
- Grupo de Investigación Traslacional en Oncología, Instituto Nacional de Cancerología (INC), Bogotá, Colombia
- Departamento de Microbiología, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
30
|
Jimenez J, Dubey P, Carter B, Koomen JM, Markowitz J. A metabolic perspective on nitric oxide function in melanoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189038. [PMID: 38061664 PMCID: PMC11380350 DOI: 10.1016/j.bbcan.2023.189038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/17/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
Nitric oxide (NO) generated from nitric oxide synthase (NOS) exerts a dichotomous effect in melanoma, suppressing or promoting tumor progression. This dichotomy is thought to depend on the intracellular NO concentration and the cell type in which it is generated. Due to its central role in the metabolism of multiple critical constituents involved in signaling and stress, it is crucial to explore NO's contribution to the metabolic dysfunction of melanoma. This review will discuss many known metabolites linked to NO production in melanoma. We discuss the synthesis of these metabolites, their role in biochemical pathways, and how they alter the biological processes observed in the melanoma tumor microenvironment. The metabolic pathways altered by NO and the corresponding metabolites reinforce its dual role in melanoma and support investigating this effect for potential avenues of therapeutic intervention.
Collapse
Affiliation(s)
- John Jimenez
- Department of Cutaneous Oncology, Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; Department of Oncologic Sciences, University of South Florida Morsani School of Medicine, Tampa, FL 33612, USA
| | - Parul Dubey
- Department of Cutaneous Oncology, Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Bethany Carter
- Department of Cutaneous Oncology, Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; Flow Cytometry Core Facility, Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - John M Koomen
- Molecular Oncology, Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Joseph Markowitz
- Department of Cutaneous Oncology, Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; Department of Oncologic Sciences, University of South Florida Morsani School of Medicine, Tampa, FL 33612, USA.
| |
Collapse
|
31
|
Song Q, Gao H, Sun S, Li Y, Wu X, Yang J, Wang B, Zhang Y, Wang L. Two-pronged microenvironmental modulation of metal-oxidase cascade catalysis and metabolic intervention for synergistic tumor immunotherapy. Acta Biomater 2024; 173:378-388. [PMID: 37925121 DOI: 10.1016/j.actbio.2023.10.037] [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: 06/19/2023] [Revised: 10/26/2023] [Accepted: 10/29/2023] [Indexed: 11/06/2023]
Abstract
Immunotherapy is an emerging treatment modality for tumors after surgery, radiotherapy, and chemotherapy. Despite the potential for eliminating primary tumor cells and depressing cancer metastasis, immunotherapy has huge challenges including low tumor immunogenicity and undesirable immunosuppressive tumor microenvironment (TME). Herein, the two-pronged microenvironmental modulation nanoplatform is developed to overcome these limitations. Specifically, hollow mesoporous MnO2 (HM) nanoparticles with pH responsive property are prepared and modified with glucose oxidase (GOX) by amide bond, which are further loaded with a potent glutaminase inhibitor CB839 to obtain HM-GOX/CB839. Under the low pH values in TME, HM was disintegrated, thereby releasing Mn2+, GOX and CB839. On the one hand, Mn2+ can convert H2O2 that increased by GOX catalysis in tumors into highly toxic hydroxyl radicals (•OH) and further induce immunogenic cell death (ICD) through the metal-oxidase cascade catalytic reaction, enhancing immunogenicity. On the other hand, GOX and CB839 can block glycolytic and glutamine metabolism pathways, respectively, which effectively reduce the number of immunosuppressive cells and reshape TME, improving anti-tumor immune efficacy. It is demonstrated that HM-GOX/CB839 can effectively activate the body's immunity and inhibit tumor growth and metastasis, providing a potential strategy for comprehensive tumor therapy. STATEMENT OF SIGNIFICANCE: Integrated microenvironmental modulation of metal-oxidase cascade catalysis and metabolic intervention offers a potential avenue for tumor immunotherapy. Under this premise, we constructed a two-pronged microenvironmental modulation nanoplatform (HM-GOX/CB839). On the one hand, the metal oxidase cascade could catalyze the generation of hydroxyl radicals (•OH) and induce immunogenic cell death (ICD), enhancing immunogenicity; on the other hand, metabolic intervention reprogrammed tumor microenvironment to relieve immunosuppression and thereby enhancing anti-tumor immune response. The resulting data demonstrated that HM-GOX/CB839 effectively inhibited tumor growth and metastasis, providing therapeutic potential for cancer immunotherapy.
Collapse
Affiliation(s)
- Qingling Song
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Hui Gao
- Department of Pharmacy, The First Hospital of Yulin (The Second Affiliated Hospital of Yan'an University), China
| | - Shuxin Sun
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Yao Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Xiaocui Wu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Junfei Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Baojin Wang
- Gynecology, the Third Affiliated Hospital of Zhengzhou University, China; Henan International Joint Laboratory of Ovarian Malignant Tumor, China.
| | - Yun Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China.
| | - Lei Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China.
| |
Collapse
|
32
|
Hu T, Allam M, Cai S, Henderson W, Yueh B, Garipcan A, Ievlev AV, Afkarian M, Beyaz S, Coskun AF. Single-cell spatial metabolomics with cell-type specific protein profiling for tissue systems biology. Nat Commun 2023; 14:8260. [PMID: 38086839 PMCID: PMC10716522 DOI: 10.1038/s41467-023-43917-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Metabolic reprogramming in cancer and immune cells occurs to support their increasing energy needs in biological tissues. Here we propose Single Cell Spatially resolved Metabolic (scSpaMet) framework for joint protein-metabolite profiling of single immune and cancer cells in male human tissues by incorporating untargeted spatial metabolomics and targeted multiplexed protein imaging in a single pipeline. We utilized the scSpaMet to profile cell types and spatial metabolomic maps of 19507, 31156, and 8215 single cells in human lung cancer, tonsil, and endometrium tissues, respectively. The scSpaMet analysis revealed cell type-dependent metabolite profiles and local metabolite competition of neighboring single cells in human tissues. Deep learning-based joint embedding revealed unique metabolite states within cell types. Trajectory inference showed metabolic patterns along cell differentiation paths. Here we show scSpaMet's ability to quantify and visualize the cell-type specific and spatially resolved metabolic-protein mapping as an emerging tool for systems-level understanding of tissue biology.
Collapse
Affiliation(s)
- Thomas Hu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Mayar Allam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Shuangyi Cai
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Walter Henderson
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Brian Yueh
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | - Anton V Ievlev
- Oak Ridge National Laboratory, Center for Nanophase Materials Sciences, Oak Ridge, TN, USA
| | - Maryam Afkarian
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, CA, USA
| | - Semir Beyaz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Ahmet F Coskun
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
33
|
Zheng S, Li H, Li Y, Chen X, Shen J, Chen M, Zhang C, Wu J, Sun Q. The emerging role of glycolysis and immune evasion in gastric cancer. Cancer Cell Int 2023; 23:317. [PMID: 38071310 PMCID: PMC10710727 DOI: 10.1186/s12935-023-03169-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/27/2023] [Indexed: 08/21/2024] Open
Abstract
Gastric cancer (GC) is the fifth most common malignancy and the third leading cause of cancer-related deaths worldwide. Similar to other types of tumors, GC cells undergo metabolic reprogramming and switch to a "predominantly glycolytic" metabolic pattern to promote its survival and metastasis, also known as "the Warburg effect", which is characterized by enhanced glucose uptake and lactate production. A large number of studies have shown that targeting cancer cells to enhanced glycolysis is a promising strategy, that can make cancer cells more susceptible to other conventional treatment methods of treatment, including chemotherapy, radiotherapy and immunotherapy, and so on. Therefore, this review summarizes the metabolic characteristics of glycolysis in GC cells and focuses on how abnormal lactate concentration can lead to immunosuppression through its effects on the differentiation, metabolism, and function of infiltrating immune cells, and how targeting this phenomenon may be a potential strategy to improve the therapeutic efficacy of GC.
Collapse
Affiliation(s)
- Shanshan Zheng
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Huaizhi Li
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Yaqi Li
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Xu Chen
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, China
| | - Junyu Shen
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Menglin Chen
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Cancan Zhang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, China
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Jian Wu
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, China.
| | - Qingmin Sun
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangsu, China.
| |
Collapse
|
34
|
Chen C, Jung A, Yang A, Monroy I, Zhang Z, Chaurasiya S, Deshpande S, Priceman S, Fong Y, Park AK, Woo Y. Chimeric Antigen Receptor-T Cell and Oncolytic Viral Therapies for Gastric Cancer and Peritoneal Carcinomatosis of Gastric Origin: Path to Improving Combination Strategies. Cancers (Basel) 2023; 15:5661. [PMID: 38067366 PMCID: PMC10705752 DOI: 10.3390/cancers15235661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 02/12/2024] Open
Abstract
Precision immune oncology capitalizes on identifying and targeting tumor-specific antigens to enhance anti-tumor immunity and improve the treatment outcomes of solid tumors. Gastric cancer (GC) is a molecularly heterogeneous disease where monoclonal antibodies against human epidermal growth factor receptor 2 (HER2), vascular endothelial growth factor (VEGF), and programmed cell death 1 (PD-1) combined with systemic chemotherapy have improved survival in patients with unresectable or metastatic GC. However, intratumoral molecular heterogeneity, variable molecular target expression, and loss of target expression have limited antibody use and the durability of response. Often immunogenically "cold" and diffusely spread throughout the peritoneum, GC peritoneal carcinomatosis (PC) is a particularly challenging, treatment-refractory entity for current systemic strategies. More adaptable immunotherapeutic approaches, such as oncolytic viruses (OVs) and chimeric antigen receptor (CAR) T cells, have emerged as promising GC and GCPC treatments that circumvent these challenges. In this study, we provide an up-to-date review of the pre-clinical and clinical efficacy of CAR T cell therapy for key primary antigen targets and provide a translational overview of the types, modifications, and mechanisms for OVs used against GC and GCPC. Finally, we present a novel, summary-based discussion on the potential synergistic interplay between OVs and CAR T cells to treat GCPC.
Collapse
Affiliation(s)
- Courtney Chen
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Audrey Jung
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Annie Yang
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Isabel Monroy
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA; (I.M.); (S.P.)
| | - Zhifang Zhang
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Shyambabu Chaurasiya
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Supriya Deshpande
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Saul Priceman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA; (I.M.); (S.P.)
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Yuman Fong
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Anthony K. Park
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA; (I.M.); (S.P.)
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Yanghee Woo
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
35
|
Shorer O, Yizhak K. Metabolic predictors of response to immune checkpoint blockade therapy. iScience 2023; 26:108188. [PMID: 37965137 PMCID: PMC10641254 DOI: 10.1016/j.isci.2023.108188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/23/2023] [Accepted: 10/10/2023] [Indexed: 11/16/2023] Open
Abstract
Metabolism of immune cells in the tumor microenvironment (TME) plays a critical role in cancer patient response to immune checkpoint inhibitors (ICI). Yet, a metabolic characterization of immune cells in the TME of patients treated with ICI is lacking. To bridge this gap we performed a semi-supervised analysis of ∼1700 metabolic genes using single-cell RNA-seq data of > 1 million immune cells from ∼230 samples of cancer patients treated with ICI. When clustering cells based on their metabolic gene expression, we found that similar immunological cellular states are found in different metabolic states. Most importantly, we found metabolic states that are significantly associated with patient response. We then built a metabolic predictor based on a dozen gene signature, which significantly differentiates between responding and non-responding patients across different cancer types (AUC = 0.8-0.92). Taken together, our results demonstrate the power of metabolism in predicting patient response to ICI.
Collapse
Affiliation(s)
- Ofir Shorer
- Department of Cell Biology and Cancer Science, The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 3525422, Israel
| | - Keren Yizhak
- Department of Cell Biology and Cancer Science, The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 3525422, Israel
- The Taub Faculty of Computer Science, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
36
|
Li Y, Liu Y, Kang Z, Guo J, Liu N. Tumor microenvironment heterogeneity in bladder cancer identifies biologically distinct subtypes predicting prognosis and anti-PD-L1 responses. Sci Rep 2023; 13:19563. [PMID: 37949863 PMCID: PMC10638294 DOI: 10.1038/s41598-023-44028-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 10/03/2023] [Indexed: 11/12/2023] Open
Abstract
Bladder cancer (BCa) is heterogeneous in the tumour microenvironment (TME). However, the role of the TME in BCa in modulating the response to immunotherapy has not been fully explored. We therefore analysed fractions of immune cells using CIBERSORTx and clustered BCa into subtypes. We also analyzed weighted correlation networks to generate immunotherapy-related hub genes that we used to construct a prediction model using multivariate Cox and LASSO regression analyses. We found that BCa comprised three subtypes (C1‒C3). The prognosis of the patients was the most favourable and the response rate to anti-programmed death ligand 1 (PD-L1) was the highest in C1 among the three subtypes. Immune cells, including CD8+, CD4+ memory activated, and follicular helper T cells, activated NK cells, and M1 macrophages infiltrated the C1 subtype. The C2 subtype was enriched in M0 macrophages and activated mast cells, and the C3 subtype was enriched in B and resting immune cells. Mechanistically, the enhanced immunogenicity of subtypes C1 and C2 correlated positively with a higher response rate, whereas the dysregulated ECM-related pathways in the C2 subtype and glycolytic and fatty acid metabolic pathways in the C3 subtype impaired the responses of patients to anti-PD-L1 therapy. We also constructed a TME-related signature based on 18 genes that performed well in terms of overall survival. In conclusion, we determined prognoses and anti-PD-L1 responses by analysing TME heterogeneity in BCa.
Collapse
Affiliation(s)
- YaFei Li
- Department of Urology, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan Province, China
| | - Yi Liu
- Department of Urology, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan Province, China.
| | - Zhengjun Kang
- Department of Urology, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan Province, China.
| | - Jianhua Guo
- Department of Urology, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan Province, China
| | - Nan Liu
- Department of Urology, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan Province, China
| |
Collapse
|
37
|
Borde S, Matosevic S. Metabolic adaptation of NK cell activity and behavior in tumors: challenges and therapeutic opportunities. Trends Pharmacol Sci 2023; 44:832-848. [PMID: 37770314 DOI: 10.1016/j.tips.2023.08.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/30/2023]
Abstract
The adaptation of natural killer (NK) cells to conditions in the microenvironment of tumors is deeply affected by their metabolic activity, itself a result of nutrient availability and the metabolism of the cancer cells themselves. Elevated rates of glycolysis and lipid metabolism in cancers not only lead to the accumulation of immunosuppressive byproducts but also contribute to an environment of elevated concentrations of extracellular metabolites. This results in altered NK cell bioenergetics through changes in transcriptional and translational profiles, ultimately affecting their pharmacology and impairing NK cell responses. However, understanding the metabolic processes that drive alterations in immunological signaling on NK cells remains both difficult and vastly underexplored. We discuss the varied and complex drivers of NK cell metabolism in homeostasis and the tumor microenvironment (TME), challenges associated with their targetability, and unexplored therapeutic opportunities.
Collapse
Affiliation(s)
- Shambhavi Borde
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN, USA
| | - Sandro Matosevic
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN, USA; Center for Cancer Research, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
38
|
Chen Y, Li H, Fan Y. Shaping the tumor immune microenvironment of SCLC: Mechanisms, and opportunities for immunotherapy. Cancer Treat Rev 2023; 120:102606. [PMID: 37579532 DOI: 10.1016/j.ctrv.2023.102606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/25/2023] [Accepted: 08/05/2023] [Indexed: 08/16/2023]
Abstract
Small-cell lung cancer (SCLC) is a very aggressive neuroendocrine tumor with a poor prognosis. Whereas immunotherapy has emerged as a promising approach for treating SCLC, its unique tumor immune microenvironment (TIME) might limit patient responses. To fully characterize the TIME and understand the mechanism of its formation with respect to SCLC is crucial. The recent rapid development of multi-omics technologies has rapidly advanced knowledge of TIME features and the regulatory mechanisms associated with SCLC. This review summarizes the TIME features of SCLC as well as shaping the TIME according to the genomics, epigenomics, and metabolomics of tumors. Future opportunities and challenges for immunotherapy are also discussed.
Collapse
Affiliation(s)
- Yunfei Chen
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Postgraduate Training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China
| | - Hui Li
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Postgraduate Training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China.
| | - Yun Fan
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Postgraduate Training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
39
|
Su WY, Tian LY, Guo LP, Huang LQ, Gao WY. PI3K signaling-regulated metabolic reprogramming: From mechanism to application. Biochim Biophys Acta Rev Cancer 2023; 1878:188952. [PMID: 37499988 DOI: 10.1016/j.bbcan.2023.188952] [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: 06/01/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/29/2023]
Abstract
Oncogenic signaling involved in tumor metabolic reprogramming. Tumorigenesis was not only determined by the mutations or deletion of oncogenes but also accompanied by the reprogramming of cellular metabolism. Metabolic alterations play a crucial regulatory role in the development and progression of tumors. Oncogenic PI3K/AKT signaling mediates the metabolic switch in cancer cells and immune cells in the tumor microenvironment. PI3K/AKT and its downstream effector branch off and connect to multiple steps of metabolism, such as glucose, lipids, and amino acids. Thus, PI3K inhibitor could effectively regulate metabolic pathway and impede the oncogenic process and some key metabolic proteins or critical enzymes also constitute biomarkers for tumor diagnosis and treatment. In the current review, we summarize the significant effect of PI3K/AKT signaling toward tumor metabolism, it enables us to obtain the better understanding for this interaction and develop more effective therapeutic strategies targeting cancer cell metabolism.
Collapse
Affiliation(s)
- Wen Ya Su
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Lu Yao Tian
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Lan Pin Guo
- National Resource Center for Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Lu Qi Huang
- National Resource Center for Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Wen Yuan Gao
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China.
| |
Collapse
|
40
|
Liu Y, Zhang X, Cheng F, Cao W, Geng Y, Chen Z, Wei W, Zhang L. Xanthatin induce DDP-resistance lung cancer cells apoptosis through regulation of GLUT1 mediated ROS accumulation. Drug Dev Res 2023; 84:1266-1278. [PMID: 37260173 DOI: 10.1002/ddr.22084] [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: 11/02/2022] [Revised: 05/09/2023] [Accepted: 05/19/2023] [Indexed: 06/02/2023]
Abstract
Chemoresistance to cisplatin (DDP) therapy is a major obstacle that needs to be overcome in treating lung cancer patients. Xanthatin has been reported to exhibit an antitumor effect on various cancers, but the function of xanthatin in DDP-resistance lung cancer remains unclear. The study aimed to explore the effect and mechanisms of xanthatin on proliferation, apoptosis, and migration in DDP-resistance lung cancer cells. In the present study, xanthatin suppresses the expression of glucose transporter 1 (GLUT1), attenuates the pentose phosphate pathway (PPP), and causes ROS accumulation and apoptosis, thereby mitigating the antioxidative capacity in DDP-resistance cells. Previous studies have shown that GLUT1 is associated with resistance to platinum drugs. We found that GLUT1 was significantly increased in the DDP-resistant lung cancer cell line compared to the parental cell line, and xanthatin significantly downregulated GLUT1 expression in DDP-resistant lung cancer cells. Notably, overexpression of GLUT1 significantly reduced the production of ROS and increased cellular NADPH/NADP+ and GSH/GSSG ratios. Thus, these results suggest that xanthatin induces DDP-resistance lung cancer cells apoptosis through regulation of GLUT1-mediated ROS accumulation. These findings might provide a possible strategy for the clinical treatment of DDP-resistant lung cancer.
Collapse
Affiliation(s)
- Yunxiao Liu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Xinge Zhang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Fenting Cheng
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Cao
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yadi Geng
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhaolin Chen
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Wei
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Lei Zhang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
41
|
Hu X, Ma Z, Xu B, Li S, Yao Z, Liang B, Wang J, Liao W, Lin L, Wang C, Zheng S, Wu Q, Huang Q, Yu L, Wang F, Shi M. Glutamine metabolic microenvironment drives M2 macrophage polarization to mediate trastuzumab resistance in HER2-positive gastric cancer. Cancer Commun (Lond) 2023; 43:909-937. [PMID: 37434399 PMCID: PMC10397568 DOI: 10.1002/cac2.12459] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 01/04/2023] [Accepted: 06/21/2023] [Indexed: 07/13/2023] Open
Abstract
BACKGROUND Trastuzumab is a first-line targeted therapy for human epidermal growth factor receptor-2 (HER2)-positive gastric cancer. However, the inevitable occurrence of acquired trastuzumab resistance limits the drug benefit, and there is currently no effective reversal measure. Existing researches on the mechanism of trastuzumab resistance mainly focused on tumor cells themselves, while the understanding of the mechanisms of environment-mediated drug resistance is relatively lacking. This study aimed to further explore the mechanisms of trastuzumab resistance to identify strategies to promote survival in these patients. METHODS Trastuzumab-sensitive and trastuzumab-resistant HER2-positive tumor tissues and cells were collected for transcriptome sequencing. Bioinformatics were used to analyze cell subtypes, metabolic pathways, and molecular signaling pathways. Changes in microenvironmental indicators (such as macrophage, angiogenesis, and metabolism) were verified by immunofluorescence (IF) and immunohistochemical (IHC) analyses. Finally, a multi-scale agent-based model (ABM) was constructed. The effects of combination treatment were further validated in nude mice to verify these effects predicted by the ABM. RESULTS Based on transcriptome sequencing, molecular biology, and in vivo experiments, we found that the level of glutamine metabolism in trastuzumab-resistant HER2-positive cells was increased, and glutaminase 1 (GLS1) was significantly overexpressed. Meanwhile, tumor-derived GLS1 microvesicles drove M2 macrophage polarization. Furthermore, angiogenesis promoted trastuzumab resistance. IHC showed high glutamine metabolism, M2 macrophage polarization, and angiogenesis in trastuzumab-resistant HER2-positive tumor tissues from patients and nude mice. Mechanistically, the cell division cycle 42 (CDC42) promoted GLS1 expression in tumor cells by activating nuclear factor kappa-B (NF-κB) p65 and drove GLS1 microvesicle secretion through IQ motif-containing GTPase-activating protein 1 (IQGAP1). Based on the ABM and in vivo experiments, we confirmed that the combination of anti-glutamine metabolism, anti-angiogenesis, and pro-M1 polarization therapy had the best effect in reversing trastuzumab resistance in HER2-positive gastric cancer. CONCLUSIONS This study revealed that tumor cells secrete GLS1 microvesicles via CDC42 to promote glutamine metabolism, M2 macrophage polarization, and pro-angiogenic function of macrophages, leading to acquired trastuzumab resistance in HER2-positive gastric cancer. A combination of anti-glutamine metabolism, anti-angiogenesis, and pro-M1 polarization therapy may provide a new insight into reversing trastuzumab resistance.
Collapse
Affiliation(s)
- Xingbin Hu
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Zhenfeng Ma
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Beibei Xu
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Shulong Li
- School of Biomedical EngineeringSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Zhiqi Yao
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Bishan Liang
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Jiao Wang
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Wangjun Liao
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Li Lin
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Chunling Wang
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Siting Zheng
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Qijing Wu
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Qiong Huang
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Le Yu
- School of Pharmaceutical SciencesSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Fenghua Wang
- Department of Medical OncologySun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer MedicineGuangzhouGuangdongP. R. China
| | - Min Shi
- Department of OncologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| |
Collapse
|
42
|
Wang X, Bai Y, Zhou Z, Ye B, Chen Z, Chen X, Wu W. Relationship Between Infiltration of CD163+ TAMs, FoxP3+ Tregs, or CD66b+ TANs and Cell Differentiation in Colorectal Cancer Tissues. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2023; 34:747-752. [PMID: 37232465 PMCID: PMC10441166 DOI: 10.5152/tjg.2023.22064] [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: 02/08/2022] [Accepted: 10/09/2022] [Indexed: 05/27/2023]
Abstract
BACKGROUND/AIMS There are many studies on immune cell infiltration in colorectal cancer, including FoxP3+-regulatory T cells, CD66b+ tumorassociated neutrophils, and CD163+ tumor-associated macrophages. These studies mainly focus on the relationship between cell infiltration and tumor progression, prognosis, and so on, while the relationship between tumor cell differentiation and cell infiltration is poorly understood. We aimed to explore the relationship between cell infiltration and tumor cell differentiation. MATERIALS AND METHODS The tissue microarray and immunohistochemistry were used to determine the infiltration of FoxP3+-regulatory T cells, CD66b+ tumor-associated neutrophils, and CD163+ tumor-associated macrophages in 673 colorectal cancer samples from the Second Affiliated Hospital, Wenzhou Medical University (2001-2009). Kruskal-Wallis test was used to assess the positive cell infiltration in colorectal cancer tissues with tumor cells of varying degrees of differentiation. RESULTS The number of CD163+ tumor-associated macrophages, FoxP3+-regulatory T cells, and CD66b+ tumor-associated neutrophils in colorectal cancer tissues was different, and the level of CD163+ tumor-associated macrophages was the highest while the level of FoxP3+-regulatory T cells was the least. There were significant differences in the cell infiltration of colorectal cancer tissue cells with different levels of differentiation (P < .05). The highest infiltration of CD163+ tumor-associated macrophages (154.07 ± 6.95) and FoxP3+-regulatory T cells (20.14 ± 2.07) were in the poorly differentiated colorectal cancer tissues, while the higher infiltration of CD66b+ tumor-associated neutrophils was in the moderately or well-differentiated colorectal cancer tissues (36.70 ± 1.10 and 36.09 ± 1.06, respectively). CONCLUSION Infiltration of CD163+ tumor-associated macrophages, FoxP3+-regulatory T cells, and CD66b+ tumor-associated neutrophils in colorectal cancer tissues may be related to the differentiation of tumor cells.
Collapse
Affiliation(s)
- Xiaobo Wang
- Department of Traumatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yongyu Bai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zhuha Zhou
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Bailiang Ye
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zhejing Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xiaolei Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Wenyi Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
43
|
Wilczyński JR, Wilczyński M, Paradowska E. "DEPHENCE" system-a novel regimen of therapy that is urgently needed in the high-grade serous ovarian cancer-a focus on anti-cancer stem cell and anti-tumor microenvironment targeted therapies. Front Oncol 2023; 13:1201497. [PMID: 37448521 PMCID: PMC10338102 DOI: 10.3389/fonc.2023.1201497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/07/2023] [Indexed: 07/15/2023] Open
Abstract
Ovarian cancer, especially high-grade serous type, is the most lethal gynecological malignancy. The lack of screening programs and the scarcity of symptomatology result in the late diagnosis in about 75% of affected women. Despite very demanding and aggressive surgical treatment, multiple-line chemotherapy regimens and both approved and clinically tested targeted therapies, the overall survival of patients is still unsatisfactory and disappointing. Research studies have recently brought some more understanding of the molecular diversity of the ovarian cancer, its unique intraperitoneal biology, the role of cancer stem cells, and the complexity of tumor microenvironment. There is a growing body of evidence that individualization of the treatment adjusted to the molecular and biochemical signature of the tumor as well as to the medical status of the patient should replace or supplement the foregoing therapy. In this review, we have proposed the principles of the novel regimen of the therapy that we called the "DEPHENCE" system, and we have extensively discussed the results of the studies focused on the ovarian cancer stem cells, other components of cancer metastatic niche, and, finally, clinical trials targeting these two environments. Through this, we have tried to present the evolving landscape of treatment options and put flesh on the experimental approach to attack the high-grade serous ovarian cancer multidirectionally, corresponding to the "DEPHENCE" system postulates.
Collapse
Affiliation(s)
- Jacek R Wilczyński
- Department of Gynecological Surgery and Gynecological Oncology, Medical University of Lodz, Lodz, Poland
| | - Miłosz Wilczyński
- Department of Gynecological, Endoscopic and Oncological Surgery, Polish Mother's Health Center-Research Institute, Lodz, Poland
- Department of Surgical and Endoscopic Gynecology, Medical University of Lodz, Lodz, Poland
| | - Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, Lodz, Poland
| |
Collapse
|
44
|
Zhang X, Dai M, Li S, Li M, Cheng B, Ma T, Zhou Z. The emerging potential role of p62 in cancer treatment by regulating metabolism. Trends Endocrinol Metab 2023:S1043-2760(23)00106-6. [PMID: 37349161 DOI: 10.1016/j.tem.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 06/24/2023]
Abstract
p62 is an important multifunctional adaptor protein participating in autophagy and many other activities. Many studies have revealed that p62 is highly expressed in multiple cancers and decreasing its level can effectively lower the proliferation ability of cancer cells. Moreover, much research has highlighted the significant role of the regulation of cancer cell metabolism in helping to treat tumors. Recent reports demonstrate that p62 could regulate cancer cell metabolism through various mechanisms. However, the relationship between p62 and cancer cell metabolism as well as the related mechanisms has not been fully elucidated. In this review, we describe glucose, glutamine, and fatty acid metabolism in tumor cells and some signaling pathways that can regulate cancer metabolism and are mediated by p62.
Collapse
Affiliation(s)
- Xiaochuan Zhang
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
| | - Mengge Dai
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Shaotong Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Meng Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Bing Cheng
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Ting Ma
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China.
| | - Zheng Zhou
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
45
|
Yin L, Chen GL, Xiang Z, Liu YL, Li XY, Bi JW, Wang Q. Current progress in chimeric antigen receptor-modified T cells for the treatment of metastatic breast cancer. Biomed Pharmacother 2023; 162:114648. [PMID: 37023621 DOI: 10.1016/j.biopha.2023.114648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Breast cancer is the leading cancer in women. Around 20-30% breast cancer patients undergo invasion or metastasis after radical surgical resection and eventually die. Number of breast cancer patients show poor sensitivity toward treatments despite the advances in chemotherapy, endocrine therapy, and molecular targeted treatments. Therapeutic resistance and tumor recurrence or metastasis develop with the ongoing treatments. Conducive treatment strategies are thus required. Chimeric antigen receptor (CAR)-modified T-cell therapy has progressed as a part of tumor immunotherapy. However, CAR-T treatment has not been effective in solid tumors because of tumor microenvironment complexity, inhibitory effects of extracellular matrix, and lacking ideal tumor antigens. Herein, the prospects of CAR-T cell therapy for metastatic breast cancer are discussed, and the targets for CAR-T therapy in breast cancer (HER-2, C-MET, MSLN, CEA, MUC1, ROR1, EGFR) at clinical level are reviewed. Moreover, solutions are proposed for the challenges of breast cancer CAR-T therapy regarding off-target effects, heterogeneous antigen expression by tumor cells and immunosuppressive tumor microenvironment. Ideas for improving the therapeutics of CAR-T cell therapy in metastatic breast cancer are suggested.
Collapse
Affiliation(s)
- Li Yin
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, 250023 Jinan, China; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, China; Shandong University of Traditional Chinese Medicine, 250355 Jinan, China
| | - Gui-Lai Chen
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, 250023 Jinan, China; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, China
| | - Zhuo Xiang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, 250023 Jinan, China; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, China
| | - Yu-Lin Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, 250023 Jinan, China; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, China
| | - Xing-Yu Li
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 266003 Qingdao, China
| | - Jing-Wang Bi
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, 250023 Jinan, China; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, China.
| | - Qiang Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, 250023 Jinan, China; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, China; Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 266003 Qingdao, China.
| |
Collapse
|
46
|
Plaza-Diaz J, Álvarez-Mercado AI. The Interplay between Microbiota and Chemotherapy-Derived Metabolites in Breast Cancer. Metabolites 2023; 13:703. [PMID: 37367861 PMCID: PMC10301694 DOI: 10.3390/metabo13060703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023] Open
Abstract
The most common cancer in women is breast cancer, which is also the second leading cause of death in this group. It is, however, important to note that some women will develop or will not develop breast cancer regardless of whether certain known risk factors are present. On the other hand, certain compounds are produced by bacteria in the gut, such as short-chain fatty acids, secondary bile acids, and other metabolites that may be linked to breast cancer development and mediate the chemotherapy response. Modeling the microbiota through dietary intervention and identifying metabolites directly associated with breast cancer and its complications may be useful to identify actionable targets and improve the effect of antiangiogenic therapies. Metabolomics is therefore a complementary approach to metagenomics for this purpose. As a result of the combination of both techniques, a better understanding of molecular biology and oncogenesis can be obtained. This article reviews recent literature about the influence of bacterial metabolites and chemotherapy metabolites in breast cancer patients, as well as the influence of diet.
Collapse
Affiliation(s)
- Julio Plaza-Diaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain;
- Instituto de Investigación Biosanitaria ibs.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| | - Ana Isabel Álvarez-Mercado
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain;
- Instituto de Investigación Biosanitaria ibs.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- Institute of Nutrition and Food Technology, Biomedical Research Center, University of Granada, 18016 Armilla, Spain
| |
Collapse
|
47
|
Fatima Z, Abonofal A, Stephen B. Targeting Cancer Metabolism to Improve Outcomes with Immune Checkpoint Inhibitors. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2023; 6:91-102. [PMID: 37214204 PMCID: PMC10195018 DOI: 10.36401/jipo-22-27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 05/24/2023]
Abstract
Immune checkpoint inhibitors have revolutionized the treatment paradigm of several cancers. However, not all patients respond to treatment. Tumor cells reprogram metabolic pathways to facilitate growth and proliferation. This shift in metabolic pathways creates fierce competition with immune cells for nutrients in the tumor microenvironment and generates by-products harmful for immune cell differentiation and growth. In this review, we discuss these metabolic alterations and the current therapeutic strategies to mitigate these alterations to metabolic pathways that can be used in combination with checkpoint blockade to offer a new path forward in cancer management.
Collapse
Affiliation(s)
- Zainab Fatima
- Department of Hospice and Palliative Care, Virginia Commonwealth University, Richmond, VA, USA
| | - Abdulrahman Abonofal
- Department of Medicine, Section of Hematology/Oncology, West Virginia University, Morgantown, WV, USA
| | - Bettzy Stephen
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
48
|
Rogers G, Barker S, Sharma M, Khakoo S, Utz M. Operando NMR metabolomics of a microfluidic cell culture. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2023; 349:107405. [PMID: 36842430 DOI: 10.1016/j.jmr.2023.107405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/09/2023] [Accepted: 02/12/2023] [Indexed: 06/18/2023]
Abstract
In this work we demonstrate the use of microfluidic NMR for in situ culture and quantitative analysis of metabolism in hepatocellular carcinoma (HCC) cell lines. A hydrothermal heating system is used to enable continuous in situ NMR observation of HCC cell culture over a 24 h incubation period. This technique is nondestructive, non-invasive and can measure millimolar concentrations at microlitre volumes, within a few minutes and in precisely controlled culture conditions. This is sufficient to observe changes in primary energy metabolism, using around 500-3500 cells per device, and with a time resolution of 17 min. The ability to observe intracellular responses in a time-resolved manner provides a more detailed view of a biological system and how it reacts to stimuli. This capability will allow detailed metabolomic studies of cell-culture based cancer models, enabling quantification of metabolic reporgramming, the metabolic tumor microenvironment, and the metabolic interplay between cancer- and immune cells.
Collapse
Affiliation(s)
- Genevieve Rogers
- School of Medicine, University of Southampton, Tremona Road, Southampton, SO17 1BJ Hampshire, UK
| | - Sylwia Barker
- School of Chemistry, University of Southampton, Highfield Campus, Southampton, SO17 1BJ Hampshire, UK
| | - Manvendra Sharma
- School of Chemistry, University of Southampton, Highfield Campus, Southampton, SO17 1BJ Hampshire, UK
| | - Salim Khakoo
- School of Medicine, University of Southampton, Tremona Road, Southampton, SO17 1BJ Hampshire, UK
| | - Marcel Utz
- School of Chemistry, University of Southampton, Highfield Campus, Southampton, SO17 1BJ Hampshire, UK.
| |
Collapse
|
49
|
Heaton AR, Rehani PR, Hoefges A, Lopez AF, Erbe AK, Sondel PM, Skala MC. Single cell metabolic imaging of tumor and immune cells in vivo in melanoma bearing mice. Front Oncol 2023; 13:1110503. [PMID: 37020875 PMCID: PMC10067577 DOI: 10.3389/fonc.2023.1110503] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 03/02/2023] [Indexed: 03/22/2023] Open
Abstract
Introduction Metabolic reprogramming of cancer and immune cells occurs during tumorigenesis and has a significant impact on cancer progression. Unfortunately, current techniques to measure tumor and immune cell metabolism require sample destruction and/or cell isolations that remove the spatial context. Two-photon fluorescence lifetime imaging microscopy (FLIM) of the autofluorescent metabolic coenzymes nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) and flavin adenine dinucleotide (FAD) provides in vivo images of cell metabolism at a single cell level. Methods Here, we report an immunocompetent mCherry reporter mouse model for immune cells that express CD4 either during differentiation or CD4 and/or CD8 in their mature state and perform in vivo imaging of immune and cancer cells within a syngeneic B78 melanoma model. We also report an algorithm for single cell segmentation of mCherry-expressing immune cells within in vivo images. Results We found that immune cells within B78 tumors exhibited decreased FAD mean lifetime and an increased proportion of bound FAD compared to immune cells within spleens. Tumor infiltrating immune cell size also increased compared to immune cells from spleens. These changes are consistent with a shift towards increased activation and proliferation in tumor infiltrating immune cells compared to immune cells from spleens. Tumor infiltrating immune cells exhibited increased FAD mean lifetime and increased protein-bound FAD lifetime compared to B78 tumor cells within the same tumor. Single cell metabolic heterogeneity was observed in both immune and tumor cells in vivo. Discussion This approach can be used to monitor single cell metabolic heterogeneity in tumor cells and immune cells to study promising treatments for cancer in the native in vivo context.
Collapse
Affiliation(s)
- Alexa R. Heaton
- Morgridge Institute for Research, Madison, WI, United States
- Department of Human Oncology, University of Wisconsin, Madison, WI, United States
| | - Peter R. Rehani
- Morgridge Institute for Research, Madison, WI, United States
| | - Anna Hoefges
- Department of Human Oncology, University of Wisconsin, Madison, WI, United States
| | - Angelica F. Lopez
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, United States
| | - Amy K. Erbe
- Department of Human Oncology, University of Wisconsin, Madison, WI, United States
| | - Paul M. Sondel
- Department of Human Oncology, University of Wisconsin, Madison, WI, United States
- Department of Pediatrics, University of Wisconsin, Madison, WI, United States
| | - Melissa C. Skala
- Morgridge Institute for Research, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
50
|
Nava Lauson CB, Tiberti S, Corsetto PA, Conte F, Tyagi P, Machwirth M, Ebert S, Loffreda A, Scheller L, Sheta D, Mokhtari Z, Peters T, Raman AT, Greco F, Rizzo AM, Beilhack A, Signore G, Tumino N, Vacca P, McDonnell LA, Raimondi A, Greenberg PD, Huppa JB, Cardaci S, Caruana I, Rodighiero S, Nezi L, Manzo T. Linoleic acid potentiates CD8 + T cell metabolic fitness and antitumor immunity. Cell Metab 2023; 35:633-650.e9. [PMID: 36898381 DOI: 10.1016/j.cmet.2023.02.013] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 12/19/2022] [Accepted: 02/15/2023] [Indexed: 03/11/2023]
Abstract
The metabolic state represents a major hurdle for an effective adoptive T cell therapy (ACT). Indeed, specific lipids can harm CD8+ T cell (CTL) mitochondrial integrity, leading to defective antitumor responses. However, the extent to which lipids can affect the CTL functions and fate remains unexplored. Here, we show that linoleic acid (LA) is a major positive regulator of CTL activity by improving metabolic fitness, preventing exhaustion, and stimulating a memory-like phenotype with superior effector functions. We report that LA treatment enhances the formation of ER-mitochondria contacts (MERC), which in turn promotes calcium (Ca2+) signaling, mitochondrial energetics, and CTL effector functions. As a direct consequence, the antitumor potency of LA-instructed CD8 T cells is superior in vitro and in vivo. We thus propose LA treatment as an ACT potentiator in tumor therapy.
Collapse
Affiliation(s)
- Carina B Nava Lauson
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Milano, Italy
| | - Silvia Tiberti
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Milano, Italy
| | - Paola A Corsetto
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Federica Conte
- Institute for Systems Analysis and Computer Science "Antonio Ruberti," National Research Council, Rome, Italy
| | - Punit Tyagi
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Milano, Italy
| | - Markus Machwirth
- Department of Paediatric Haematology, Oncology and Stem Cell Transplantation, University Hospital of Würzburg, Würzburg, Germany
| | - Stefan Ebert
- Department of Paediatric Haematology, Oncology and Stem Cell Transplantation, University Hospital of Würzburg, Würzburg, Germany
| | - Alessia Loffreda
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, San Raffaele Vita-Salute University, Milano, Italy
| | - Lukas Scheller
- Interdisciplinary Center for Clinical Research (IZKF), Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
| | - Dalia Sheta
- Interdisciplinary Center for Clinical Research (IZKF), Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
| | - Zeinab Mokhtari
- Interdisciplinary Center for Clinical Research (IZKF), Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
| | - Timo Peters
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Ayush T Raman
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Francesco Greco
- Fondazione Pisana per la Scienza, ONLUS, San Giuliano Terme, Italy; Institute of Life Sciences, Sant' Anna School of Advanced Studies, Pisa, Italy
| | - Angela M Rizzo
- Department of Paediatric Haematology, Oncology and Stem Cell Transplantation, University Hospital of Würzburg, Würzburg, Germany
| | - Andreas Beilhack
- Interdisciplinary Center for Clinical Research (IZKF), Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
| | - Giovanni Signore
- Fondazione Pisana per la Scienza, ONLUS, San Giuliano Terme, Italy
| | - Nicola Tumino
- Immunology Research Area, Innate Lymphoid Cells Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Paola Vacca
- Immunology Research Area, Innate Lymphoid Cells Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Liam A McDonnell
- Fondazione Pisana per la Scienza, ONLUS, San Giuliano Terme, Italy
| | - Andrea Raimondi
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, San Raffaele Vita-Salute University, Milano, Italy
| | - Philip D Greenberg
- Clinical Research Division and Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Johannes B Huppa
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Simone Cardaci
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ignazio Caruana
- Department of Paediatric Haematology, Oncology and Stem Cell Transplantation, University Hospital of Würzburg, Würzburg, Germany
| | - Simona Rodighiero
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Milano, Italy
| | - Luigi Nezi
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Milano, Italy
| | - Teresa Manzo
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Milano, Italy.
| |
Collapse
|