1
|
Larraz-Prieto B, Lind LH, Olesen JB, Azfer A, Hansen MS, Frost M, Jafari A, Ralston SH, Søe K, Alonso N. CXCR4 is a response gene for parathyroid hormone which affects osteoblast and osteoclast function in vitro. Bone Joint Res 2025; 14:463-476. [PMID: 40376984 DOI: 10.1302/2046-3758.145.bjr-2024-0167.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/18/2025] Open
Abstract
Aims To investigate the role of CXCR4 in response to teriparatide (TPTD) treatment in osteoblasts and osteoclasts. Methods Primary murine and human osteoblasts and osteoclasts, MC3T3 cell lines, and hMSC-TERT4 cell lines were treated with TPTD and/or AMD3100, a pharmacological inhibitor of CXCR4. Changes in gene expression, osteoblast viability, mobility, mineralization capacity, and alkaline phosphatase activity were investigated. Osteoclastogenesis and cell fusion were also assessed in response to both treatments. Results TPTD increased messenger RNA levels of CXCR4 in all stages of both murine and human osteoblast differentiation. Mineralization analysis showed that CXCR4 was involved in bone matrix formation in response to TPTD. Alkaline phosphatase activity was also impaired by CXCR4 inhibition at early stages of osteoblast differentiation, while it was promoted at late stages, suggesting that CXCR4 could produce a delay in osteoblast maturation. Moreover, we also found a direct activation of osteoclastogenesis after TPTD treatment in murine and human osteoclasts. This process seems to involve CXCR4 activity, since AMD3100-induced CXCR4 inhibition led to a reduction in both murine and human osteoclastogenesis. This process, however, could not be prevented by TPTD treatment. Conclusion Our results suggest that CXCR4 is a responsive gene to TPTD treatment, involved in the regulation of osteoblast and osteoclast generation and function. Further in vivo studies are required to confirm this role, and to determine whether pharmacological strategies targeting CXCR4 could potentially improve the treatment outcome for osteoporotic patients.
Collapse
Affiliation(s)
- Beatriz Larraz-Prieto
- Rheumatic Disease Unit, Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Louise Hjorth Lind
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | | | - Asim Azfer
- Rheumatic Disease Unit, Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Morten Svarer Hansen
- Department of Endocrinology and Metabolism, Molecular Endocrinology Laboratory (KMEB), Odense University Hospital, Odense, Denmark
| | - Morten Frost
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Endocrinology and Metabolism, Molecular Endocrinology Laboratory (KMEB), Odense University Hospital, Odense, Denmark
- Steno Diabetes Center Odense, Odense, Denmark
| | - Abbas Jafari
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Stuart H Ralston
- Rheumatic Disease Unit, Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Kent Søe
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Nerea Alonso
- Rheumatic Disease Unit, Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| |
Collapse
|
2
|
Liu Y, Chen X, Zhang W, Yu B, Cen Y, Liu Q, Tang Y, Li S. A CXCR4-targeted immunomodulatory nanomedicine for photodynamic amplified immune checkpoint blockade therapy against breast cancer. Acta Biomater 2025; 197:400-415. [PMID: 40154764 DOI: 10.1016/j.actbio.2025.03.049] [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: 11/19/2024] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
The therapeutic efficacy of immune checkpoint blockade (ICB) is critically compromised by inadequate T lymphocyte infiltration, low T cell-induced pro-inflammatory responses, and the accumulation of immunosuppressive cells within the tumor microenvironment (TME). In this work, a chimeric peptide-engineered immunomodulatory nanomedicine (designated as CXNP-CeBM) is developed for photodynamic amplified ICB therapy against breast cancer. CXNP-CeBM is composed of a CXCR4-targeting peptide ((C16)2-KLGASWHRPDK) loaded with the photosensitizer of Ce6 and the PD-1/PD-L1 inhibitor of BMS8. CXNP-CeBM specifically recognizes CXCR4 on breast cancer, thus suppressing CXCR4-mediated signaling pathways and enhancing the intracellular delivery of therapeutic agents. The photodynamic therapy (PDT) of CXNP-CeBM damages primary tumor cells to initiate immunogenic cell death (ICD), leading to the release of high mobility group box 1 (HMGB1) and the exposure of calreticulin (CRT). Simultaneously, the interruption of CXCR4 signaling reduces tumor fibrosis, promotes T-cell infiltration, and decreases the number of immunosuppressive cells, thereby enhancing the immunotherapeutic effect of ICB. Treatment with CXNP-CeBM would activate systemic anti-tumor immunity, leading to effective inhibition of both primary and lung metastatic tumors, while maintaining low systemic toxicity. This work provides a reliable strategy for the delivery of multi-synergistic agents, effectively activating breast cancer immunity through a multifaceted mechanism. STATEMENT OF SIGNIFICANCE: Although immune checkpoint blockade (ICB) has shown great potential for malignant tumor therapy, its efficacy is compromised by immunosuppressive microenvironments. Herein, a CXCR4-targeted immunomodulatory nanomedicine (CXNP-CeBM) was constructed for photodynamic amplified ICB therapy of breast cancer. CXNP-CeBM could selectively deliver photosensitizers and PD-1/PD-L1 inhibitors to breast cancer cells that overexpressed the chemokine receptor CXCR4, while interrupting CXCR4 signaling to reduce tumor fibrosis, promote T-cell infiltration, and decrease the number of immunosuppressive cells. Moreover, CXNP-CeBM induced photodynamic therapy to trigger immunogenic cell death while downregulating the PD-L1 level to destroy immune evasion mechanisms, thus activating immunological cascades to treat both primary and lung metastatic tumors. This study provided a multi-synergistic strategy for breast cancer immunotherapy through a multifaceted mechanism.
Collapse
Affiliation(s)
- Yibin Liu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Xiayun Chen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Wei Zhang
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Baixue Yu
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yi Cen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Qianqian Liu
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Youzhi Tang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China.
| | - Shiying Li
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China.
| |
Collapse
|
3
|
Zhang Z, Zhang Q, Wang Y. CAF-mediated tumor vascularization: From mechanistic insights to targeted therapies. Cell Signal 2025; 132:111827. [PMID: 40288665 DOI: 10.1016/j.cellsig.2025.111827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Cancer-associated fibroblasts (CAFs) are a major component of the tumor microenvironment (TME) and play a crucial role in tumor progression. The biological properties of tumors, such as drug resistance, vascularization, immunosuppression, and metastasis are closely associated with CAFs. During tumor development, CAFs contribute to tumor progression by remodeling the extracellular matrix (ECM), inhibiting immune cell function, promoting angiogenesis, and facilitating tumor cell growth, invasion, and metastasis. Studies have shown that CAFs can promote endothelial cell proliferation by directly secreting cytokines such as vascular endothelial growth factor (VEGF) and fibroblast Growth Factor (FGF), as well as through exosomes. CAFs also secrete the chemokine stromal cell-derived factor 1 (SDF-1) to recruit endothelial progenitor cells (EPCs) into the peripheral blood and guide their migration to the tumor periphery. Additionally, CAFs can induce tumor cells to transform into "endothelial cells" that participate in vascular wall formation. However, the precise mechanisms remain to be further investigated. Due to their widespread presence in various solid tumors and their tumor-promoting function, CAFs are emerging as therapeutic targets. In this review, we summarize the specific mechanisms through which CAFs promote angiogenesis and outline current therapeutic strategies targeting CAF-induced vascularization, ongoing clinical trials targeting CAFs, and discuss potential future treatment approaches. We hope this will contribute to the advancement of CAF-targeted tumor treatment strategies.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Qing Zhang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Yang Wang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
4
|
Braun T, Bisht A, Zhu C, Idrees M, Alabeedi F, Kujan O. Diagnostic, prognostic, and metastatic value of chemokines as biomarkers for oral squamous cell carcinoma and their precursor lesions - A systematic review. Crit Rev Oncol Hematol 2025; 211:104738. [PMID: 40268074 DOI: 10.1016/j.critrevonc.2025.104738] [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: 11/08/2024] [Revised: 02/26/2025] [Accepted: 04/18/2025] [Indexed: 04/25/2025] Open
Abstract
Oral cancer remains a significant public health concern, with many patients diagnosed at advanced stages and facing poor prognoses. Despite advances in cancer research, diagnosis has seen only limited improvements, with biopsies still being the primary reliable method. This systematic review investigates the role of chemokines as potential biomarkers for early detection, prognosis, and metastasis in oral squamous cell carcinoma (OSCC) and oral potentially malignant disorders (OPMDs). Through an extensive literature search of MEDLINE, EMBASE, PubMed, and Scopus, 3350 articles were initially identified. After eliminating duplicates and screening for eligibility, 50 high-quality studies were included, offering a comprehensive overview of chemokine research in OSCC and OPMDs. Key findings indicate that CCR7 shows significant promise as a diagnostic, prognostic, and metastatic marker, although its function in precancerous conditions remains inadequately understood. CXCL10 and CCL22 were also highlighted for their strong prognostic and metastatic relevance, while CXCR4 and CXCL12 were identified as critical indicators of OSCC metastasis. Other chemokines, such as CXCR2, CCR4, XCR1, CXCL13, and CCL2 can aid OSCC differentiation and staging. However, the review emphasises the limitations of small patient cohorts and the lack of longitudinal research, stressing the need for further studies. Additionally, there is a pressing gap in research addressing chemokines as biomarkers for OPMDs. Rigorous validation is crucial to establish these biomarkers' reliability and clinical utility across various stages of oral cancer development.
Collapse
Affiliation(s)
- Timothy Braun
- UWA Dental School, The University of Western Australia, Nedlands, WA, Australia
| | - Abhimanyu Bisht
- UWA Dental School, The University of Western Australia, Nedlands, WA, Australia
| | - Christopher Zhu
- UWA Dental School, The University of Western Australia, Nedlands, WA, Australia
| | - Majdy Idrees
- UWA Dental School, The University of Western Australia, Nedlands, WA, Australia
| | - Faris Alabeedi
- UWA Dental School, The University of Western Australia, Nedlands, WA, Australia
| | - Omar Kujan
- UWA Dental School, The University of Western Australia, Nedlands, WA, Australia.
| |
Collapse
|
5
|
Khaleel AQ, Altalbawy FMA, Jabir MS, F Hasan T, Jain V, Abbot V, Nakash P, Kumar MR, Mustafa YF, Jawad MA. CXCR4/CXCL12 blockade therapy; a new horizon in TNBC therapy. Med Oncol 2025; 42:161. [PMID: 40216617 DOI: 10.1007/s12032-025-02705-5] [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: 11/30/2024] [Accepted: 03/29/2025] [Indexed: 05/03/2025]
Abstract
The only subtype of breast cancer (BC) without specific therapy is triple-negative breast cancer (TNBC), which represents 15-20% of incidence cases of BC. TNBC encompasses transformed and nonmalignant cells, including cancer-associated fibroblasts (CAF), endothelial vasculature, and tumor-infiltrating cells. These nonmalignant cells, soluble factors (e.g., cytokines), and the extracellular matrix (ECM) form the tumor microenvironment (TME). The TME is made up of these nonmalignant cells, ECM, and soluble components, including cytokines. Direct cell-to-cell contact and soluble substances like cytokines (e.g., chemokines) may facilitate interaction between cancer cells and the surrounding TME. Through growth-promoting cytokines, TME not only enables the development of cancer but also confers therapy resistance. New treatment targets will probably be suggested by comprehending the processes behind tumor development and progression as well as the functions of chemokines in TNBC. In this light, several investigations have shown the pivotal function of the C-X-C motif chemokine ligand 12 (CXCL12 or SDF-1) axis and chemokine receptor type 4 (CXCR4) in the pathophysiology of TNBC. This review provides an overview of the CXCR4/CXCL12 axis' function in TNBC development, metastasis, angiogenesis, and treatment resistance. A synopsis of current literature on targeting the CXCR4/CXCL12 axis for treating and managing TNBC has also been provided.
Collapse
Affiliation(s)
- Abdulrahman Qais Khaleel
- Department of Medical Instruments Engineering, Al-Maarif University College, Al Anbar, 31001, Iraq.
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | - Majid S Jabir
- Department of Applied Sciences, University of Technology, Baghdad, Iraq
| | - Thikra F Hasan
- College of Health&Medical Technology, Uruk University, Baghdad, Iraq
| | - Vicky Jain
- Department of Chemistry, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - Vikrant Abbot
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Prashant Nakash
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - M Ravi Kumar
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | | |
Collapse
|
6
|
Dai X, Xi M, Li J. Cancer metastasis: molecular mechanisms and therapeutic interventions. MOLECULAR BIOMEDICINE 2025; 6:20. [PMID: 40192949 PMCID: PMC11977077 DOI: 10.1186/s43556-025-00261-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 04/10/2025] Open
Abstract
The metastatic cascade is a complicated process where cancer cells travel across multiple organs distant from their primary site of onset. Despite the wide acceptance of the 'seed and soil' theory, mechanisms driving metastasis organotropism remain mystery. Using breast cancer of different subtypes as the disease model, we characterized the 'metastatic profile of cancer cells' and the 'redox status of the organ microenvironment' as the primary determinants of cancer metastasis organotropism. Mechanically, we identified a positive correlation between cancer metabolic plasticity and stemness, and proposed oxidative stress as the selection power of cancer cells succeeding the metastasis cascade. Therapeutically, we proposed the use of pro-oxidative therapeutics in ablating cancer cells taking advantages of this fragile moment during metastasis. We comprehensively reviewed current pro-oxidative strategies for treating cancers that cover the first line chemo- and radio-therapies, approaches relying on naturally existing power including magnetic field, electric field, light and sound, nanoparticle-based anti-cancer composites obtained through artificial design, as well as cold atmospheric plasma as an innovative pro-oxidative multi-modal modality. We discussed possible combinations of pro-oxidative approaches with existing therapeutics in oncology prior to the forecast of future research directions. This paper identified the fundamental mechanics driving metastasis organotropism and proposed intervention strategies accordingly. Insights provided here may offer clues for the design of innovative solutions that may open a new paradigm for cancer treatment.
Collapse
Affiliation(s)
- Xiaofeng Dai
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China.
| | - Ming Xi
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Jitian Li
- Molecular Biology Lab, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Henan Province, Zhengzhou, 450000, China
| |
Collapse
|
7
|
Guo Y, Zhang Z, Huang H, Wu Y, Yin L, Zhou Y, Ding F, Hong S, Steinmetz NF, Cai H. Targeting S100A8/A9-NCF1 axis in tumor microenvironment to prevent tumor metastasis by self-assembled peptide nanofibers. Mol Ther 2025; 33:1502-1518. [PMID: 40040282 PMCID: PMC11997502 DOI: 10.1016/j.ymthe.2025.02.042] [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: 10/27/2024] [Revised: 01/31/2025] [Accepted: 02/27/2025] [Indexed: 03/06/2025] Open
Abstract
The immunosuppressive microenvironment plays a crucial role in driving and accelerating tumor metastasis. S100A8/A9, produced by myeloid-derived suppressor cells, is a potential therapeutic target for metastatic cancer due to its role in promoting premetastatic niche formation. Previous studies have revealed that the S100A9-targeted peptide (H6, MEWSLEKGYTIK) fused to the Fc region of mouse IgG2b antibodies exhibits antitumor effects; however, the mechanism remains unclear. Here, dual-function peptide nanofibers (H6-Q11) were constructed, consisting of peptide H6 and self-assembly peptide (Q11, QQKFQFQFEQQ), which achieved high avidity for S100A9. In vivo studies showed that H6-Q11 nanofibers significantly prolonged lung retention and inhibited pulmonary metastasis from melanoma and breast cancer without obvious toxicity. Immunological analyses indicated that treatment with H6-Q11 nanofibers decreased the infiltration of immunosuppressive cells while promoting the recruitment of immune effector cells to the lungs, potentially correlated with disturbances of S100A8/A9-NCF1 signaling in the tumor microenvironment. Our findings support a potential clinical application of S100A9-targeted peptide nanofibers as candidate nanomedicine for inhibiting tumor metastasis.
Collapse
Affiliation(s)
- Yajing Guo
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchanglu Road, Guangming District, Shenzhen 518107, China
| | - Zhifei Zhang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchanglu Road, Guangming District, Shenzhen 518107, China
| | - Hongxia Huang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchanglu Road, Guangming District, Shenzhen 518107, China
| | - Ye Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchanglu Road, Guangming District, Shenzhen 518107, China
| | - Lixin Yin
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchanglu Road, Guangming District, Shenzhen 518107, China
| | - Yang Zhou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchanglu Road, Guangming District, Shenzhen 518107, China
| | - Feiqing Ding
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchanglu Road, Guangming District, Shenzhen 518107, China
| | - Sheng Hong
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchanglu Road, Guangming District, Shenzhen 518107, China
| | - Nicole F Steinmetz
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Center for Nano Immuno-Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory University of California, San Diego, La Jolla, CA 92093, USA
| | - Hui Cai
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, 66 Gongchanglu Road, Guangming District, Shenzhen 518107, China.
| |
Collapse
|
8
|
Spahn MA, Loy TV, Celen S, Koole M, Deroose CM, Cawthorne C, Vanduffel W, Schols D, Bormans G, Cleeren F. Selective PET imaging of CXCR4 using the Al 18F-labeled antagonist LY2510924. Eur J Nucl Med Mol Imaging 2025; 52:1723-1738. [PMID: 39658737 PMCID: PMC11928405 DOI: 10.1007/s00259-024-07025-w] [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: 08/29/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND [68Ga]PentixaFor detects C-X-C chemokine receptor type 4 (CXCR4) overexpression in various malignancies, such as multiple myeloma and non-Hodgkin lymphomas, as well as in endocrine and inflammatory disorders. This study aimed to develop an Al18F-labeled radiotracer derived from LY2510924 for CXCR4-targeted imaging, leveraging the physical and logistical advantages of fluorine-18. METHODS We designed a CXCR4-specific radioprobe, [18F]AlF-NOTA-SC, based on LY2510924 by incorporating a triglutamate linker and NOTA chelator to enable Al18F-labeling. The in vitro CXCR4 affinity was assessed using cell-based binding assays. Subsequently, in vivo pharmacokinetics and tumor uptake of [18F]AlF-NOTA-SC were assessed in naïve mice and mice with xenografts derived from U87.CD4/U87.CD4.CXCR4 and MM.1 S cells. Finally, biodistribution was determined in a non-human primate using PET-MR. RESULTS Compared to Ga-PentixaFor, AlF-NOTA-SC demonstrated similar in vitro affinity for human CXCR4. [18F]AlF-NOTA-SC was produced with a decay-corrected radiochemical yield of 21.0 ± 7.1% and an apparent molar activity of 16.4 ± 3.6 GBq/µmol. In [18F]AlF-NOTA-SC binding assays on U87.CD4.CXCR4 cells, the total bound fraction was 7.1 ± 0.5% (58% blocking by AMD3100). In naïve mice, the radiotracer did not accumulate in any organs; however, it showed a significant CXCR4-specific uptake in xenografted tumors (SUVmeanU87.CD4 = 0.04 ± 0.00 (n = 3); SUVmeanU87.CD4.CXCR4 = 3.04 ± 0.65 (n = 3); SUVmeanMM.1 S = 1.95 ± 0.11 (n = 3)). In a non-human primate, [18F]AlF-NOTA-SC accumulated in CXCR4 expressing organs, such as the spleen and bone marrow. CONCLUSION [18F]AlF-NOTA-SC exhibited CXCR4-specific uptake in vitro and in vivo, with fast and persistent tumor accumulation, making it a strong candidate for clinical translation as an 18F-alternative to [68Ga]PentixaFor.
Collapse
Affiliation(s)
- Muriel Aline Spahn
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Tom Van Loy
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular Structural and Translational Virology Research Group, Leuven, B-3000, Belgium
| | - Sofie Celen
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Christophe M Deroose
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Christopher Cawthorne
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Wim Vanduffel
- Laboratory for Neuro- and Psychophysiology, KU Leuven Medical School, Leuven, Belgium
| | - Dominique Schols
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular Structural and Translational Virology Research Group, Leuven, B-3000, Belgium
| | - Guy Bormans
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Frederik Cleeren
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
9
|
Puig-Gámez M, Van Attekum M, Theis T, Dick A, Park JE. Transcriptional signature of rapidly responding NK cells reveals S1P5 and CXCR4 as anti-tumor response disruptors. Sci Rep 2025; 15:10769. [PMID: 40155684 PMCID: PMC11953373 DOI: 10.1038/s41598-025-95211-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/19/2025] [Indexed: 04/01/2025] Open
Abstract
Natural killer (NK) cells are prototypic cytotoxic innate lymphocytes that can kill target cells, such as tumor cells, in the absence of antigen-restriction. Peripheral NK cells exhibit a high degree of heterogeneity. Here, we set out to broadly assess intrinsic modulators of NK cell degranulation in an unbiased manner. We stimulated human primary blood-borne NK cells pre-treated with different cytokine regimens with the HCT116 human colon cancer cell line and used detection of lysosome-associated membrane glycoprotein 1 (LAMP1) as an identifier of rapid NK cell degranulation. RNA sequencing of FACS-sorted LAMP1hi NK cells showed CXCR4 and S1PR5 were top down-regulated genes. Using compounds that modulate activity of CXCR4 and S1P receptor family members S1P1 and S1P5, we confirmed they play an important immunosuppressive role in NK cell cytotoxicity. Mechanistically, engagement of CXCR4 and S1P1/5 receptors triggered phosphorylation of p42 and Ca2+ influx. CXCR4 activation promoted S1P5 upregulation and vice versa, and joint activation of both receptors amplified the defect NK cell degranulation. Intriguingly, in tumor samples the expression of both receptors and the synthesis of their ligands themselves appear to be coordinately regulated. Together, these data suggest that specifically and simultaneously targeting CXCR4 and S1P5 activity in the tumor microenvironment (TME) could be a beneficial strategy to unleash full cytotoxic potential of cytotoxic NK effector cells in the tumor.
Collapse
Affiliation(s)
- Marta Puig-Gámez
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, 88387, Biberach an der Riss, Germany
| | - Martijn Van Attekum
- Department of Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88387, Biberach an der Riss, Germany
| | - Theodor Theis
- Department of Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, 88387, Biberach an der Riss, Germany
| | - Alec Dick
- Department of Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88387, Biberach an der Riss, Germany
| | - John E Park
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, 88387, Biberach an der Riss, Germany.
| |
Collapse
|
10
|
Smaldone G, Di Matteo F, Castelluccio R, Napolitano V, Miranda MR, Manfra M, Campiglia P, Vestuto V. Targeting the CXCR4/CXCL12 Axis in Cancer Therapy: Analysis of Recent Advances in the Development of Potential Anticancer Agents. Molecules 2025; 30:1380. [PMID: 40142155 PMCID: PMC11945090 DOI: 10.3390/molecules30061380] [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: 02/20/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
Cancer, a leading cause of premature death, arises from genetic and epigenetic mutations that transform normal cells into tumor cells, enabling them to proliferate, evade cell death, and stimulate angiogenesis. Recent evidence indicates that chemokines are essential in tumor development, activating receptors that promote proliferation, invasion, and metastasis. The CXCR4/CXCL12 signaling pathway is gaining attention as a promising target for cancer therapy. CXCR4, a chemokine receptor, is often overexpressed in various types of cancer, including kidney, lung, brain, prostate, breast, pancreas, ovarian, and melanomas. When it binds to its endogenous ligand, CXCL12, it promotes cell survival, proliferation, and migration, crucial mechanisms for the retention of hematopoietic stem cells in the bone marrow and the movement of lymphocytes. The extensive expression of CXCR4 in cancer, coupled with the constant presence of CXCL12 in various organs, drives the activation of this axis, which in turn facilitates angiogenesis, tumor progression, and metastasis. Given the detrimental role of the CXCR4/CXCL12 axis, the search for drugs acting selectively against this protein represents an open challenge. This review aims to summarize the recent advancements in the design and development of CXCR4 antagonists as potential anticancer agents.
Collapse
Affiliation(s)
- Gerardina Smaldone
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (G.S.); (F.D.M.); (R.C.); (V.N.); (P.C.)
| | - Francesca Di Matteo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (G.S.); (F.D.M.); (R.C.); (V.N.); (P.C.)
| | - Roberta Castelluccio
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (G.S.); (F.D.M.); (R.C.); (V.N.); (P.C.)
| | - Valeria Napolitano
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (G.S.); (F.D.M.); (R.C.); (V.N.); (P.C.)
| | - Maria Rosaria Miranda
- PhD Program in Drug Discovery and Development, University of Salerno, 84084 Fisciano, Italy;
| | - Michele Manfra
- Department of Health Science, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy;
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (G.S.); (F.D.M.); (R.C.); (V.N.); (P.C.)
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (G.S.); (F.D.M.); (R.C.); (V.N.); (P.C.)
| |
Collapse
|
11
|
Worden AN, Pittard EG, Stern M, Uline MJ, Potts JD. The Role of the CXCL12/CXCR4 Signaling Pathway in Regulating Cellular Migration. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2025; 31:ozaf011. [PMID: 40095909 DOI: 10.1093/mam/ozaf011] [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: 11/07/2024] [Revised: 02/06/2025] [Accepted: 02/17/2025] [Indexed: 03/19/2025]
Abstract
We investigated the CXCL12/CXCR4 signaling pathway as a regulator of adipose-derived stem cell (ADSC) self-assembling toroidal constructs using collagen hydrogels. ADSCs formed toroid rings when cultured on hydrogel surfaces but failed to do so when mixed within the matrix. Gene expression profiling revealed significant upregulation of the CXCL12/CXCR4 pathway in toroid-forming conditions, supported by immunofluorescence studies that confirmed CXCL12 presence in toroids but not in mixed-in cultures. Early toroid formation was marked by the emergence of CXCL12 expression, correlating with cell migration. Targeted inhibition experiments identified the PI3K pathway as a critical regulator, delaying cell migration by ∼16 h, while N-Cadherin, Ras/Raf, and ERK1/2 inhibition either reduced or halted migration over extended periods. Through Western blot analysis, altered expression of α-Smooth muscle actin and focal adhesion kinase under PI3K inhibition was highlighted thus emphasizing their roles in toroid formation. Lastly, initial coculture studies with 4T1 breast cancer cells unexpectedly showed CXCL12 localization primarily in 4T1 cells within mixed toroids, suggesting modified chemotactic signaling. Our findings establish CXCL12/CXCR4 as crucial for ADSC toroid formation and reveal the pathway's complex involvement in cellular organization and migration, presenting a robust model for exploring cell-cell and cell-matrix interactions relevant to tissue engineering and cancer research.
Collapse
Affiliation(s)
- Austin N Worden
- Department of Cell Biology and Anatomy, University of South Carolina, School of Medicine, 6311 Garners Ferry Rd., Columbia, SC 29209, USA
| | - Emma Grace Pittard
- Department of Cell Biology and Anatomy, University of South Carolina, School of Medicine, 6311 Garners Ferry Rd., Columbia, SC 29209, USA
| | - Matt Stern
- Biology Department, Winthrop University, 701 Oakland Ave., Rock Hill, SC 29733, USA
| | - Mark J Uline
- Chemical Engineering Department, University of South Carolina, 301 Main St., Columbia, SC 29208, USA
- Biomedical Engineering Program, University of South Carolina, 300 Main St., Columbia, SC 29208, USA
| | - Jay D Potts
- Department of Cell Biology and Anatomy, University of South Carolina, School of Medicine, 6311 Garners Ferry Rd., Columbia, SC 29209, USA
- Biomedical Engineering Program, University of South Carolina, 300 Main St., Columbia, SC 29208, USA
| |
Collapse
|
12
|
Lee YI, Yang Y, Ham S, Shim JE, Lee SG, Lee SH, Kim TG, Lee WJ, Kim DY, Lee JH. Heterogeneity in Keloid Scars: Influence of Mechanical Stretching on Keloids Arising from Different Anatomical Sites. J Invest Dermatol 2025; 145:710-713.e7. [PMID: 39245138 DOI: 10.1016/j.jid.2024.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 09/10/2024]
Affiliation(s)
- Young In Lee
- Department of Dermatology, Yonsei University College of Medicine, Seoul, Republic of Korea; Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Scar Laser and Plastic Surgery Center, Yonsei Cancer Hospital, Seoul, Republic of Korea
| | - Yohan Yang
- Bioinformatics Collaboration Unit, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seoyoon Ham
- Department of Dermatology, Yonsei University College of Medicine, Seoul, Republic of Korea; Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jung Eun Shim
- Bioinformatics Collaboration Unit, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang Gyu Lee
- Department of Dermatology, Yonsei University College of Medicine, Seoul, Republic of Korea; Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Si-Hyung Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Tae-Gyun Kim
- Department of Dermatology, Yonsei University College of Medicine, Seoul, Republic of Korea; Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Won Jai Lee
- Scar Laser and Plastic Surgery Center, Yonsei Cancer Hospital, Seoul, Republic of Korea; Department of Plastic Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Do-Young Kim
- Department of Dermatology, Yonsei University College of Medicine, Seoul, Republic of Korea; Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Ju Hee Lee
- Department of Dermatology, Yonsei University College of Medicine, Seoul, Republic of Korea; Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Scar Laser and Plastic Surgery Center, Yonsei Cancer Hospital, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Menon SR, Sahu S, Mitra A, Chakraborty A, Malhotra G, Kamaldeep, Tawate M, Lad S, Rakshit S, Upadhye T, Ray MK, Banerjee S. On the automated radiosynthesis of pharmaceutical grade [ 68Ga]Ga-Pentixafor, its pre-clinical evaluation, clinical application and radiation dosimetry aspects. Sci Rep 2025; 15:6476. [PMID: 39987209 PMCID: PMC11846852 DOI: 10.1038/s41598-024-84096-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/19/2024] [Indexed: 02/24/2025] Open
Abstract
The current study outlines a consistent and reproducible protocol for the routine clinical dose preparation of [68Ga]Ga-Pentixafor using the Eckert and Ziegler 'Modular-Lab Standard' non-cassette based automated module, that can be effectively used in the hospital radiopharmacy unit of a high volume nuclear medicine centre. The pre-clinical studies (including in-vitro cell line studies, in-vivo PET/CT imaging and pre-clinical dosimetry) were conducted to show the promising potential of the product for clinical use in targeting CXCR4 tumor overexpression. PET/CT image of SCID mouse bearing lymphoma xenograft tumor, at 2 h post-injection, clearly delineated the tumor. The pre-clinical dosimetry results show the suitability of the product for clinical use in patients. [68Ga]Ga-Pentixafor when administered to patients with primary aldosteronism exhibited distinct uptake in the adrenal nodules. The clinical PET/CT scan of the patients demonstrated the potential use of CXCR4 targeted imaging as a promising surgical decision-making tool for patients with primary aldosteronism.
Collapse
Affiliation(s)
- Sreeja Raj Menon
- Health Physics Division, Bhabha Atomic Research Centre, Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sudeep Sahu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Maharashtra, Mumbai, India
| | - Arpit Mitra
- Radiopharmaceutical Laboratory, Board of Radiation and Isotope Technology, Navi Mumbai, Maharashtra, India
| | - Avik Chakraborty
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Maharashtra, Mumbai, India
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Gaurav Malhotra
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Maharashtra, Mumbai, India
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Kamaldeep
- Health Physics Division, Bhabha Atomic Research Centre, Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Megha Tawate
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Maharashtra, Mumbai, India
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sangita Lad
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Maharashtra, Mumbai, India
| | - Sutapa Rakshit
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Maharashtra, Mumbai, India
| | - Trupti Upadhye
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Maharashtra, Mumbai, India
| | - Mukti Kanta Ray
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Maharashtra, Mumbai, India
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sharmila Banerjee
- Homi Bhabha National Institute, Mumbai, Maharashtra, India.
- Radiological Research Unit, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra, 410210, India.
| |
Collapse
|
14
|
Cong T, Morse KW, Sosa BR, Lane JM, Rodeo SA, Greenblatt MB. Skeletal Stem Cells: A Basis for Orthopaedic Pathology and Tissue Repair. J Bone Joint Surg Am 2025; 107:418-426. [PMID: 39693451 PMCID: PMC11839314 DOI: 10.2106/jbjs.24.00905] [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] [Indexed: 12/20/2024]
Abstract
➢ Skeletal stem cells (SSCs) continually replenish mature cell populations to support skeletal homeostasis.➢ SSCs repopulate by self-renewal, have multilineage potential, and are long-lived in vivo.➢ SSCs express specific combinations of cell surface markers that reflect their lineage identity.➢ SSCs adapt to their anatomic environment to support regional differences in skeletal behavior and pathology.
Collapse
Affiliation(s)
- Ting Cong
- Department of Orthopaedic Surgery, UPMC Sports Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Department of Orthopedic Surgery, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Kyle W Morse
- Hospital for Special Surgery, New York, NY
- Department of Orthopaedic Surgery, Weill Cornell Medicine, New York, NY
| | - Branden R Sosa
- Hospital for Special Surgery, New York, NY
- Department of Orthopaedic Surgery, Weill Cornell Medicine, New York, NY
| | - Joseph M Lane
- Hospital for Special Surgery, New York, NY
- Department of Orthopaedic Surgery, Weill Cornell Medicine, New York, NY
| | - Scott A Rodeo
- Hospital for Special Surgery, New York, NY
- Department of Orthopaedic Surgery, Weill Cornell Medicine, New York, NY
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
- Research Division, Hospital for Special Surgery, New York, NY
| |
Collapse
|
15
|
Duan L, Cao S, Zhao F, Du X, Gao Z, Wang X, Bian F. Effects of FAP+ fibroblasts on cell proliferation migration and immunoregulation of esophageal squamous carcinoma cells through the CXCL12/CXCR4 axis. Mol Cell Biochem 2025:10.1007/s11010-025-05226-x. [PMID: 39934460 DOI: 10.1007/s11010-025-05226-x] [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: 10/25/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Cancer-associated fibroblasts (CAFs) secrete and synthesize fibroblast activation protein (FAP), which could promote proliferation and immunosuppression of multiple cancers including esophageal squamous cell carcinoma (ESCC). CXCL12/CXCR4 signaling could be revitalized by CAFs in cancer cells. Nevertheless, the significance of this interaction in ESCC has yet to be elucidated. Herein, we investigated whether FAP+ CAF cells could promote ESCC cells proliferation, migration and regulate immunity through the CXCL12/CXCR4 pathway in vitro and in vivo. The protein expression level of FSP1, FAP, CD8+ and Ki-67 in different sample was estimated by IHC and western blot. qPCR was used to quantify the mRNA level of FSP1, FAP, CD8+ and Ki-67 in different sample. The cell viability, proliferation, migration and invasion of different sample were evaluated by CCK-8, EdU staining, wound healing assay and Transwell assay, respectively. The ELISA was carried out to measure the protein level of IFN-γ, TNF-α, GZMB and IL-2. ESCC xenograft mice model was established to assess the impact of FAP+ CAF. FSP1, FAP, CD8+ and Ki-67 are greatly up-regulated in hESCC tissues. Through CXCL12/CXCR4 axis, FAP-positive CAF was capable of promoting the cell proliferation, migration and invasion of ESCC tumor cells and preventing the CD8+ T cells from secreting cytokine. Blocking this signaling with selective CXCR4 antagonist could counteract the effects caused by high-expression of FAP. FAP+ CAFs could inhibit the occurrence and development of tumors. These results indicated that FAP-positive CAF have an impact on cell proliferation migration and immunoregulation of ESCC through the CXCL12/CXCR4 axis.
Collapse
Affiliation(s)
- Lijuan Duan
- Central Laboratory, Anyang Cancer Hospital, No.1, Huanbin North Road, Beiguan District, Anyang, 455000, Henan Province, People's Republic of China.
- Henan Provincial Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455000, Henan Province, People's Republic of China.
| | - Shasha Cao
- Central Laboratory, Anyang Cancer Hospital, No.1, Huanbin North Road, Beiguan District, Anyang, 455000, Henan Province, People's Republic of China
- Henan Provincial Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455000, Henan Province, People's Republic of China
| | - Fang Zhao
- Central Laboratory, Anyang Cancer Hospital, No.1, Huanbin North Road, Beiguan District, Anyang, 455000, Henan Province, People's Republic of China
- Henan Provincial Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455000, Henan Province, People's Republic of China
| | - Xianjuan Du
- Department of Pathology, Anyang Cancer Hospital, Anyang, 455000, Henan Province, People's Republic of China
| | - Zhaowei Gao
- Central Laboratory, Anyang Cancer Hospital, No.1, Huanbin North Road, Beiguan District, Anyang, 455000, Henan Province, People's Republic of China
- Henan Provincial Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455000, Henan Province, People's Republic of China
| | - Xiaoxiao Wang
- Central Laboratory, Anyang Cancer Hospital, No.1, Huanbin North Road, Beiguan District, Anyang, 455000, Henan Province, People's Republic of China
| | - Fang Bian
- Department of Pathology, Anyang Cancer Hospital, Anyang, 455000, Henan Province, People's Republic of China
| |
Collapse
|
16
|
Saotome K, McGoldrick LL, Ho JH, Ramlall TF, Shah S, Moore MJ, Kim JH, Leidich R, Olson WC, Franklin MC. Structural insights into CXCR4 modulation and oligomerization. Nat Struct Mol Biol 2025; 32:315-325. [PMID: 39313635 PMCID: PMC11832422 DOI: 10.1038/s41594-024-01397-1] [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: 09/12/2023] [Accepted: 08/28/2024] [Indexed: 09/25/2024]
Abstract
Activation of the chemokine receptor CXCR4 by its chemokine ligand CXCL12 regulates diverse cellular processes. Previously reported crystal structures of CXCR4 revealed the architecture of an inactive, homodimeric receptor. However, many structural aspects of CXCR4 remain poorly understood. Here, we use cryo-electron microscopy to investigate various modes of human CXCR4 regulation. CXCL12 activates CXCR4 by inserting its N terminus deep into the CXCR4 orthosteric pocket. The binding of US Food and Drug Administration-approved antagonist AMD3100 is stabilized by electrostatic interactions with acidic residues in the seven-transmembrane-helix bundle. A potent antibody blocker, REGN7663, binds across the extracellular face of CXCR4 and inserts its complementarity-determining region H3 loop into the orthosteric pocket. Trimeric and tetrameric structures of CXCR4 reveal modes of G-protein-coupled receptor oligomerization. We show that CXCR4 adopts distinct subunit conformations in trimeric and tetrameric assemblies, highlighting how oligomerization could allosterically regulate chemokine receptor function.
Collapse
Affiliation(s)
- Kei Saotome
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA.
| | | | - Jo-Hao Ho
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | - Sweta Shah
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | - Jee Hae Kim
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | | | | |
Collapse
|
17
|
Hu H, He R, Liu M, Zhou H, Tan L, Ai Q, Wang Q, Zeng L, Qu W. C-X-C Motif Chemokine 12 Was Identified as a Potential Gene Target in the Treatment of Crohn's Disease. Int J Gen Med 2024; 17:6219-6228. [PMID: 39703796 PMCID: PMC11656194 DOI: 10.2147/ijgm.s487505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024] Open
Abstract
Object The present study aimed to identify hub genes associated with the treatment and control of active and inactive Crohn's disease (CD). Methods Differentially expressed genes (DEGs) were identified in normal, active CD, and inactive CD samples from GSE95095 dataset. Intersection genes screened by Venn diagram in DEGs. Subsequently, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were conducted on the intersection genes. The protein-protein interaction (PPI) network was used to screen of hub gene. The expression and mRNA levels of CXCL12 in CD and ROC curves in GSE95095 dataset. Signaling pathways of hub genes and their correlation with immune cells were analyzed by gene set enrichment analysis (GSEA), EPIC, and ESTIMATE, respectively. Finally, immunohistochemistry (IHC) and Reverse Transcription-Polymerase Chain Reaction (RT-PCR) were used to detect the expression of the hub gene in normal, inactive, and active CD tissues. Results In GSE95095 dataset, CXCL12 was identified as the most hub gene by limma analysis, Venn diagram and A protein-protein interaction (PPI) network. CXCL12 expression was highest in active CD (p < 0.001) followed by inactive CD (p < 0.01). Subsequently, it was validated through IHC and RT-PCR in normal intestinal mucosal, active CD, and inactive CD. CXCL12 was overexpressed in active and inactive CD (IHC: p < 0.001 and RT-PCR: p < 0.001, respectively). CXCL12 expression in active CD was determined via analysis with receiver operating characteristic (ROC) curves. The specificity and sensitivity were 0.875 and 0.625, respectively, the accuracy was 72.92%, the area under the curve (AUC) was 0.780, and the 95% confidence interval (CI) was in the range of 0.648-0.912. CXCL12 expression was closely correlated with various immune cells. Conclusion CXCL12 is overexpressed in active CD and is closely correlated with various immune cells. We propose that CXCL12 as a potential target genes for the treatment and management of both active and inactive CD.
Collapse
Affiliation(s)
- Hongsai Hu
- Department of Gastroenterology, Zhuzhou Hospital Affiliated to Xiangya Medical College, Central South University, Zhuzhou, 412000, People’s Republic of China
| | - Rong He
- Department of Gastroenterology, Zhuzhou Hospital Affiliated to Xiangya Medical College, Central South University, Zhuzhou, 412000, People’s Republic of China
| | - Minji Liu
- Department of Gastroenterology, Zhuzhou Hospital Affiliated to Xiangya Medical College, Central South University, Zhuzhou, 412000, People’s Republic of China
| | - Hongbing Zhou
- Department of Gastroenterology, Zhuzhou Hospital Affiliated to Xiangya Medical College, Central South University, Zhuzhou, 412000, People’s Republic of China
| | - Lin Tan
- Department of Gastroenterology, Zhuzhou Hospital Affiliated to Xiangya Medical College, Central South University, Zhuzhou, 412000, People’s Republic of China
| | - Qiongjia Ai
- Department of Gastroenterology, Zhuzhou Hospital Affiliated to Xiangya Medical College, Central South University, Zhuzhou, 412000, People’s Republic of China
| | - Qian Wang
- Department of Gastroenterology, Zhuzhou Hospital Affiliated to Xiangya Medical College, Central South University, Zhuzhou, 412000, People’s Republic of China
| | - Luwei Zeng
- Department of Gastroenterology, Zhuzhou Hospital Affiliated to Xiangya Medical College, Central South University, Zhuzhou, 412000, People’s Republic of China
| | - Weiming Qu
- Department of Gastroenterology, Zhuzhou Hospital Affiliated to Xiangya Medical College, Central South University, Zhuzhou, 412000, People’s Republic of China
| |
Collapse
|
18
|
Portale F, Carriero R, Iovino M, Kunderfranco P, Pandini M, Marelli G, Morina N, Lazzeri M, Casale P, Colombo P, De Simone G, Camisaschi C, Lugli E, Basso G, Cibella J, Marchini S, Bordi M, Meregalli G, Garbin A, Dambra M, Magrini E, Rackwitz W, Cecconi F, Corbelli A, Fiordaliso F, Eitler J, Tonn T, Di Mitri D. C/EBPβ-dependent autophagy inhibition hinders NK cell function in cancer. Nat Commun 2024; 15:10343. [PMID: 39609420 PMCID: PMC11604937 DOI: 10.1038/s41467-024-54355-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 11/05/2024] [Indexed: 11/30/2024] Open
Abstract
NK cells are endowed with tumor killing ability, nevertheless most cancers impair NK cell functionality, and cell-based therapies have limited efficacy in solid tumors. How cancers render NK cell dysfunctional is unclear, and overcoming resistance is an important immune-therapeutic aim. Here, we identify autophagy as a central regulator of NK cell anti-tumor function. Analysis of differentially expressed genes in tumor-infiltrating versus non-tumor NK cells from our previously published scRNA-seq data of advanced human prostate cancer shows deregulation of the autophagic pathway in tumor-infiltrating NK cells. We confirm this by flow cytometry in patients and in diverse cancer models in mice. We further demonstrate that exposure of NK cells to cancer deregulates the autophagic process, decreases mitochondrial polarization and impairs effector functions. Mechanistically, CCAAT enhancer binding protein beta (C/EBPβ), downstream of CXCL12-CXCR4 interaction, acts as regulator of NK cell metabolism. Accordingly, inhibition of CXCR4 and C/EBPβ restores NK cell fitness. Finally, genetic and pharmacological activation of autophagy improves NK cell effector and cytotoxic functions, which enables tumour control by NK and CAR-NK cells. In conclusion, our study identifies autophagy as an intracellular checkpoint in NK cells and introduces autophagy regulation as an approach to strengthen NK-cell-based immunotherapies.
Collapse
Affiliation(s)
- Federica Portale
- IRCCS Humanitas Research Hospital, Tumor Microenviroment Unit, 20089, Via Manzoni 56, Rozzano, Milan, Italy
| | - Roberta Carriero
- IRCCS Humanitas Research Hospital, Bioinformatics Unit, 20089, Via Manzoni 56, Rozzano, Milan, Italy
| | - Marta Iovino
- IRCCS Humanitas Research Hospital, Tumor Microenviroment Unit, 20089, Via Manzoni 56, Rozzano, Milan, Italy
| | - Paolo Kunderfranco
- IRCCS Humanitas Research Hospital, Bioinformatics Unit, 20089, Via Manzoni 56, Rozzano, Milan, Italy
| | - Marta Pandini
- IRCCS Humanitas Research Hospital, Tumor Microenviroment Unit, 20089, Via Manzoni 56, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, 20072, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan, Italy
| | - Giulia Marelli
- IRCCS Humanitas Research Hospital, Tumor Microenviroment Unit, 20089, Via Manzoni 56, Rozzano, Milan, Italy
| | - Nicolò Morina
- IRCCS Humanitas Research Hospital, Tumor Microenviroment Unit, 20089, Via Manzoni 56, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, 20072, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan, Italy
| | - Massimo Lazzeri
- IRCCS Humanitas Research Hospital, Urology Unit, 20089, Via Manzoni 56, Rozzano, Milan, Italy
| | - Paolo Casale
- IRCCS Humanitas Research Hospital, Urology Unit, 20089, Via Manzoni 56, Rozzano, Milan, Italy
| | - Piergiuseppe Colombo
- IRCCS Humanitas Research Hospital, Department of Pathology, 20089, Via Manzoni 56, Rozzano, Milan, Italy
| | - Gabriele De Simone
- IRCCS Humanitas Research Hospital, Flow Cytometry Core, 20089, Via Manzoni 56, Rozzano, Milan, Italy
| | - Chiara Camisaschi
- IRCCS Humanitas Research Hospital, Flow Cytometry Core, 20089, Via Manzoni 56, Rozzano, Milan, Italy
| | - Enrico Lugli
- IRCCS Humanitas Research Hospital, Flow Cytometry Core, 20089, Via Manzoni 56, Rozzano, Milan, Italy
| | - Gianluca Basso
- IRCCS Humanitas Research Hospital, Genomics Unit, 20089, Via Manzoni 56, Rozzano, Milan, Italy
| | - Javier Cibella
- IRCCS Humanitas Research Hospital, Genomics Unit, 20089, Via Manzoni 56, Rozzano, Milan, Italy
| | - Sergio Marchini
- IRCCS Humanitas Research Hospital, Genomics Unit, 20089, Via Manzoni 56, Rozzano, Milan, Italy
| | - Matteo Bordi
- Department of Basic Biological science, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Greta Meregalli
- IRCCS Humanitas Research Hospital, Tumor Microenviroment Unit, 20089, Via Manzoni 56, Rozzano, Milan, Italy
| | - Anna Garbin
- IRCCS Humanitas Research Hospital, Tumor Microenviroment Unit, 20089, Via Manzoni 56, Rozzano, Milan, Italy
| | - Monica Dambra
- IRCCS Humanitas Research Hospital, Immunopathology Lab, 20089, Via Manzoni 56, Rozzano, Milan, Italy
| | - Elena Magrini
- IRCCS Humanitas Research Hospital, Immunopathology Lab, 20089, Via Manzoni 56, Rozzano, Milan, Italy
| | - Wiebke Rackwitz
- Experimental Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Francesco Cecconi
- Department of Basic Biological science, Università Cattolica del Sacro Cuore, Rome, Italy
- IRCCS, Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Alessandro Corbelli
- Unit of Bio-imaging, Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Fabio Fiordaliso
- Unit of Bio-imaging, Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Jiri Eitler
- Experimental Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
| | - Torsten Tonn
- Experimental Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
| | - Diletta Di Mitri
- IRCCS Humanitas Research Hospital, Tumor Microenviroment Unit, 20089, Via Manzoni 56, Rozzano, Milan, Italy.
- Department of Biomedical Sciences, Humanitas University, 20072, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan, Italy.
| |
Collapse
|
19
|
Roustaei H, Vosoughi H, Askari E, Aziz Kalantari B, Norouzbeigi N, Anvari K, Beheshti M, Aryana K. [ 68 Ga]Ga-CXCR4 PET/CT imaging in high-grade glioma for assessment of CXCR4 receptor expression. Eur J Radiol 2024; 180:111694. [PMID: 39213763 DOI: 10.1016/j.ejrad.2024.111694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE Gliomas account for 75 % of primary malignant CNS tumors. High-grade glioma (CNS WHO grades 3 and 4) have an unfavorable treatment response and poor outcome. CXCR4 is a G protein-coupled receptor that plays an important part in the signaling pathway between cancer cells and tumor microenvironment. CXCR4 overexpression has been shown in a variety of cancers. In this study, we evaluate the potential value of [68Ga]Ga-Pentixafor as a PET/CT CXCR4-probe for in vivo assessment of CXCR4 expression in patients with high-grade glioma and its correlation with tumor grade. MATERIALS AND METHODS [68Ga]Ga-CXCR4 PET/CT was performed in the prospective single-center study in treatment-naïve biopsy-proven patients with high-grade glioma. The acquired images were analyzed qualitatively and semi-quantitatively. RESULT A total of 26 patients (mean age: 53.3±14.4 years, 11 women, 15 men) were enrolled. CNS WHO grade 3 pathology was seen in 19 % (5/26) of the sample. The patient-based sensitivity of 68Ga-CXCR4 was 96.2 %. Overall, 28 pathologic lesions were detected, leading to a lesion-based sensitivity of 96.4 %. The median (IQR) SUVmax of grade 4 lesions was substantially greater than the grade 3(3.03(2.5-3.7) vs. 1.51(1.2-1.8), p = 0.0145).). The highest tracer activity of organs -beside bladder as the main excretion reservoir-was in lymphoid tissue of Waldeyer's ring (mean SUVmax: 7.41), and spleen (mean SUVmax: 6.62). CONCLUSION In conclusion, this new application for [68Ga]Ga-Pentixafor PET tracer exhibits excellent visual and semi-quantitative diagnostic properties. Further studies are warranted.
Collapse
Affiliation(s)
- Hessamoddin Roustaei
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Habibeh Vosoughi
- Nuclear Medicine Department, Razavi Hospital, Imam Reza International University, Mashhad, Iran
| | - Emran Askari
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Nasim Norouzbeigi
- Nuclear Medicine Department, Razavi Hospital, Imam Reza International University, Mashhad, Iran
| | - Kazem Anvari
- Cancer Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Beheshti
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Kamran Aryana
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
20
|
Dong L, Hu S, Li X, Pei S, Jin L, Zhang L, Chen X, Min A, Yin M. SPP1 + TAM Regulates the Metastatic Colonization of CXCR4 + Metastasis-Associated Tumor Cells by Remodeling the Lymph Node Microenvironment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400524. [PMID: 39236316 PMCID: PMC11600252 DOI: 10.1002/advs.202400524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 05/06/2024] [Indexed: 09/07/2024]
Abstract
Lymph node metastasis, the initial step in distant metastasis, represents a primary contributor to mortality in patients diagnosed with oral squamous cell carcinoma (OSCC). However, the underlying mechanisms of lymph node metastasis in OSCC remain incompletely understood. Here, the transcriptomes of 56 383 single cells derived from paired tissues of six OSCC patients are analyzed. This study founds that CXCR4+ epithelial cells, identified as highly malignant disseminated tumor cells (DTCs), exhibited a propensity for lymph node metastasis. Importantly, a distinct subset of tumor-associated macrophages (TAMs) characterized by exclusive expression of phosphoprotein 1 (SPP1) is discovered. These TAMs may remodel the metastatic lymph node microenvironment by potentially activating fibroblasts and promoting T cell exhaustion through SPP1-CD44 and CD155-CD226 ligand-receptor interactions, thereby facilitating colonization and proliferation of disseminated tumor cells. The research advanced the mechanistic understanding of metastatic tumor microenvironment (TME) and provided a foundation for the development of personalized treatments for OSCC patients with metastasis.
Collapse
Affiliation(s)
- Liang Dong
- Department of DermatologyHunan Engineering Research Center of Skin Health and DiseaseHunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalCentral South UniversityChangshaHunan410008China
- Clinical Research Center (CRC)Medical Pathology Center (MPC)Cancer Early Detection and Treatment Center (CEDTC)Chongqing University Three Gorges HospitalChongqing UniversityChongqing404100China
- Translational Medicine Research Center (TMRC)School of Medicine Chongqing UniversityChongqing404100China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Shujun Hu
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
- Department of Oral and Maxillofacial SurgeryCenter of StomatologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- Research Center of Oral and Maxillofacail TumorXiangya HospitalCentral South UniversityChangshaHunan410008China
- Insititute of Oral Cancer and Precancerous LesionsCentral South UniversityChangshaHunan410008China
| | - Xin Li
- Clinical Research Center (CRC)Medical Pathology Center (MPC)Cancer Early Detection and Treatment Center (CEDTC)Chongqing University Three Gorges HospitalChongqing UniversityChongqing404100China
- Translational Medicine Research Center (TMRC)School of Medicine Chongqing UniversityChongqing404100China
| | - Shiyao Pei
- Department of DermatologyHunan Engineering Research Center of Skin Health and DiseaseHunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
- Department of DermatologyThird Xiangya HospitalCentral South UniversityChangsha410008China
| | - Liping Jin
- Department of DermatologyHunan Engineering Research Center of Skin Health and DiseaseHunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Lining Zhang
- Clinical Research Center (CRC)Medical Pathology Center (MPC)Cancer Early Detection and Treatment Center (CEDTC)Chongqing University Three Gorges HospitalChongqing UniversityChongqing404100China
- Translational Medicine Research Center (TMRC)School of Medicine Chongqing UniversityChongqing404100China
| | - Xiang Chen
- Department of DermatologyHunan Engineering Research Center of Skin Health and DiseaseHunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Anjie Min
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
- Department of Oral and Maxillofacial SurgeryCenter of StomatologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- Research Center of Oral and Maxillofacail TumorXiangya HospitalCentral South UniversityChangshaHunan410008China
- Insititute of Oral Cancer and Precancerous LesionsCentral South UniversityChangshaHunan410008China
| | - Mingzhu Yin
- Department of DermatologyHunan Engineering Research Center of Skin Health and DiseaseHunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalCentral South UniversityChangshaHunan410008China
- Clinical Research Center (CRC)Medical Pathology Center (MPC)Cancer Early Detection and Treatment Center (CEDTC)Chongqing University Three Gorges HospitalChongqing UniversityChongqing404100China
- Translational Medicine Research Center (TMRC)School of Medicine Chongqing UniversityChongqing404100China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| |
Collapse
|
21
|
Osborne OM, Daftari M, Naranjo O, Johar AN, Brooks S, Colbert BM, Torices S, Lewis E, Sendaydiego J, Drexler G, Bashti M, Margetts AV, Tuesta LM, Mason C, Bilbao D, Vontell R, Griswold AJ, Dykxhoorn DM, Toborek M. Post-stroke hippocampal neurogenesis is impaired by microvascular dysfunction and PI3K signaling in cerebral amyloid angiopathy. Cell Rep 2024; 43:114848. [PMID: 39392753 PMCID: PMC11562893 DOI: 10.1016/j.celrep.2024.114848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/05/2024] [Accepted: 09/23/2024] [Indexed: 10/13/2024] Open
Abstract
Ischemic stroke and cerebral amyloid angiopathy (CAA) pose significant challenges in an aging population, particularly in post-stroke recovery. Using the 5xFAD mouse model, we explore the relationship between CAA, ischemic stroke, and tissue recovery. We hypothesize that amyloid-beta accumulation worsens stroke outcomes by inducing blood-brain barrier (BBB) dysfunction, leading to impaired neurogenesis. Our findings show that CAA exacerbates stroke outcomes, with mice exhibiting constricted BBB microvessels, reduced cerebral blood flow, and impaired tissue recovery. Transcriptional analysis shows that endothelial cells and neural progenitor cells (NPCs) in the hippocampus exhibit differential gene expression in response to CAA and stroke, specifically targeting the phosphatidylinositol 3-kinase (PI3K) pathway. In vitro experiments with human NPCs validate these findings, showing that disruption of the CXCL12-PIK3C2A-CREB3L2 axis impairs neurogenesis. Notably, PI3K pathway activation restores neurogenesis, highlighting a potential therapeutic approach. These results suggest that CAA combined with stroke induces microvascular dysfunction and aberrant neurogenesis through this specific pathway.
Collapse
Affiliation(s)
- Olivia M Osborne
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Manav Daftari
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Oandy Naranjo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Adarsh N Johar
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Samantha Brooks
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Brett M Colbert
- Medical Scientist Training Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Elizabeth Lewis
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jet Sendaydiego
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Gillian Drexler
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Malek Bashti
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alexander V Margetts
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Luis M Tuesta
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Neurology and Evelyn F. McKnight Brain Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Christian Mason
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Daniel Bilbao
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Regina Vontell
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, USA; Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Anthony J Griswold
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA; John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Derek M Dykxhoorn
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA; John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
22
|
Liao S, Deng J, Deng M, Chen C, Han F, Ye K, Wu C, Pan L, Lai M, Tang Z, Zhang H. AFDN Deficiency Promotes Liver Tropism of Metastatic Colorectal Cancer. Cancer Res 2024; 84:3158-3172. [PMID: 39047222 DOI: 10.1158/0008-5472.can-23-3140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 05/09/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024]
Abstract
Liver metastasis is a major cause of morbidity and mortality in patients with colorectal cancer. A better understanding of the biological mechanisms underlying liver tropism and metastasis in colorectal cancer could help to identify improved prevention and treatment strategies. In this study, we performed genome-wide CRISPR loss-of-function screening in a mouse colorectal cancer model and identified deficiency of AFDN, a protein involved in establishing and maintaining cell-cell contacts, as a driver of liver metastasis. Elevated AFDN expression was correlated with prolonged survival in patients with colorectal cancer. AFDN-deficient colorectal cancer cells preferentially metastasized to the liver but not in the lungs. AFDN loss in colorectal cancer cells at the primary site promoted cancer cell migration and invasion by disrupting tight intercellular junctions. Additionally, CXCR4 expression was increased in AFDN-deficient colorectal cancer cells via the JAK-STAT signaling pathway, which reduced the motility of AFDN-deficient colorectal cancer cells and facilitated their colonization of the liver. Collectively, these data shed light on the mechanism by which AFDN deficiency promotes liver tropism in metastatic colorectal cancer. Significance: A CRISPR screen reveals AFDN loss as a mediator of liver tropism in colorectal cancer metastasis by decreasing tight junctions in the primary tumor and increasing interactions between cancer cells and hepatocytes.
Collapse
Affiliation(s)
- Shaoxia Liao
- Department of Pathology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences (2019RU042), Hangzhou, China
| | - Jingwen Deng
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengli Deng
- Department of Pathology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Chaoyi Chen
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Fengyan Han
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Kehong Ye
- Department of Pathology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Chenxia Wu
- Department of Pathology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Lvyuan Pan
- Department of Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, China
| | - Maode Lai
- Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences (2019RU042), Hangzhou, China
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhe Tang
- Department of Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, China
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Honghe Zhang
- Department of Pathology and International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences (2019RU042), Hangzhou, China
- Department of Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, China
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
23
|
Li X, Ohler ZW, Day A, Bassel L, Grosskopf A, Afsari B, Tagawa T, Custer W, Mangusan R, Lurain K, Yarchoan R, Ziegelbauer J, Ramaswami R, Krug LT. Mapping herpesvirus-driven impacts on the cellular milieu and transcriptional profile of Kaposi sarcoma in patient-derived mouse models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.27.615429. [PMID: 39386738 PMCID: PMC11463583 DOI: 10.1101/2024.09.27.615429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Kaposi sarcoma (KS) is defined by aberrant angiogenesis driven by Kaposi sarcoma herpesvirus (KSHV)-infected spindle cells with endothelial characteristics. KS research is hindered by rapid loss of KSHV infection upon explant culture of tumor cells. Here, we establish patient-derived KS xenografts (PDXs) upon orthotopic implantation of cutaneous KS biopsies in immunodeficient mice. KS tumors were maintained in 27/28 PDX until experimental endpoint, up to 272 days in the first passage of recipient mice. KSHV latency associated nuclear antigen (LANA)+ endothelial cell density increased by a mean 4.3-fold in 14/15 PDX analyzed by IHC at passage 1 compared to respective input biopsies, regardless of implantation variables and clinical features of patients. The Ki-67 proliferation marker colocalized with LANA more frequently in PDXs. Spatial transcriptome analysis revealed increased expression of viral transcripts from latent and lytic gene classes in the PDX. The expanded KSHV+ regions of the PDX maintained signature gene expression of KS tumors, with enrichment in pathways associated with angiogenesis and endothelium development. Cells with characteristics of tumor-associated fibroblasts derived from PDX were propagated for 15 passages. These fibroblast-like cells were permissive for de novo KSHV infection, and one lineage produced CXCL12, a cancer-promoting chemokine. Spatial analysis revealed that fibroblasts are a likely source of CXCL12 signaling to CXCR4 that was upregulated in KS regions. The reproducible expansion of KSHV-infected endothelial cells in PDX from multiple donors and recapitulation of a KS tumor gene signature supports the application of patient-derived KS mouse models for studies of pathogenesis and novel therapies.
Collapse
Affiliation(s)
- Xiaofan Li
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| | - Zoë Weaver Ohler
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute; Frederick, MD
| | - Amanda Day
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute; Frederick, MD
| | - Laura Bassel
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute; Frederick, MD
| | - Anna Grosskopf
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| | - Bahman Afsari
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| | - Takanobu Tagawa
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| | - Wendi Custer
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute; Frederick, MD
| | - Ralph Mangusan
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| | - Kathryn Lurain
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| | - Robert Yarchoan
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| | - Joseph Ziegelbauer
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| | - Ramya Ramaswami
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| | - Laurie T. Krug
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute; Bethesda, MD
| |
Collapse
|
24
|
Ocak B, Sahin AB, Ertürk I, Korkmaz M, Erdem D, Cakıroglu U, Karaca M, Dirican A, Olmez OF, Goktas Aydın S, Gökyer A, Kücükarda A, Gülmez A, Yumuk PF, Demircan NC, Oyman A, Sakalar T, Karatas F, Demir H, Yasin AI, Deligonul A, Dakiki B, Goktug MR, Avcı O, Tacar SY, Turhal NS, Deniz GI, Kacan T, Cubukcu E, Evrensel T. Can Cytoreductive Nephrectomy Improve Outcomes of Nivolumab Treatment in Patients with Metastatic Clear-Cell Renal Carcinoma? Curr Oncol 2024; 31:5195-5205. [PMID: 39330012 PMCID: PMC11431784 DOI: 10.3390/curroncol31090384] [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: 08/04/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
Background: This study aimed to investigate the effect of cytoreductive nephrectomy (CN) on the survival outcomes of nivolumab used as a subsequent therapy after the failure of at least one anti-vascular endothelial growth factor (VEGF) agent in patients with metastatic clear-cell renal-cell carcinoma (ccRCC). Methods: We included 106 de novo metastatic ccRCC patients who received nivolumab after progression on at least one anti-VEGF agent. Multivariate Cox regression analysis was performed to investigate the factors affecting survival in patients receiving nivolumab. Results: Of the 106 de novo metastatic ccRCC patients, 83 (78.3%) underwent CN. There were no statistical differences between the two groups in terms of age, gender, Eastern Cooperative Oncology Group (ECOG) score, tumor size, International Metastatic RCC Database Consortium (IMDC) risk group, number of previous treatment lines, first-line anti-VEGF therapy, or metastasis sites (p = 0.137, p = 0.608, p = 0.100, p = 0.376, p = 0.185, p = 0.776, p = 0.350, and p = 0.608, respectively). The patients who received nivolumab with CN had a longer time to treatment discontinuation (TTD) [14.5 months, 95% confidence interval (CI): 8.6-20.3] than did those without CN 6.7 months (95% CI: 3.9-9.5) (p = 0.001). The median overall survival (OS) was 22.7 months (95% CI: 16.1-29.4). The patients with CN had a median OS of 22.9 months (95% CI: 16.3-29.4), while those without CN had a median OS of 8.1 months (95% CI: 5.6-10.5) (p = 0.104). In the multivariate analysis, CN [hazard ratio (HR): 0.521; 95% CI: 0.297-0.916; p = 0.024] and the IMDC risk score (p = 0.011) were statistically significant factors affecting TTD; however, the IMDC risk score (p = 0.006) was the only significant factor for overall survival. Conclusions: Our study showed that the TTD of nivolumab was longer in metastatic ccRCC patients who underwent cytoreductive nephrectomy.
Collapse
Affiliation(s)
- Birol Ocak
- Department of Medical Oncology, Bursa Yuksek Ihtisas Training and Research Hospital, University of Health Sciences, Bursa 16350, Turkey;
| | - Ahmet Bilgehan Sahin
- Department of Medical Oncology, School of Medicine, Bursa Uludag University, Bursa 16059, Turkey; (A.B.S.); (A.D.); (B.D.); (M.R.G.); (E.C.); (T.E.)
| | - Ismail Ertürk
- Department of Medical Oncology, Gulhane School of Medicine, University of Health Sciences, Ankara 06018, Turkey;
| | - Mustafa Korkmaz
- Department of Medical Oncology, School of Medicine, Necmettin Erbakan University, Konya 42090, Turkey;
| | - Dilek Erdem
- Department of Medical Oncology, VM Medical Park Samsun Hospital, Samsun 55200, Turkey;
| | - Umut Cakıroglu
- Department of Medical Oncology, Van Training and Research Hospital, University of Health Sciences, Van 65300, Turkey;
| | - Mustafa Karaca
- Department of Medical Oncology, Antalya Training and Research Hospital, University of Health Sciences, Antalya 07100, Turkey;
| | - Ahmet Dirican
- Department of Medical Oncology, School of Medicine, Celal Bayar University, Manisa 45140, Turkey;
| | - Omer Fatih Olmez
- Department of Medical Oncology, Medipol University Hospital, Istanbul 34810, Turkey; (O.F.O.); (S.G.A.)
| | - Sabin Goktas Aydın
- Department of Medical Oncology, Medipol University Hospital, Istanbul 34810, Turkey; (O.F.O.); (S.G.A.)
| | - Ali Gökyer
- Department of Medical Oncology, Department of Internal Medicine, School of Medicine, Trakya University, Edirne 22130, Turkey; (A.G.); (A.K.)
| | - Ahmet Kücükarda
- Department of Medical Oncology, Department of Internal Medicine, School of Medicine, Trakya University, Edirne 22130, Turkey; (A.G.); (A.K.)
| | - Ahmet Gülmez
- Department of Medical Oncology, School of Medicine, Inonu University, Malatya 44280, Turkey;
| | - Perran Fulden Yumuk
- Department of Medical Oncology, School of Medicine, Marmara University, Istanbul 34854, Turkey; (P.F.Y.); (N.C.D.)
| | - Nazim Can Demircan
- Department of Medical Oncology, School of Medicine, Marmara University, Istanbul 34854, Turkey; (P.F.Y.); (N.C.D.)
| | - Abdilkerim Oyman
- Department of Medical Oncology, Umraniye Training and Research Hospital, University of Health Sciences, Istanbul 34764, Turkey;
| | - Teoman Sakalar
- Department of Medical Oncology, Necip Fazıl City Hospital, Kahramanmaraş 46050, Turkey
| | - Fatih Karatas
- Department of Medical Oncology, Faculty of Medicine, Karabuk University, Karabuk 78100, Turkey;
| | - Hacer Demir
- Department of Medical Oncology, Afyonkarahisar Health Sciences University, Afyon 03030, Turkey;
| | - Ayse Irem Yasin
- Department of Medical Oncology, Faculty of Medicine, Bezmialem Vakif University, Istanbul 34093, Turkey;
| | - Adem Deligonul
- Department of Medical Oncology, School of Medicine, Bursa Uludag University, Bursa 16059, Turkey; (A.B.S.); (A.D.); (B.D.); (M.R.G.); (E.C.); (T.E.)
| | - Bahar Dakiki
- Department of Medical Oncology, School of Medicine, Bursa Uludag University, Bursa 16059, Turkey; (A.B.S.); (A.D.); (B.D.); (M.R.G.); (E.C.); (T.E.)
| | - Mehmet Refik Goktug
- Department of Medical Oncology, School of Medicine, Bursa Uludag University, Bursa 16059, Turkey; (A.B.S.); (A.D.); (B.D.); (M.R.G.); (E.C.); (T.E.)
| | - Okan Avcı
- Department of Medical Oncology, Tekirdağ Namık Kemal University, Tekirdağ 34093, Turkey; (O.A.); (S.Y.T.)
| | - Seher Yildiz Tacar
- Department of Medical Oncology, Tekirdağ Namık Kemal University, Tekirdağ 34093, Turkey; (O.A.); (S.Y.T.)
| | - Nazım Serdar Turhal
- Department of Medical Oncology, Anadolu Health Center, Kocaeli 2255, Turkey;
| | - Gülhan Ipek Deniz
- Department of Medical Oncology, Sisli Hamidiye Etfal Training and Research Hospital, University of Health Sciences, Istanbul 34371, Turkey;
| | - Turgut Kacan
- Department of Medical Oncology, Bursa Yuksek Ihtisas Training and Research Hospital, University of Health Sciences, Bursa 16350, Turkey;
| | - Erdem Cubukcu
- Department of Medical Oncology, School of Medicine, Bursa Uludag University, Bursa 16059, Turkey; (A.B.S.); (A.D.); (B.D.); (M.R.G.); (E.C.); (T.E.)
| | - Türkkan Evrensel
- Department of Medical Oncology, School of Medicine, Bursa Uludag University, Bursa 16059, Turkey; (A.B.S.); (A.D.); (B.D.); (M.R.G.); (E.C.); (T.E.)
| |
Collapse
|
25
|
Zou D, Xin X, Xu Y, Xu H, Huang L, Xu T. Improving the efficacy of immunotherapy for colorectal cancer: Targeting tumor microenvironment-associated immunosuppressive cells. Heliyon 2024; 10:e36446. [PMID: 39262952 PMCID: PMC11388603 DOI: 10.1016/j.heliyon.2024.e36446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 09/13/2024] Open
Abstract
Currently, immune checkpoint inhibitors (ICIs) have changed the treatment paradigm for many malignant tumors. As the most common digestive tract malignancy, colorectal cancer (CRC) shows a good response to ICIs only in a small subset of patients with MSI-H/dMMR CRC. In contrast, patients with MSS/pMMR CRC show minimal response to ICIs. The results of the REGONIVO study suggest that targeting the tumor microenvironment (TME) to improve immunotherapy outcomes in MSS/pMMR CRC patients is a feasible strategy. Therefore, this article focuses on exploring the feasibility of targeting the TME to enhance immunotherapy outcomes in CRC, collecting recent basic research on targeting the TME to enhance immunotherapy outcomes in CRC and analyzing ongoing clinical trials to provide a theoretical basis and future research directions for improving immunotherapy outcomes in MSS/pMMR CRC.
Collapse
Affiliation(s)
- Daoyang Zou
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Xi Xin
- Ganzhou People's Hospital, Ganzhou, 341000, China
| | - Yunxian Xu
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Huangzhen Xu
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Linyan Huang
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Tianwen Xu
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| |
Collapse
|
26
|
Drouillard D, Halyko M, Cinquegrani E, McAllister D, Peterson FC, Marchese A, Dwinell MB. CXCL12 chemokine dimer signaling modulates acute myelogenous leukemia cell migration through altered receptor internalization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.26.609725. [PMID: 39253415 PMCID: PMC11383031 DOI: 10.1101/2024.08.26.609725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Acute myeloid leukemia (AML) is a malignancy of immature myeloid blast cells with stem-like and chemoresistant cells being retained in the bone marrow through CXCL12-CXCR4 signaling. Current CXCR4 inhibitors mobilize AML cells into the bloodstream where they become more chemosensitive have failed to improve patient survival, likely reflecting persistent receptor localization on target cells. Here we characterize the signaling properties of CXCL12-locked dimer (CXCL12-LD), a bioengineered variant of the dimeric CXCL12 structure. CXCL12-LD binding resulted in lower levels of G protein, β-arrestin, and intracellular calcium mobilization, consistent with the locked dimer being a partial agonist of CXCR4. Further, CXCL12-LD failed to induce chemotaxis in AML cells. Despite these partial agonist properties, CXCL12-LD increased CXCR4 internalization compared to wildtype and locked-monomer forms of CXCL12. Analysis of a previously published AML transcriptomic data showed CXCR4 positive AML cells co-express genes involved in chemoresistance and maintenance of a blast-like state. The CXCL12-LD partial agonist effectively mobilized stem cells into the bloodstream in mice suggesting a potential role for their use in targeting CXCR4. Together, our results suggest that enhanced internalization by CXCL12-LD partial agonist signaling can avoid pharmacodynamic tolerance and may identify new avenues to better target GPCRs.
Collapse
Affiliation(s)
- Donovan Drouillard
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee WI, USA
- Center for Immunology, Medical College of Wisconsin, Milwaukee WI, USA
| | - Michael Halyko
- Center for Immunology, Medical College of Wisconsin, Milwaukee WI, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee WI, USA
| | | | - Donna McAllister
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee WI, USA
- Center for Immunology, Medical College of Wisconsin, Milwaukee WI, USA
| | | | - Adriano Marchese
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee WI, USA
| | - Michael B. Dwinell
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee WI, USA
- Center for Immunology, Medical College of Wisconsin, Milwaukee WI, USA
- Department of Surgery, Medical College of Wisconsin, Milwaukee WI, USA
| |
Collapse
|
27
|
Ahuja S, Lazar IM. Proteomic insights into breast cancer response to brain cell-secreted factors. Sci Rep 2024; 14:19351. [PMID: 39169222 PMCID: PMC11339284 DOI: 10.1038/s41598-024-70386-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024] Open
Abstract
The most devastating feature of cancer cells is their ability to metastasize to distant sites in the body. HER2 + and TN breast cancers frequently metastasize to the brain and stay potentially dormant for years until favorable conditions support their proliferation. The sheltered and delicate nature of the brain prevents, however, early disease detection and effective delivery of therapeutic drugs. Moreover, the challenges associated with the acquisition of brain biopsies add compounding difficulties to exploring the mechanistic aspects of tumor development. To provide insights into the determinants of cancer cell behavior at the brain metastatic site, this study was aimed at exploring the early response of HER2 + breast cancer cells (SKBR3) to factors present in the brain perivascular niche. The neural microenvironment was simulated by using the secretome of a set of brain cells that come first in contact with the cancer cells upon crossing the blood brain barrier, i.e., endothelial cells, astrocytes, and microglia. Cytokine microarrays were used to investigate the secretome mediators of intercellular communication, and proteomic technologies for assessing the changes in the behavior of cancer cells upon exposure to the brain cell-secreted factors. The cytokines detected in the brain secretomes were supportive of inflammatory conditions, while the SKBR3 cells secreted numerous cancer-promoting growth factors that were either absent or present in lower abundance in the brain cell cultures, indicating that upon exposure the SKBR3 cells may have been deprived of favorable conditions for optimal growth. Altogether, the results suggest that the exposure of SKBR3 cells to the brain cell-secreted factors altered their growth potential and drove them toward a state of quiescence, with broader overall outcomes that affected cellular metabolism, adhesion and immune response processes. The findings of this study underscore the key role played by the neural niche in shaping the behavior of metastasized cancer cells, provide insights into the cellular cross-talk that may lead cancer cells into dormancy, and highlight novel opportunities for the development of metastatic breast cancer therapeutic strategies.
Collapse
Affiliation(s)
- Shreya Ahuja
- Department of Biological Sciences, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA
| | - Iulia M Lazar
- Department of Biological Sciences, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA.
- Fralin Life Sciences Institute, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA.
- Carilion School of Medicine, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA.
- Division of Systems Biology/AIS, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA.
| |
Collapse
|
28
|
Anderson AN, Conley P, Klocke CD, Sengupta SK, Pang A, Farley HC, Gillingham AR, Dawson AD, Fan Y, Jones JA, Gibbs SL, Skalet AH, Wu G, Wong MH. Detection of neoplastic-immune hybrid cells with metastatic properties in uveal melanoma. Biomark Res 2024; 12:67. [PMID: 39030653 PMCID: PMC11264923 DOI: 10.1186/s40364-024-00609-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/18/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Uveal melanoma is the most common non-cutaneous melanoma and is an intraocular malignancy affecting nearly 7,000 individuals per year worldwide. Of these, approximately 50% will progress to metastatic disease for which there are currently no effective curative therapies. Despite advances in molecular profiling and metastatic stratification of uveal melanoma tumors, little is known regarding their underlying biology of metastasis. Our group has identified a disseminated neoplastic cell population characterized by co-expression of immune and melanoma proteins, circulating hybrid cells (hybrids), in patients with uveal melanoma. Compared to circulating tumor cells, which lack expression of immune proteins, hybrids are detected at an increased prevalence in peripheral blood and can be used as a non-invasive biomarker to predict metastatic progression. METHODS To ascertain mechanisms underlying enhanced hybrid cell dissemination we identified hybrid cells within primary uveal melanoma tumors using single cell RNA sequencing (n = 8) and evaluated their gene expression and predicted ligand-receptor interactions in relation to other melanoma and immune cells within the primary tumor. We then verified expression of upregulated hybrid pathways within patient-matched tumor and peripheral blood hybrids (n = 4) using cyclic immunofluorescence and quantified their protein expression relative to other non-hybrid tumor and disseminated tumor cells. RESULTS Among the top upregulated genes and pathways in hybrid cells were those involved in enhanced cell motility and cytoskeletal rearrangement, immune evasion, and altered cellular metabolism. In patient-matched tumor and peripheral blood, we verified gene expression by examining concordant protein expression for each pathway category: TMSB10 (cell motility), CD74 (immune evasion) and GPX1 (metabolism). Both TMSB10 and GPX1 were expressed on significantly higher numbers of disseminated hybrid cells compared to circulating tumor cells, and CD74 and GPX1 were expressed on more disseminated hybrids than tumor-resident hybrids. Lastly, we identified that hybrid cells express ligand-receptor signaling pathways implicated in promoting metastasis including GAS6-AXL, CXCL12-CXCR4, LGALS9-P4HB and IGF1-IGFR1. CONCLUSION These findings highlight the importance of TMSB10, GPX1 and CD74 for successful hybrid cell dissemination and survival in circulation. Our results contribute to the understanding of uveal melanoma tumor progression and interactions between tumor cells and immune cells in the tumor microenvironment that may promote metastasis.
Collapse
Affiliation(s)
- Ashley N Anderson
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Patrick Conley
- Department of Medical Informatics and Clinical Epidemiology, OHSU, Portland, OR, USA
| | - Christopher D Klocke
- Department of Medical Informatics and Clinical Epidemiology, OHSU, Portland, OR, USA
| | - Sidharth K Sengupta
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Amara Pang
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Hannah C Farley
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
- Department of Biomedical Engineering, OHSU, Portland, OR, USA
| | - Abigail R Gillingham
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Aubrey D Dawson
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Yichen Fan
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Jocelyn A Jones
- Department of Biomedical Engineering, OHSU, Portland, OR, USA
| | - Summer L Gibbs
- Department of Biomedical Engineering, OHSU, Portland, OR, USA
- Knight Cancer Institute, OHSU, Portland, OR, USA
| | - Alison H Skalet
- Casey Eye Institute, OHSU, Portland, OR, USA
- Knight Cancer Institute, OHSU, Portland, OR, USA
| | - Guanming Wu
- Department of Medical Informatics and Clinical Epidemiology, OHSU, Portland, OR, USA
- Knight Cancer Institute, OHSU, Portland, OR, USA
| | - Melissa H Wong
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA.
- Knight Cancer Institute, OHSU, Portland, OR, USA.
| |
Collapse
|
29
|
Yen JH, Chang CC, Hsu HJ, Yang CH, Mani H, Liou JW. C-X-C motif chemokine ligand 12-C-X-C chemokine receptor type 4 signaling axis in cancer and the development of chemotherapeutic molecules. Tzu Chi Med J 2024; 36:231-239. [PMID: 38993827 PMCID: PMC11236080 DOI: 10.4103/tcmj.tcmj_52_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/14/2024] [Accepted: 04/18/2024] [Indexed: 07/13/2024] Open
Abstract
Chemokines are small, secreted cytokines crucial in the regulation of a variety of cell functions. The binding of chemokine C-X-C motif chemokine ligand 12 (CXCL12) (stromal cell-derived factor 1) to a G-protein-coupled receptor C-X-C chemokine receptor type 4 (CXCR4) triggers downstream signaling pathways with effects on cell survival, proliferation, chemotaxis, migration, and gene expression. Intensive and extensive investigations have provided evidence suggesting that the CXCL12-CXCR4 axis plays a pivotal role in tumor development, survival, angiogenesis, metastasis, as well as in creating tumor microenvironment, thus implying that this axis is a potential target for the development of cancer therapies. The structures of CXCL12 and CXCR4 have been resolved with experimental methods such as X-ray crystallography, NMR, or cryo-EM. Therefore, it is possible to apply structure-based computational approaches to discover, design, and modify therapeutic molecules for cancer treatments. Here, we summarize the current understanding of the roles played by the CXCL12-CXCR4 signaling axis in cellular functions linking to cancer progression and metastasis. This review also provides an introduction to protein structures of CXCL12 and CXCR4 and the application of computer simulation and analysis in understanding CXCR4 activation and antagonist binding. Furthermore, examples of strategies and current progress in CXCL12-CXCR4 axis-targeted development of therapeutic anticancer inhibitors are discussed.
Collapse
Affiliation(s)
- Jui-Hung Yen
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Chun-Chun Chang
- Department of Laboratory Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Laboratory Medicine and Biotechnology, Tzu Chi University, Hualien, Taiwan
| | - Hao-Jen Hsu
- Department of Biomedical Sciences and Engineering, Tzu Chi University, Hualien, Taiwan
| | - Chin-Hao Yang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Hemalatha Mani
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Je-Wen Liou
- Department of Laboratory Medicine and Biotechnology, Tzu Chi University, Hualien, Taiwan
- Department of Biomedical Sciences and Engineering, Tzu Chi University, Hualien, Taiwan
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
30
|
Virgili AC, Salazar J, Gallardo A, López-Pousa A, Terés R, Bagué S, Orellana R, Fumagalli C, Mangues R, Alba-Castellón L, Unzueta U, Casanova I, Sebio A. CXCR4 Expression as a Prognostic Biomarker in Soft Tissue Sarcomas. Diagnostics (Basel) 2024; 14:1195. [PMID: 38893721 PMCID: PMC11172351 DOI: 10.3390/diagnostics14111195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/30/2024] [Accepted: 06/05/2024] [Indexed: 06/21/2024] Open
Abstract
Poor long-term survival in localized high-risk soft tissue sarcomas (STSs) of the extremities and trunk highlights the need to identify new prognostic factors. CXCR4 is a chemokine receptor involved in tumor progression, angiogenesis, and metastasis. The aim of this study was to evaluate the association between CXCR4 expression in tumor tissue and survival in STSs patients treated with neoadjuvant therapy. CXCR4 expression was retrospectively determined by immunohistochemical analysis in serial specimens including initial biopsies, tumors post-neoadjuvant treatment, and tumors after relapse. We found that a positive cytoplasmatic expression of CXCR4 in tumors after neoadjuvant treatment was a predictor of poor recurrence-free survival (RFS) (p = 0.003) and overall survival (p = 0.019) in synovial sarcomas. We also found that positive nuclear CXCR4 expression in the initial biopsies was associated with poor RFS (p = 0.022) in undifferentiated pleomorphic sarcomas. In conclusion, our study adds to the evidence that CXCR4 expression in tumor tissue is a promising prognostic factor for STSs.
Collapse
Affiliation(s)
- Anna C. Virgili
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (A.C.V.); (A.L.-P.); (R.T.)
- Department of Medicine, Faculty of Medicine, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Translational Medical Oncology Laboratory, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain
| | - Juliana Salazar
- Translational Medical Oncology Laboratory, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain
| | - Alberto Gallardo
- Department of Pathology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (A.G.); (S.B.); (R.O.); (C.F.)
| | - Antonio López-Pousa
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (A.C.V.); (A.L.-P.); (R.T.)
- Translational Medical Oncology Laboratory, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain
| | - Raúl Terés
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (A.C.V.); (A.L.-P.); (R.T.)
- Translational Medical Oncology Laboratory, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain
| | - Silvia Bagué
- Department of Pathology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (A.G.); (S.B.); (R.O.); (C.F.)
| | - Ruth Orellana
- Department of Pathology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (A.G.); (S.B.); (R.O.); (C.F.)
| | - Caterina Fumagalli
- Department of Pathology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (A.G.); (S.B.); (R.O.); (C.F.)
| | - Ramon Mangues
- Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (R.M.); (L.A.-C.); (U.U.); (I.C.)
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Cerdanyola del Vallès, 08193 Barcelona, Spain
- Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
| | - Lorena Alba-Castellón
- Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (R.M.); (L.A.-C.); (U.U.); (I.C.)
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Cerdanyola del Vallès, 08193 Barcelona, Spain
- Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
| | - Ugutz Unzueta
- Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (R.M.); (L.A.-C.); (U.U.); (I.C.)
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Cerdanyola del Vallès, 08193 Barcelona, Spain
- Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
| | - Isolda Casanova
- Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (R.M.); (L.A.-C.); (U.U.); (I.C.)
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, Cerdanyola del Vallès, 08193 Barcelona, Spain
- Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Spain
| | - Ana Sebio
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (A.C.V.); (A.L.-P.); (R.T.)
- Translational Medical Oncology Laboratory, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain
| |
Collapse
|
31
|
Deng T, Wang F, Zhang L, Ning T, Sun Y, Ge S, Bai M, Lu Y, Li H, Ba Y. Combinational zimberelimab plus lenvatinib and chemotherapy for alpha-fetoprotein elevated, advanced gastric cancer patients (AFPGC): a phase 1 dose-escalation study. Cancer Immunol Immunother 2024; 73:154. [PMID: 38833154 PMCID: PMC11150360 DOI: 10.1007/s00262-024-03743-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 05/20/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Alpha-fetoprotein elevated gastric cancer (AFPGC) got growing interests for its aggressive nature and unfavorable prognosis. Here, a phase 1 dose escalation study was conducted to evaluate safety and efficacy of zimberelimab (GLS-010, anti-PD-1) plus lenvatinib and chemotherapy (XELOX) as the first-line treatment for AFPGC. METHODS Histologically confirmed HER2-negative, advanced GC patients with elevated serum AFP level (≥ 20 ng/ml) were screened. Using a 3 + 3 dose escalation design, patients were administered varying doses of lenvatinib (12, 16, 20 mg) with GLS-010 and XELOX. The primary endpoints were safety and determination of recommended phase II dose (RP2D). Secondary endpoints included overall response rate (ORR), progression-free survival (PFS) and disease control rate. RESULTS Nine patients were enrolled with no dose-limiting toxicities observed. Most frequent treatment-related AEs were fatigue (55.6%), hand-foot syndrome (55.6%) and rash (55.6%), and no grade ≥ 4 AEs were reported. All patients exhibited disease control with ORR reaching 33.3%. The median PFS and OS reached 7.67 months (95% CI 4.07-11.27) and 13.17 months (95% CI 2.78-23.56), respectively. Serum AFP level was found correlated with therapeutic responses. Further 16s rRNA sequencing analysis demonstrated altered gut microbiota with elevated abundance of Lachnospiraceae bacterium-GAM79 and Roseburia hominis A2-183. CONCLUSIONS GLS-010 plus lenvatinib and XELOX demonstrated a manageable safety profile with promising efficacy for AFPGC. With RP2D of lenvatinib determined as 16 mg, further expansion cohort is now ongoing. Translational investigation suggested that serum AFP can be indictive for therapeutic responses and certain microbiota species indicating favorable responses to immunotherapy was elevated after the combinational treatment.
Collapse
Affiliation(s)
- Ting Deng
- Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, China
| | - Feixue Wang
- Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, China
| | - Le Zhang
- Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, China
| | - Tao Ning
- Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, China
| | - Yansha Sun
- Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, China
| | - Shaohua Ge
- Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, China
| | - Ming Bai
- Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, China
| | - Yao Lu
- Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, China
| | - Hongli Li
- Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, China
| | - Yi Ba
- Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, China.
- Department of Medical Oncology, Department of Cancer Center, Peking Union Medical College Hospital, Beijing, China.
| |
Collapse
|
32
|
Giorgiutti S, Rottura J, Korganow AS, Gies V. CXCR4: from B-cell development to B cell-mediated diseases. Life Sci Alliance 2024; 7:e202302465. [PMID: 38519141 PMCID: PMC10961644 DOI: 10.26508/lsa.202302465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 03/24/2024] Open
Abstract
Chemokine receptors are members of the G protein-coupled receptor superfamily. The C-X-C chemokine receptor type 4 (CXCR4), one of the most studied chemokine receptors, is widely expressed in hematopoietic and immune cell populations. It is involved in leukocyte trafficking in lymphoid organs and inflammatory sites through its interaction with its natural ligand CXCL12. CXCR4 assumes a pivotal role in B-cell development, ranging from early progenitors to the differentiation of antibody-secreting cells. This review emphasizes the significance of CXCR4 across the various stages of B-cell development, including central tolerance, and delves into the association between CXCR4 and B cell-mediated disorders, from immunodeficiencies such as WHIM (warts, hypogammaglobulinemia, infections, and myelokathexis) syndrome to autoimmune diseases such as systemic lupus erythematosus. The potential of CXCR4 as a therapeutic target is discussed, especially through the identification of novel molecules capable of modulating specific pockets of the CXCR4 molecule. These insights provide a basis for innovative therapeutic approaches in the field.
Collapse
Affiliation(s)
- Stéphane Giorgiutti
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, Strasbourg, France
- INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Faculty of Medicine, Université de Strasbourg, Strasbourg, France
| | - Julien Rottura
- INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Anne-Sophie Korganow
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, Strasbourg, France
- INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Faculty of Medicine, Université de Strasbourg, Strasbourg, France
| | - Vincent Gies
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Systemic Autoimmune Diseases (CNR RESO), Tertiary Center for Primary Immunodeficiency, Strasbourg University Hospital, Strasbourg, France
- INSERM UMR - S1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Faculty of Pharmacy, Université de Strasbourg, Illkirch, France
| |
Collapse
|
33
|
Takkar S, Sharma G, Kaushal JB, Abdullah KM, Batra SK, Siddiqui JA. From orphan to oncogene: The role of GPR35 in cancer and immune modulation. Cytokine Growth Factor Rev 2024; 77:56-66. [PMID: 38514303 PMCID: PMC11793123 DOI: 10.1016/j.cytogfr.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024]
Abstract
G protein-coupled receptors (GPCRs) are well-studied and the most traceable cell surface receptors for drug discovery. One of the intriguing members of this family is G protein-coupled receptors 35 (GPR35), which belongs to the class A rhodopsin-like family of GPCRs identified over two decades ago. GPR35 presents interesting features such as ubiquitous expression and distinct isoforms. Moreover, functional and genome-wide association studies on its widespread expression have linked GPR35 with pathophysiological disease progression. Various pieces of evidence have been accumulated regarding the independent or endogenous ligand-dependent role of GPR35 in cancer progression and metastasis. In the current scenario, the relationship of this versatile receptor and its putative endogenous ligands for the activation of oncogenic signal transduction pathways at the cellular level is an active area of research. These intriguing features offered by GPR35 make it an oncological target, justifying its uniqueness at the physiological and pathophysiological levels concerning other GPCRs. For pharmacologically targeting receptor-induced signaling, few potential competitive antagonists have been discovered that offer high selectivity at a human level. In addition to its fascinating features, targeting GPR35 at rodent and human orthologue levels is distinct, thus contributing to the sub-species selectivity. Strategies to modulate these issues will help us understand and truly target GPR35 at the therapeutic level. In this article, we have provided prospects on each topic mentioned above and suggestions to overcome the challenges. This review discusses the molecular mechanism and signal transduction pathways activated by endogenous ligands or spontaneous auto-activation of GPR35 that contributes towards disease progression. Furthermore, we have highlighted the GPR35 structure, ubiquitous expression, its role in immunomodulation, and at the pathophysiological level, especially in cancer, indicating its status as a versatile receptor. Subsequently, we discussed the various proposed ligands and their mechanism of interaction with GPR35. Additionally, we have summarized the GPR35 antagonist that provides insights into the opportunities for therapeutically targeting this receptor.
Collapse
Affiliation(s)
- Simran Takkar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Gunjan Sharma
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jyoti B Kaushal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - K M Abdullah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Jawed A Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
34
|
Chaudary N, Hill RP, Milosevic M. Targeting the CXCL12/CXCR4 pathway to reduce radiation treatment side effects. Radiother Oncol 2024; 194:110194. [PMID: 38447871 DOI: 10.1016/j.radonc.2024.110194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/08/2024]
Abstract
High precision, image-guided radiotherapy (RT) has increased the therapeutic ratio, enabling higher tumor and lower normal tissue doses, leading to improved patient outcomes. Nevertheless, some patients remain at risk of developing serious side effects.In many clinical situations, the radiation tolerance of normal tissues close to the target volume limits the dose that can safely be delivered and thus the potential for tumor control and cure. This is particularly so in patients being re-treated for tumor progression or a second primary tumor within a previous irradiated volume, scenarios that are becoming more frequent in clinical practice.Various normal tissue 'radioprotective' drugs with the potential to reduce side effects have been studied previously. Unfortunately, most have failed to impact clinical practice because of lack of therapeutic efficacy, concern about concurrent tumor protection or excessive drug-related toxicity. This review highlights the evidence indicating that targeting the CXCL12/CXCR4 pathway can mitigate acute and late RT-induced injury and reduce treatment side effects in a manner that overcomes these previous translational challenges. Pre-clinical studies involving a broad range of normal tissues commonly affected in clinical practice, including skin, lung, the gastrointestinal tract and brain, have shown that CXCL12 signalling is upregulated by RT and attracts CXCR4-expressing inflammatory cells that exacerbate acute tissue injury and late fibrosis. These studies also provide convincing evidence that inhibition of CXCL12/CXCR4 signalling during or after RT can reduce or prevent RT side effects, warranting further evaluation in clinical studies. Greater dialogue with the pharmaceutical industry is needed to prioritize the development and availability of CXCL12/CXCR4 inhibitors for future RT studies.
Collapse
Affiliation(s)
- Naz Chaudary
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Richard P Hill
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Michael Milosevic
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
35
|
Huang Y, Qin Y, He Y, Qiu D, Zheng Y, Wei J, Zhang L, Yang DH, Li Y. Advances in molecular targeted drugs in combination with CAR-T cell therapy for hematologic malignancies. Drug Resist Updat 2024; 74:101082. [PMID: 38569225 DOI: 10.1016/j.drup.2024.101082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/03/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
Molecular targeted drugs and chimeric antigen receptor (CAR) T cell therapy represent specific biological treatments that have significantly improved the efficacy of treating hematologic malignancies. However, they face challenges such as drug resistance and recurrence after treatment. Combining molecular targeted drugs and CAR-T cells could regulate immunity, improve tumor microenvironment (TME), promote cell apoptosis, and enhance sensitivity to tumor cell killing. This approach might provide a dual coordinated attack on cancer cells, effectively eliminating minimal residual disease and overcoming therapy resistance. Moreover, molecular targeted drugs can directly or indirectly enhance the anti-tumor effect of CAR-T cells by inducing tumor target antigen expression, reversing CAR-T cell exhaustion, and reducing CAR-T cell associated toxic side effects. Therefore, combining molecular targeted drugs with CAR-T cells is a promising and novel tactic for treating hematologic malignancies. In this review article, we focus on analyzing the mechanism of therapy resistance and its reversal of CAR-T cell therapy resistance, as well as the synergistic mechanism, safety, and future challenges in CAR-T cell therapy in combination with molecular targeted drugs. We aim to explore the benefits of this combination therapy for patients with hematologic malignancies and provide a rationale for subsequent clinical studies.
Collapse
Affiliation(s)
- Yuxian Huang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China.
| | - Yinjie Qin
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China
| | - Yingzhi He
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China
| | - Dezhi Qiu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China
| | - Yeqin Zheng
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China
| | - Jiayue Wei
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China
| | - Lenghe Zhang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China
| | - Dong-Hua Yang
- New York College of Traditional Chinese Medicine, Mineola, NY, USA.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, Guangdong, China.
| |
Collapse
|
36
|
Qian J, Huang C, Wang M, Liu Y, Zhao Y, Li M, Zhang X, Gao X, Zhang Y, Wang Y, Huang J, Li J, Zhou Q, Liu R, Wang X, Cui J, Yang Y. Nuclear translocation of metabolic enzyme PKM2 participates in high glucose-promoted HCC metastasis by strengthening immunosuppressive environment. Redox Biol 2024; 71:103103. [PMID: 38471282 PMCID: PMC10945175 DOI: 10.1016/j.redox.2024.103103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/14/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Although some cohort studies have indicated a close association between diabetes and HCC, the underlying mechanism about the contribution of diabetes to HCC progression remains largely unknown. In the study, we applied a novel HCC model in SD rat with diabetes and a series of high glucose-stimulated cell experiments to explore the effect of a high glucose environment on HCC metastasis and its relevant mechanism. Our results uncovered a novel regulatory mechanism by which nuclear translocation of metabolic enzyme PKM2 mediated high glucose-promoted HCC metastasis. Specifically, high glucose-increased PKM2 nuclear translocation downregulates chemerin expression through the redox protein TRX1, and then strengthens immunosuppressive environment to promote HCC metastasis. To the best of our knowledge, this is the first report to elucidate the great contribution of a high glucose environment to HCC metastasis from a new perspective of enhancing the immunosuppressive microenvironment. Simultaneously, this work also highlights a previously unidentified non-metabolic role of PKM2 and opens a novel avenue for cross research and intervention for individuals with HCC and comorbid diabetes.
Collapse
Affiliation(s)
- Jiali Qian
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuxin Huang
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Mimi Wang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Ying Liu
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yingying Zhao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Miao Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Xi Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Xiangyu Gao
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yawen Zhang
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi Wang
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinya Huang
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiajun Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Qiwen Zhou
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Rui Liu
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xuanchun Wang
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiefeng Cui
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China.
| | - Yehong Yang
- Department of endocrinology, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
37
|
Xu Y, Miller CP, Tykodi SS, Akilesh S, Warren EH. Signaling crosstalk between tumor endothelial cells and immune cells in the microenvironment of solid tumors. Front Cell Dev Biol 2024; 12:1387198. [PMID: 38726320 PMCID: PMC11079179 DOI: 10.3389/fcell.2024.1387198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/11/2024] [Indexed: 05/12/2024] Open
Abstract
Tumor-associated endothelial cells (TECs) are crucial mediators of immune surveillance and immune escape in the tumor microenvironment (TME). TECs driven by angiogenic growth factors form an abnormal vasculature which deploys molecular machinery to selectively promote the function and recruitment of immunosuppressive cells while simultaneously blocking the entry and function of anti-tumor immune cells. TECs also utilize a similar set of signaling regulators to promote the metastasis of tumor cells. Meanwhile, the tumor-infiltrating immune cells further induce the TEC anergy by secreting pro-angiogenic factors and prevents further immune cell penetration into the TME. Understanding the complex interactions between TECs and immune cells will be needed to successfully treat cancer patients with combined therapy to achieve vasculature normalization while augmenting antitumor immunity. In this review, we will discuss what is known about the signaling crosstalk between TECs and tumor-infiltrating immune cells to reveal insights and strategies for therapeutic targeting.
Collapse
Affiliation(s)
- Yuexin Xu
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Chris P. Miller
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Scott S. Tykodi
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Shreeram Akilesh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
- Kidney Research Institute, University of Washington, Seattle, WA, United States
| | - Edus H. Warren
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, WA, United States
| |
Collapse
|
38
|
Mizejewski GJ. The Role of Ion Channels and Chemokines in Cancer Growth and Metastasis: A Proposed Mode of Action Using Peptides in Cancer Therapy. Cancers (Basel) 2024; 16:1531. [PMID: 38672613 PMCID: PMC11048196 DOI: 10.3390/cancers16081531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Metastasis (Met) largely contributes to the major cause of cancer deaths throughout the world, rather than the growth of the tumor mass itself. The present report brings together several of the pertinent contributors to cancer growth and metastatic processes from an activity standpoint. Such biological activities include the following: (1) cell adherence and detachment; (2) cell-to-cell contact; (3) contact inhibition; (4) the cell interfacing with the extracellular matrix (ECM); (5) tumor cell-to-stroma communication networks; (6) chemotaxis; and (7) cell membrane potential. Moreover, additional biochemical factors that contribute to cancer growth and metastasis have been shown to comprise the following: (a) calcium levels in the extracellular matrix and in intracellular compartments; (b) cation voltage and ATP-regulated potassium channels; (c) selective and non-selective cation channels; and (d) chemokines (cytokines) and their receptors, such as CXCL12 (SDF-1) and its receptor/binding partner, CXCR4. These latter molecular components represent a promising group of an interacting and synchronized set of candidates ideal for peptide therapeutic targeting for cancer growth and metastasis. Such peptides can be obtained from naturally occurring proteins such as alpha-fetoprotein (AFP), an onco-fetal protein and clinical biomarker.
Collapse
Affiliation(s)
- Gerald J. Mizejewski
- Division of Translational Medicine, Molecular Diagnostics Laboratory, Albany, NY 12201, USA; ; Tel.: +518-486-5900; Fax: +518-402-5002
- Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, NY 12201, USA
- Biggs Laboratory, Empire State Plaza, Albany, NY 12237, USA
| |
Collapse
|
39
|
MacLean MR, Walker OL, Arun RP, Fernando W, Marcato P. Informed by Cancer Stem Cells of Solid Tumors: Advances in Treatments Targeting Tumor-Promoting Factors and Pathways. Int J Mol Sci 2024; 25:4102. [PMID: 38612911 PMCID: PMC11012648 DOI: 10.3390/ijms25074102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer stem cells (CSCs) represent a subpopulation within tumors that promote cancer progression, metastasis, and recurrence due to their self-renewal capacity and resistance to conventional therapies. CSC-specific markers and signaling pathways highly active in CSCs have emerged as a promising strategy for improving patient outcomes. This review provides a comprehensive overview of the therapeutic targets associated with CSCs of solid tumors across various cancer types, including key molecular markers aldehyde dehydrogenases, CD44, epithelial cellular adhesion molecule, and CD133 and signaling pathways such as Wnt/β-catenin, Notch, and Sonic Hedgehog. We discuss a wide array of therapeutic modalities ranging from targeted antibodies, small molecule inhibitors, and near-infrared photoimmunotherapy to advanced genetic approaches like RNA interference, CRISPR/Cas9 technology, aptamers, antisense oligonucleotides, chimeric antigen receptor (CAR) T cells, CAR natural killer cells, bispecific T cell engagers, immunotoxins, drug-antibody conjugates, therapeutic peptides, and dendritic cell vaccines. This review spans developments from preclinical investigations to ongoing clinical trials, highlighting the innovative targeting strategies that have been informed by CSC-associated pathways and molecules to overcome therapeutic resistance. We aim to provide insights into the potential of these therapies to revolutionize cancer treatment, underscoring the critical need for a multi-faceted approach in the battle against cancer. This comprehensive analysis demonstrates how advances made in the CSC field have informed significant developments in novel targeted therapeutic approaches, with the ultimate goal of achieving more effective and durable responses in cancer patients.
Collapse
Affiliation(s)
- Maya R. MacLean
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
| | - Olivia L. Walker
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
| | - Raj Pranap Arun
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
| | - Wasundara Fernando
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
- Department of Biology, Acadia University, Wolfville, NS B4P 2R6, Canada
| | - Paola Marcato
- Department of Pathology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.M.); (O.L.W.); (R.P.A.); (W.F.)
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Nova Scotia Health Authority, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
40
|
Barton LJ, Roa-de la Cruz L, Lehmann R, Lin B. The journey of a generation: advances and promises in the study of primordial germ cell migration. Development 2024; 151:dev201102. [PMID: 38607588 PMCID: PMC11165723 DOI: 10.1242/dev.201102] [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] [Indexed: 04/13/2024]
Abstract
The germline provides the genetic and non-genetic information that passes from one generation to the next. Given this important role in species propagation, egg and sperm precursors, called primordial germ cells (PGCs), are one of the first cell types specified during embryogenesis. In fact, PGCs form well before the bipotential somatic gonad is specified. This common feature of germline development necessitates that PGCs migrate through many tissues to reach the somatic gonad. During their journey, PGCs must respond to select environmental cues while ignoring others in a dynamically developing embryo. The complex multi-tissue, combinatorial nature of PGC migration is an excellent model for understanding how cells navigate complex environments in vivo. Here, we discuss recent findings on the migratory path, the somatic cells that shepherd PGCs, the guidance cues somatic cells provide, and the PGC response to these cues to reach the gonad and establish the germline pool for future generations. We end by discussing the fate of wayward PGCs that fail to reach the gonad in diverse species. Collectively, this field is poised to yield important insights into emerging reproductive technologies.
Collapse
Affiliation(s)
- Lacy J. Barton
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| | - Lorena Roa-de la Cruz
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| | - Ruth Lehmann
- Whitehead Institute and Department of Biology, MIT, 455 Main Street, Cambridge, MA 02142, USA
| | - Benjamin Lin
- Department of Biochemistry & Cell Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| |
Collapse
|
41
|
Moulin C, Beaupain B, Suarez F, Bertrand Y, Beaussant SC, Fischer A, Durin J, Ranta D, Espéli M, Bachelerie F, Bellanné-Chantelot C, Molina T, Emile JF, Balabanian K, Deback C, Donadieu J. CXCR4 WHIM syndrome is a cancer predisposition condition for virus-induced malignancies. Br J Haematol 2024; 204:1383-1392. [PMID: 38442908 DOI: 10.1111/bjh.19373] [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: 11/23/2023] [Revised: 01/31/2024] [Accepted: 02/20/2024] [Indexed: 03/07/2024]
Abstract
Warts, hypogammaglobulinaemia, infections and myelokathexis syndrome (WHIMS) is a rare combined primary immunodeficiency caused by the gain of function of the CXCR4 chemokine receptor. We present the prevalence of cancer in WHIMS patients based on data from the French Severe Chronic Neutropenia Registry and an exhaustive literature review. The median follow-up of the 14 WHIMS 'patients was 28.5 years. A central review and viral evaluation of pathological samples were organized, and we conducted a thorough literature review to identify all reports of WHIMS cases. Six French patients were diagnosed with cancer at a median age of 37.6 years. The 40-year risk of malignancy was 39% (95% confidence interval [CI]: 6%-74%). We observed two human papillomavirus (HPV)-induced vulvar carcinomas, three lymphomas (two Epstein-Barr virus [EBV]-related) and one basal cell carcinoma. Among the 155 WHIMS cases from the literature, 22 cancers were reported in 16 patients, with an overall cancer 40-year risk of 23% (95% CI: 13%-39%). Malignancies included EBV-associated lymphoproliferative disorders and HPV-positive genital and anal cancers as in the French cohort. Worldwide, nine cases of malignancy were associated with HPV and four with EBV. Immunocompromised WHIMS patients appear to be particularly susceptible to developing early malignancy, mainly HPV-induced carcinomas, followed by EBV-related lymphomas.
Collapse
Affiliation(s)
- Clémentine Moulin
- Université Paris-Cité, INSERM U1160, Institut de Recherche Saint-Louis, Paris, France
| | - Blandine Beaupain
- Centre de référence des neutropénies chroniques, Registre des neutropénies chroniques, APHP, Hôpital Trousseau Paris, Paris, France
| | - Felipe Suarez
- Service d'hématologie, Hôpital Necker Enfants Malades, APHP, Paris, France
| | - Yves Bertrand
- Institut d'hémato oncologie Pédiatrique, Hospice Civil de Lyon, Paris, France
| | - Sarah Cohen Beaussant
- Centre de référence des neutropénies chroniques, Registre des neutropénies chroniques, APHP, Hôpital Trousseau Paris, Paris, France
| | - Alain Fischer
- Centre de référence des déficits immunitaires héréditaires, Unité d'Immuno-Hématologie Pédiatrique, Hôpital Necker Enfants Malades, APHP, Paris, France
| | - Julie Durin
- Centre de référence des neutropénies chroniques, Registre des neutropénies chroniques, APHP, Hôpital Trousseau Paris, Paris, France
| | - Dana Ranta
- Service d'hématologie, CHU Nancy, Nancy, France
| | - Marion Espéli
- Université Paris-Cité, INSERM U1160, Institut de Recherche Saint-Louis, Paris, France
| | - Françoise Bachelerie
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| | | | - Thierry Molina
- Service d'anatomie pathologique Hôpital Necker Enfants Malades, APHP, Paris, France
| | - Jean François Emile
- Service d'anatomie pathologique Hôpital Ambroise Paré, APHP, Boulogne-Billancourt, France
| | - Karl Balabanian
- Université Paris-Cité, INSERM U1160, Institut de Recherche Saint-Louis, Paris, France
| | - Claire Deback
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
- Laboratoire de Virologie, Hôpitaux Universitaires Paris-Saclay, Hôpital Paul Brousse, AP-HP, Villejuif, France
| | - Jean Donadieu
- Centre de référence des neutropénies chroniques, Registre des neutropénies chroniques, APHP, Hôpital Trousseau Paris, Paris, France
| |
Collapse
|
42
|
Iyer M, Ravichandran N, Karuppusamy PA, Gnanarajan R, Yadav MK, Narayanasamy A, Vellingiri B. Molecular insights and promise of oncolytic virus based immunotherapy. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 140:419-492. [PMID: 38762277 DOI: 10.1016/bs.apcsb.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
Discovering a therapeutic that can counteract the aggressiveness of this disease's mechanism is crucial for improving survival rates for cancer patients and for better understanding the most different types of cancer. In recent years, using these viruses as an anticancer therapy has been thought to be successful. They mostly work by directly destroying cancer cells, activating the immune system to fight cancer, and expressing exogenous effector genes. For the treatment of tumors, oncolytic viruses (OVs), which can be modified to reproduce only in tumor tissues and lyse them while preserving the healthy non-neoplastic host cells and reinstating antitumor immunity which present a novel immunotherapeutic strategy. OVs can exist naturally or be created in a lab by altering existing viruses. These changes heralded the beginning of a new era of less harmful virus-based cancer therapy. We discuss three different types of oncolytic viruses that have already received regulatory approval to treat cancer as well as clinical research using oncolytic adenoviruses. The primary therapeutic applications, mechanism of action of oncolytic virus updates, future views of this therapy will be covered in this chapter.
Collapse
Affiliation(s)
- Mahalaxmi Iyer
- Department of Microbiology, Central University of Punjab, Bathinda, India
| | - Nandita Ravichandran
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | | | - Roselin Gnanarajan
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Mukesh Kumar Yadav
- Department of Microbiology, Central University of Punjab, Bathinda, India
| | - Arul Narayanasamy
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, Tamil Nadu, India.
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India.
| |
Collapse
|
43
|
Wang P, Jie Y, Yao L, Sun YM, Jiang DP, Zhang SQ, Wang XY, Fan Y. Cells in the liver microenvironment regulate the process of liver metastasis. Cell Biochem Funct 2024; 42:e3969. [PMID: 38459746 DOI: 10.1002/cbf.3969] [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: 10/05/2023] [Revised: 01/31/2024] [Accepted: 02/26/2024] [Indexed: 03/10/2024]
Abstract
The research of liver metastasis is a developing field. The ability of tumor cells to invade the liver depends on the complicated interactions between metastatic cells and local subpopulations in the liver (including Kupffer cells, hepatic stellate cells, liver sinusoidal endothelial cells, and immune-related cells). These interactions are mainly mediated by intercellular adhesion and the release of cytokines. Cell populations in the liver microenvironment can play a dual role in the progression of liver metastasis through different mechanisms. At the same time, we can see the participation of liver parenchymal cells and nonparenchymal cells in the process of liver metastasis of different tumors. Therefore, the purpose of this article is to summarize the relationship between cellular components of liver microenvironment and metastasis and emphasize the importance of different cells in the occurrence or potential regression of liver metastasis.
Collapse
Affiliation(s)
- Pei Wang
- Cancer Institute, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yu Jie
- Cancer Institute, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Lin Yao
- Cancer Institute, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yi-Meng Sun
- Cancer Institute, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Da-Peng Jiang
- Cancer Institute, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Shi-Qi Zhang
- Department of Gastroenterology, The Affiliated Suqian First People's Hospital of Xuzhou Medical University, Suqian, Jiangsu, China
| | - Xiao-Yan Wang
- Department of Gastroenterology, The Affiliated Suqian First People's Hospital of Xuzhou Medical University, Suqian, Jiangsu, China
| | - Yu Fan
- Cancer Institute, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
44
|
Anastasiadou DP, Quesnel A, Duran CL, Filippou PS, Karagiannis GS. An emerging paradigm of CXCL12 involvement in the metastatic cascade. Cytokine Growth Factor Rev 2024; 75:12-30. [PMID: 37949685 DOI: 10.1016/j.cytogfr.2023.10.003] [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: 10/15/2023] [Accepted: 10/20/2023] [Indexed: 11/12/2023]
Abstract
The chemokine CXCL12, also known as stromal cell-derived factor 1 (SDF1), has emerged as a pivotal regulator in the intricate molecular networks driving cancer progression. As an influential factor in the tumor microenvironment, CXCL12 plays a multifaceted role that spans beyond its traditional role as a chemokine inducing invasion and metastasis. Indeed, CXCL12 has been assigned functions related to epithelial-to-mesenchymal transition, cancer cell stemness, angiogenesis, and immunosuppression, all of which are currently viewed as specialized biological programs contributing to the "metastatic cascade" among other cancer hallmarks. Its interaction with its cognate receptor, CXCR4, initiates a cascade of events that not only shapes the metastatic potential of tumor cells but also defines the niches within the secondary organs that support metastatic colonization. Given the profound implications of CXCL12 in the metastatic cascade, understanding its mechanistic underpinnings is of paramount importance for the targeted elimination of rate-limiting steps in the metastatic process. This review aims to provide a comprehensive overview of the current knowledge surrounding the role of CXCL12 in cancer metastasis, especially its molecular interactions rationalizing its potential as a therapeutic target.
Collapse
Affiliation(s)
- Dimitra P Anastasiadou
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA; Tumor Microenvironment & Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, USA
| | - Agathe Quesnel
- School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3BX, United Kingdom; National Horizons Centre, Teesside University, Darlington DL1 1HG, United Kingdom
| | - Camille L Duran
- Tumor Microenvironment & Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, USA; Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA; Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Panagiota S Filippou
- School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3BX, United Kingdom; National Horizons Centre, Teesside University, Darlington DL1 1HG, United Kingdom
| | - George S Karagiannis
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA; Tumor Microenvironment & Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, USA; Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA; Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
45
|
Dai Y, Yu C, Zhou L, Cheng L, Ni H, Liang W. Chemokine receptor CXCR4 interacts with nuclear receptor Nur77 and promote glioma invasion and progression. Brain Res 2024; 1822:148647. [PMID: 37890573 DOI: 10.1016/j.brainres.2023.148647] [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: 09/01/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 10/29/2023]
Abstract
BACKGROUND Glioma is the most common primary brain tumor. It is prone to progress and have high rate of mortality regardless of radiation or chemotherapy due to its invasive growth features. Chemokine and its receptor CXCL12 and CXCR4 play important roles in cancer metastasis. METHODS In this study, we investigate the role of CXCR4 in the progression of glioma by various molecular technologies, including qRT-PCR, Western blotting, wound closure assay, transwell assay et al. RESULTS: It was found that CXCR4 was overexpressed in glioma tissues. The expression of CXCR4 was correlated with patients' overall survival. Wound closure assay and transwell invasion assay showed that inhibition of CXCR4 significantly reduced the expression of biomarkers related to the formation of invadopodium, leading to decrease the invasion and migration of glioma tumor cells. Knocking down the nuclear receptor Nur77 remarkably decreased CXCR4 expression and reduced glioma cell invasion and migration. The reduction of glioma cell invasion and migration were observed after Nur77 inhibitor treatment. CONCLUSION Taken together, these results indicated that CXCR4 is critical in promoting glioma migration and invasion. Inhibition of Nur77 reduces CXCR4 related cancer progression.
Collapse
Affiliation(s)
- Yuxiang Dai
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, China
| | - Chen Yu
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, China
| | - Lu Zhou
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, China
| | - Longyang Cheng
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, China
| | - Hongbin Ni
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, China
| | - Weibang Liang
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, China.
| |
Collapse
|
46
|
Laureano RS, Vanmeerbeek I, Sprooten J, Govaerts J, Naulaerts S, Garg AD. The cell stress and immunity cycle in cancer: Toward next generation of cancer immunotherapy. Immunol Rev 2024; 321:71-93. [PMID: 37937803 DOI: 10.1111/imr.13287] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/05/2023] [Accepted: 10/20/2023] [Indexed: 11/09/2023]
Abstract
The cellular stress and immunity cycle is a cornerstone of organismal homeostasis. Stress activates intracellular and intercellular communications within a tissue or organ to initiate adaptive responses aiming to resolve the origin of this stress. If such local measures are unable to ameliorate this stress, then intercellular communications expand toward immune activation with the aim of recruiting immune cells to effectively resolve the situation while executing tissue repair to ameliorate any damage and facilitate homeostasis. This cellular stress-immunity cycle is severely dysregulated in diseased contexts like cancer. On one hand, cancer cells dysregulate the normal cellular stress responses to reorient them toward upholding growth at all costs, even at the expense of organismal integrity and homeostasis. On the other hand, the tumors severely dysregulate or inhibit various components of organismal immunity, for example, by facilitating immunosuppressive tumor landscape, lowering antigenicity, and increasing T-cell dysfunction. In this review we aim to comprehensively discuss the basis behind tumoral dysregulation of cellular stress-immunity cycle. We also offer insights into current understanding of the regulators and deregulators of this cycle and how they can be targeted for conceptualizing successful cancer immunotherapy regimen.
Collapse
Affiliation(s)
- Raquel S Laureano
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Isaure Vanmeerbeek
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jenny Sprooten
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jannes Govaerts
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Stefan Naulaerts
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
47
|
Safak O, Wang S, Mota Reyes C, Gürcinar IH, Tokalov S, Cevik NC, Görgülü K, Yilmaz BS, Erdogan E, Ye L, Li Q, Sever EA, Özer S, Süyen G, Friess H, Ceyhan GO, Istvanffy R, Algül H, Demir IE. Dynamics and cytokinic regulation of immune cell infiltration in genetically engineered mouse models of pancreatic cancer dictate the sensitivity to immunotherapy. Cancer Commun (Lond) 2024; 44:178-182. [PMID: 37877813 PMCID: PMC10794007 DOI: 10.1002/cac2.12496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 09/20/2023] [Accepted: 10/10/2023] [Indexed: 10/26/2023] Open
Affiliation(s)
- Okan Safak
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Shenghan Wang
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Carmen Mota Reyes
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Ibrahim Halil Gürcinar
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Sergey Tokalov
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Nedim Can Cevik
- Department of General Surgery, HPB‐Unit, School of MedicineAcibadem Mehmet Ali Aydinlar UniversityIstanbulIstanbulTurkey
| | - Kivanc Görgülü
- Comprehensive Cancer Center München, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Bengi Su Yilmaz
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Emre Erdogan
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Linhan Ye
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Qiaolin Li
- Department of Hematology, Oncology and Tumor ImmunologyCharité University Medicine Berlin, Campus Virchow ClinicBerlinBerlinGermany
| | - Elif Arik Sever
- Department of General Surgery, HPB‐Unit, School of MedicineAcibadem Mehmet Ali Aydinlar UniversityIstanbulIstanbulTurkey
| | - Samed Özer
- Graduate School of Health SciencesAcibadem Mehmet Ali Aydinlar UniversityIstanbulIstanbulTurkey
| | - Güldal Süyen
- Department of PhysiologyAcibadem Mehmet Ali Aydinlar UniversityIstanbulIstanbulTurkey
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Güralp Onur Ceyhan
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
- Department of General Surgery, HPB‐Unit, School of MedicineAcibadem Mehmet Ali Aydinlar UniversityIstanbulIstanbulTurkey
| | - Rouzanna Istvanffy
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Hana Algül
- Comprehensive Cancer Center München, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
| | - Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der IsarTechnical University of Munich, School of MedicineMunichBavariaGermany
- Department of General Surgery, HPB‐Unit, School of MedicineAcibadem Mehmet Ali Aydinlar UniversityIstanbulIstanbulTurkey
- Else Kröner Clinician Scientist Professorship for Translational Pancreatic SurgeryMunichGermany
- Neural Influences in Cancer (NIC) International Research ConsortiumMunichGermany
| |
Collapse
|
48
|
Hassanzadeh L, Erfani M, Jokar S, Shariatpanahi M. Design of a New 99mTc-radiolabeled Cyclo-peptide as Promising Molecular Imaging Agent of CXCR 4 Receptor: Molecular Docking, Synthesis, Radiolabeling, and Biological Evaluation. Curr Radiopharm 2024; 17:77-90. [PMID: 37921191 DOI: 10.2174/0118744710249305231017073022] [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: 02/16/2023] [Revised: 07/09/2023] [Accepted: 08/28/2023] [Indexed: 11/04/2023]
Abstract
INTRODUCTION C-X-C Chemokine receptor type 4 (CXCR4) is often overexpressed or overactivated in different types and stages of cancer disease. Therefore, it is considered a promising target for imaging and early detection of primary tumors and metastasis. In the present research, a new cyclo-peptide radiolabelled with 99mTc, 99mTc-Cyclo [D-Phe-D-Tyr-Lys (HYNIC)- D-Arg-2-Nal-Gly-Lys(iPr)], was designed based on the parental LY251029 peptide, as a potential in vivo imaging agent of CXCR4-expressing tumors. METHODS The radioligand was successfully prepared using the method of Fmoc solid-phase peptide synthesis and was evaluated in biological assessment. Molecular docking findings revealed high affinity (binding energy of -9.7 kcal/mol) and effective interaction of Cyclo [D-Phe- D-Tyr-Lys (HYNIC)-D-Arg-2-Nal-Gly-Lys(iPr)] in the binding pocket of CXCR4 receptor (PDB code: 3OE0) as well. RESULT The synthesized peptide and its purity were assessed by both reversed-phase high-performance liquid chromatography (RP-HPLC) and mass spectroscopy. High stability (95%, n = 3) in human serum and favorable affinity (Kd = 28.70 ± 13.56 nM and Bmax = 1.896 ± 0.123 fmol/mg protein) in the B16-F10 cell line resulted. Biodistribution evaluation findings and planar image interpretation of mice both showed high affinity and selectivity of the radiotracer to the CXCR4 receptors. CONCLUSION Therefore, the findings indicate this designed radioligand could be used as a potential SPECT imaging agent in highly proliferated CXCR4 receptor tumors.
Collapse
Affiliation(s)
- Leila Hassanzadeh
- Department of Nuclear Medicine, School of Medicine, Rajaie Cardiovascular, Medical & Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Imaging Technology, Molecular Imaging, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mostafa Erfani
- Radiation Application Research School, Nuclear Science and Technology Research Institute, (NSTRI), P.O. Box: 14395-836, Tehran, Iran
| | - Safura Jokar
- Department of Nuclear Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Marjan Shariatpanahi
- Department of Pharmacology and Toxicology, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
49
|
Venkataraman K, Salehi T, Carroll RP. Immune Checkpoint Inhibitors in Recipients of Renal Allografts. Semin Nephrol 2024; 44:151500. [PMID: 38548484 DOI: 10.1016/j.semnephrol.2024.151500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Kidney transplant recipients are at increased risk of malignancy as a result of immunosuppression and are increasingly exposed to checkpoint inhibitors (CPIs). However, CPI therapy can precipitate allograft rejection. This review aims to summarize the current literature describing the epidemiology, immunological mechanisms, diagnosis, and treatment of CPI-associated allograft rejection.Initial studies of CPIs suggested allograft rejection post commencement of CPIs occured commonly (40-60%), occurring between 2 and 6 weeks after CPI initiation, with a cancer response rate approaching 50%. More recent studies with predefined, structured immunosuppressive regimens have seen rejection rates of 0-12.5%, with rejection occurring later. Allograft biopsy remains the mainstay of diagnosis; however, noninvasive tools are emerging, including donor-derived cell-free DNA, urinary chemokine assessment, and defining alloreactive T-cell clones prior to or during CPI therapy.
Collapse
Affiliation(s)
- Karthik Venkataraman
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, Australia
| | - Tania Salehi
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, Australia
| | - Robert P Carroll
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, Australia; Australian Red Cross Lifeblood Service, Department of Health Sciences, University of South Australia, Adelaide, Australia.
| |
Collapse
|
50
|
Li Q, Fan J, Zhou Z, Ma Z, Che Z, Wu Y, Yang X, Liang P, Li H. AID-induced CXCL12 upregulation enhances castration-resistant prostate cancer cell metastasis by stabilizing β-catenin expression. iScience 2023; 26:108523. [PMID: 38162032 PMCID: PMC10755053 DOI: 10.1016/j.isci.2023.108523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 11/14/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
Prostate cancer (PCa) is one of the most common malignant diseases of urinary system and has poor prognosis after progression to castration-resistant prostate cancer (CRPC), and increased cytosine methylation heterogeneity is associated with the more aggressive phenotype of PCa cell line. Activation-induced cytidine deaminase (AID) is a multifunctional enzyme and contributes to antibody diversification. However, the dysregulation of AID participates in the progression of multiple diseases and related with certain oncogenes through demethylation. Nevertheless, the role of AID in PCa remains elusive. We observed a significant upregulation of AID expression in PCa samples, which exhibited a negative correlation with E-cadherin expression. Furthermore, AID expression is remarkably higher in CRPC cells than that in HSPC cells, and AID induced the demethylation of CXCL12, which is required to stabilize the Wnt signaling pathway executor β-catenin and EMT procedure. Our study suggests that AID drives CRPC metastasis by demethylation and can be a potential therapeutic target for CRPC.
Collapse
Affiliation(s)
- Qi Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
- Department of Urology, TianYou Hospital affiliated to Wuhan University of Science & Technology, Wuhan, Hubei Province, China
| | - Jinfeng Fan
- Department of Urology, the First Affiliated Hospital of Hainan Medical College, Haikou, Hainan Province, China
| | - Zhiyan Zhou
- Department of Urology, the First Affiliated Hospital of Hainan Medical College, Haikou, Hainan Province, China
| | - Zhe Ma
- The First Hospital of Tsinghua University, Beijing, China
| | - Zhifei Che
- Department of Urology, the First Affiliated Hospital of Hainan Medical College, Haikou, Hainan Province, China
| | - Yaoxi Wu
- Department of Urology, the First Affiliated Hospital of Hainan Medical College, Haikou, Hainan Province, China
| | - Xiangli Yang
- Department of Urology, TianYou Hospital affiliated to Wuhan University of Science & Technology, Wuhan, Hubei Province, China
| | - Peiyu Liang
- Department of Urology, the First Affiliated Hospital of Hainan Medical College, Haikou, Hainan Province, China
| | - Haoyong Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|