1
|
Nour MA, Rajabivahid M, Mehdi MSS, Tahmasebi S, Dashtgol SN, Dehghani-Ghorbi M, Vanan AG, Ghorbaninezhad F. A new era in melanoma immunotherapy: focus on DCs metabolic reprogramming. Cancer Cell Int 2025; 25:149. [PMID: 40234886 PMCID: PMC12001691 DOI: 10.1186/s12935-025-03781-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/03/2025] [Indexed: 04/17/2025] Open
Abstract
Melanoma, being one of the most dangerous forms of skin cancer, is characterized by its aggressive and metastatic nature, with the potential to develop resistance to various treatments. This resistance makes the disease challenging to treat, emphasizing the need for new treatment strategies. Within the tumor microenvironment (TME), melanoma cells exploit metabolic shifts, particularly glycolysis, to create an immunosuppressive TME that prevents dendritic cells (DCs) from functioning properly. Essential metabolic alterations such as lactate and lipid accumulation, and lack of tryptophan disrupt DC maturation, antigen presentation, and T cell activation. In recent years, melanoma immunotherapy has increasingly focused on reprogramming the metabolism of DCs. This review paper aims to provide insights into the metabolic suppression of melanoma-associated DCs, allowing the design of therapeutic strategies based on metabolic interventions to promote or restore DC function. This contribution reviews the metabolic reprogramming of DCs as a new approach for melanoma immunotherapy.
Collapse
Affiliation(s)
- Mina Afrashteh Nour
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mansour Rajabivahid
- Department of Internal Medicine, Valiasr Hospital, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Marjan Sadat Seyed Mehdi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Safa Tahmasebi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepideh Nasirzadeh Dashtgol
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Tehran Medical Branch, Islamic Azad University, Tehran, Iran
| | - Mahmoud Dehghani-Ghorbi
- Hematology-Oncology Department, Imam Hossein Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ahmad Ghorbani Vanan
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Farid Ghorbaninezhad
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Parvanian S, Ge X, Garris CS. Recent developments in myeloid immune modulation in cancer therapy. Trends Cancer 2025; 11:365-375. [PMID: 39794212 DOI: 10.1016/j.trecan.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/13/2025]
Abstract
Myeloid cells play a crucial dual role in cancer progression and response to therapy, promoting tumor growth, enabling immune suppression, and contributing to metastatic spread. The ability of these cells to modulate the immune system has made them attractive targets for therapeutic strategies aimed at shifting their function from tumor promotion to fostering antitumor immunity. Therapeutic approaches targeting myeloid cells focus on modifying their numbers, genetics, metabolism, and interactions within the tumor microenvironment. These strategies aim to reverse their suppressive functions and redirect them to support antitumor immune responses by inhibiting immunosuppressive pathways, targeting specific receptors, and promoting their differentiation into less immunosuppressive phenotypes. Here, we discuss recent approaches to clinically target tumor myeloid cells, focusing on reprogramming myeloid cells to promote antitumor immunity.
Collapse
Affiliation(s)
- Sepideh Parvanian
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114, USA
| | - Xinying Ge
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114, USA; Master's Program in Immunology Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA
| | - Christopher S Garris
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114, USA; Department of Pathology, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114, USA.
| |
Collapse
|
3
|
Fan S, Wang W, Che W, Xu Y, Jin C, Dong L, Xia Q. Nanomedicines Targeting Metabolic Pathways in the Tumor Microenvironment: Future Perspectives and the Role of AI. Metabolites 2025; 15:201. [PMID: 40137165 PMCID: PMC11943624 DOI: 10.3390/metabo15030201] [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: 01/30/2025] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Tumor cells engage in continuous self-replication by utilizing a large number of resources and capabilities, typically within an aberrant metabolic regulatory network to meet their own demands. This metabolic dysregulation leads to the formation of the tumor microenvironment (TME) in most solid tumors. Nanomedicines, due to their unique physicochemical properties, can achieve passive targeting in certain solid tumors through the enhanced permeability and retention (EPR) effect, or active targeting through deliberate design optimization, resulting in accumulation within the TME. The use of nanomedicines to target critical metabolic pathways in tumors holds significant promise. However, the design of nanomedicines requires the careful selection of relevant drugs and materials, taking into account multiple factors. The traditional trial-and-error process is relatively inefficient. Artificial intelligence (AI) can integrate big data to evaluate the accumulation and delivery efficiency of nanomedicines, thereby assisting in the design of nanodrugs. Methods: We have conducted a detailed review of key papers from databases, such as ScienceDirect, Scopus, Wiley, Web of Science, and PubMed, focusing on tumor metabolic reprogramming, the mechanisms of action of nanomedicines, the development of nanomedicines targeting tumor metabolism, and the application of AI in empowering nanomedicines. We have integrated the relevant content to present the current status of research on nanomedicines targeting tumor metabolism and potential future directions in this field. Results: Nanomedicines possess excellent TME targeting properties, which can be utilized to disrupt key metabolic pathways in tumor cells, including glycolysis, lipid metabolism, amino acid metabolism, and nucleotide metabolism. This disruption leads to the selective killing of tumor cells and disturbance of the TME. Extensive research has demonstrated that AI-driven methodologies have revolutionized nanomedicine development, while concurrently enabling the precise identification of critical molecular regulators involved in oncogenic metabolic reprogramming pathways, thereby catalyzing transformative innovations in targeted cancer therapeutics. Conclusions: The development of nanomedicines targeting tumor metabolic pathways holds great promise. Additionally, AI will accelerate the discovery of metabolism-related targets, empower the design and optimization of nanomedicines, and help minimize their toxicity, thereby providing a new paradigm for future nanomedicine development.
Collapse
Affiliation(s)
| | | | | | | | | | - Lei Dong
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (S.F.); (W.W.); (W.C.); (Y.X.); (C.J.)
| | - Qin Xia
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (S.F.); (W.W.); (W.C.); (Y.X.); (C.J.)
| |
Collapse
|
4
|
Wu Z, Wang H, Zheng Z, Lin Y, Bian L, Geng H, Huang X, Zhu J, Jing H, Zhang Y, Ji C, Zhai B. IDO1 inhibition enhances CLDN18.2-CAR-T cell therapy in gastrointestinal cancers by overcoming kynurenine-mediated metabolic suppression in the tumor microenvironment. J Transl Med 2025; 23:275. [PMID: 40045363 PMCID: PMC11884131 DOI: 10.1186/s12967-025-06276-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/20/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-T cell therapy has achieved remarkable success in hematologic malignancies but faces significant limitations in gastrointestinal tumors due to the immunosuppressive tumor microenvironment (TME). Indoleamine 2,3-dioxygenase 1 (IDO1), a key enzyme in the TME, suppresses T cell efficacy by catalyzing tryptophan degradation to kynurenine (Kyn), leading to T cell exhaustion and reduced cytotoxicity. This study investigates the role of IDO1 inhibition in overcoming metabolic suppression by kynurenine and enhancing Claudin18.2 (CLDN18.2) CAR-T cell therapy in gastric and pancreatic adenocarcinoma models. METHODS We evaluated the impact of genetic knockdown and pharmacological inhibition of IDO1 (using epacadostat) on CAR-T cell functionality, including cytokine production and exhaustion marker expression. The effects of fludarabine and cyclophosphamide preconditioning on IDO1 expression, CAR-T cell infiltration, and antitumor activity was also examined. In vivo tumor models of gastric and pancreatic adenocarcinomas were used to assess the efficacy of combining IDO1 inhibition with CLDN18.2-CAR-T therapy. RESULTS IDO1 inhibition significantly enhanced CAR-T cell function by increasing cytokine production, reducing exhaustion markers by decreasing TOX expression and improving tumor cell lysis. Preconditioning with fludarabine and cyclophosphamide further suppressed IDO1 expression in the TME, facilitating enhanced CAR-T cell infiltration. In vivo studies demonstrated that combining IDO1 inhibition with CAR-T therapy led to robust tumor growth suppression and prolonged survival in gastric and pancreatic tumor models. CONCLUSIONS Targeting IDO1 represents a promising strategy to overcome immunosuppressive barriers in gastrointestinal cancers, improving the efficacy of CLDN18.2-CAR-T therapy. These findings highlight the potential for integrating IDO1 inhibition into CAR-T treatment regimens to address resistance in treatment-refractory cancers.
Collapse
Affiliation(s)
- Zhaorong Wu
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hongye Wang
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhigang Zheng
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yan Lin
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Linke Bian
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Haigang Geng
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | | | - Jiufei Zhu
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hongshu Jing
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yi Zhang
- Department of Urology, Fujian Renmin Hospital, Fuzhou, 350001, Fujian, China
| | - Chen Ji
- Department of Urology, Fujian Renmin Hospital, Fuzhou, 350001, Fujian, China.
| | - Bo Zhai
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Mini-Invasive Interventional Therapy Center, Shanghai East Hospital, Tongji University, Shanghai, 200025, China.
| |
Collapse
|
5
|
Yamamoto Y, Goto N, Kambara K, Fujigaki S, Fujigaki H, Takemura M, Nabeshima T, Tomita A, Saito K. Usefulness of the 3-hydroxykynurenine/kynurenic acid ratio as a diagnostic biomarker for diffuse larger B-cell lymphoma. Ann Clin Biochem 2025; 62:109-117. [PMID: 39439179 DOI: 10.1177/00045632241297873] [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: 10/25/2024]
Abstract
ObjectivesReports have shown that the kynurenine pathway, one of the pathways by which tryptophan is metabolized, is activated in patients with diffuse large B-cell lymphoma (DLBCL). Activation of the kynurenine pathway triggers the production of various metabolites, such as kynurenine (Kyn), 3-hydroxykynurenine (3-HK), 3-hydroxyanthranilic acid (3-HAA), kynurenic acid (KA), and anthranilic acid (AA), which contribute to immune tolerance. The current study aimed to investigate the changes in metabolites of kynurenine pathway in DLBCL patients and evaluate their performance predicting DLBCL.MethodsChanges in metabolites of kynurenine pathway were examined using high-performance liquid chromatography in 35 DLBCL patients (age 61.2 ± 13.5 years) and 44 healthy controls (age 58.5 ± 12.5 years).ResultsDLBCL patients had significantly higher levels of 3-HK, AA, and 3-HAA but lower levels of tryptophan (Trp) and KA compared to healthy controls. Given that the ratio of each metabolite represents the change in the Kyn pathway, the 3-HK/KA ratio was examined. Notably, DLBCL patients had a significantly higher 3-HK/KA ratio compared to healthy controls. In DLBCL, the area under the receiver operative characteristic (ROC) curve for 3-HK/KA (0.999) was higher than that for lactate dehydrogenase (0.885) and comparable to that for soluble interleukin-2 receptor (sIL-2R) (0.997). Based on ROC curve analysis, the 3-HK/KA ratio was found to be useful biomarker for the diagnosis of DLBCL.ConclusionOur results suggest that the 3-HK/KA ratio is a clinically useful biomarker of DLBCL. Moreover, its combination with existing markers, such as sIL-2R, can improve its effectiveness of diagnosing DLBCL.
Collapse
Affiliation(s)
- Yasuko Yamamoto
- Advanced Diagnostic System Development, Fujita Health University Graduate School of Health Sciences, Toyoake, Japan
| | - Naoe Goto
- Department of Hematology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Kengo Kambara
- Medical Systems Research & Development Center, Medical Systems Business Division, FUJIFILM Corporation, Amagasaki, Japan
| | - Suwako Fujigaki
- Advanced Diagnostic System Development, Fujita Health University Graduate School of Health Sciences, Toyoake, Japan
| | - Hidetsugu Fujigaki
- Advanced Diagnostic System Development, Fujita Health University Graduate School of Health Sciences, Toyoake, Japan
| | - Masao Takemura
- Advanced Diagnostic System Development, Fujita Health University Graduate School of Health Sciences, Toyoake, Japan
| | - Toshitaka Nabeshima
- Laboratory of Health and Medical Science Innovation, Fujita Health University, Toyoake, Japan
- Japanese Drug Organization of Appropriate Use and Research, Nagoya, Japan
| | - Akihiro Tomita
- Department of Hematology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Kuniaki Saito
- Advanced Diagnostic System Development, Fujita Health University Graduate School of Health Sciences, Toyoake, Japan
- Laboratory of Health and Medical Science Innovation, Fujita Health University, Toyoake, Japan
- Japanese Drug Organization of Appropriate Use and Research, Nagoya, Japan
| |
Collapse
|
6
|
Liu Y, Li C, Cui X, Liu C, Xiao P, Yang W. Kynureninase induce cuproptosis resistance in gastric cancer progression through downregulating lipotic acid synthetase mediated non-canonical mechanism. Cell Signal 2025; 127:111565. [PMID: 39681223 DOI: 10.1016/j.cellsig.2024.111565] [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: 09/04/2024] [Revised: 11/15/2024] [Accepted: 12/09/2024] [Indexed: 12/18/2024]
Abstract
BACKGROUND Gastric cancer (GC) is among the most malignant tumors, with the lowest five-year survival rate, and limited treatment options. Kynureninase (KYNU), is a key molecule in tryptophan metabolism and promotes tumor progression and immunosuppression. Cuproptosis is a non-apoptotic cell death mechanism, primarily due to oxidative stress caused by copper ion accumulation, that is related to tumor progression and drug resistance. KYNU can inhibit ferroptosis of tumor cells by alleviating oxidative stress. Here, we explored whether KYNU can regulate the biological behavior of GC and cuproptosis. METHODS Expression, prognostic association, and functional analysis of KYNU in GC and tumor-adjacent tissues were analyzed using data from The Cancer Genome Atlas and clinical specimens. Effects of KYNU on proliferation, invasion, metastasis, and cuproptosis of GC cells were detected by CCK8, clone formation, Transwell, and flow cytometry assays. Elesclomol (ES) combined with CuCl2 were used to induce cuproptosis in GC cells. 3-hydroxyanthranilic acid (3-HA) was used to indicate KYNU function. Key cuproptosis genes were detected by qPCR and WB. The effects of KYNU on GC cell behavior and cuproptosis through lipoic acid synthetase (LIAS) were verified by stable overexpression and knockdown of LIAS. RESULTS KYNU is highly expressed in GC, and high KYNU expression is an independent predictor of poor prognosis in patients with GC. KYNU can promote GC cell proliferation, invasion, metastasis, and cuproptosis resistance. 3-HA had a certain inhibitory effect on the expression of LIAS, but it was not significant. KYNU had no effect on the intracellular 3-HA level. KYNU expression was negatively correlated with that of LIAS, and promoted GC cell proliferation, invasion, metastasis, and cuproptosis resistance by downregulating LIAS. CONCLUSIONS KYNU can promote GC proliferation, invasion, metastasis, and cuproptosis resistance.This effect is not associated with its metabolite 3-HA, but is achieved by a non classical mechanisms that downregulating the expression of LIAS, a key gene of cuproptosis.
Collapse
Affiliation(s)
- Yuanda Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China.
| | - Xilun Cui
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chang Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Pengtuo Xiao
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China.
| |
Collapse
|
7
|
Elmer DP, Stockmaier G, Grund-Gröschke S, Strobl V, Dang HH, Wiederstein M, Licha D, Strobl A, Eglseer A, Sternberg C, Tesanovic S, Gruber W, Wolff F, Moriggl R, Risch A, Reischl R, Huber CG, Krenn PW, Fortelny N, Horejs-Hoeck J, Aberger F. Cooperative Hedgehog/GLI and JAK/STAT signaling drives immunosuppressive tryptophan/kynurenine metabolism via synergistic induction of IDO1 in skin cancer. Cell Commun Signal 2025; 23:91. [PMID: 39962447 PMCID: PMC11834474 DOI: 10.1186/s12964-025-02101-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 02/09/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Pharmacological targeting of Hedgehog (HH)/GLI has proven effective for certain blood, brain and skin cancers including basal cell carcinoma (BCC). However, limited response rates and the development of drug resistance call for improved anti-HH therapies that take synergistic crosstalk mechanisms and immune evasion strategies into account. In previous work, we demonstrated that cooperation of HH/GLI and Interleukin 6 (IL6)/STAT3 signaling drives BCC growth. Whether synergistic HH-IL6 signaling promotes BCC via the activation of immune evasion mechanisms remained unclear. METHODS HH-IL6 regulated immunosuppressive genes such as indoleamine 2,3-dioxygenase 1 (IDO1) were identified by gene expression profiling. IDO1 expression was evaluated in human BCC and melanoma models by qPCR and Western blot analyses. The cis-regulatory region of IDO1 was interrogated for HH-IL6-regulated GLI and STAT transcription factor binding and epigenetic modifications by targeted chromatin-immunoprecipitation and bisulfite pyrosequencing. Functional analyses of the immunosuppressive effects of IDO1 involved HPLC-MS measurements of its metabolites and the assessment of T cell proliferation via flow cytometry. Bioinformatic analyses of GLI-STAT cooperation were conducted on published bulk and single-cell RNA-seq data of human BCC and melanoma patients. RESULTS We identified IDO1 as a target gene of cooperative GLI-STAT activity in BCC and melanoma. GLI1 and STAT3 transcription factors synergistically enhanced IDO1 expression by jointly binding to the cis-regulatory region of IDO1 and by increasing active chromatin marks at the histone level. In human melanoma cells, inhibition of GLI1 expression prevented the induction of IDO1 expression in response to IL6/STAT3 and IFNγ/STAT1 signaling. Pharmacological targeting of HH/GLI signaling reduced IDO1 expression, resulting in decreased production of the immunosuppressive metabolite kynurenine. Further, inhibition of GLI1 enhanced the efficacy of the selective IDO1 inhibitor epacadostat and rescued T cell proliferation by attenuating IDO1/kynurenine-mediated immunosuppression. Elevated expression of IDO1 correlated with active HH/GLI and JAK/STAT signaling in skin cancer patients supporting the clinical relevance of the mechanistic data presented. CONCLUSIONS These results identify the immunosuppressive IDO1-kynurenine pathway as a novel pro-tumorigenic target of oncogenic GLI and STAT1/STAT3 cooperation. Our data suggest simultaneous pharmacological targeting of these signaling axes as rational combination therapy in melanoma and non-melanoma skin cancers.
Collapse
Affiliation(s)
- Dominik P Elmer
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Georg Stockmaier
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Sandra Grund-Gröschke
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Victoria Strobl
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Hieu-Hoa Dang
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Markus Wiederstein
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - David Licha
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Anna Strobl
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Anna Eglseer
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Christina Sternberg
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
- Institute of Biochemistry, University of Kiel, Kiel, Germany
| | - Suzana Tesanovic
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Wolfgang Gruber
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Florian Wolff
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Richard Moriggl
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris Lodron University of Salzburg, Salzburg, Austria
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Angela Risch
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris Lodron University of Salzburg, Salzburg, Austria
| | - Roland Reischl
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Christian G Huber
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris Lodron University of Salzburg, Salzburg, Austria
| | - Peter W Krenn
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - Nikolaus Fortelny
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris Lodron University of Salzburg, Salzburg, Austria
| | - Jutta Horejs-Hoeck
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria
- Center for Tumor Biology and Immunology, Paris Lodron University of Salzburg, Salzburg, Austria
| | - Fritz Aberger
- Department of Biosciences and Medical Biology, Cancer Cluster Salzburg, Paris Lodron University of Salzburg, Hellbrunner Strasse 34, Salzburg, 5020, Austria.
- Center for Tumor Biology and Immunology, Paris Lodron University of Salzburg, Salzburg, Austria.
| |
Collapse
|
8
|
Hennes E, Lucas B, Scholes NS, Cheng XF, Scott DC, Bischoff M, Reich K, Gasper R, Lucas M, Xu TT, Pulvermacher LM, Dötsch L, Imrichova H, Brause A, Naredla KR, Sievers S, Kumar K, Janning P, Gersch M, Murray PJ, Schulman BA, Winter GE, Ziegler S, Waldmann H. Identification of a Monovalent Pseudo-Natural Product Degrader Class Supercharging Degradation of IDO1 by its native E3 KLHDC3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.10.602857. [PMID: 39026748 PMCID: PMC11257573 DOI: 10.1101/2024.07.10.602857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Targeted protein degradation (TPD) modulates protein function beyond inhibition of enzyme activity or protein-protein interactions. Most degrader drugs function by directly mediating proximity between a neosubstrate and hijacked E3 ligase. Here, we identified pseudo-natural products derived from (-)-myrtanol, termed iDegs that inhibit and induce degradation of the immunomodulatory enzyme indoleamine-2,3-dioxygenase 1 (IDO1) by a distinct mechanism. iDegs boost IDO1 ubiquitination and degradation by the cullin-RING E3 ligase CRL2KLHDC3, which we identified to natively mediate ubiquitin-mediated degradation of IDO1. Therefore, iDegs increase IDO1 turnover using the native proteolytic pathway. In contrast to clinically explored IDO1 inhibitors, iDegs reduce formation of kynurenine by both inhibition and induced degradation of the enzyme and, thus, would also modulate non-enzymatic functions of IDO1. This unique mechanism of action may open up new therapeutic opportunities for the treatment of cancer beyond classical inhibition of IDO1.
Collapse
Affiliation(s)
- Elisabeth Hennes
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
- Technische Universität Dortmund, Fakultät Chemie und Chemische Biologie, Otto-Hahn-Straße 6, 44221 Dortmund, Germany
| | - Belén Lucas
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Natalie S. Scholes
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Xiu-Fen Cheng
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Daniel C. Scott
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Matthias Bischoff
- Compound Management and Screening Center Otto-Hahn-Str.11, 44227 Dortmund, Germany
| | - Katharina Reich
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Raphael Gasper
- Max-Planck-Institut für Molekulare Physiologie, Zentrale Einheit für Kristallographie und Biophysik, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - María Lucas
- Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-CSIC, C/Albert Einstein 22, PCTCAN, 39011 Santander, Spain
| | - Teng Teng Xu
- Immunoregulation Research Group, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Lisa-Marie Pulvermacher
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Lara Dötsch
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
- Technische Universität Dortmund, Fakultät Chemie und Chemische Biologie, Otto-Hahn-Straße 6, 44221 Dortmund, Germany
| | - Hana Imrichova
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Alexandra Brause
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Kesava Reddy Naredla
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Sonja Sievers
- Compound Management and Screening Center Otto-Hahn-Str.11, 44227 Dortmund, Germany
| | - Kamal Kumar
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Petra Janning
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Malte Gersch
- Technische Universität Dortmund, Fakultät Chemie und Chemische Biologie, Otto-Hahn-Straße 6, 44221 Dortmund, Germany
- Chemical Genomics Centre, Max-Planck-Institut für Molekulare Physiologie, 44227 Dortmund, Germany
| | - Peter J. Murray
- Immunoregulation Research Group, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Brenda A. Schulman
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Georg E. Winter
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Slava Ziegler
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Herbert Waldmann
- Max-Planck-Institut für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
- Technische Universität Dortmund, Fakultät Chemie und Chemische Biologie, Otto-Hahn-Straße 6, 44221 Dortmund, Germany
| |
Collapse
|
9
|
Song L, Xue F, Li T, Zhang Q, Xu X, He C, Zhao B, Han XX, Cai L. Differential Diagnosis of Urinary Cancers by Surface-Enhanced Raman Spectroscopy and Machine Learning. Anal Chem 2025; 97:27-32. [PMID: 39757799 DOI: 10.1021/acs.analchem.4c05287] [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: 01/07/2025]
Abstract
Bladder, kidney, and prostate cancers are prevalent urinary cancers, and developing efficient detection methods is of significance for the early diagnosis of them. However, noninvasive and sensitive detection of urinary cancers still challenges traditional techniques. In this study, we developed a SERS-based method to analyze serum samples from patients with urinary cancers. Rapid, label-free, and highly sensitive detection of human sera is achieved by cleaning and aggregating silver nanoparticles. Furthermore, a long short-term memory deep learning algorithm is used to distinguish serum spectra, and the performance of the model is evaluated by comparing the accuracy, sensitivity, specificity, and receiver operating characteristic curves. Taking advantage of SERS and machine learning in sensitivity and data processing, the three urinary cancers are clearly classified. This is the first attempt to exploit the SERS-machine learning strategy to discriminate multiple urinary cancers with clinical serum samples, and our results showed the potential application of this method in the early diagnosis and screening of cancers.
Collapse
Affiliation(s)
- Li Song
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, P. R. China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Fei Xue
- Department of Laboratory Medicine, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Tingmiao Li
- Department of Laboratory Medicine, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Qian Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, P. R. China
| | - Xuesong Xu
- Department of Laboratory Medicine, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Chengyan He
- Department of Laboratory Medicine, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Bing Zhao
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Xiao Xia Han
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Linjun Cai
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, P. R. China
| |
Collapse
|
10
|
Clay R, Li K, Jin L. Metabolic Signaling in the Tumor Microenvironment. Cancers (Basel) 2025; 17:155. [PMID: 39796781 PMCID: PMC11719658 DOI: 10.3390/cancers17010155] [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: 10/30/2024] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Cancer cells must reprogram their metabolism to sustain rapid growth. This is accomplished in part by switching to aerobic glycolysis, uncoupling glucose from mitochondrial metabolism, and performing anaplerosis via alternative carbon sources to replenish intermediates of the tricarboxylic acid (TCA) cycle and sustain oxidative phosphorylation (OXPHOS). While this metabolic program produces adequate biosynthetic intermediates, reducing agents, ATP, and epigenetic remodeling cofactors necessary to sustain growth, it also produces large amounts of byproducts that can generate a hostile tumor microenvironment (TME) characterized by low pH, redox stress, and poor oxygenation. In recent years, the focus of cancer metabolic research has shifted from the regulation and utilization of cancer cell-intrinsic pathways to studying how the metabolic landscape of the tumor affects the anti-tumor immune response. Recent discoveries point to the role that secreted metabolites within the TME play in crosstalk between tumor cell types to promote tumorigenesis and hinder the anti-tumor immune response. In this review, we will explore how crosstalk between metabolites of cancer cells, immune cells, and stromal cells drives tumorigenesis and what effects the competition for resources and metabolic crosstalk has on immune cell function.
Collapse
Affiliation(s)
| | | | - Lingtao Jin
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (R.C.); (K.L.)
| |
Collapse
|
11
|
Sugiyama S, Yumimoto K, Nakayama KI. Immune Cell Profiling Reveals a Common Pattern in Premetastatic Niche Formation Across Various Cancer Types. Cancer Med 2025; 14:e70557. [PMID: 39740041 DOI: 10.1002/cam4.70557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/05/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Metastasis is the major cause of cancer-related mortality. The premetastatic niche is a promising target for its prevention. However, the generality and cellular dynamics in premetastatic niche formation have remained unclear. AIMS This study aimed to elucidate the generality and cellular dynamics in premetastatic niche formation. MATERIALS AND METHODS We performed comprehensive flow cytometric analysis of lung and peripheral immune cells at three time points (early premetastatic, late premetastatic, and micrometastatic phases) for mice with subcutaneous implants of three types of cancer cells (breast cancer, lung cancer, or melanoma cells). The immuno-cell profiles were then used to predict the metastatic phase by machine learning. RESULTS We found a common pattern of changes in both lung and peripheral immune cell profiles across the three cancer types, including a decrease in the proportion of eosinophils in the early premetastatic phase, an increase in that of regulatory T cells in the late premetastatic phase, and an increase in that of polymorphonuclear myeloid-derived suppressor cells and a decrease in that of B cells in the micrometastatic phase. Machine learning using immune cell profiles could predict the metastatic phase with approximately 75% accuracy. DISCUSSION Validation of our findings in humans will require data on the presence or absence of micrometastases in patients and the accumulation of comprehensive and temporal information on immune cells. In addition, blood proteins, extracellular vesicles, DNA, RNA, or metabolites may be useful for more accurate prediction. CONCLUSION The discovery of generalities in premetastatic niche formation allow prediction of metastatic phase and provide a basis for the development of methods for early detection and prevention of cancer metastasis in a cancer type-independent manner.
Collapse
Affiliation(s)
- Shigeaki Sugiyama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Department of Molecular Biology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Kanae Yumimoto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Dynamic Chemical Life Science Laboratory, Department of Chemistry, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Anticancer Strategies Laboratory, Advanced Research Initiative, Institute of Science Tokyo, Tokyo, Japan
| |
Collapse
|
12
|
Aden D, Sureka N, Zaheer S, Chaurasia JK, Zaheer S. Metabolic Reprogramming in Cancer: Implications for Immunosuppressive Microenvironment. Immunology 2025; 174:30-72. [PMID: 39462179 DOI: 10.1111/imm.13871] [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: 05/18/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
Cancer is a complex and heterogeneous disease characterised by uncontrolled cell growth and proliferation. One hallmark of cancer cells is their ability to undergo metabolic reprogramming, which allows them to sustain their rapid growth and survival. This metabolic reprogramming creates an immunosuppressive microenvironment that facilitates tumour progression and evasion of the immune system. In this article, we review the mechanisms underlying metabolic reprogramming in cancer cells and discuss how these metabolic alterations contribute to the establishment of an immunosuppressive microenvironment. We also explore potential therapeutic strategies targeting metabolic vulnerabilities in cancer cells to enhance immune-mediated anti-tumour responses. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT02044861, NCT03163667, NCT04265534, NCT02071927, NCT02903914, NCT03314935, NCT03361228, NCT03048500, NCT03311308, NCT03800602, NCT04414540, NCT02771626, NCT03994744, NCT03229278, NCT04899921.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical Science and Research, New Delhi, India
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
13
|
Salmi T, Ameur D, Dali-Sahi M, Dib J, Amraoui N, Kachekouche Y, Dennouni-Medjati N. Exploration of plasma tryptophan levels along with Ki-67 expression binomial investigation for forecasting tumor aggressiveness within invasive ductal breast cancer. J Mol Histol 2024; 56:52. [PMID: 39708255 DOI: 10.1007/s10735-024-10333-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/07/2024] [Indexed: 12/23/2024]
Abstract
Ki-67 is a histological marker indicating cancer aggressiveness, while tryptophan (TRP) depletion modulates immune responses, including tumor aggressiveness. The study evaluates Ki-67's predictive value in relation to plasma TRP levels in invasive ductal carcinoma of breast cancer, aiming to improve understanding of tumor characteristics and clinical behavior. A study involving 165 women, measured plasma TRP levels and Ki-67 and analyzed their relationship with tumor aggressiveness markers using statistical analyses and predictive models. Our study highlighted a significant correlation between decreased plasma levels of TRP and a high mitotic index, measured by the Ki-67 marker (Pearson correlation coefficient r = - 0.402; p = 0.011). Tryptophan levels below 40 µmol/L were associated with a Ki-67 level above 15%, suggesting more active tumor growth in patients. Additionally, several risk factors for BC were identified within the studied population. The demographic and clinical characteristics of the participants include an average age of 63 years, plasma glucose levels above 1.2 g/L, and plasma TRP levels below 40 µmol/L, which are associated with an increased risk of BC. Furthermore, various polynomial logistic regression models indicate that TRP levels may be predicted based on Ki-67 expression, providing a promising approach to refine prognostic assessments. The study showed a correlation between low levels of tryptophan (TRP) and a high Ki-67 mitotic index in breast cancer patients, particularly in invasive ductal carcinoma, which is strongly linked to the aggressiveness of the disease. The integration of these markers into routine practice remains a technical and economic challenge.
Collapse
MESH Headings
- Tryptophan/blood
- Tryptophan/metabolism
- Humans
- Ki-67 Antigen/blood
- Ki-67 Antigen/metabolism
- Ki-67 Antigen/analysis
- Female
- Breast Neoplasms/blood
- Breast Neoplasms/pathology
- Breast Neoplasms/diagnosis
- Middle Aged
- Carcinoma, Ductal, Breast/blood
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/diagnosis
- Carcinoma, Ductal, Breast/metabolism
- Aged
- Biomarkers, Tumor/blood
- Adult
- Prognosis
- Mitotic Index
- Aged, 80 and over
Collapse
Affiliation(s)
- Takwa Salmi
- Department of Biology, University of Tlemcen, 22, Rue Abi Ayed Abdelkrim, Fg Pasteur, B.P 119, 13000, Tlemcen, Algeria
- Analytical Chemistry and Electrochemistry Laboratory, University of Tlemcen, 13000, Tlemcen, Algeria
| | - Djilali Ameur
- Departement of Physics, University of Tlemcen, 22, Rue Abi Ayed Abdelkrim, Fg Pasteur, B.P 119, 13000, Tlemcen, Algeria.
- Laboratory of Theoretical Physics, Faculty of Sciences, University of Tlemcen, 13000, Tlemcen, Algeria.
| | - Majda Dali-Sahi
- Department of Biology, University of Tlemcen, 22, Rue Abi Ayed Abdelkrim, Fg Pasteur, B.P 119, 13000, Tlemcen, Algeria
- Analytical Chemistry and Electrochemistry Laboratory, University of Tlemcen, 13000, Tlemcen, Algeria
| | - Joanna Dib
- Analytical Chemistry and Electrochemistry Laboratory, University of Tlemcen, 13000, Tlemcen, Algeria
- Departement of Mathematics, University of Tlemcen, 22, Rue Abi Ayed Abdelkrim, Fg Pasteur, B.P 119, 13000, Tlemcen, Algeria
| | - Nawel Amraoui
- Department of Biology, University of Tlemcen, 22, Rue Abi Ayed Abdelkrim, Fg Pasteur, B.P 119, 13000, Tlemcen, Algeria
| | - Youssouf Kachekouche
- Department of Biology, Faculty of Nature and Life Sciences, University of Chlef, Chlef, Algeria
| | - Nouria Dennouni-Medjati
- Department of Biology, University of Tlemcen, 22, Rue Abi Ayed Abdelkrim, Fg Pasteur, B.P 119, 13000, Tlemcen, Algeria
- Analytical Chemistry and Electrochemistry Laboratory, University of Tlemcen, 13000, Tlemcen, Algeria
| |
Collapse
|
14
|
Sharma N, Bhat SH, Mathew B, Yadav M, Tripathi G, Bindal V, Yadav S, Sharma N, Pandey S, Hemati H, Bohra D, Rana R, Sharma NK, Falari S, Pamecha V, Maras JS. Bile molecular landscape provides pathological insight and classifies signatures predictive of carcinoma of the gall bladder. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200904. [PMID: 39640865 PMCID: PMC11617464 DOI: 10.1016/j.omton.2024.200904] [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: 06/26/2024] [Revised: 10/01/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Carcinoma of the gall bladder (CAGB) has a poor prognosis. Molecular analysis of bile could classify indicators of CAGB. Bile samples (n = 87; training cohort) were screened for proteomics and metabolomics signatures of cancer detection. In bile, CAGB showed distinct proteomic (217 upregulated, 258 downregulated) and metabolomic phenotypes (111 upregulated, 505 downregulated, p < 0.05, fold change > 1.5, false discovery rate <0.01) linked to significantly increased inflammation (coagulation, arachidonic acid, bile acid) and alternate energy pathways (pentose-phosphate pathway, amino acids, lipid metabolism); and decreased glycolysis, cholesterol metabolism, PPAR, RAS, and RAP1 signaling, oxidative phosphorylation, and others compared to gallstone or healthy controls (p < 0.05). Bile proteins/metabolites signatures showed significant correlation (r 2 > 0.5, p < 0.05) with clinical parameters. Metabolite/protein signature-based probability of detection for CAGB (cancer) was >90% (p < 0.05), with area under the receiver operating characteristic curve >0.94. Validation of the top four metabolites-toluene, 5,6-DHET, creatine, and phenylacetaldehyde-in separate cohorts (n = 80; bile [T1] and paired plasma [T2]) showed accuracy (99%) and sensitivity/specificity (>98%) for CAGB detection. Tissue validation showed bile 5,6-DHET positively correlated with tissue PCNA (proliferation), and caspase-3 linked to cancer development (r 2 >0.5, p < 0.05). In conclusion, the bile molecular landscape provides critical molecular understanding and outlines metabolomic indicator panels for early CAGB detection.
Collapse
Affiliation(s)
- Nupur Sharma
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Sadam H. Bhat
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Babu Mathew
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Manisha Yadav
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Gaurav Tripathi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Vasundhra Bindal
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Sanju Yadav
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Neha Sharma
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Sushmita Pandey
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Hami Hemati
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Deepika Bohra
- Department of Research, GRIPMER, New Delhi 110060, India
| | - Rashmi Rana
- Department of Research, GRIPMER, New Delhi 110060, India
| | - Narendra Kumar Sharma
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Rajasthan 304022, India
| | - Sanyam Falari
- Department of Liver Transplant and HepatoPancreato Biliary Surgery, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Viniyendra Pamecha
- Department of Liver Transplant and HepatoPancreato Biliary Surgery, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Jaswinder Singh Maras
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| |
Collapse
|
15
|
Li H, Liu J, Wang J, Li Z, Yu J, Huang X, Wan B, Meng X, Zhang X. Improving the Anti-Tumor Effect of Indoleamine 2,3-Dioxygenase Inhibitor CY1-4 by CY1-4 Nano-Skeleton Drug Delivery System. J Funct Biomater 2024; 15:372. [PMID: 39728172 DOI: 10.3390/jfb15120372] [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: 11/12/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
Background: CY1-4, 9-nitropyridine [2',3':4,5] pyrimido [1,2-α] indole -5,11- dione, is an indoleamine 2,3-dioxygenase (IDO) inhibitor and a poorly water-soluble substance. It is very important to increase the solubility of CY1-4 to improve its bioavailability and therapeutic effect. In this study, the mesoporous silica nano-skeleton carrier material Sylysia was selected as the carrier to load CY1-4, and then the CY1-4 nano-skeleton drug delivery system (MSNM@CY1-4) was prepared by coating the hydrophilic polymer material Hydroxypropyl methylcellulose (HPMC) and the lipid material Distearoylphosphatidyl-ethanolamine-poly(ethylene glycol)2000 (DSPE-PEG2000) to improve the anti-tumor effect of CY1-4. Methods: The solubility and dissolution of MSNM@CY1-4 were investigated, and its bioavailability, anti-tumor efficacy, IDO inhibitory ability and immune mechanism were evaluated in vivo. Results: CY1-4 was loaded in MSNM@CY1-4 in an amorphous form, and MSNM@CY1-4 could significantly improve the solubility (up to about 200 times) and dissolution rate of CY1-4. In vivo studies showed that the oral bioavailability of CY1-4 in 20 mg/kg MSNM@CY1-4 was about 23.9-fold more than that in 50 mg/kg CY1-4 suspension. In B16F10 tumor-bearing mice, MSNM@CY1-4 significantly inhibited tumor growth, prolonged survival time, significantly inhibited IDO activity in blood and tumor tissues, and reduced Tregs in tumor tissues and tumor-draining lymph nodes to improve anti-tumor efficacy. Conclusions: The nano-skeleton drug delivery system (MSNM@CY1-4) constructed in this study is a potential drug delivery platform for improving the anti-tumor effect of oral poorly water-soluble CY1-4.
Collapse
Affiliation(s)
- Hui Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Junwei Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jingru Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhuoyue Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jianming Yu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xu Huang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bingchuan Wan
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xiangbao Meng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xuan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Ningbo Institute of Marine Medicine, Peking University, Ningbo 315832, China
| |
Collapse
|
16
|
Rosario SR, Long MD, Chilakapati S, Gomez EC, Battaglia S, Singh PK, Wang J, Wang K, Attwood K, Hess SM, McGray AJR, Odunsi K, Segal BH, Paragh G, Liu S, Wargo JA, Zsiros E. Integrative multi-omics analysis uncovers tumor-immune-gut axis influencing immunotherapy outcomes in ovarian cancer. Nat Commun 2024; 15:10609. [PMID: 39638782 PMCID: PMC11621351 DOI: 10.1038/s41467-024-54565-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024] Open
Abstract
Recurrent ovarian cancer patients, especially those resistant to platinum, lack effective curative treatments. To address this, we conducted a phase 2 clinical trial (NCT02853318) combining pembrolizumab with bevacizumab, to increase T cell infiltration into the tumor, and oral cyclophosphamide, to reduce the number of regulatory T cells. The trial accrued 40 heavily pretreated recurrent ovarian cancer patients. The primary endpoint, progression free survival, was extended to a median of 10.2 months. The secondary endpoints demonstrated an objective response rate of 47.5%, and disease control in 30% of patients for over a year while maintaining a good quality of life. We performed comprehensive molecular, immune, microbiome, and metabolic profiling on samples of trial patients. Here, we show increased T and B cell clusters and distinct microbial patterns with amino acid and lipid metabolism are linked to exceptional clinical responses. This study suggests the immune milieu and host-microbiome can be leveraged to improve antitumor response in future immunotherapy trials.
Collapse
Affiliation(s)
- Spencer R Rosario
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Mark D Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Shanmuga Chilakapati
- New England Inflammation and Tissue Protection Institute, Northeastern University, Boston, MA, 02111, USA
| | - Eduardo Cortes Gomez
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Sebastiano Battaglia
- Computational Biology Office of Translational Research, Janssen Pharmaceuticals, Buffalo, NY, 14263, USA
| | - Prashant K Singh
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Katy Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Kristopher Attwood
- Department of Clinical Research, American College of Radiology, Reston, VA, 20191, USA
| | - Suzanne M Hess
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - A J Robert McGray
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Kunle Odunsi
- Department of Obstetrics and Gynecology, University of Chicago Comprehensive Cancer Center, Chicago, IL, 60637, USA
| | - Brahm H Segal
- Department of Internal Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Gyorgy Paragh
- Department of Dermatology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Jennifer A Wargo
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Emese Zsiros
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| |
Collapse
|
17
|
Suvieri C, Belladonna ML, Volpi C. The Two Sides of Indoleamine 2,3-Dioxygenase 2 (IDO2). Cells 2024; 13:1894. [PMID: 39594642 PMCID: PMC11593294 DOI: 10.3390/cells13221894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) and IDO2 originated from gene duplication before vertebrate divergence. While IDO1 has a well-defined role in immune regulation, the biological role of IDO2 remains unclear. Discovered in 2007, IDO2 is located near the IDO1 gene. Because of their high sequence similarity, IDO2 was initially thought to be a tryptophan (Trp)-degrading enzyme like IDO1. Differently from what expected, IDO2 displays extremely low catalytic activity toward Trp. Nevertheless, many studies, often contradictory, have tried to demonstrate that IDO2 modulates immune responses by catabolizing Trp into kynurenine, an unconvincing hypothesis linked to an incomplete understanding of IDO2's activity. In this study, we review IDO2's functional role beyond Trp metabolism. IDO2's evolutionary persistence across species, despite being almost inactive as an enzyme, suggests it has some relevant biological importance. IDO2 expression in human normal cells is poor, but significant in various cancers, with two prevalent SNPs. Overall, the comparison of IDO2 to IDO1 as a Trp-degrading enzyme may have led to misunderstandings about IDO2's true physiological and pathological roles. New insights suggest that IDO2 might function more as a signaling molecule, particularly in cancer contexts, and further studies could reveal its potential as a target for cancer therapy.
Collapse
Affiliation(s)
| | | | - Claudia Volpi
- Section of Pharmacology, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy; (C.S.); (M.L.B.)
| |
Collapse
|
18
|
Yan J, Chen D, Ye Z, Zhu X, Li X, Jiao H, Duan M, Zhang C, Cheng J, Xu L, Li H, Yan D. Molecular mechanisms and therapeutic significance of Tryptophan Metabolism and signaling in cancer. Mol Cancer 2024; 23:241. [PMID: 39472902 PMCID: PMC11523861 DOI: 10.1186/s12943-024-02164-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024] Open
Abstract
Tryptophan (Trp) metabolism involves three primary pathways: the kynurenine (Kyn) pathway (KP), the 5-hydroxytryptamine (serotonin, 5-HT) pathway, and the indole pathway. Under normal physiological conditions, Trp metabolism plays crucial roles in regulating inflammation, immunity, and neuronal function. Key rate-limiting enzymes such as indoleamine-2,3-dioxygenase (IDO), Trp-2,3-dioxygenase (TDO), and kynurenine monooxygenase (KMO) drive these metabolic processes. Imbalances in Trp metabolism are linked to various cancers and often correlate with poor prognosis and adverse clinical characteristics. Dysregulated Trp metabolism fosters tumor growth and immune evasion primarily by creating an immunosuppressive tumor microenvironment (TME). Activation of the KP results in the production of immunosuppressive metabolites like Kyn, which modulate immune responses and promote oncogenesis mainly through interaction with the aryl hydrocarbon receptor (AHR). Targeting Trp metabolism therapeutically has shown significant potential, especially with the development of small-molecule inhibitors for IDO1, TDO, and other key enzymes. These inhibitors disrupt the immunosuppressive signals within the TME, potentially restoring effective anti-tumor immune responses. Recently, IDO1 inhibitors have been tested in clinical trials, showing the potential to enhance the effects of existing cancer therapies. However, mixed results in later-stage trials underscore the need for a deeper understanding of Trp metabolism and its complex role in cancer. Recent advancements have also explored combining Trp metabolism inhibitors with other treatments, such as immune checkpoint inhibitors, chemotherapy, and radiotherapy, to enhance therapeutic efficacy and overcome resistance mechanisms. This review summarizes the current understanding of Trp metabolism and signaling in cancer, detailing the oncogenic mechanisms and clinical significance of dysregulated Trp metabolism. Additionally, it provides insights into the challenges in developing Trp-targeted therapies and future research directions aimed at optimizing these therapeutic strategies and improving patient outcomes.
Collapse
Affiliation(s)
- Jing Yan
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan, Zhengzhou, China
| | - Di Chen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Zi Ye
- Department of Scientific Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xuqiang Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Xueyuan Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Henan Jiao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Mengjiao Duan
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan, Zhengzhou, China
| | - Chaoli Zhang
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan, Zhengzhou, China
| | - Jingliang Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan, Zhengzhou, China
| | - Lixia Xu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Hongjiang Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.
| | - Dongming Yan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
19
|
Wang S, Gao S, Lin S, Fang X, Zhang H, Qiu M, Zheng K, Ji Y, Xiao B, Zhang X. Integrated analysis of bulk and single-cell RNA sequencing reveals the impact of nicotinamide and tryptophan metabolism on glioma prognosis and immunotherapy sensitivity. BMC Neurol 2024; 24:419. [PMID: 39468708 PMCID: PMC11514892 DOI: 10.1186/s12883-024-03924-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Nicotinamide and tryptophan metabolism play important roles in regulating tumor synthesis metabolism and signal transduction functions. However, their comprehensive impact on the prognosis and the tumor immune microenvironment of glioma is still unclear. The purpose of this study was to investigate the association of nicotinamide and tryptophan metabolism with prognosis and immune status of gliomas and to develop relevant models for predicting prognosis and sensitivity to immunotherapy in gliomas. METHODS Bulk and single-cell transcriptome data from TCGA, CGGA and GSE159416 were obtained for this study. Gliomas were classified based on nicotinamide and tryptophan metabolism, and PPI network associated with differentially expressed genes was established. The core genes were identified and the risk model was established by machine learning techniques, including univariate Cox regression and LASSO regression. Then the risk model was validated with data from the CGGA. Finally, the effects of genes in the risk model on the biological behavior of gliomas were verified by in vitro experiments. RESULTS The high nicotinamide and tryptophan metabolism is associated with poor prognosis and high levels of immune cell infiltration in glioma. Seven of the core genes related to nicotinamide and tryptophan metabolism were used to construct a risk model, and the model has good predictive ability for prognosis, immune microenvironment, and response to immune checkpoint therapy of glioma. We also confirmed that high expression of TGFBI can lead to an increased level of migration, invasion, and EMT of glioma cells, and the aforementioned effect of TGFBI can be reduced by FAK inhibitor PF-573,228. CONCLUSIONS Our study evaluated the effects of nicotinamide and tryptophan metabolism on the prognosis and tumor immune microenvironment of glioma, which can help predict the prognosis and sensitivity to immunotherapy of glioma.
Collapse
Affiliation(s)
- Sen Wang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Shen Gao
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Shaochong Lin
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xiaofeng Fang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Haopeng Zhang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Man Qiu
- Department of Neurosurgery, Xinyang Central Hospital, Xinyang, 464000, China
| | - Kai Zheng
- Department of Neurosurgery, Xianyang First People's Hospital, Xianyang, 712000, China
| | - Yupeng Ji
- Department of Cardiovascular Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Baijun Xiao
- Department of Neurosurgery, Pingshan People's Hospital, Shenzhen, 518118, China
| | - Xiangtong Zhang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
20
|
Panfili E, Rezzi SJ, Adamo A, Mazzoletti D, Massarotti A, Miggiano R, Fallarini S, Ambrosino S, Coletti A, Molinaro P, Milella M, Paiella S, Macchiarulo A, Ugel S, Pirali T, Pallotta MT. Identification of a Compound Inhibiting Both the Enzymatic and Nonenzymatic Functions of Indoleamine 2,3-Dioxygenase 1. ACS Pharmacol Transl Sci 2024; 7:3056-3070. [PMID: 39421661 PMCID: PMC11480892 DOI: 10.1021/acsptsci.4c00265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 10/19/2024]
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) plays a key role in tumor immune escape. Besides being a metabolic enzyme that catalyzes the first step of tryptophan catabolism, it also acts as a signal-transducing protein, whose partnering with tyrosine phosphatase Src homology 2 (SH2) domain-containing protein tyrosine phosphatase substrate (SHPs) and phosphatidylinositol-3-kinase (PI3K) regulatory subunit p85 promotes the establishment of a sustained immunosuppressive phenotype. While IDO1 inhibitors typically interfere with its enzymatic activity, we aimed to discover a more effective modulator capable of blocking not only the enzymatic but also the signaling-mediated functions of IDO1. By virtual screening, we identified the compound VS-15, which selectively binds the heme-free form of IDO1, inhibits its enzymatic activity, and reduces the IDO1-mediated signaling pathway by negatively interfering with its partnership with SHPs and PI3K regulatory subunit p85 as well as with the IDO1 anchoring to the early endosomes in tumor cells. Moreover, VS-15 counteracts the TGF-β-mediated immunosuppressive phenotype in dendritic cells and reduces the level of inhibition of T cell proliferation by suppressive monocytes isolated from patients affected by pancreatic cancer. Herein, we describe the discovery and characterization of a small molecule with an unprecedented mechanism of action, capable of inhibiting both the enzymatic and nonenzymatic activities of IDO1 by binding to its apo-form. These results pave the way for the development of next-generation IDO1 inhibitors with a unique competitive advantage over the currently available modulators, thereby opening therapeutic opportunities in cancer immunotherapy.
Collapse
Affiliation(s)
- Eleonora Panfili
- Pharmacology
Section, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy
| | - Sarah Jane Rezzi
- Department
of Pharmaceutical Sciences, University of
Piemonte Orientale, Novara 28100, Italy
| | - Annalisa Adamo
- Immunology
Section, Department of Medicine, University
and Hospital Trust of Verona, Verona 37134, Italy
| | - Daniele Mazzoletti
- Department
of Pharmaceutical Sciences, University of
Piemonte Orientale, Novara 28100, Italy
| | - Alberto Massarotti
- Department
of Pharmaceutical Sciences, University of
Piemonte Orientale, Novara 28100, Italy
| | - Riccardo Miggiano
- Department
of Pharmaceutical Sciences, University of
Piemonte Orientale, Novara 28100, Italy
| | - Silvia Fallarini
- Department
of Pharmaceutical Sciences, University of
Piemonte Orientale, Novara 28100, Italy
| | - Sara Ambrosino
- Pharmacology
Section, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy
| | - Alice Coletti
- Department
of Pharmaceutical Sciences, University of
Perugia, Perugia 06132, Italy
| | - Pasquale Molinaro
- Pharmacology
Section, Department of Neuroscience, Reproductive and Dentistry Sciences,
School of Medicine, Federico II University
of Naples, Naples 80131, Italy
| | - Michele Milella
- Department
of Engineering for Innovative Medicine and Hospital of Trust of Verona, Oncology Section, Verona 37134, Italy
| | - Salvatore Paiella
- General
and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona 37134, Italy
| | - Antonio Macchiarulo
- Department
of Pharmaceutical Sciences, University of
Perugia, Perugia 06132, Italy
| | - Stefano Ugel
- Immunology
Section, Department of Medicine, University
and Hospital Trust of Verona, Verona 37134, Italy
| | - Tracey Pirali
- Department
of Pharmaceutical Sciences, University of
Piemonte Orientale, Novara 28100, Italy
| | - Maria Teresa Pallotta
- Pharmacology
Section, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy
| |
Collapse
|
21
|
Liu H, Zhu K, Yang C. The Intersection between Tryptophan-Kynurenine Pathway Metabolites and Immune Inflammation, Hormones, and Gut Microbiota in Perinatal Depression. ACTAS ESPANOLAS DE PSIQUIATRIA 2024; 52:733-740. [PMID: 39403906 PMCID: PMC11475056 DOI: 10.62641/aep.v52i5.1748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Perinatal depression is a prevalent mental disorder among pregnant women, characterized by sleep disturbances, appetite changes, negative emotions, cognitive impairment, and suicidal or homicidal tendencies. These symptoms severely compromise personal well-being, disrupt family life, and burden society. Early detection and intervention are thus crucial. The tryptophan-kynurenine (TRP-KYN) pathway is central to the inflammatory hypothesis of depression and has gained significant attention in perinatal depression research. This pathway encompasses numerous metabolic enzymes and neuroactive metabolites that interact with other physiological systems, influencing neurotransmitter synthesis and neuronal development. Through these interactions, the TRP-KYN pathway exerts psychotropic effects. This article reviews the key metabolites and enzymes of the TRP-KYN pathway and examines its intersection with immune inflammation, hormones, and gut microbiota.
Collapse
Affiliation(s)
- Huiyan Liu
- Department of Anesthesiology, Liuyang Maternal and Child Health Hospital, 410300 Liuyang, Hunan, China
| | - Kuangyi Zhu
- Department of Neonatology, Liuyang Maternal and Child Health Hospital, 410300 Liuyang, Hunan, China
| | - Cheng Yang
- Department of Obstetrical, Liuyang Maternal and Child Health Hospital, 410300 Liuyang, Hunan, China
| |
Collapse
|
22
|
Dexheimer TS, Coussens NP, Silvers T, Jones EM, Chen L, Fang J, Morris J, Moscow JA, Doroshow JH, Teicher BA. Combination screen in multi-cell type tumor spheroids reveals interaction between aryl hydrocarbon receptor antagonists and E1 ubiquitin-activating enzyme inhibitor. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100186. [PMID: 39362362 PMCID: PMC11562894 DOI: 10.1016/j.slasd.2024.100186] [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: 04/03/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that regulates genes of drug transporters and metabolic enzymes to detoxify small molecule xenobiotics. It has a complex role in cancer biology, influencing both the progression and suppression of tumors by modulating malignant properties of tumor cells and anti-tumor immunity, depending on the specific tumor type and developmental stage. This has led to the discovery and development of selective AhR modulators, including BAY 2416964 which is currently in clinical trials. To identify small molecule anticancer agents that might be combined with AhR antagonists for cancer therapy, a high-throughput combination screen was performed using multi-cell type tumor spheroids grown from malignant cells, endothelial cells, and mesenchymal stem cells. The AhR selective antagonists BAY 2416964, GNF351, and CH-223191 were tested individually and in combination with twenty-five small molecule anticancer agents. As single agents, BAY 2416964 and CH-223191 showed minimal activity, whereas GNF351 reduced the viability of some spheroid models at concentrations greater than 1 µM. The activity of most combinations aligned well with the single agent activity of the combined agent, without apparent contributions from the AhR antagonist. All three AhR antagonists sensitized tumor spheroids to TAK-243, an E1 ubiquitin-activating enzyme inhibitor. These combinations were active in spheroids containing bladder, breast, ovary, kidney, pancreas, colon, and lung tumor cell lines. The AhR antagonists also potentiated pevonedistat, a selective inhibitor of the NEDD8-activating enzyme E1 regulatory subunit, in several tumor spheroid models. In contrast, the AhR antagonists did not enhance the cytotoxicity of the proteasome inhibitor bortezomib.
Collapse
Affiliation(s)
- Thomas S Dexheimer
- Target Validation and Screening Laboratory, Molecular Pharmacology Laboratory, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, 1050 Boyles Street, Frederick, MD 21702, USA.
| | - Nathan P Coussens
- Target Validation and Screening Laboratory, Molecular Pharmacology Laboratory, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, 1050 Boyles Street, Frederick, MD 21702, USA
| | - Thomas Silvers
- Target Validation and Screening Laboratory, Molecular Pharmacology Laboratory, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, 1050 Boyles Street, Frederick, MD 21702, USA
| | - Eric M Jones
- Target Validation and Screening Laboratory, Molecular Pharmacology Laboratory, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, 1050 Boyles Street, Frederick, MD 21702, USA
| | - Li Chen
- Molecular Characterization Laboratory, Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Jianwen Fang
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joel Morris
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jeffrey A Moscow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Beverly A Teicher
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
23
|
Peng Y, Wang L, Yang J, Wu Q, Sun X, Zhang J, Yu Y, Zhang L, Gao J, Zhou Q, Zhu H, Yin F. Integrated analyses reveal IDO1 as a prognostic biomarker coexpressed with PD-1 on tumor-associated macrophages in esophageal squamous cell carcinoma. Front Pharmacol 2024; 15:1466779. [PMID: 39351094 PMCID: PMC11439782 DOI: 10.3389/fphar.2024.1466779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/27/2024] [Indexed: 10/04/2024] Open
Abstract
Background Inhibition of indolamine-2,3-dioxygenase 1 (IDO1) has been proposed as a promising strategy for cancer immunotherapy; however, it has failed in clinical trials. Macrophages in the tumor microenvironment (TME) contribute to immune escape and serve as potential therapeutic targets. This study investigated the expression pattern of IDO1 in TME and its impact on prognosis and therapeutic response of patients with esophageal squamous cell carcinoma (ESCC). Methods RNA sequencing data from 95 patients with ESCC from The Cancer Genome Atlas (TCGA) database were used to explore the prognostic value of IDO1. Bioinformatics tools were used to estimate scores for stromal and immune cells in tumour tissues, abundance of eight immune cell types in TME, and sensitivity of chemotherapeutic drugs and immune checkpoint (IC) blockage. The results were validated using digitalized immunohistochemistry and multiplexed immunofluorescence in ESCC tissue samples obtained from our clinical center. Results TCGA and validation data suggested that high expression of IDO1 was associated with poor patient survival, and IDO1 was an independent prognostic factor. IDO1 expression positively correlated with macrophages in TME and PDCD1 within diverse IC genes. Single-cell RNA sequencing data analysis and multiplexed immunofluorescence verified the coexpression of IDO1 and PD-1 in tumor-associated macrophages (TAMs). Patients with high IDO1 expression showed increased sensitivity to various chemotherapeutic drugs, while were more likely to resist IC blockage. Conclusion This study identifies IDO1 as an independent prognostic indicator of OS in patients with ESCC, reveals a compelling connection of IDO1, PD-1, and TAMs, and explores the sensitivity of patients with high IDO1 expression to chemotherapeutic drugs and their resistance to IC blockade. These findings open new avenues for potential targets in ESCC immunotherapy.
Collapse
Affiliation(s)
- Yaojun Peng
- Department of Emergency, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
- Medical School of Chinese People's Liberation Army (PLA), Beijing, China
| | - Lingxiong Wang
- Lab of the Oncology Department, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
- Institute of Oncology, The Fifth Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Juan Yang
- Department of Cardiothoracic Surgery, Tianjin Fourth Center Hospital, Tianjin, China
| | - Qiyan Wu
- Lab of the Oncology Department, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
- Institute of Oncology, The Fifth Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Xiaoxuan Sun
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Oncology Surgery, Tianjin Cancer Hospital Airport Free Trade Zone Hospital, Tianjin, China
| | - Jinying Zhang
- Department of Basic Medicine, Medical School of Chinese People's Liberation Army (PLA), Beijing, China
| | - Yanju Yu
- Institute of Oncology, The Fifth Medical Centre, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Liping Zhang
- Department of Emergency, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jie Gao
- Department of Oncology, The Second Medical Center and National Clinical Research Center of Geriatric Disease, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Qing Zhou
- Department of Gastroenterology, The Second Medical Center and National Clinical Research Center of Geriatric Disease, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Haiyan Zhu
- Department of Emergency, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Fan Yin
- Department of Oncology, The Second Medical Center and National Clinical Research Center of Geriatric Disease, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
24
|
Varzieva VG, Mesonzhnik NV, Ilgisonis IS, Belenkov YN, Kozhevnikova MV, Appolonova SA. Metabolomic biomarkers of multiple myeloma: A systematic review. Biochim Biophys Acta Rev Cancer 2024; 1879:189151. [PMID: 38986721 DOI: 10.1016/j.bbcan.2024.189151] [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: 11/21/2023] [Accepted: 07/03/2024] [Indexed: 07/12/2024]
Abstract
Multiple myeloma (MM) is an incurable malignancy of clonal plasma cells. Various diagnostic methods are used in parallel to accurately determine stage and severity of the disease. Identifying a biomarker or a panel of biomarkers could enhance the quality of medical care that patients receive by adopting a more personalized approach. Metabolomics utilizes high-throughput analytical platforms to examine the levels and quantities of biochemical compounds in biosamples. The aim of this review was to conduct a systematic literature search for potential metabolic biomarkers that may aid in the diagnosis and prognosis of MM. The review was conducted in accordance with PRISMA recommendations and was registered in PROSPERO. The systematic search was performed in PubMed, CINAHL, SciFinder, Scopus, The Cochrane Library and Google Scholar. Studies were limited to those involving people with clinically diagnosed MM and healthy controls as comparators. Articles had to be published in English and had no restrictions on publication date or sample type. The quality of articles was assessed according to QUADOMICS criteria. A total of 709 articles were collected during the literature search. Of these, 436 were excluded based on their abstract, with 26 more removed after a thorough review of the full text. Finally, 16 articles were deemed relevant and were subjected to further analysis of their data. A number of promising candidate biomarkers was discovered. Follow-up studies with large sample sizes are needed to determine their suitability for clinical applications.
Collapse
Affiliation(s)
- Valeria G Varzieva
- Department of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), Vernadskogo pr., 96, 119571 Moscow, Russia; Centre of Biopharmaceutical Analysis and Metabolomics, Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University (Sechenov University), Nakhimovsky pr., 45, 117418 Moscow, Russia.
| | - Natalia V Mesonzhnik
- Centre of Biopharmaceutical Analysis and Metabolomics, Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University (Sechenov University), Nakhimovsky pr., 45, 117418 Moscow, Russia.
| | - Irina S Ilgisonis
- Hospital Therapy No. 1 Department, Sechenov First Moscow State Medical University (Sechenov University), Bol'shaya Pirogovskaya st. 6/1, 119435 Moscow, Russia
| | - Yuri N Belenkov
- Hospital Therapy No. 1 Department, Sechenov First Moscow State Medical University (Sechenov University), Bol'shaya Pirogovskaya st. 6/1, 119435 Moscow, Russia
| | - Maria V Kozhevnikova
- Hospital Therapy No. 1 Department, Sechenov First Moscow State Medical University (Sechenov University), Bol'shaya Pirogovskaya st. 6/1, 119435 Moscow, Russia
| | - Svetlana A Appolonova
- Department of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), Vernadskogo pr., 96, 119571 Moscow, Russia; Centre of Biopharmaceutical Analysis and Metabolomics, Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University (Sechenov University), Nakhimovsky pr., 45, 117418 Moscow, Russia
| |
Collapse
|
25
|
Dong Y, Luo J, Pei M, Liu S, Gao Y, Zhou H, Nueraihemaiti Y, Zhan X, Xie T, Yao X, Guan X, Xu Y. Biomimetic Hydrogel-Mediated Mechano-Immunometabolic Therapy for Inhibition of ccRCC Recurrence After Surgery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308734. [PMID: 38884220 PMCID: PMC11321661 DOI: 10.1002/advs.202308734] [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: 11/14/2023] [Revised: 05/18/2024] [Indexed: 06/18/2024]
Abstract
The unique physical tumor microenvironment (TME) and aberrant immune metabolic status are two obstacles that must be overcome in cancer immunotherapy to improve clinical outcomes. Here, an in situ mechano-immunometabolic therapy involving the injection of a biomimetic hydrogel is presented with sequential release of the anti-fibrotic agent pirfenidone, which softens the stiff extracellular matrix, and small interfering RNA IDO1, which disrupts kynurenine-mediated immunosuppressive metabolic pathways, together with the multi-kinase inhibitor sorafenib, which induces immunogenic cell death. This combination synergistically augmented tumor immunogenicity and induced anti-tumor immunity. In mouse models of clear cell renal cell carcinoma, a single-dose peritumoral injection of a biomimetic hydrogel facilitated the perioperative TME toward a more immunostimulatory landscape, which prevented tumor relapse post-surgery and prolonged mouse survival. Additionally, the systemic anti-tumor surveillance effect induced by local treatment decreased lung metastasis by inhibiting epithelial-mesenchymal transition conversion. The versatile localized mechano-immunometabolic therapy can serve as a universal strategy for conferring efficient tumoricidal immunity in "cold" tumor postoperative interventions.
Collapse
Affiliation(s)
- Yunze Dong
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Jun Luo
- Department of UrologyShanghai Fourth People's HospitalSchool of MedicineTongji UniversityShanghai200434P. R. China
| | - Mingliang Pei
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| | - Shuai Liu
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Yuchen Gao
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Hongmin Zhou
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Yimingniyizi Nueraihemaiti
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Xiangcheng Zhan
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Tiancheng Xie
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Xudong Yao
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Xin Guan
- Department of UltrasoundInstitute of Ultrasound in Medicine and EngineeringZhongshan HospitalFudan UniversityShanghai200032P. R. China
| | - Yunfei Xu
- Department of UrologyShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| |
Collapse
|
26
|
Mor A, Tankiewicz-Kwedlo A, Ciwun M, Lewkowicz J, Pawlak D. Kynurenines as a Novel Target for the Treatment of Inflammatory Disorders. Cells 2024; 13:1259. [PMID: 39120289 PMCID: PMC11311768 DOI: 10.3390/cells13151259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/09/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
This review discusses the potential of targeting the kynurenine pathway (KP) in the treatment of inflammatory diseases. The KP, responsible for the catabolism of the amino acid tryptophan (TRP), produces metabolites that regulate various physiological processes, including inflammation, cell cycle, and neurotransmission. These metabolites, although necessary to maintain immune balance, may accumulate excessively during inflammation, leading to systemic disorders. Key KP enzymes such as indoleamine 2,3-dioxygenase 1 (IDO1), indoleamine 2,3-dioxygenase 2 (IDO2), tryptophan 2,3-dioxygenase (TDO), and kynurenine 3-monooxygenase (KMO) have been considered promising therapeutic targets. It was highlighted that both inhibition and activation of these enzymes may be beneficial, depending on the specific inflammatory disorder. Several inflammatory conditions, including autoimmune diseases, for which modulation of KP activity holds therapeutic promise, have been described in detail. Preclinical studies suggest that this modulation may be an effective treatment strategy for diseases for which treatment options are currently limited. Taken together, this review highlights the importance of further research on the clinical application of KP enzyme modulation in the development of new therapeutic strategies for inflammatory diseases.
Collapse
Affiliation(s)
- Adrian Mor
- Department of Pharmacodynamics, Medical University of Bialystok, A. Mickiewicza 2C, 15-222 Bialystok, Poland; (A.M.); (M.C.); (D.P.)
| | - Anna Tankiewicz-Kwedlo
- Department of Pharmacodynamics, Medical University of Bialystok, A. Mickiewicza 2C, 15-222 Bialystok, Poland; (A.M.); (M.C.); (D.P.)
| | - Marianna Ciwun
- Department of Pharmacodynamics, Medical University of Bialystok, A. Mickiewicza 2C, 15-222 Bialystok, Poland; (A.M.); (M.C.); (D.P.)
| | - Janina Lewkowicz
- Department of Internal Medicine and Metabolic Diseases, Medical University of Bialystok, M. Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland;
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, A. Mickiewicza 2C, 15-222 Bialystok, Poland; (A.M.); (M.C.); (D.P.)
| |
Collapse
|
27
|
Lim BY, Guo Z, Lim JQ, Ko TK, Lee ECY, Kannan B, Lee JY, Lim AH, Li Z, Ng CCY, Busmanis I, Chan JY. Whole genome sequencing of HER2-positive metastatic extramammary Paget's disease: a case report. Orphanet J Rare Dis 2024; 19:223. [PMID: 38831459 PMCID: PMC11149212 DOI: 10.1186/s13023-024-03169-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/30/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Extramammary Paget's disease (EMPD) is a rare cancer that occurs within the epithelium of the skin, arising predominantly in areas with high apocrine gland concentration such as the vulva, scrotum, penis and perianal regions. Here, we aim to integrate clinicopathological data with genomic analysis of aggressive, rapidly-progressing de novo metastatic EMPD responding to HER2-directed treatment in combination with other agents, to attain a more comprehensive understanding of the disease landscape. METHODS Immunohistochemical staining on the scrotal wall tumor and bone marrow metastasis demonstrated HER2 overexpression. Whole genome sequencing of the tumor and matched blood was performed. RESULTS Notable copy number gains (log2FC > 0.9) on chromosomes 7 and 8 were detected (n = 81), with 92.6% of these unique genes specifically located on chromosome 8. Prominent cancer-associated genes include ZNF703, HOOK3, DDHD2, LSM1, NSD3, ADAM9, BRF2, KAT6A and FGFR1. Interestingly, ERBB2 gene did not exhibit high copy number gain (log2FC = 0.4) although 90% of tumor cells stained HER2-positive. Enrichment in pathways associated with transforming growth factor-beta (TGFβ) (FDR = 0.0376, Enrichment Ratio = 8.12) and fibroblast growth factor receptor (FGFR1) signaling (FDR = 0.0082, Enrichment Ratio = 2.3) was detected. Amplicon structure analysis revealed that this was a simple-linear amplification event. CONCLUSION Whole genome sequencing revealed the underlying copy number variation landscape in HER2-positive metastatic EMPD. The presence of alternative signalling pathways and genetic variants suggests potential interactions with HER2 signalling, which possibly contributed to the HER2 overexpression and observed response to HER2-directed therapy combined with other agents in a comprehensive treatment regimen.
Collapse
Affiliation(s)
- Boon Yee Lim
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Zexi Guo
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Jing Quan Lim
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Tun Kiat Ko
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | | | - Bavani Kannan
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Jing Yi Lee
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Abner Herbert Lim
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Zhimei Li
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | | | - Inny Busmanis
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Jason Yongsheng Chan
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore.
- Duke-NUS Medical School, Singapore, Singapore.
- Divison of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore.
| |
Collapse
|
28
|
Li B, Zu M, Jiang A, Cao Y, Wu J, Shahbazi MA, Shi X, Reis RL, Kundu SC, Xiao B. Magnetic natural lipid nanoparticles for oral treatment of colorectal cancer through potentiated antitumor immunity and microbiota metabolite regulation. Biomaterials 2024; 307:122530. [PMID: 38493672 DOI: 10.1016/j.biomaterials.2024.122530] [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: 11/15/2023] [Revised: 02/10/2024] [Accepted: 03/08/2024] [Indexed: 03/19/2024]
Abstract
The therapeutic efficacy of oral nanotherapeutics against colorectal cancer (CRC) is restricted by inadequate drug accumulation, immunosuppressive microenvironment, and intestinal microbiota imbalance. To overcome these challenges, we elaborately constructed 6-gingerol (Gin)-loaded magnetic mesoporous silicon nanoparticles and functionalized their surface with mulberry leaf-extracted lipids (MLLs) and Pluronic F127 (P127). In vitro experiments revealed that P127 functionalization and alternating magnetic fields (AMFs) promoted internalization of the obtained P127-MLL@Gins by colorectal tumor cells and induced their apoptosis/ferroptosis through Gin/ferrous ion-induced oxidative stress and magneto-thermal effect. After oral administration, P127-MLL@Gins safely passed to the colorectal lumen, infiltrated the mucus barrier, and penetrated into the deep tumors under the influence of AMFs. Subsequently, the P127-MLL@Gin (+ AMF) treatment activated antitumor immunity and suppressed tumor growth. We also found that this therapeutic modality significantly increased the abundance of beneficial bacteria (e.g., Bacillus and unclassified-c-Bacilli), reduced the proportions of harmful bacteria (e.g., Bacteroides and Alloprevotella), and increased lipid oxidation metabolites. Strikingly, checkpoint blockers synergistically improved the therapeutic outcomes of P127-MLL@Gins (+ AMF) against orthotopic and distant colorectal tumors and significantly prolonged mouse life spans. Overall, this oral therapeutic platform is a promising modality for synergistic treatment of CRC.
Collapse
Affiliation(s)
- Baoyi Li
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Menghang Zu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Aodi Jiang
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Yingui Cao
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Jiaxue Wu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands; W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
| | - Xiaoxiao Shi
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China.
| | - Rui L Reis
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco, Guimarães, 4805-017, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, 4800-058, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco, Guimarães, 4805-017, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, 4800-058, Portugal
| | - Bo Xiao
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
29
|
Green GBH, Cox-Holmes AN, Backan O, Valbak O, Potier ACE, Chen D, Morrow CD, Willey CD, McFarland BC. Exploring Gut Microbiota Alterations with Trimethoprim-Sulfamethoxazole and Dexamethasone in a Humanized Microbiome Mouse Model. Microorganisms 2024; 12:1015. [PMID: 38792844 PMCID: PMC11124107 DOI: 10.3390/microorganisms12051015] [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: 04/16/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Along with the standard therapies for glioblastoma, patients are commonly prescribed trimethoprim-sulfamethoxazole (TMP-SMX) and dexamethasone for preventing infections and reducing cerebral edema, respectively. Because the gut microbiota impacts the efficacy of cancer therapies, it is important to understand how these medications impact the gut microbiota of patients. Using mice that have been colonized with human microbiota, this study sought to examine how TMP-SMX and dexamethasone affect the gut microbiome. Two lines of humanized microbiota (HuM) Rag1-/- mice, HuM1Rag and HuM2Rag, were treated with either TMP-SMX or dexamethasone via oral gavage once a day for a week. Fecal samples were collected pre-treatment (pre-txt), one week after treatment initiation (1 wk post txt), and three weeks post-treatment (3 wk post txt), and bacterial DNA was analyzed using 16S rRNA-sequencing. The HuM1Rag mice treated with TMP-SMX had significant shifts in alpha diversity, beta diversity, and functional pathways at all time points, whereas in the HuM2Rag mice, it resulted in minimal changes in the microbiome. Likewise, dexamethasone treatment resulted in significant changes in the microbiome of the HuM1Rag mice, whereas the microbiome of the HuM2Rag mice was mostly unaffected. The results of our study show that routine medications used during glioblastoma treatment can perturb gut microbiota, with some microbiome compositions being more sensitive than others, and these treatments could potentially affect the overall efficacy of standard-of-care therapy.
Collapse
Affiliation(s)
- George B. H. Green
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
| | - Alexis N. Cox-Holmes
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
| | - Olivia Backan
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
- Undergraduate Cancer Biology Program, Birmingham, AL 35294, USA
| | - Olivia Valbak
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
- Undergraduate Cancer Biology Program, Birmingham, AL 35294, USA
| | - Anna Claire E. Potier
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
- Undergraduate Cancer Biology Program, Birmingham, AL 35294, USA
| | | | - Casey D. Morrow
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
| | - Christopher D. Willey
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Braden C. McFarland
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
| |
Collapse
|
30
|
Ma S, Ming Y, Wu J, Cui G. Cellular metabolism regulates the differentiation and function of T-cell subsets. Cell Mol Immunol 2024; 21:419-435. [PMID: 38565887 PMCID: PMC11061161 DOI: 10.1038/s41423-024-01148-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 02/23/2024] [Indexed: 04/04/2024] Open
Abstract
T cells are an important component of adaptive immunity and protect the host from infectious diseases and cancers. However, uncontrolled T cell immunity may cause autoimmune disorders. In both situations, antigen-specific T cells undergo clonal expansion upon the engagement and activation of antigens. Cellular metabolism is reprogrammed to meet the increase in bioenergetic and biosynthetic demands associated with effector T cell expansion. Metabolites not only serve as building blocks or energy sources to fuel cell growth and expansion but also regulate a broad spectrum of cellular signals that instruct the differentiation of multiple T cell subsets. The realm of immunometabolism research is undergoing swift advancements. Encapsulating all the recent progress within this concise review in not possible. Instead, our objective is to provide a succinct introduction to this swiftly progressing research, concentrating on the metabolic intricacies of three pivotal nutrient classes-lipids, glucose, and amino acids-in T cells. We shed light on recent investigations elucidating the roles of these three groups of metabolites in mediating the metabolic and immune functions of T cells. Moreover, we delve into the prospect of "editing" metabolic pathways within T cells using pharmacological or genetic approaches, with the aim of synergizing this approach with existing immunotherapies and enhancing the efficacy of antitumor and antiinfection immune responses.
Collapse
Affiliation(s)
- Sicong Ma
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China
| | - Yanan Ming
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China
| | - Jingxia Wu
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China.
| | - Guoliang Cui
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China.
| |
Collapse
|
31
|
Gkountana GV, Wang L, Giacomini M, Hyytiäinen A, Juurikka K, Salo T, Al-Samadi A. IDO1 correlates with the immune landscape of head and neck squamous cell carcinoma: a study based on bioinformatics analyses. FRONTIERS IN ORAL HEALTH 2024; 5:1335648. [PMID: 38736462 PMCID: PMC11082366 DOI: 10.3389/froh.2024.1335648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/12/2024] [Indexed: 05/14/2024] Open
Abstract
Background Head and neck squamous cell carcinoma (HNSCCs) is a common cancer type with a high mortality rate and poor prognosis. Recent studies have focused on the role of immune checkpoints in HNSCC progression and in their potential use as prognostic markers and immunotherapeutic candidates. Some immune checkpoints, such as PD-1 and PD-L1, have been studied thoroughly in HNSCC. Other molecules, such as indoleamine 2,3-dioxygenase 1 (IDO1), have been investigated minimally. Methods IDO1 expression, prognostic potential, and association with the immune profile of HNSCC were explored using online databases, including GEPIA, UALCAN, TIMER2.0, cBioPortal, and LinkedOmics, which utilize TCGA datasets and are freely available for use. For validation purposes, seven pairs of primary and metastatic HNSCC were immunostained for IDO1. Results Our analysis revealed significantly higher expression of IDO1 in HNSCC, especially in HPV+ SCCs compared with healthy control tissue. However, IDO1 expression showed weak to no prognostic potential for overall and disease-free survival in HNSCC. IDO1 expression in HNSCC was positively correlated with several immune-related molecules, including most of the immune checkpoints. Additionally, GO enrichment analysis revealed that several immune-related pathways are positively correlated with IDO1 expression in HNSCC, such as response to type I interferon and lymphocyte-mediated immunity pathways. Finally, IDO1 expression positively correlated with infiltration of most of the immune cells in HNSCC, such as CD4+ T cells, CD8+ T cells, M1 and M2 macrophages, dendritic cells, and B cells. Conclusion IDO1 expression is closely correlated with the immune profile of the HNSCC. This observation should be explored further to elucidate the potential of targeting IDO1 as a novel immunotherapeutic approach for HNSCC.
Collapse
Affiliation(s)
- Georgia Vasiliki Gkountana
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program (TRIMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Lezhou Wang
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Martina Giacomini
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Aini Hyytiäinen
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program (TRIMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Krista Juurikka
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program (TRIMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Ahmed Al-Samadi
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program (TRIMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Institute of Dentistry, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
32
|
Jin E, Yin Z, Zheng X, Yan C, Xu K, Eunice FY, Gao Y. Potential of Targeting TDO2 as the Lung Adenocarcinoma Treatment. J Proteome Res 2024; 23:1341-1350. [PMID: 38421152 DOI: 10.1021/acs.jproteome.3c00746] [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: 03/02/2024]
Abstract
Tryptophan catabolism plays an important role in the metabolic reconnection in cancer cells to support special demands of tumor initiation and progression. The catabolic product of the tryptophan pathway, kynurenine, has the capability of suppressing the immune reactions of tumor cells. In this study, we conducted internal and external cohort studies to reveal the importance of tryptophan 2,3-dioxygenase (TDO) for lung adenocarcinoma (LUAD). Our study further demonstrated that the TDO2 expression was associated with the proliferation, survival, and invasion of LUAD cells, and targeting TDO2 for LUAD tumors could be a potential therapeutic strategy.
Collapse
Affiliation(s)
- Er Jin
- Department of Respiratory Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310002 Zhejiang Province, China
| | - Zhidong Yin
- Department of Pathology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009 Zhejiang Province, China
| | - Xiuxiu Zheng
- Department of Respiratory Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310002 Zhejiang Province, China
| | - Chenhong Yan
- Department of Respiratory Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310002 Zhejiang Province, China
| | - Kai Xu
- Department of Respiratory Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310002 Zhejiang Province, China
| | - Fouejio Yemele Eunice
- Department of Respiratory Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310002 Zhejiang Province, China
| | - Yue Gao
- Department of Geriatric, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006 Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Major Chronic Disease in the Elderly, Hangzhou 310006 Zhejiang Province, China
| |
Collapse
|
33
|
Yu HJ, Jang E, Woo A, Han IW, Jeon HG, Linh VTN, Park SG, Jung HS, Lee MY. Cancer screening through surface-enhanced Raman spectroscopy fingerprinting analysis of urinary metabolites using surface-carbonized silver nanowires on a filter membrane. Anal Chim Acta 2024; 1292:342233. [PMID: 38309850 DOI: 10.1016/j.aca.2024.342233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/05/2023] [Accepted: 01/09/2024] [Indexed: 02/05/2024]
Abstract
BACKGROUND Label-free surface-enhanced Raman spectroscopy (SERS)-based metabolic profiling has great potential for early cancer diagnosis, but further advancements in analytical methods and clinical evidence studies are required for clinical applications. To improve the cancer diagnostic accuracy of label-free SERS spectral analysis of complex biological fluids, it is necessary to obtain specifically enhanced SERS signals of cancer-related metabolites present at low concentrations. RESULTS This study presents a novel 3D SERS sensor, comprising a surface-carbonized silver nanowire (AgNW)-stacked filter membrane, alongside an optimized urine/methanol/chloroform extraction technique, which specifically changes the molecular adsorption and orientation of aromatic metabolites onto SERS substrates. By analyzing the pretreated urine samples on the surface-carbonized AgNW 3D SERS sensor, distinct and highly enhanced SERS peaks derived from semi-polar aromatic metabolites were observed for pancreatic cancer and prostate cancer samples compared with normal controls. Urine metabolite analysis using SERS fingerprinting successfully differentiated pancreatic cancer and prostate cancer groups from normal control group: normal control (n = 56), pancreatic cancer (n = 40), and prostate cancer (n = 39). SIGNIFICANCE AND NOVELTY We confirmed the clinical feasibility of performing fingerprint analysis of urinary metabolites based on the surface-carbonized AgNW 3D SERS sensor and methanol/chloroform extraction for noninvasive cancer screening. This technology holds potential for large-scale screening owing to its high accuracy, and cost effective, simple and rapid detection method.
Collapse
Affiliation(s)
- Ho-Jae Yu
- Medical Device Research Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Eunji Jang
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea
| | - Ayoung Woo
- Medical Device Research Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - In Woong Han
- Division of Hepato Biliary Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| | - Hwang Gyun Jeon
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| | - Vo Thi Nhat Linh
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea
| | - Sung-Gyu Park
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea
| | - Ho Sang Jung
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea.
| | - Min-Young Lee
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea.
| |
Collapse
|
34
|
Vilbois S, Xu Y, Ho PC. Metabolic interplay: tumor macrophages and regulatory T cells. Trends Cancer 2024; 10:242-255. [PMID: 38135571 DOI: 10.1016/j.trecan.2023.11.007] [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: 09/06/2023] [Revised: 11/19/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023]
Abstract
The tumor microenvironment (TME) contains a complex cellular ecosystem where cancer, stromal, vascular, and immune cells interact. Macrophages and regulatory T cells (Tregs) are critical not only for maintaining immunological homeostasis and tumor growth but also for monitoring the functional states of other immune cells. Emerging evidence reveals that metabolic changes in macrophages and Tregs significantly influence their pro-/antitumor functions through the regulation of signaling cascades and epigenetic reprogramming. Hence, they are increasingly recognized as therapeutic targets in cancer immunotherapy. Specific metabolites in the TME may also affect their pro-/antitumor functions by intervening with the metabolic machinery. We discuss how metabolites influence the immunosuppressive phenotypes of tumor-associated macrophages (TAMs) and Tregs. We then describe how TAMs and Tregs, independently or collaboratively, utilize metabolic mechanisms to suppress the activity of CD8+ T cells. Finally, we highlight promising metabolic interventions that can improve the outcome of current cancer therapies.
Collapse
Affiliation(s)
- Stefania Vilbois
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Yingxi Xu
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland.
| | - Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland.
| |
Collapse
|
35
|
McPhedran SJ, Carleton GA, Lum JJ. Metabolic engineering for optimized CAR-T cell therapy. Nat Metab 2024; 6:396-408. [PMID: 38388705 DOI: 10.1038/s42255-024-00976-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 01/04/2024] [Indexed: 02/24/2024]
Abstract
The broad effectiveness of T cell-based therapy for treating solid tumour cancers remains limited. This is partly due to the growing appreciation that immune cells must inhabit and traverse a metabolically demanding tumour environment. Accordingly, recent efforts have centred on using genome-editing technologies to augment T cell-mediated cytotoxicity by manipulating specific metabolic genes. However, solid tumours exhibit numerous characteristics restricting immune cell-mediated cytotoxicity, implying a need for metabolic engineering at the pathway level rather than single gene targets. This emerging concept has yet to be put into clinical practice as many questions concerning the complex interplay between metabolic networks and T cell function remain unsolved. This Perspective will highlight key foundational studies that examine the relevant metabolic pathways required for effective T cell cytotoxicity and persistence in the human tumour microenvironment, feasible strategies for metabolic engineering to increase the efficiency of chimeric antigen receptor T cell-based approaches, and the challenges lying ahead for clinical implementation.
Collapse
Affiliation(s)
- Sarah J McPhedran
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, British Columbia, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Gillian A Carleton
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, British Columbia, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Julian J Lum
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, British Columbia, Canada.
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada.
| |
Collapse
|
36
|
Sugiyama S, Yumimoto K, Fujinuma S, Nakayama KI. Identification of effective CCR2 inhibitors for cancer therapy using humanized mice. J Biochem 2024; 175:195-204. [PMID: 37947138 DOI: 10.1093/jb/mvad086] [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: 10/22/2023] [Revised: 10/28/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023] Open
Abstract
C-C chemokine receptor type 2 (CCR2) is the receptor for C-C motif chemokine 2 (CCL2) and is associated with various inflammatory diseases and cancer metastasis. Although many inhibitors for CCR2 have been developed, it remains unresolved which inhibitors are the most effective in the clinical setting. In the present study, we compared 10 existing human CCR2 antagonists in a calcium influx assay using human monocytic leukemia cells. Among them, MK0812 was found to be the most potent inhibitor of human CCR2. Furthermore, we generated a human CCR2B knock-in mouse model to test the efficacy of MK0812 against a lung metastasis model of breast cancer. Oral administration of MK0812 to humanized mice did indeed reduce the number of monocytic myeloid-derived suppressor cells and the rate of lung metastasis. These results suggest that MK0812 is the most promising candidate among the commercially available CCR2 inhibitors. We propose that combining these two screening methods may provide an excellent experimental method for identifying effective drugs that inhibit human CCR2.
Collapse
Affiliation(s)
- Shigeaki Sugiyama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Kanae Yumimoto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Shun Fujinuma
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
- Anticancer Strategies Laboratory, TMDU Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| |
Collapse
|
37
|
Obrador E, Moreno-Murciano P, Oriol-Caballo M, López-Blanch R, Pineda B, Gutiérrez-Arroyo JL, Loras A, Gonzalez-Bonet LG, Martinez-Cadenas C, Estrela JM, Marqués-Torrejón MÁ. Glioblastoma Therapy: Past, Present and Future. Int J Mol Sci 2024; 25:2529. [PMID: 38473776 PMCID: PMC10931797 DOI: 10.3390/ijms25052529] [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: 12/23/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Glioblastoma (GB) stands out as the most prevalent and lethal form of brain cancer. Although great efforts have been made by clinicians and researchers, no significant improvement in survival has been achieved since the Stupp protocol became the standard of care (SOC) in 2005. Despite multimodality treatments, recurrence is almost universal with survival rates under 2 years after diagnosis. Here, we discuss the recent progress in our understanding of GB pathophysiology, in particular, the importance of glioma stem cells (GSCs), the tumor microenvironment conditions, and epigenetic mechanisms involved in GB growth, aggressiveness and recurrence. The discussion on therapeutic strategies first covers the SOC treatment and targeted therapies that have been shown to interfere with different signaling pathways (pRB/CDK4/RB1/P16ink4, TP53/MDM2/P14arf, PI3k/Akt-PTEN, RAS/RAF/MEK, PARP) involved in GB tumorigenesis, pathophysiology, and treatment resistance acquisition. Below, we analyze several immunotherapeutic approaches (i.e., checkpoint inhibitors, vaccines, CAR-modified NK or T cells, oncolytic virotherapy) that have been used in an attempt to enhance the immune response against GB, and thereby avoid recidivism or increase survival of GB patients. Finally, we present treatment attempts made using nanotherapies (nanometric structures having active anti-GB agents such as antibodies, chemotherapeutic/anti-angiogenic drugs or sensitizers, radionuclides, and molecules that target GB cellular receptors or open the blood-brain barrier) and non-ionizing energies (laser interstitial thermal therapy, high/low intensity focused ultrasounds, photodynamic/sonodynamic therapies and electroporation). The aim of this review is to discuss the advances and limitations of the current therapies and to present novel approaches that are under development or following clinical trials.
Collapse
Affiliation(s)
- Elena Obrador
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Paz Moreno-Murciano
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
| | - María Oriol-Caballo
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Rafael López-Blanch
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Begoña Pineda
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Julia Lara Gutiérrez-Arroyo
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Alba Loras
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Luis G. Gonzalez-Bonet
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon, Spain;
| | - Conrado Martinez-Cadenas
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - José M. Estrela
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain
| | | |
Collapse
|
38
|
Li X, Zhang HS. Amino acid metabolism, redox balance and epigenetic regulation in cancer. FEBS J 2024; 291:412-429. [PMID: 37129434 DOI: 10.1111/febs.16803] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/11/2023] [Accepted: 04/21/2023] [Indexed: 05/03/2023]
Abstract
Amino acids act as versatile nutrients driving cell growth and survival, especially in cancer cells. Amino acid metabolism comprises numerous metabolic networks and is closely linked with intracellular redox balance and epigenetic regulation. Reprogrammed amino acid metabolism has been recognized as a ubiquitous feature in tumour cells. This review outlines the metabolism of several primary amino acids in cancer cells and highlights the pivotal role of amino acid metabolism in sustaining redox homeostasis and regulating epigenetic modification in response to oxidative and genetic stress in cancer cells.
Collapse
Affiliation(s)
- Xiang Li
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Hong-Sheng Zhang
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| |
Collapse
|
39
|
Griffith BD, Frankel TL. The Aryl Hydrocarbon Receptor: Impact on the Tumor Immune Microenvironment and Modulation as a Potential Therapy. Cancers (Basel) 2024; 16:472. [PMID: 38339226 PMCID: PMC10854841 DOI: 10.3390/cancers16030472] [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: 12/11/2023] [Revised: 01/18/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a ubiquitous nuclear receptor with a broad range of functions, both in tumor cells and immune cells within the tumor microenvironment (TME). Activation of AhR has been shown to have a carcinogenic effect in a variety of organs, through induction of cellular proliferation and migration, promotion of epithelial-to-mesenchymal transition, and inhibition of apoptosis, among other functions. However, the impact on immune cell function is more complicated, with both pro- and anti-tumorigenic roles identified. Although targeting AhR in cancer has shown significant promise in pre-clinical studies, there has been limited efficacy in phase III clinical trials to date. With the contrasting roles of AhR activation on immune cell polarization, understanding the impact of AhR activation on the tumor immune microenvironment is necessary to guide therapies targeting the AhR. This review article summarizes the state of knowledge of AhR activation on the TME, limitations of current findings, and the potential for modulation of the AhR as a cancer therapy.
Collapse
Affiliation(s)
- Brian D. Griffith
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Timothy L. Frankel
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA;
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
40
|
Tyagi R, Yadav K, Srivastava N, Sagar R. Applications of Pyrrole and Pyridine-based Heterocycles in Cancer Diagnosis and Treatment. Curr Pharm Des 2024; 30:255-277. [PMID: 38711394 DOI: 10.2174/0113816128280082231205071504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/23/2023] [Indexed: 05/08/2024]
Abstract
BACKGROUND The escalation of cancer worldwide is one of the major causes of economy burden and loss of human resources. According to the American Cancer Society, there will be 1,958,310 new cancer cases and 609,820 projected cancer deaths in 2023 in the United States. It is projected that by 2040, the burden of global cancer is expected to rise to 29.5 million per year, causing a death toll of 16.4 million. The hemostasis regulation by cellular protein synthesis and their targeted degradation is required for normal cell growth. The imbalance in hemostasis causes unbridled growth in cells and results in cancer. The DNA of cells needs to be targeted by chemotherapeutic agents for cancer treatment, but at the same time, their efficacy and toxicity also need to be considered for successful treatment. OBJECTIVE The objective of this study is to review the published work on pyrrole and pyridine, which have been prominent in the diagnosis and possess anticancer activity, to obtain some novel lead molecules of improved cancer therapeutic. METHODS A literature search was carried out using different search engines, like Sci-finder, Elsevier, ScienceDirect, RSC etc., for small molecules based on pyrrole and pyridine helpful in diagnosis and inducing apoptosis in cancer cells. The research findings on the application of these compounds from 2018-2023 were reviewed on a variety of cell lines, such as breast cancer, liver cancer, epithelial cancer, etc. Results: In this review, the published small molecules, pyrrole and pyridine and their derivatives, which have roles in the diagnosis and treatment of cancers, were discussed to provide some insight into the structural features responsible for diagnosis and treatment. The analogues with the chromeno-furo-pyridine skeleton showed the highest anticancer activity against breast cancer. The compound 5-amino-N-(1-(pyridin-4- yl)ethylidene)-1H-pyrazole-4-carbohydrazides was highly potent against HEPG2 cancer cell. Redaporfin is used for the treatment of cholangiocarcinoma, biliary tract cancer, cisplatin-resistant head and neck squamous cell carcinoma, and pigmentation melanoma, and it is in clinical trials for phase II. These structural features present a high potential for designing novel anticancer agents for diagnosis and drug development. CONCLUSION Therefore, the N- and C-substituted pyrrole and pyridine-based novel privileged small Nheterocyclic scaffolds are potential molecules used in the diagnosis and treatment of cancer. This review discusses the reports on the synthesis of such molecules during 2018-2023. The review mainly discusses various diagnostic techniques for cancer, which employ pyrrole and pyridine heterocyclic scaffolds. Furthermore, the anticancer activity of N- and C-substituted pyrrole and pyridine-based scaffolds has been described, which works against different cancer cell lines, such as MCF-7, A549, A2780, HepG2, MDA-MB-231, K562, HT- 29, Caco-2 cells, Hela, Huh-7, WSU-DLCL2, HCT-116, HBL-100, H23, HCC827, SKOV3, etc. This review will help the researchers to obtain a critical insight into the structural aspects of pyrrole and pyridine-based scaffolds useful in cancer diagnosis as well as treatment and design pathways to develop novel drugs in the future.
Collapse
Affiliation(s)
- Rajdeep Tyagi
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110007, India
| | - Kanchan Yadav
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110007, India
| | - Nitin Srivastava
- Department of Chemistry, Amity University Lucknow Campus, Lucknow, Uttar Pradesh 226028, India
| | - Ram Sagar
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110007, India
| |
Collapse
|
41
|
Peng X, Zheng J, Liu T, Zhou Z, Song C, Geng Y, Wang Z, Huang Y. Tumor Microenvironment Heterogeneity, Potential Therapeutic Avenues, and Emerging Therapies. Curr Cancer Drug Targets 2024; 24:288-307. [PMID: 37537777 DOI: 10.2174/1568009623666230712095021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/30/2023] [Accepted: 06/08/2023] [Indexed: 08/05/2023]
Abstract
OBJECTIVE This review describes the comprehensive portrait of tumor microenvironment (TME). Additionally, we provided a panoramic perspective on the transformation and functions of the diverse constituents in TME, and the underlying mechanisms of drug resistance, beginning with the immune cells and metabolic dynamics within TME. Lastly, we summarized the most auspicious potential therapeutic strategies. RESULTS TME is a unique realm crafted by malignant cells to withstand the onslaught of endogenous and exogenous therapies. Recent research has revealed many small-molecule immunotherapies exhibiting auspicious outcomes in preclinical investigations. Furthermore, some pro-immune mechanisms have emerged as a potential avenue. With the advent of nanosystems and precision targeting, targeted therapy has now transcended the "comfort zone" erected by cancer cells within TME. CONCLUSION The ceaseless metamorphosis of TME fosters the intransigent resilience and proliferation of tumors. However, existing therapies have yet to surmount the formidable obstacles posed by TME. Therefore, scientists should investigate potential avenues for therapeutic intervention and design innovative pharmacological and clinical technologies.
Collapse
Affiliation(s)
- Xintong Peng
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Jingfan Zheng
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Tianzi Liu
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Ziwen Zhou
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Chen Song
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Yan Geng
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Zichuan Wang
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Yan Huang
- Department of Oncology, Affiliated Hospital of Weifang Medical University, Weifang, China
| |
Collapse
|
42
|
Zheng Y, Yao Y, Ge T, Ge S, Jia R, Song X, Zhuang A. Amino acid metabolism reprogramming: shedding new light on T cell anti-tumor immunity. J Exp Clin Cancer Res 2023; 42:291. [PMID: 37924140 PMCID: PMC10623764 DOI: 10.1186/s13046-023-02845-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 09/28/2023] [Indexed: 11/06/2023] Open
Abstract
Metabolic reprogramming of amino acids has been increasingly recognized to initiate and fuel tumorigenesis and survival. Therefore, there is emerging interest in the application of amino acid metabolic strategies in antitumor therapy. Tremendous efforts have been made to develop amino acid metabolic node interventions such as amino acid antagonists and targeting amino acid transporters, key enzymes of amino acid metabolism, and common downstream pathways of amino acid metabolism. In addition to playing an essential role in sustaining tumor growth, new technologies and studies has revealed amino acid metabolic reprograming to have wide implications in the regulation of antitumor immune responses. Specifically, extensive crosstalk between amino acid metabolism and T cell immunity has been reported. Tumor cells can inhibit T cell immunity by depleting amino acids in the microenvironment through nutrient competition, and toxic metabolites of amino acids can also inhibit T cell function. In addition, amino acids can interfere with T cells by regulating glucose and lipid metabolism. This crucial crosstalk inspires the exploitation of novel strategies of immunotherapy enhancement and combination, owing to the unprecedented benefits of immunotherapy and the limited population it can benefit. Herein, we review recent findings related to the crosstalk between amino acid metabolism and T cell immunity. We also describe possible approaches to intervene in amino acid metabolic pathways by targeting various signaling nodes. Novel efforts to combine with and unleash potential immunotherapy are also discussed. Hopefully, some strategies that take the lead in the pipeline may soon be used for the common good.
Collapse
Affiliation(s)
- Yue Zheng
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 20025, P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, P. R. China
| | - Yiran Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 20025, P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, P. R. China
| | - Tongxin Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 20025, P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, P. R. China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 20025, P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, P. R. China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 20025, P. R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, P. R. China.
| | - Xin Song
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 20025, P. R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, P. R. China.
| | - Ai Zhuang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 20025, P. R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, P. R. China.
| |
Collapse
|
43
|
Weidle UH, Nopora A. Hepatocellular Carcinoma: Up-regulated Circular RNAs Which Mediate Efficacy in Preclinical In Vivo Models. Cancer Genomics Proteomics 2023; 20:500-521. [PMID: 37889063 PMCID: PMC10614070 DOI: 10.21873/cgp.20401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/22/2023] [Accepted: 08/30/2023] [Indexed: 10/28/2023] Open
Abstract
Hepatocellular carcinoma (HCC) ranges as number two with respect to the incidence of tumors and is associated with a dismal prognosis. The therapeutic efficacy of approved multi-tyrosine kinase inhibitors and checkpoint inhibitors is modest. Therefore, the identification of new therapeutic targets and entities is of paramount importance. We searched the literature for up-regulated circular RNAs (circRNAs) which mediate efficacy in preclinical in vivo models of HCC. Our search resulted in 14 circRNAs which up-regulate plasma membrane transmembrane receptors, while 5 circRNAs induced secreted proteins. Two circRNAs facilitated replication of Hepatitis B or C viruses. Three circRNAs up-regulated high mobility group proteins. Six circRNAs regulated components of the ubiquitin system. Seven circRNAs induced GTPases of the family of ras-associated binding proteins (RABs). Three circRNAs induced redox-related proteins, eight of them up-regulated metabolic enzymes and nine circRNAs induced signaling-related proteins. The identified circRNAs up-regulate the corresponding targets by sponging microRNAs. Identified circRNAs and their targets have to be validated by standard criteria of preclinical drug development. Identified targets can potentially be inhibited by small molecules or antibody-based moieties and circRNAs can be inhibited by small-interfering RNAs (siRNAs) or short hairpin RNAs (shRNAs) for therapeutic purposes.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Adam Nopora
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
44
|
Yousf S, Malla JA, Sardesai DM, Sharma S, Talukdar P, Chugh J. Mapping metabolic perturbations induced by glutathione activatable synthetic ion channels in human breast cancer cells. J Pharm Biomed Anal 2023; 235:115605. [PMID: 37531734 DOI: 10.1016/j.jpba.2023.115605] [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: 06/05/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023]
Abstract
Ion channels and transporters play key roles in various biological processes, including cell proliferation and programmed cell death. Recently, we reported that 2,4-dinitrobenzene-sulfonyl-protected N1,N3-dihexy-2-hydroxyisophthalamide (1) forms ion channels upon activation by glutathione (GSH) and results in the induction of apoptosis by depleting the intracellular GSH reservoir in cancer cells. However, the detailed molecular events leading to the induction of apoptosis by these synthetic transport systems in cancer cells still need to be uncovered. Along these lines, we investigated the alterations in cellular metabolites and the associated metabolic pathways by performing untargeted global metabolic profiling of breast cancer cells - MCF-7 - using 1H NMR-based metabolomics. The evaluation of spectral profiles from MCF-7 cells exposed to 1 and their comparison with those corresponding to untreated (control) cells identified 14 significantly perturbed signature metabolites. These metabolites belonged mostly to antioxidant defence, energy metabolism, amino acid biosynthesis, and lipid metabolism pathways and included GSH, o-phosphocholine, malate, and aspartate, to name a few. These results would help us gain deeper insights into the molecular mechanism underlying 1-mediated cytotoxicity of MCF-7 cells and eventually help identify potential novel therapeutic targets for more effective cancer management.
Collapse
Affiliation(s)
- Saleem Yousf
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Dr. Homi Bhabha Road, Pashan, Pune 411008, India.
| | - Javid A Malla
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Dr. Homi Bhabha Road, Pashan, Pune 411008, India
| | - Devika M Sardesai
- Department of Biotechnology, Savitribai Phule Pune University (SPPU), Ganeshkhind, Pune 411007, India
| | - Shilpy Sharma
- Department of Biotechnology, Savitribai Phule Pune University (SPPU), Ganeshkhind, Pune 411007, India
| | - Pinaki Talukdar
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Dr. Homi Bhabha Road, Pashan, Pune 411008, India
| | - Jeetender Chugh
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Dr. Homi Bhabha Road, Pashan, Pune 411008, India.
| |
Collapse
|
45
|
Basson C, Serem JC, Hlophe YN, Bipath P. An in vitro investigation of l-kynurenine, quinolinic acid, and kynurenic acid on B16 F10 melanoma cell cytotoxicity and morphology. Cell Biochem Funct 2023; 41:912-922. [PMID: 37661337 DOI: 10.1002/cbf.3843] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 08/03/2023] [Accepted: 08/20/2023] [Indexed: 09/05/2023]
Abstract
The metastatic behavior of melanoma has accentuated the need for specific therapy targets. Compounds, namely l-kynurenine ( l-kyn), quinolinic acid (Quin), and kynurenic acid (KA) previously displayed antiproliferative and cytotoxic effects in vitro against cancer cells. Despite the growing interest in these compounds there are limited studies examining the in vitro effects on melanoma. In B16 F10 melanoma cells, RAW 264.7 macrophage cells, and HaCat keratinocyte cells, postexposure to the compounds, crystal violet staining was used to determine the half-maximal inhibitory concentration (IC50 ), whereas polarization-optical transmitted light differential interference contrast and light microscopy after hematoxylin and eosin (H&E) staining was used to assess morphological changes. l-kyn, Quin, and KA-induced cytotoxicity in all cell lines, with l-kyn being the most cytotoxic compound. l-kyn and KA at IC50 -induced morphological changes in B16 F10, RAW 264.7, and HaCat cell lines, whereas Quin had effects on B16 F10 and RAW 264.7 cells but did not affect HaCat cells. l-kyn, Quin, and KA each display different levels of cytotoxicity, which were cell line specific. l-kyn was shown to be the most potent compound against all cell lines and may offer future treatment strategies when combined with other viable treatments against melanoma.
Collapse
Affiliation(s)
- Charlise Basson
- Department of Physiology, School of Medicine, University of Pretoria, Pretoria, South Africa
| | - June Cheptoo Serem
- Department of Anatomy, School of Medicine, University of Pretoria, Pretoria, South Africa
| | - Yvette Nkondo Hlophe
- Department of Physiology, School of Medicine, University of Pretoria, Pretoria, South Africa
| | - Priyesh Bipath
- Department of Physiology, School of Medicine, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
46
|
Li T, Shi J, Wang L, Qin X, Zhou R, Dong M, Ren F, Li X, Zhang Z, Chen Y, Liu Y, Piao Y, Shi Y, Xu S, Chen J, Li J. Thymol targeting interleukin 4 induced 1 expression reshapes the immune microenvironment to sensitize the immunotherapy in lung adenocarcinoma. MedComm (Beijing) 2023; 4:e355. [PMID: 37655051 PMCID: PMC10466095 DOI: 10.1002/mco2.355] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/28/2023] [Accepted: 08/06/2023] [Indexed: 09/02/2023] Open
Abstract
Immune checkpoint blockades are the most promising therapy in lung adenocarcinoma (LUAD). However, the response rate remains limited, underscoring the urgent need for effective sensitizers. Interleukin 4 induced 1 (IL4I1) is reported to have immunoinhibitory and tumor-promoting effects in several cancers. However, the targetable value of IL4I1 in sensitizing the immunotherapy is not clear, and there is a lack of effective small molecules that specifically target IL4I1. Here, we show that silencing IL4I1 significantly remodels the immune microenvironment via inhibiting aryl hydrocarbon receptor (AHR) signaling, thereby enhancing the efficacy of anti-PD-1 antibody in LUAD, which suggests that IL4I1 is a potential drug target for the combination immunotherapy. We then identify thymol as the first small molecule targeting IL4I1 transcription through a drug screening. Thymol inhibits the IL4I1 expression and blocks AHR signaling in LUAD cells. Thymol treatment restores the antitumor immune response and suppresses the progression of LUAD in an orthotopic mouse model. Strikingly, the combination treatment of thymol with anti-PD-1 antibody shows significant tumor regression in LUAD mice. Thus, we demonstrate that thymol is an effective small molecule to sensitize the PD-1 blockade in LUAD via targeting IL4I1, which provides a novel strategy for the immunotherapy of LUAD.
Collapse
Affiliation(s)
- Tong Li
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor MicroenvironmentLung Cancer InstituteTianjin Medical University General HospitalTianjinChina
| | - Jie Shi
- School of MedicineNankai UniversityTianjinChina
| | | | - Xuan Qin
- Department of Thyroid and Neck TumorTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Rui Zhou
- School of MedicineNankai UniversityTianjinChina
| | - Ming Dong
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor MicroenvironmentLung Cancer InstituteTianjin Medical University General HospitalTianjinChina
| | - Fan Ren
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor MicroenvironmentLung Cancer InstituteTianjin Medical University General HospitalTianjinChina
| | - Xin Li
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor MicroenvironmentLung Cancer InstituteTianjin Medical University General HospitalTianjinChina
| | - Zihe Zhang
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor MicroenvironmentLung Cancer InstituteTianjin Medical University General HospitalTianjinChina
| | - Yanan Chen
- School of MedicineNankai UniversityTianjinChina
| | - Yanhua Liu
- School of MedicineNankai UniversityTianjinChina
| | | | - Yi Shi
- School of MedicineNankai UniversityTianjinChina
| | - Song Xu
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor MicroenvironmentLung Cancer InstituteTianjin Medical University General HospitalTianjinChina
| | - Jun Chen
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor MicroenvironmentLung Cancer InstituteTianjin Medical University General HospitalTianjinChina
| | - Jia Li
- School of MedicineNankai UniversityTianjinChina
| |
Collapse
|
47
|
Laubach K, Turan T, Mathew R, Wilsbacher J, Engelhardt J, Samayoa J. Tumor-intrinsic metabolic reprogramming and how it drives resistance to anti-PD-1/PD-L1 treatment. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:611-641. [PMID: 37842241 PMCID: PMC10571065 DOI: 10.20517/cdr.2023.60] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/15/2023] [Accepted: 08/29/2023] [Indexed: 10/17/2023]
Abstract
The development of immune checkpoint blockade (ICB) therapies has been instrumental in advancing the field of immunotherapy. Despite the prominence of these treatments, many patients exhibit primary or acquired resistance, rendering them ineffective. For example, anti-programmed cell death protein 1 (anti-PD-1)/anti-programmed cell death ligand 1 (anti-PD-L1) treatments are widely utilized across a range of cancer indications, but the response rate is only 10%-30%. As such, it is necessary for researchers to identify targets and develop drugs that can be used in combination with existing ICB therapies to overcome resistance. The intersection of cancer, metabolism, and the immune system has gained considerable traction in recent years as a way to comprehensively study the mechanisms that drive oncogenesis, immune evasion, and immunotherapy resistance. As a result, new research is continuously emerging in support of targeting metabolic pathways as an adjuvant to ICB to boost patient response and overcome resistance. Due to the plethora of studies in recent years highlighting this notion, this review will integrate the relevant articles that demonstrate how tumor-derived alterations in energy, amino acid, and lipid metabolism dysregulate anti-tumor immune responses and drive resistance to anti-PD-1/PD-L1 therapy.
Collapse
Affiliation(s)
- Kyra Laubach
- Computational Oncology, AbbVie, South San Francisco, CA 94080, USA
- Immuno-Oncology, AbbVie, South San Francisco, CA 94080, USA
| | - Tolga Turan
- Computational Oncology, AbbVie, South San Francisco, CA 94080, USA
| | - Rebecca Mathew
- Immuno-Oncology, AbbVie, South San Francisco, CA 94080, USA
| | | | | | - Josue Samayoa
- Computational Oncology, AbbVie, South San Francisco, CA 94080, USA
| |
Collapse
|
48
|
Zhu Z, Yang Z, Zhu C, Hu Z, Jiang Z, Gong J, Yuan Y, Chen X, Jin Y, Yin Y. Development of a DNA aptamer targeting IDO1 with anti-tumor effects. iScience 2023; 26:107367. [PMID: 37520707 PMCID: PMC10374466 DOI: 10.1016/j.isci.2023.107367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/29/2023] [Accepted: 07/10/2023] [Indexed: 08/01/2023] Open
Abstract
Immune checkpoint blockade has become an effective approach to reverse the immune tolerance of tumor cells. Indoleamine 2,3-dioxygenase 1 (IDO1) is frequently upregulated in many types of cancers and contributes to the establishment of an immunosuppressive cancer microenvironment, which has been thought to be a potential target for cancer therapy. However, the development of IDO1 inhibitors for clinical application is still limited. Here, we isolated a DNA aptamer with a strong affinity and inhibitory activity against IDO1, designated as IDO-APT. By conjugating with nanoparticles, in situ injection of IDO-APT to CT26 tumor-bearing mice significantly suppresses the activity of regulatory T cells and promotes the function of CD8+ T cells, leading to tumor suppression and prolonged survival. Therefore, this functional IDO1-specific aptamer with potent anti-tumor effects may serve as a potential therapeutic strategy in cancer immunotherapy. Our data provide an alternative way to target IDO1 in addition to small molecule inhibitors.
Collapse
Affiliation(s)
- Zhenyu Zhu
- Institute of Systems Biomedicine, Department of Pathology, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Zeliang Yang
- Institute of Systems Biomedicine, Department of Pathology, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Chuanda Zhu
- Institute of Systems Biomedicine, Department of Pathology, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Zixi Hu
- Institute of Systems Biomedicine, Department of Pathology, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Zhongyu Jiang
- Institute of Systems Biomedicine, Department of Pathology, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Jingjing Gong
- Institute of Systems Biomedicine, Department of Pathology, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yuyao Yuan
- Institute of Systems Biomedicine, Department of Pathology, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Xi Chen
- Institute of Systems Biomedicine, Department of Pathology, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yan Jin
- Institute of Systems Biomedicine, Department of Pathology, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yuxin Yin
- Institute of Systems Biomedicine, Department of Pathology, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
- Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, China
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China
| |
Collapse
|
49
|
Abdel-Aal FAM, Kamel RM, Abdeltawab AA, Mohamed FA, Mohamed AMI. Polypyrrole/carbon dot nanocomposite as an electrochemical biosensor for liquid biopsy analysis of tryptophan in the human serum of normal and breast cancer women. Anal Bioanal Chem 2023; 415:4985-5001. [PMID: 37401962 PMCID: PMC10386971 DOI: 10.1007/s00216-023-04784-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/14/2023] [Accepted: 05/30/2023] [Indexed: 07/05/2023]
Abstract
Liquid biopsy analysis represents a suitable alternative analysis procedure in several cases where no tumor tissue is available or in poor patient conditions. Amino acids can play a crucial role in aiding cancer diagnosis. Monitoring of tryptophan (Trp) catabolism can aid in tracking cancer progression. Therefore, a novel nanocomposite was fabricated using overoxidized polypyrrole film doped with nano-carbon dots (nano-CDs) on the pencil graphite electrode (PGE) surface for sensitive evaluation of Trp in human serum. Using square wave voltammetry (SWV), the overoxidized polypyrrole/carbon dots/pencil graphite electrode (Ov-Ox PPy/CDs/PGE) achieved excellent electrochemical catalytic activity for evaluating Trp. The modified electrode, known as Ov-Ox PPy/CDs/PGE, demonstrated superior electrochemical catalytic activity compared to bare PGE, CDs/PGE, PPy/PGE, and PPy/CDs/PGE for evaluation of Trp. The method's excellent sensitivity was confirmed by the low limits of detection (LOD = 0.003 μmol L-1) and limit of quantitation (LOQ = 0.009 μmol L-1). The biosensor that was developed can measure tryptophan (Trp) levels in the serum of both healthy individuals and female breast cancer patients with high accuracy and sensitivity. The results indicate that there is a significant difference, as shown by the F-test, between healthy individuals and those with breast cancer. This suggests that Trp amino acid could be an essential biomarker for cancer diagnosis. Consequently, liquid biopsy analysis presents a valuable opportunity for early disease detection, particularly for cancer.
Collapse
Affiliation(s)
- Fatma A M Abdel-Aal
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt.
| | - Rania M Kamel
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| | - Asmaa A Abdeltawab
- Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Assuit University, Assiut, 71526, Egypt
| | - Fardous A Mohamed
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| | - Abdel-Maaboud I Mohamed
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| |
Collapse
|
50
|
Cortellino S, Longo VD. Metabolites and Immune Response in Tumor Microenvironments. Cancers (Basel) 2023; 15:3898. [PMID: 37568713 PMCID: PMC10417674 DOI: 10.3390/cancers15153898] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/27/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
The remodeled cancer cell metabolism affects the tumor microenvironment and promotes an immunosuppressive state by changing the levels of macro- and micronutrients and by releasing hormones and cytokines that recruit immunosuppressive immune cells. Novel dietary interventions such as amino acid restriction and periodic fasting mimicking diets can prevent or dampen the formation of an immunosuppressive microenvironment by acting systemically on the release of hormones and growth factors, inhibiting the release of proinflammatory cytokines, and remodeling the tumor vasculature and extracellular matrix. Here, we discuss the latest research on the effects of these therapeutic interventions on immunometabolism and tumor immune response and future scenarios pertaining to how dietary interventions could contribute to cancer therapy.
Collapse
Affiliation(s)
- Salvatore Cortellino
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy;
| | - Valter D. Longo
- IFOM, The AIRC Institute of Molecular Oncology, 20139 Milan, Italy
- Longevity Institute, Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|