1
|
Slisarenko M, Alaghehbandan R, Rogala J, Farcas M, Skopal J, Svajdler M, Fiala O, Stránský P, Vesela AB, Pitra T, Hora M, Michal M, Hes O, Pivovarcikova K. Reactivity of carbonic anhydrase IX (CA IX) across the spectrum of renal cell carcinomas with sarcomatoid differentiation. Ann Diagn Pathol 2025; 79:152517. [PMID: 40516201 DOI: 10.1016/j.anndiagpath.2025.152517] [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: 04/22/2025] [Revised: 05/27/2025] [Accepted: 06/02/2025] [Indexed: 06/16/2025]
Abstract
Carbonic anhydrase IX (CA IX) is traditionally considered to be an immunomarker of clear cell renal cell carcinoma (RCC). However, CA IX expression has also been documented in other RCCs subtypes. Discrimination between clear cell RCC and non-clear cell RCC is crucial for further patient management. The aim of this study was to assess CA IX immunoreactivity across the spectrum of RCC with sarcomatoid differentiation. The expression of CA IX was evaluated in 32 cases of RCCs with sarcomatoid differentiation (12 clear cell RCC, 7 papillary RCC and 13 chromophobe RCC). Seven urothelial carcinomas (UC) with sarcomatoid differentiation (originally from renal pelvis) and 23 soft tissue tumors were also included as a control cohort. The sensitivity for CA IX in sarcomatoid component of clear cell RCC was 91.7 % (moderate/strong CA IX staining in >60 % of sarcomatoid component). However, the CA IX specificity was rather low (45 %), as a significant proportion of sarcomatoid components in different renal cell carcinoma subtypes also stained with CA IX (2/7 papillary RCC, 9/14 chromophobe RCC). When urothelial carcinoma and soft tissue tumors were included in the evaluation, the specificity of CA IX staining for sarcomatoid clear cell RCC reached 60 %. In conclusion, CA IX shows decent sensitivity for sarcomatoid clear cell RCC, but with low specificity, hence limiting its diagnostic utility as a reliable marker of in tumors with predominant sarcomatoid component.
Collapse
Affiliation(s)
- Maryna Slisarenko
- Department of Pathology, Medical Laboratory CSD LAB, Kyiv, Ukraine; Department of Pathology, Charles University in Prague, Faculty of Medicine in Plzeň, Pilsen, Czech Republic
| | - Reza Alaghehbandan
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Department of Anatomic Pathology, Cleveland Clinic, Cleveland, OH, USA
| | - Joanna Rogala
- Department of Pathology, Regional Specialist Hospital Wroclaw, Poland
| | - Mihaela Farcas
- Department of Pathology, Charles University in Prague, Faculty of Medicine in Plzeň, Pilsen, Czech Republic; Onco Team Diagnostic, București, Romania
| | - Josef Skopal
- Department of Pathology, Charles University in Prague, Faculty of Medicine in Plzeň, Pilsen, Czech Republic
| | - Marian Svajdler
- Department of Pathology, Charles University in Prague, Faculty of Medicine in Plzeň, Pilsen, Czech Republic; Cytopathos s. r. o., Bratislava, Slovakia
| | - Ondrej Fiala
- Department of Oncology and Radiotherapeutics, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic; Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Petr Stránský
- Department of Urology, Charles University in Prague, Faculty of Medicine in Plzeň, Pilsen, Czech Republic
| | - Adriena Bartos Vesela
- Department of Urology, Charles University in Prague, Faculty of Medicine in Plzeň, Pilsen, Czech Republic
| | - Tomas Pitra
- Department of Urology, Charles University in Prague, Faculty of Medicine in Plzeň, Pilsen, Czech Republic
| | - Milan Hora
- Department of Urology, Charles University in Prague, Faculty of Medicine in Plzeň, Pilsen, Czech Republic
| | - Michal Michal
- Department of Pathology, Charles University in Prague, Faculty of Medicine in Plzeň, Pilsen, Czech Republic
| | - Ondrej Hes
- Department of Pathology, Charles University in Prague, Faculty of Medicine in Plzeň, Pilsen, Czech Republic
| | - Kristyna Pivovarcikova
- Department of Pathology, Charles University in Prague, Faculty of Medicine in Plzeň, Pilsen, Czech Republic.
| |
Collapse
|
2
|
Elsayad KA, Elmasry GF, Mahmoud ST, Awadallah FM, Giovannuzzi S, Supuran CT. Development of novel amino-benzenesulfonamide derivatives and their analogues as carbonic anhydrase inhibitors: Design, synthesis, anticancer activity assessment, and pharmacokinetic studies using UPLC-MS/MS. Bioorg Chem 2025; 159:108335. [PMID: 40086186 DOI: 10.1016/j.bioorg.2025.108335] [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: 01/06/2025] [Revised: 02/23/2025] [Accepted: 02/28/2025] [Indexed: 03/16/2025]
Abstract
The present study outlines the design and synthesis of dual-tail analogues of SLC-0111 as carbonic anhydrase inhibitors (CAIs) targeting tumor isoforms IX and XII 4a-h and 5a-h, along with pharmacokinetic studies. The synthesized compounds were evaluated for their inhibitory activity against four carbonic anhydrase isoforms (hCA I, II, IX, and XII), revealing potent activity, particularly against hCA IX and XII. Notably, compounds 4b, 5a, and 5b demonstrated strong inhibition of hCA IX with Ki values of 20.4, 12.9, and 18.2 nM, respectively, compared to acetazolamide (AAZ), which has a Ki of 25 nM. Additionally, compounds 5a, 5b, 5c, and 5d showed selective inhibition of hCA XII, with Ki values of 26.6, 8.7, 17.2, and 10.9 nM, respectively, relative to AAZ (Ki = 5.7 nM). Moreover, both series were tested for their anti-proliferative activity following the US-NCI protocol against a panel of more than fifty cancer cell lines. Compound 5h met the activity criteria and was automatically scheduled for further evaluation at five concentrations with 10-fold dilutions, revealing high toxicity toward leukemia and lower toxicity against melanoma. In addition, the MTT cytotoxicity assay was performed on 5f, 5d and acetazolamide using WI-38 cells. Furthermore, an in vivo pharmacokinetic study was conducted using UPLC-MS/MS on the most potent derivative, 5d, demonstrating a comparable pharmacokinetic profile compared to the reference drug acetazolamide. Furthermore, molecular docking prediction studies were conducted for the most active compounds, 5d and 5h, to elucidate their interactions with the active site hot spots of the CA isoform.
Collapse
Affiliation(s)
- Khaled A Elsayad
- Pharmacy Department, Cairo University Hospitals, Cairo University, Cairo 11662, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, 11562 Cairo, Egypt.
| | - Ghada F Elmasry
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, 11562 Cairo, Egypt
| | - Sally T Mahmoud
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, 11562 Cairo, Egypt; Pharmaceutical Chemistry Department, School of Pharmacy, New Giza University, New Giza, km 22 Cairo- Alexandria Desert Road, Cairo, Egypt.
| | - Fadi M Awadallah
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, 11562 Cairo, Egypt
| | - Simone Giovannuzzi
- Department NEUROFARBA - Pharmaceutical and Nutraceutical section, University of Firenze, Università Degli Studi di Firenze, Sesto Fiorentino, Italy
| | - Claudiu T Supuran
- Department NEUROFARBA - Pharmaceutical and Nutraceutical section, University of Firenze, Università Degli Studi di Firenze, Sesto Fiorentino, Italy
| |
Collapse
|
3
|
Grossmannova K, Belvoncikova P, Puzderova B, Simko V, Csaderova L, Pastorek J, Barathova M. Carbonic anhydrase IX downregulation linked to disruption of HIF-1, NFκB and STAT3 pathways as a new mechanism of ibuprofen anti-cancer effect. PLoS One 2025; 20:e0323635. [PMID: 40408503 PMCID: PMC12101644 DOI: 10.1371/journal.pone.0323635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 04/10/2025] [Indexed: 05/25/2025] Open
Abstract
Numerous studies have highlighted the anti-cancer effects of nonsteroidal anti-inflammatory drugs (NSAIDs), although the underlying mechanisms remain unclear. This study focuses on elucidating the impact of the NSAID ibuprofen (IBU) on cancer cells exposed to hypoxia, as the hypoxic microenvironment significantly influences tumor progression, metastatic potential, and therapy resistance. Given that carbonic anhydrase IX (CA IX) is a key hypoxia-associated protein and a promising therapeutic target due to its tumor-specific expression, we primarily examined the impact of IBU on CA IX and the transcription factors regulating CA IX expression. We found that IBU downregulates expression and protein level of CA IX in hypoxic colon carcinoma and head and neck cancer cells, resulting in a reduction of membranous CA IX. To elucidate the mechanism of this phenomenon, we analyzed the key CA IX-regulating transcription factor HIF-1 and found decreased levels of the HIF-1α subunit in IBU-treated cells, leading to its impaired binding to the CA9 promotor. Analysis of another transcription factor involved in CA IX expression, NFκB, showed suppressed NFκB pathway under IBU treatment. Moreover, we demonstrated IBU-mediated induction in apoptosis in cancer cells, as well as a decrease in their ability to migrate. Our study is the first to demonstrate that ibuprofen downregulates carbonic anhydrase IX expression in hypoxic colon and head and neck tumor cells by decreasing HIF-1α levels. Additionally, ibuprofen impairs key transcription factors NFκB and STAT3, leading to reduced adaptation to hypoxic stress, decreased tumor cell viability, and migration. This indicates its potential as a therapeutic agent in combination therapy for colon carcinoma or head and neck cancer.
Collapse
Affiliation(s)
- Katarina Grossmannova
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Petra Belvoncikova
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Barbora Puzderova
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Veronika Simko
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lucia Csaderova
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | - Monika Barathova
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
4
|
Ren S, Zhu J, Shan G, Liang J, Bian Y, Lin H, Shi H, Pan B, Zhao G, Yang H, Huang X, Zhan C, Ge D, Bi G. Transcription factor ZNF266 suppresses cancer progression by modulating CA9-mediated intracellular pH alteration in lung adenocarcinoma. Respir Res 2025; 26:191. [PMID: 40389968 PMCID: PMC12090625 DOI: 10.1186/s12931-025-03278-7] [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: 01/11/2025] [Accepted: 05/15/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Lung cancer remains the leading cause of cancer-related mortality globally, with lung adenocarcinoma (LUAD) being the most prevalent subtype. Despite extensive research efforts, the role of transcription factors in LUAD progression remains largely uncharacterized. In this study, we focused on ZNF266, a transcription factor whose impacts on LUAD have not been investigated. METHODS Using high-throughput sequencing data, we observed a significant downregulation of ZNF266 expression in LUAD tissues. To validate this finding, we conducted a retrospective analysis of nearly three thousand LUAD patients' data from public databases and our institution. Functional studies were performed using cell lines, organoids, and xenograft models to assess the role of ZNF266 in LUAD progression. RNA sequencing, chromatin immunoprecipitation, DNA pull-down assays, and dual-luciferase reporter assays were employed to elucidate the underlying mechanism. Additionally, adeno-associated virus (AAV)-mediated overexpression of ZNF266 was used to evaluate its therapeutic potential. RESULTS Patients with low ZNF266 expression had poorer prognosis compared to those with high expression. ZNF266 inhibits the malignant phenotypes of LUAD, including proliferation, migration, and invasion. Mechanistically, ZNF266 binds to the promoter region of CA9, suppressing its transcription. This leads to a reduction in intracellular pH and subsequent inhibition of the mTOR signaling pathway, which is crucial for cancer cell growth and survival. Furthermore, AAV-mediated overexpression of ZNF266 significantly inhibited tumor growth in patient-derived xenograft models. CONCLUSIONS Our study demonstrated that ZNF266 inhibits LUAD progression in a pH-dependent manner via modulating CA9 expression, uncovering its therapeutic significance for LUAD treatment.
Collapse
Affiliation(s)
- Shencheng Ren
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Junkan Zhu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jiaqi Liang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yunyi Bian
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Han Lin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Haochun Shi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Binyang Pan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Guangyin Zhao
- Department of Thoracic Surgery, Shanghai Geriatric Medical Center, Fudan University, Shanghai, 201104, China
| | - Huiqin Yang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaolong Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Di Ge
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
5
|
Kalniņa Z, Liekniņa I, Koteloviča S, Petrovska R, Žvinys G, Petrosiute A, Zubrienė A, Laugalis MT, Skeltona V, Jansons J, Kreishmane M, Čapkauskaitė E, Matulis D, Tārs K. Development of 4T1 breast cancer mouse model system for preclinical carbonic anhydrase IX studies. FEBS Open Bio 2025. [PMID: 40371725 DOI: 10.1002/2211-5463.70052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/10/2025] [Accepted: 04/30/2025] [Indexed: 05/16/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive type of breast cancer, for which targeted treatment is currently lacking. Carbonic anhydrase IX (CAIX) is a known cancer target due to its selective overexpression in hypoxia, a hallmark of many solid cancers including TNBC. This study aimed to develop a robust murine TNBC cell line 4T1-based model system that could be used in the comprehensive preclinical evaluation of targeting CAIX. The model is based on the original 4T1 breast cancer cell line and two genetically edited versions of it-one with biallelic CRISPR/Cas9-mediated Car9 inactivation and another with constitutively expressed Car9, thus ensuring negative and positive controls for CAIX production in the model system, respectively. The generated cell lines were validated for CAIX production and characterised functionally in vitro and in vivo after orthotopic implantation in syngeneic BALB/c mice. Results demonstrated significantly reduced primary tumour growth and metastatic progression rates in animals with CAIX-deficient tumours, while the CAIX-expressing tumours had vascularised phenotypes with prominent central areas of coagulative necrosis. The differential CAIX expression levels in the model were preserved during tumour growth in syngeneic mice, as verified by in vivo imaging using a novel high-affinity CAIX-specific near-infrared (NIR) fluorescent imaging probe, GZ22-4. Constitutive overexpression of autologous CAIX did not elicit specific autoantibody responses in vivo, demonstrating the suitability of this model for evaluating the efficacy of anti-CAIX vaccination as a therapeutic strategy. The in vivo study was repeated as an independent experiment and demonstrated good robustness of the developed model.
Collapse
Affiliation(s)
- Zane Kalniņa
- Latvian Biomedical Research and Study Centre, Riga, Latvia
- Faculty of Medicine and Life Sciences, University of Latvia, Riga, Latvia
| | - Ilva Liekniņa
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | | | | | - Gediminas Žvinys
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Lithuania
| | - Agne Petrosiute
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Lithuania
| | - Asta Zubrienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Lithuania
| | | | - Vendija Skeltona
- Faculty of Medicine and Life Sciences, University of Latvia, Riga, Latvia
| | - Juris Jansons
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | | | - Edita Čapkauskaitė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Lithuania
| | - Daumantas Matulis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Lithuania
| | - Kaspars Tārs
- Latvian Biomedical Research and Study Centre, Riga, Latvia
- Faculty of Medicine and Life Sciences, University of Latvia, Riga, Latvia
| |
Collapse
|
6
|
Napolitano R, Adamo A, Biondi L, Cordaro A, Iaboni M, Stummo F, La Cava F, Arena F, Faletto D, Chianale F, Reitano E, Poggi L, Blasi F. Novel Near Infrared Dyes Targeting Carbonic Anhydrase IX for Fluorescence Imaging Applications. Invest Radiol 2025:00004424-990000000-00335. [PMID: 40334098 DOI: 10.1097/rli.0000000000001204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
OBJECTIVES Fluorescence tumor-targeted imaging is a sensitive technique that may assist surgeons to remove residual cancer tissue during resection. Carbonic anhydrase IX (CAIX) is a tumor-associated cell-surface glycoprotein, upregulated in hypoxic environments, and a suitable biomarker to develop targeted dyes for fluorescence-guided surgery. This study describes design, synthesis, in vitro and in vivo assessment of novel CAIX-targeting fluorescent probes based on the well-known drug acetazolamide, addressing the contribution of both targeting moiety and fluorophore structure on imaging efficacy. MATERIALS AND METHODS All the CAIX-targeting heptamethine cyanines synthesized and described in the present work were characterized in terms of their optical properties in different media. The affinity to human serum albumin was evaluated by UV-VIS spectrophotometry. The affinity to the CA catalytic site was determined on a recombinant bovine CAII enzyme (bCAII), with a fluorescent-based assay. Human colon adenocarcinoma HT-29 cells, highly expressing CAIX, were used for the in vitro characterization, including cell binding, uptake and competition assays, by flow cytometry. Finally, the in vivo tumor targeting efficacy of a selected group of probes was assessed by Optical Imaging in a mouse subcutaneous tumor from HT-29 cells, characterized by both expression of CAIX and a hypoxic tumor microenvironment. RESULTS First, a family of CAIX-targeting probes was prepared by functionalizing a novel glucamine-bearing heptamethine cyanine (Dye1) with a modified acetazolamide moiety, whose acetyl group was replaced with i) aminooctanoic acid C8, ii) phenylalanine, iii) amino-PEG2-acid and iv) the longer linker 4a, or 2 commercially available benzenesulfonamides. From the in vitro screening of this first group of compounds, the C8-AZA targeting moiety was selected due to its highest affinity. Indeed, Dye1-C8-AZA exhibited the lowest KD values for both bCAII (6.1 ± 1.6 nM) and CAIX-expressing HT-29 cells (58 ± 9 nM), even lower than HypoxyFluor-1 (HF-1), a CAIX-targeted dye already reported in the literature. Then, other heptamethine cyanines (Dye2-Dye5, linear or cyclic, with different substituents on the indolenines and different conjugation position of the targeting vector) were functionalized with C8-AZA and fully characterized both in vitro and in vivo, to evaluate the combinatory effect of vector and fluorophore on the performance of the resulting probes. The different chemical features of the cyanines influenced the optical properties, solubility, binding with albumin, biodistribution, and imaging efficacy of the probes, while leaving unaffected the high affinity to the target. When tested in vivo for the visualization of CAIX-expressing HT-29 tumors, all C8-AZA probes showed high and specific tumor accumulation, often superior to HF-1. CONCLUSIONS Several CAIX-targeting probes were synthesized to test the combinatory effect of different molecular vectors and dyes on the biological properties. All probes containing the C8-AZA targeting moiety displayed higher affinity and specificity to the target, while imaging efficacy in vivo was strongly influenced also by the structure of the labelling dye. All probes, and among them especially Dye1-C8-AZA, displayed efficient in vivo tumor accumulation. These results support further studies toward clinical testing of CAIX as suitable target for tumor fluorescence imaging and pave the way for future clinical applications.
Collapse
|
7
|
Li Y, Shi M, Bie B, Tian H, Li J, Li Z, Sun J. NRF1-Induced lncRNA DDX11-AS1 Contributes to the Progression of Hepatocellular Carcinoma via Activating CA9 Expression and the MEK/ERK Pathway. J Hepatocell Carcinoma 2025; 12:891-908. [PMID: 40356690 PMCID: PMC12067462 DOI: 10.2147/jhc.s516656] [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: 01/09/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Purpose DDX11 antisense RNA 1 (DDX11-AS1) has been recognized for its strong correlation with hepatocellular carcinoma (HCC). Nevertheless, the exact biological functions and fundamental molecular processes of DDX11-AS1 in HCC require further in-depth investigation. Methods A comprehensive bioinformatics analysis was carried out to explore the expression of DDX11-AS1 and its clinical implication in HCC utilizing the TCGA data. qRT-PCR was employed to validate the expression of DDX11-AS1 in HCC tissues/cell lines. RNA fluorescence in situ hybridization (RNA-FISH) was used to observe the subcellular localization of DDX11-AS1 in HCC cells. Loss-of-function experiments, both in vitro and in vivo, were executed to elucidate the biological functions of DDX11-AS1 in HCC. RNA sequencing (RNA-seq) was employed to identify genes and signaling pathways potentially regulated by DDX11-AS1. Rescue experiments were conducted to validate that carbonic anhydrase IX (CA9) mediates DDX11-AS1 promoting HCC progression. The influence of nuclear respiratory factor 1 (NRF1) on the transcription of DDX11-AS1 was investigated through dual-luciferase reporter assays and ChIP-qPCR. Results The increased expression of DDX11-AS1 is positively associated with several aggressive clinical characteristics (pathologic T stage, histologic grade, AFP level, and vascular invasion), and is closely linked to unfavorable outcomes in HCC patients, acting as a separate hazardous factor for overall survival. DDX11-AS1 is predominantly situated in the nucleus of HCC cells. DDX11-AS1 knockdown impeded the growth, migration, and invasion capabilities of HCC cells in vitro, and reduced the tumor enlargement in a subcutaneous mouse model. RNA-Seq unveiled that silencing DDX11-AS1 lessened the expression of CA9 and suppressed the activity of the MEK/ERK signaling cascade in HCC cells. Rescue experiments uncovered that CA9 acts as a downstream target facilitating the cancer-causing roles of DDX11-AS1 in HCC. Furthermore, DDX11-AS1 was revealed to be transcriptionally regulated by NRF1. Conclusion DDX11-AS1, a NRF1-induced lncRNA, facilitates HCC development by upregulating CA9 expression and activating the MEK/ERK signaling cascade.
Collapse
Affiliation(s)
- Yingnan Li
- Department of General Surgery, National-Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
- Center for Tumor and Immunology, The Precision Medical Institute, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710115, People’s Republic of China
| | - Mengjiao Shi
- Department of General Surgery, National-Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| | - Beibei Bie
- Department of Pharmacy, Medical School, Xi’an Peihua University, Xi’an, 710125, People’s Republic of China
| | - Hongwei Tian
- Department of General Surgery, National-Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| | - Jun Li
- Department of General Surgery, National-Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| | - Zongfang Li
- Department of General Surgery, National-Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
- Center for Tumor and Immunology, The Precision Medical Institute, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710115, People’s Republic of China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| | - Jin Sun
- Department of General Surgery, National-Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| |
Collapse
|
8
|
Nie T, Fang Y, Zhang R, Cai Y, Wang X, Jiao Y, Wu J. Self-healable and pH-responsive spermidine/ferrous ion complexed hydrogel Co-loaded with CA inhibitor and glucose oxidase for combined cancer immunotherapy through triple ferroptosis mechanism. Bioact Mater 2025; 47:51-63. [PMID: 39877156 PMCID: PMC11772096 DOI: 10.1016/j.bioactmat.2025.01.005] [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: 07/01/2024] [Revised: 12/24/2024] [Accepted: 01/07/2025] [Indexed: 01/31/2025] Open
Abstract
Tumor microenvironment governs various therapeutic tolerability of cancer such as ferroptosis and immunotherapy through rewiring tumor metabolic reprogramming like Warburg metabolism. Highly expressed carbonic anhydrases (CA) in tumor that maintaining the delicate metabolic homeostasis is thus the most potential target to be modulated to resolve the therapeutic tolerability. Hence, in this article, a self-healable and pH-responsive spermidine/ferrous ion hydrogel loaded with CA inhibitor (acetazolamide, ACZ) and glucose oxidase (ACZ/GOx@SPM-HA Gel) was fabricated through the Schiff-base reaction between spermidine-dextran and oxidized hyaluronic acid, along with ferrous coordination. Investigation on cancer cell lines (MOC-1) demonstrated ACZ/GOx@SPM-HA Gel may induce cellular oxidative stress and mitochondrial dysfunction through disrupting the cellular homeostasis. Moreover, with the facilitation of autophagy induced by spermidine, ACZ/GOx@SPM-HA Gel may trigger a positive feedback loop to maximally amplify cellular ferroptosis and promote DAMPs release. The anti-tumor evaluation on xenograft mice models furtherly proved the local injection of such hydrogel formulation could efficiently inhibit the tumor growth and distinctively promote the immunogenicity of tumor bed to provide a more favorable environment for immunotherapy. Overall, ACZ/GOx@SPM-HA Gel, with such feasible physiochemical properties and great biocompatibility, holds great potential in treating solid tumors with acidosis-mediated immunotherapy tolerance.
Collapse
Affiliation(s)
- Tianqi Nie
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou Medical University, Guangzhou, 510620, China
| | - Yifei Fang
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou, 511400, China
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Ruhe Zhang
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518106, China
| | - Yishui Cai
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou, 511400, China
| | - Xiaobo Wang
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518106, China
| | - Yuenong Jiao
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou Medical University, Guangzhou, 510620, China
| | - Jun Wu
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou, 511400, China
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518106, China
| |
Collapse
|
9
|
Tran RL, Li T, de la Cerda J, Schuler FW, Khaled AS, Pudakalakatti S, Bhattacharya PK, Sinharay S, Pagel MD. Potentiation of immune checkpoint blockade with a pH-sensitizer as monitored in two pre-clinical tumor models with acidoCEST MRI. Br J Cancer 2025; 132:744-753. [PMID: 39994445 PMCID: PMC11997056 DOI: 10.1038/s41416-025-02962-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 01/20/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Tumor acidosis causes resistance to immune checkpoint blockade (ICB). We hypothesized that a "pH-sensitizer" can increase tumor extracellular pH (pHe) and improve tumor control following ICB. We also hypothesized that pHe measured with acidoCEST MRI can predict improved tumor control with ICB. METHODS We tested the effects of pH-sensitizers on proton efflux rate (PER), cytotoxicity, T cell activation, tumor immunogenicity, tumor growth and survival using 4T1 and B16-F10 tumor cells. We measured in vivo tumor pHe of 4T1 and B16-F10 models with acidoCEST MRI. RESULTS Among the pH-sensitizers tested, someprazole caused the greatest reduction in PER without exhibiting cytotoxicity or reducing T cell activation. Esomeprazole improved 4T1 tumor control with ICB administered one day after the pH-sensitizer. Tumor pHe positively correlated with TCF-1 + CD4 effector and CD8 T cell intratumoral frequencies and predicted improved 4T1 tumor control with ICB. For comparison, esomeprazole had a mild effect on B16-F10 tumor pHe, and worsened tumor control with ICB and increased intratumoral myeloid and dendritic cell (DC) frequencies. CONCLUSIONS A pH-sensitizer can improve tumor control with ICB, and acidoCEST MRI can be used to measure pHe and predict tumor control, but only in the 4T1 model and not the B16-F10 model.
Collapse
Affiliation(s)
- Renee L Tran
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | - Tianzhe Li
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jorge de la Cerda
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | - F William Schuler
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | - Alia S Khaled
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Sanhita Sinharay
- Centre for Biosystems Science & Engineering, Indian Institute of Science, Bangalore, India
| | - Mark D Pagel
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
10
|
Bai Y, Osmundson EC, Donahue MJ, De Vis JB. Magnetic resonance imaging to detect tumor hypoxia in brain malignant disease: A systematic review of validation studies. Clin Transl Radiat Oncol 2025; 52:100940. [PMID: 40093743 PMCID: PMC11908384 DOI: 10.1016/j.ctro.2025.100940] [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: 10/17/2024] [Revised: 02/17/2025] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
Tumor hypoxia indicates a worse prognosis in brain malignancies; however, current gold-standard methods for assessing tumor hypoxia are invasive and often inaccessible. Magnetic Resonance Imaging (MRI) is widely available, but its validity for identifying tumor hypoxia or hypoxia-related neoangiogenesis is not well characterized. A systematic literature search was performed across PubMed and Embase Databases. The search query identified MRI studies that validated hypoxia-surrogate imaging sequences against gold-standard hypoxia or neoangiogenesis detection methods in patients with brain malignancies. Literature screen identified 23 manuscripts published between 2007 and 2022. Among conventional MRI sequences, peritumoral edema and signal change after contrast administration were associated with gold-standard oxygen-assessment methods. T2*- and T2'-derived measures were associated with gold-standard methods, while reports on quantitative measures of oxygen extraction fraction were conflicting. Fiber density, tissue cellularity, blood volume, vascular transit time, and permeability measurements were associated with gold-standard methods, whereas blood flow measurements yielded conflicting results. MRI measures are promising surrogates for tumor hypoxia or hypoxia-related neoangiogenesis. Additional studies are needed to reconcile disparate findings. Future sensitivity analyses are needed to establish the MRI methods most accurate at identifying tumor hypoxia.
Collapse
Affiliation(s)
- Y Bai
- Vanderbilt School of Medicine, Vanderbilt University, Nashville, TN, USA
| | - E C Osmundson
- Department of Radiation Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M J Donahue
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Electrical and Computer Engineering, Vanderbilt University, Nashville, TN, USA
| | - J B De Vis
- Department of Radiation Oncology, Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
11
|
Huang R, Kee L, Gont A, Meens J, Ferens FG, Irwin MS, Ailles L, Yuzwa SA, Robinson CM, Ohh M. Comparative single-cell transcriptomic profiling of patient-derived renal carcinoma cells in cellular and animal models of kidney cancer. FEBS Open Bio 2025. [PMID: 40241258 DOI: 10.1002/2211-5463.70022] [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: 09/05/2024] [Revised: 02/26/2025] [Accepted: 03/06/2025] [Indexed: 04/18/2025] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common form of kidney cancer that often displays resistance to conventional cancer therapies, including chemotherapy and radiation therapy. Targeted treatments, including immunotherapies and small molecular inhibitors, have been associated with improved outcomes. However, variations in the patient response and the development of resistance suggest that more models that better recapitulate the pathogenesis and metastatic mechanisms of ccRCC are required to improve our understanding and disease management. Here, we examined the transcriptional landscapes of in vitro cell culture as well as in vivo orthotopic and metastatic NOD/SCID-γ mouse models of ccRCC using a single patient-derived RCC243 cell line to allow unambiguous comparison between models. In our mouse model assays, RCC243 cells formed metastatic tumors, and all tumors retained clear cell morphology irrespective of model type. Notably, gene expression profiles differed markedly between the RCC243 tumor models-cell culture, orthotopic tumors, and metastatic tumors-suggesting an impact of the experimental model system and whether the tumor was orthotopic or metastatic. Furthermore, we found conserved prognostic markers between RCC243 tumor models and human ccRCC patient datasets, and genes upregulated in metastatic RCC243 were associated with worse patient outcomes.
Collapse
Affiliation(s)
- Richard Huang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
| | - Lynn Kee
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Alexander Gont
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Jalna Meens
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Fraser G Ferens
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
| | - Meredith S Irwin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Canada
- Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada
| | - Laurie Ailles
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Canada
| | - Scott A Yuzwa
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
| | - Claire M Robinson
- School of Medicine, Health Sciences Centre, University College Dublin, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Michael Ohh
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
- Department of Biochemistry, University of Toronto, Canada
| |
Collapse
|
12
|
Tarawneh N, Hussein SA, Abdalla S. Repurposing Antiepileptic Drugs for Cancer: A Promising Therapeutic Strategy. J Clin Med 2025; 14:2673. [PMID: 40283503 PMCID: PMC12027853 DOI: 10.3390/jcm14082673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/29/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Epilepsy is a neurological disorder characterized by repeated convulsions. Antiepileptic drugs (AEDs) are the main course of therapy for epilepsy. These medications are given according to each patient's personal medical history and the types of seizures they suffer. They have been employed for decades to manage epilepsy, thus delivering relief from seizures through numerous mechanisms of action. Aside from their anticonvulsant attributes, current evidence suggests that certain AEDs may display potential inhibitory effects against cancer invasion and metastasis. This review explored the complicated interactions between the modes of action of AEDs and the pathways causing cancer, and the potential impact of AEDs on the invasion and metastasis of various forms of cancer, while addressing their associated side effects. For example, valproic acid inhibits histone deacetylase, causing hyperacetylation of genes, especially those regulating cell cycle, culminating in cell cycle arrest. Topiramate inhibits carbonic anhydrase, thus disrupting the acidic microenvironment needed for cancer cells to thrive. Lacosamide increases the slow inactivation of the voltage gated Na+ channel, thus inhibiting the growth, proliferation, and metastasis of many cancers. Although drug development is a complex task due to regulatory, intellectual property, and economic challenges, researchers are exploring drug repurposing tactics to overcome these challenges and to find new therapeutic alternatives for diseases like cancer. Thus, drug repurposing is considered among the most effective ways to develop drug candidates using novel properties and therapeutic characteristics, and this review also discusses these issues.
Collapse
Affiliation(s)
- Noor Tarawneh
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan;
| | - Shaymaa A. Hussein
- Department of Biological Sciences, School of Science, The University of Jordan, Amman 11942, Jordan;
| | - Shtaywy Abdalla
- Department of Biological Sciences, School of Science, The University of Jordan, Amman 11942, Jordan;
| |
Collapse
|
13
|
Onali A, Sanna E, Lupia A, Secci D, Atzeni G, Demuru L, Angeli A, Cottiglia F, Meleddu R, Emmolo R, Corona A, Maccioni E, Supuran CT, Distinto S. Synthesis and Evaluation of Thiazolidinone-Isatin Hybrids for Selective Inhibition of Cancer-Related Carbonic Anhydrases. ACS Med Chem Lett 2025; 16:560-566. [PMID: 40236560 PMCID: PMC11995222 DOI: 10.1021/acsmedchemlett.4c00599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 04/17/2025] Open
Abstract
A small library of novel thiazolidinone-based sulfonamide derivatives was designed, synthesized and evaluated for their ability to target human carbonic anhydrase (hCA) isoforms IX and XII, which are overexpressed in malignant cells and play a key role in metastasis and therapeutic response of cancer cells. A molecular hybridization approach was employed to design the molecules by combining different moieties identified as having antitumor activity. The thiazolidinone core was functionalized with benzenesulfonamide as a zinc-binding group and different isatin derivatives to enhance the chemical profile and optimize the hydrophilic/lipophilic balance. Biological evaluation against hCA I, II, IX and XII isoforms showed promising inhibitory activities, and some compounds exhibited selectivity and high inhibitory activity against hCA IX and hCA XII while not affecting off-target hCA I and hCA II. In particular, compound 3h demonstrated high selectivity with Ki values of 57.8 nM for hCA IX and 44.3 nM for hCA XII.
Collapse
Affiliation(s)
- Alessia Onali
- Department
of Life and Environmental Sciences, University
of Cagliari, Cittadella Universitaria sp8, 09042 Monserrato, Cagliari, Italy
| | - Erica Sanna
- Department
of Life and Environmental Sciences, University
of Cagliari, Cittadella Universitaria sp8, 09042 Monserrato, Cagliari, Italy
| | - Antonio Lupia
- Department
of Life and Environmental Sciences, University
of Cagliari, Cittadella Universitaria sp8, 09042 Monserrato, Cagliari, Italy
| | - Daniela Secci
- Faculty
of Pharmacy, University of Lubiana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Giulia Atzeni
- Department
of Life and Environmental Sciences, University
of Cagliari, Cittadella Universitaria sp8, 09042 Monserrato, Cagliari, Italy
| | - Laura Demuru
- Department
of Life and Environmental Sciences, University
of Cagliari, Cittadella Universitaria sp8, 09042 Monserrato, Cagliari, Italy
| | - Andrea Angeli
- Dipartimento
NEUROFARBA, Sezione di Scienze Farmaceutiche, Università degli Studi di Firenze, 50019 Sesto Fiorentino, Florence, Italy
| | - Filippo Cottiglia
- Department
of Life and Environmental Sciences, University
of Cagliari, Cittadella Universitaria sp8, 09042 Monserrato, Cagliari, Italy
| | - Rita Meleddu
- Department
of Life and Environmental Sciences, University
of Cagliari, Cittadella Universitaria sp8, 09042 Monserrato, Cagliari, Italy
| | - Roberta Emmolo
- Department
of Life and Environmental Sciences, University
of Cagliari, Cittadella Universitaria sp8, 09042 Monserrato, Cagliari, Italy
| | - Angela Corona
- Department
of Life and Environmental Sciences, University
of Cagliari, Cittadella Universitaria sp8, 09042 Monserrato, Cagliari, Italy
| | - Elias Maccioni
- Department
of Life and Environmental Sciences, University
of Cagliari, Cittadella Universitaria sp8, 09042 Monserrato, Cagliari, Italy
| | - Claudiu T. Supuran
- Dipartimento
NEUROFARBA, Sezione di Scienze Farmaceutiche, Università degli Studi di Firenze, 50019 Sesto Fiorentino, Florence, Italy
| | - Simona Distinto
- Department
of Life and Environmental Sciences, University
of Cagliari, Cittadella Universitaria sp8, 09042 Monserrato, Cagliari, Italy
| |
Collapse
|
14
|
Hashem H, Abdelfattah S, Hassan HM, Al-Emam A, Alqarni M, Alotaibi G, Radwan IT, Kaur K, Rao DP, Bräse S, Alkhammash A. Discovery of a novel 4-pyridyl SLC-0111 analog targeting tumor-associated carbonic anhydrase isoform IX through tail-based design approach with potent anticancer activity. Front Chem 2025; 13:1571646. [PMID: 40255643 PMCID: PMC12006758 DOI: 10.3389/fchem.2025.1571646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/11/2025] [Indexed: 04/22/2025] Open
Abstract
Introduction: Carbonic anhydrase IX (CA IX) is a tumor-associated enzyme involved in cancer progression and survival. Targeting CA IX with selective inhibitors like SLC-0111 has shown therapeutic potential. This study aimed to develop a novel 4-pyridyl analog (Pyr) of SLC-0111 with enhanced anticancer activity. Methods: Pyr was synthesized using a tail-based design and characterized by NMR. Its cytotoxicity was tested against cancer and normal cell lines. CA inhibition, cell cycle effects, apoptosis induction, and protein expression changes were evaluated. Molecular docking and ADMET predictions assessed binding and drug-like properties. Results and Discussion: Pyr showed selective cytotoxicity toward cancer cells and potent CA IX inhibition. It induced G0/G1 arrest, apoptosis, and modulated p53, Bax, and Bcl-2 levels. Docking confirmed strong CA IX binding, and ADMET analysis indicated good oral bioavailability. These results support Pyr as a promising anticancer candidate.
Collapse
Affiliation(s)
- Hamada Hashem
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Sohag University, Sohag, Egypt
| | - Shadwa Abdelfattah
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Merit University (MUE), Sohag, Egypt
| | - Hesham M. Hassan
- Department of Pathology, College of Medicine, King Khalid University, Asir, Saudi Arabia
| | - Ahmed Al-Emam
- Department of Pathology, College of Medicine, King Khalid University, Asir, Saudi Arabia
| | - Mohammed Alqarni
- Department of Pharmaceutical chemistry, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Ghallab Alotaibi
- Department of Pharmacology, College of Pharmacy, Shaqra University, Shaqra, Saudi Arabia
| | - Ibrahim Taha Radwan
- Supplementary General Sciences Department, Faculty of Oral and Dental Medicine, Future University in Egypt, Cairo, Egypt
| | - Kirandeep Kaur
- Department of Chemistry, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Devendra Pratap Rao
- Coordination Chemistry Laboratory, Department of Chemistry, Dayanand Anglo-Vedic (PG) College, Kanpur, Uttar Pradesh, India
| | - Stefan Bräse
- Institute of Biological and Chemical Systems, Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Abdullah Alkhammash
- Department of Pharmacology, College of Pharmacy, Shaqra University, Shaqra, Saudi Arabia
| |
Collapse
|
15
|
Barata PC, Zarrabi KK, Bex A, Grivas P, Hermann K, Hofman MS, Li R, Lopez-Beltran A, Padani AR, Powles T, Taplin ME, Loriot Y. Novel Methods to Assess Tumor Burden and Minimal Residual Disease in Genitourinary Cancers. Eur Urol 2025; 87:412-423. [PMID: 39638730 DOI: 10.1016/j.eururo.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/23/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND AND OBJECTIVE Advances in molecular diagnostics have ushered in a new era for patients with prostate, renal, and urothelial cancers, with novel radiographic and molecular modalities for the assessment of disease burden and minimal residual disease (MRD). Conventional imaging has a limited threshold for disease detection and is often unable to discern clinically occult disease with varying risks of false-negative or false-positive findings depending on the disease state and type of imaging. METHODS We provide an overview of emerging radiographic and molecular tools in development within the genitourinary (GU) disease space. A literature review of contemporary basic, translational, and clinical research studies was performed, covering the timeframe of 1980-2024 through the MEDLINE (via PubMed) and Scopus databases. We highlight select examples of emerging technologies and biomarker-informed clinical trials, which aim to quantify disease at lower thresholds and have the potential for integrating MRD in clinical practice for GU patients. KEY FINDINGS AND LIMITATIONS The development of novel radiotracers, such as prostate-specific membrane antigen or carbonic anhydrase IX, is being evaluated in both clinical practice and trial setting, aiming to change the management of these tumors. Molecular tools including circulating tumor cells and byproducts such as plasma and urine cell-free circulating tumor DNA provide the opportunity for MRD detection. MRD capture on single-cell or cellular byproducts can serve as a conduit for genomic and transcriptomic analyses, providing insight into the molecular underpinnings and clonal evolution of disease. CONCLUSIONS AND CLINICAL IMPLICATIONS While the full potential for MRD applications has yet to be realized, we are witnessing the emergence of novel techniques aimed at MRD detection and the rapid development of elegantly designed studies implementing iterative detection of MRD as means to provide biological rationale and tailor therapeutic options in GU tumors.
Collapse
Affiliation(s)
- Pedro C Barata
- Division of Solid Tumor Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, USA.
| | - Kevin K Zarrabi
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Axel Bex
- The Royal Free London NHS Foundation Trust, London, UK; UCL Division of Surgery and Interventional Science, University College London, London, UK; Department of Urology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Petros Grivas
- Department of Medicine, Division of Hematology Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Ken Hermann
- Department of Nuclear Medicine, University of Duisburg-Essen, German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Michael S Hofman
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Roger Li
- Department of GU Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Antonio Lopez-Beltran
- Department of Morphological Sciences, Unit of Anatomic Pathology, University of Cordoba Medical School, Cordoba, Spain
| | - Anwar R Padani
- Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, London, UK
| | - Thomas Powles
- Barts Cancer Institute, Experimental Cancer Medicine Centre, Queen Mary University of London, St. Bartholomew's Hospital, London, UK
| | - Mary-Ellen Taplin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Yohann Loriot
- Department of Cancer Medicine and INSERM U981, Université Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| |
Collapse
|
16
|
Petrosiute A, Zakšauskas A, Lučiūnaitė A, Petrauskas V, Baranauskienė L, Kvietkauskaitė A, Ščerbavičienė A, Tamošiūnaitė M, Musvicaitė J, Jankūnaitė A, Žvinys G, Stančaitis L, Čapkauskaitė E, Mickevičiūtė A, Juozapaitienė V, Dudutienė V, Zubrienė A, Grincevičienė Š, Bukelskienė V, Schiöth HB, Matulienė J, Matulis D. Carbonic anhydrase IX inhibition as a path to treat neuroblastoma. Br J Pharmacol 2025; 182:1610-1629. [PMID: 39776083 DOI: 10.1111/bph.17429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/12/2024] [Accepted: 11/23/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND AND PURPOSE Tumour hypoxia frequently presents a major challenge in the treatment of neuroblastoma (NBL). The neuroblastoma cells produce carbonic anhydrase IX (CA IX), an enzyme crucial for the survival of cancer cells in low-oxygen environments. EXPERIMENTAL APPROACH We designed and synthesised a novel high-affinity inhibitor of CA IX. The highest to-date. The affinities were determined for all human catalytically active CA isozymes showing significant selectivity for CA IX over other isozymes. The inhibitor effect on neuroblastoma cancer cell growth was determined in vitro and in vivo via a mice xenograft model. KEY RESULTS The novel designed inhibitor effectively mitigated the acidification induced by CA IX and reduced spheroid growth under hypoxic conditions in the SK-N-AS cell line. It also diminished the secretion of pro-tumour chemokines IL-8 (CXCL2) and CCL2. When we combined this novel CA IX inhibitor with a compound that inhibits the chemokine receptor CCR2 protein activity, we observed a reduction in mouse tumour growth. The combined treatment also prompted tumours to exhibit adaptive resistance by producing higher levels of vascular endothelial growth factor receptors (VEGFR) and other compensatory signals. CONCLUSIONS AND IMPLICATIONS This research underscores the pivotal role of CA IX in cancer and the potential of a novel CA IX inhibitor-based combination intervention therapy for neuroblastoma treatment.
Collapse
Affiliation(s)
- Agne Petrosiute
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Audrius Zakšauskas
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Asta Lučiūnaitė
- Department of Immunology, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Vytautas Petrauskas
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Lina Baranauskienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Agnė Kvietkauskaitė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Alvilė Ščerbavičienė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Marta Tamošiūnaitė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Justina Musvicaitė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Alberta Jankūnaitė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Gediminas Žvinys
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Laimonas Stančaitis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Edita Čapkauskaitė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Aurelija Mickevičiūtė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Vaida Juozapaitienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Virginija Dudutienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Asta Zubrienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Švitrigailė Grincevičienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Virginija Bukelskienė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Helgi B Schiöth
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Jurgita Matulienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Daumantas Matulis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
17
|
Gore M, Kabekkodu SP, Chakrabarty S. Exploring the metabolic alterations in cervical cancer induced by HPV oncoproteins: From mechanisms to therapeutic targets. Biochim Biophys Acta Rev Cancer 2025; 1880:189292. [PMID: 40037419 DOI: 10.1016/j.bbcan.2025.189292] [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: 09/12/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
The role of human Papillomavirus (HPV) in metabolic reprogramming is implicated in the development and progression of cervical cancer. During carcinogenesis, cancer cells modify various metabolic pathways to generate energy and sustain their growth and development. Cervical cancer, one of the most prevalent malignancies affecting women globally, involves metabolic alterations such as increased glycolysis, elevated lactate production, and lipid accumulation. The oncoproteins, primarily E6 and E7, which are encoded by high-risk HPVs, facilitate the accumulation of several cancer markers, promoting not only the growth and development of cancer but also metastasis, immune evasion, and therapy resistance. HPV oncoproteins interact with cellular MYC (c-MYC), retinoblastoma protein (pRB), p53, and hypoxia-inducible factor 1α (HIF-1α), leading to the induction of metabolic reprogramming and favour the Warburg effect. Metabolic reprogramming enables HPV to persist for an extended period and accelerates the progression of cervical cancer. This review summarizes the role of HPV oncoproteins in metabolic reprogramming and their contributions to the development and progression of cervical cancer. Additionally, this review provides insights into how metabolic reprogramming opens avenues for novel therapeutic strategies, including the discovery of new and repurposed drugs that could be applied to treat cervical cancer.
Collapse
Affiliation(s)
- Mrudula Gore
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Sanjiban Chakrabarty
- Department of Public Health Genomics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| |
Collapse
|
18
|
Wang Y, Zhou H, Ju S, Dong X, Zheng C. The solid tumor microenvironment and related targeting strategies: a concise review. Front Immunol 2025; 16:1563858. [PMID: 40207238 PMCID: PMC11979131 DOI: 10.3389/fimmu.2025.1563858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/12/2025] [Indexed: 04/11/2025] Open
Abstract
The malignant tumor is a serious disease threatening human life. Increasing studies have confirmed that the tumor microenvironment (TME) is composed of a variety of complex components that precisely regulate the interaction of tumor cells with other components, allowing tumor cells to continue to proliferate, resist apoptosis, evade immune surveillance and clearance, and metastasis. However, the characteristics of each component and their interrelationships remain to be deeply understood. To target TME, it is necessary to deeply understand the role of various components of TME in tumor growth and search for potential therapeutic targets. Herein, we innovatively classify the TME into physical microenvironment (such as oxygen, pH, etc.), mechanical microenvironment (such as extracellular matrix, blood vessels, etc.), metabolic microenvironment (such as glucose, lipids, etc.), inflammatory microenvironment and immune microenvironment. We introduce a concise but comprehensive classification of the TME; depict the characteristics of each component in TME; summarize the existing methods for detecting each component in TME; highlight the current strategies and potential therapeutic targets for TME; discuss current challenges in presenting TME and its clinical applications; and provide our prospect on the future research direction and clinical benefits of TME.
Collapse
Affiliation(s)
- Yingliang Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, China
| | - Huimin Zhou
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuguang Ju
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, China
| | - Xiangjun Dong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, China
| |
Collapse
|
19
|
Al-Mansour FSH, Almasoudi HH, Albarrati A. Mapping molecular landscapes in triple-negative breast cancer: insights from spatial transcriptomics. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04057-3. [PMID: 40119898 DOI: 10.1007/s00210-025-04057-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
The tumor microenvironment (TME) of triple-negative breast cancer (TNBC) is a highly heterogeneous and very aggressive form of the disease that has few suitable treatment options; however, spatial transcriptomics (ST) is a powerful tool for elucidation of the TME in TNBC. Because of its spatial context preservation, ST has a unique capability to map tumor-stroma interactions, immune infiltration, and therapy resistance mechanisms (which are key to understanding TNBC progression), compared with conventional transcriptomics. This review shows the use of ST in TNBC, its utilization in spatial biomarker identification, intratumoral heterogeneity definition, molecular subtyping refinement, and prediction of immunotherapy responses. Recent insight from ST-driven insights has explained the key spatial patterns on immune evasion, chemotherapy resistance, racial disparities of TNBC, and aspects for patient stratification and therapeutic decision. With the integration of ST with the subjects of proteomics and imaging mass cytometry, this approach has been enlarged and is now applied in precision medicine and biomarker discovery. Recently, advancements in AI-based spatial analysis for tumor classification, identification of biomarkers, and creation of therapy prediction models have occurred. However, continued developments in ST technologies, computational tools, and partnerships amongst multiple centers to facilitate the integration of ST into clinical routine practice are needed to unlock novel therapeutic targets.
Collapse
Affiliation(s)
- Fares Saeed H Al-Mansour
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Hassan H Almasoudi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Ali Albarrati
- Rehabilitation Sciences Department, College of Applied Medical Sciences, King Saud University, 11451, Riyadh, Saudi Arabia.
| |
Collapse
|
20
|
Torres SW, Lan C, Harthorn A, Schmitz Z, Blanchard PL, Collins J, Hackel BJ. Molecular Determinants of Affinity and Isoform Selectivity in Protein─Small Molecule Hybrid Inhibitors of Carbonic Anhydrase. Bioconjug Chem 2025; 36:549-562. [PMID: 40030409 DOI: 10.1021/acs.bioconjchem.5c00006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Multiple studies have demonstrated the benefit of engineering hybrid ligands that combine the unique benefits of small molecules and proteins or peptides. However, the molecular complexity of hybrid ligands generates a parameter space so large it cannot be exhaustively explored. We systematically evaluated the impact of one molecular design element, conjugation site, on the discovery of functional protein-small molecule hybrids (PriSMs). We utilized a library of yeast-displayed fibronectin domain variants with amino acid and loop length diversity in the paratope and a single cysteine at one of 18 possible conjugation sites. The protein variants were coupled with maleimide-functionalized acetazolamide and sorted via competitive flow cytometry to discover potent and selective inhibitors of three isoforms of carbonic anhydrase. Deep sequencing of the resultant populations of functional PriSMs revealed an isoform-dependent distribution of conjugation site preferences. The top PriSMs showed potency and selectivity gains up to 23- and 100-fold (in this case, for CA-II vs CA-XII, with a 43-fold selectivity gain for CA-II vs CA-IX) relative to PEG2-acetazolamide alone. The presented study expands our fundamental understanding of the role of conjugation site in PriSM function and informs future PriSM engineering efforts by highlighting the benefit of conjugation site diversity in PriSM libraries.
Collapse
Affiliation(s)
- Sarah W Torres
- Department of Biomedical Engineering, University of Minnesota─Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Crystal Lan
- Department of Chemical Engineering and Materials Science, University of Minnesota─Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Abbigael Harthorn
- Department of Biomedical Engineering, University of Minnesota─Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Zachary Schmitz
- Department of Chemical Engineering and Materials Science, University of Minnesota─Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Paul L Blanchard
- Department of Chemical Engineering and Materials Science, University of Minnesota─Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Jon Collins
- Department of Biomedical Engineering, University of Minnesota─Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Benjamin J Hackel
- Department of Biomedical Engineering, University of Minnesota─Twin Cities, Minneapolis, Minnesota 55455, United States
- Department of Chemical Engineering and Materials Science, University of Minnesota─Twin Cities, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
21
|
Saravanan V, Palani SP, Chagaleti BK, Gao QZ, Valsaladevi AG, Kumaradoss KM. Molecular dynamics simulation reveals structural insights into isozyme selectivity of carbonic anhydrase XII inhibitors in hypoxic tumor microenvironment. Biochem Biophys Res Commun 2025; 753:151471. [PMID: 39965264 DOI: 10.1016/j.bbrc.2025.151471] [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: 09/19/2024] [Revised: 12/23/2024] [Accepted: 02/08/2025] [Indexed: 02/20/2025]
Abstract
Human carbonic anhydrase (CA) isoenzymes IX and XII are overexpressed in cancer cells, contributing to tumor microenvironment acidification and representing important targets for cancer therapy. In this study, we identified compound V35 (ZINC09419065) as a selective inhibitor of CA IX and CA XII with enhanced binding stability and selectivity compared to standard inhibitors. We analyzed conserved regions in CA I, CA II, CA IX, and CA XII to investigate their isozyme selectivity, revealing critical selectivity determinants at positions 95, 141, and 203. Molecular docking results indicated that V35 interacts robustly with CA XII, forming a metal ion coordination complex with Zn via HIS94, HIS96, HIS119, and THR199, similar to the interaction pattern of standard inhibitor SLC-0111. Molecular dynamics (MD) simulations conducted over 500 ns under hypoxic conditions showed that V35 has high binding stability, with root mean square deviation (RMSD) and fluctuation (RMSF) values comparable to SLC-0111, demonstrating its conformational stability in CA XII. Binding free energy calculations using the MMGBSA method showed that V35 achieves binding free energy of -44.17 kcal/mol with CA XII, closely matching SLC-0111 (-49.41 kcal/mol). Density functional theory (DFT) calculations further highlighted V35's electrostatic potential distribution, supporting its isozyme selectivity. Post-dynamics analysis indicated that the ester functional groups and the inward movement of HIS64 stabilize V35's interactions in CA XII, a feature absent in CA I.
Collapse
Affiliation(s)
- Venkatesan Saravanan
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, 603203, India
| | - Sathiya Priya Palani
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, 603203, India
| | - Bharath Kumar Chagaleti
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, 603203, India
| | - Quan-Ze Gao
- National Applied Research Laboratories, National Centre for High-Performance Computing, Hsinchu City, 30076, Taiwan
| | - Anjana Gopi Valsaladevi
- Dr APJ Abdul Kalam Lab, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, 603203, India.
| | - Kathiravan Muthu Kumaradoss
- Dr APJ Abdul Kalam Lab, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, 603203, India.
| |
Collapse
|
22
|
Choi SH, Chen YW, Panian J, Yuen K, McKay RR. Emerging innovative treatment strategies for advanced clear cell renal cell carcinoma. Oncologist 2025; 30:oyae276. [PMID: 39401004 PMCID: PMC11954509 DOI: 10.1093/oncolo/oyae276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024] Open
Abstract
Dramatic advances in biological discoveries, since the 1990s, have continued to reshape the treatment paradigm of metastatic renal cell carcinoma (RCC). Von Hippel Lindau (VHL) gene alterations are associated with pro-angiogenic activity and are central to the pathogenesis of clear cell RCC (ccRCC), the most predominant histologic subtype of RCC. Antiangiogenic strategies revolving around this VHL/HIF/VEGF axis have been shown to improve survival in metastatic ccRCC. The discovery of immune checkpoints and agents that target their inhibition introduced a new treatment paradigm for patients with RCC. While initially approved as monotherapy, studies investigating immune checkpoint inhibitor combinations have led to their approval as the new standard of care, providing durable responses and unprecedented improvements in clinical outcome. Despite these advances, the projected 14 390 deaths in 2024 from RCC underscore the need to continue efforts in expanding and optimizing treatment options for patients with metastatic RCC. This article reviews key findings that have transformed the way we understand and treat metastatic RCC, in addition to highlighting novel treatment strategies that are currently under development.
Collapse
Affiliation(s)
- Sharon H Choi
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
| | - Yu-Wei Chen
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
| | - Justine Panian
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
| | - Kit Yuen
- Department of Urology, University of California San Diego, San Diego, CA, United States
| | - Rana R McKay
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
- Department of Urology, University of California San Diego, San Diego, CA, United States
| |
Collapse
|
23
|
Moi D, Carradori S, Gallorini M, Mencarelli N, Deplano A, Angeli A, Vittorio S, Supuran CT, Onnis V. Investigation on Human Carbonic Anhydrase IX and XII Inhibitory Activity and A549 Antiproliferative Activity of a New Class of Coumarinamides. Pharmaceuticals (Basel) 2025; 18:372. [PMID: 40143148 PMCID: PMC11944513 DOI: 10.3390/ph18030372] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 02/27/2025] [Accepted: 03/01/2025] [Indexed: 03/28/2025] Open
Abstract
Background-Aggressive solid tumors are commonly characterized by both basic intracellular pH and acidic extracellular pH, which increase cell survival and proliferation. As carbonic anhydrases IX/XII are involved in this pH regulation, their inhibition is an appealing approach in cancer therapy, avoiding cancer cell survival and proliferation. Substituted coumarins are selective non-classical CA IX and CA XII inhibitors. Methods-In this study, new 7-hydroxycoumarinamides were synthesized and assayed for CA inhibition and antiproliferative activity. Results-All of the coumarinamides showed human CA IX and CA XII selective inhibition over the off-target CA I and CA II isoforms. Coumarin acts as a suicide inhibitor because its heterocyclic ring can be hydrolyzed by CA esterase activity to give the corresponding 2-hydroxycinnamic acid derivative which blocks the entrance of the active site. The 2-hydroxycinnamic acid derivatives deriving from the most potent and selective coumarinamides were docked into CA IX and XII to better understand the activity and selectivity against the two CA isoforms. The most active coumarinamides also produced a decrease of A549 cell proliferation and were able to arrest cells at the G1/S checkpoint. Conclusions-These results may open new perspectives for developing coumarin-based CA IX/XII inhibitors.
Collapse
Affiliation(s)
- Davide Moi
- Department of Life and Environmental Sciences, Unit of Pharmaceuitical, Pharmacological and Nutraceutical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, I-09042 Monserrato, CA, Italy; (D.M.); (A.D.)
| | - Simone Carradori
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, I-66100 Chieti, CH, Italy; (S.C.); (M.G.); (N.M.)
| | - Marialucia Gallorini
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, I-66100 Chieti, CH, Italy; (S.C.); (M.G.); (N.M.)
| | - Noemi Mencarelli
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, I-66100 Chieti, CH, Italy; (S.C.); (M.G.); (N.M.)
| | - Alberto Deplano
- Department of Life and Environmental Sciences, Unit of Pharmaceuitical, Pharmacological and Nutraceutical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, I-09042 Monserrato, CA, Italy; (D.M.); (A.D.)
| | - Andrea Angeli
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence, Via Ugo Schiff 6, I-50019 Sesto Fiorentino, FI, Italy; (A.A.); (C.T.S.)
| | - Serena Vittorio
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, I-20133 Milano, MI, Italy;
| | - Claudiu T. Supuran
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence, Via Ugo Schiff 6, I-50019 Sesto Fiorentino, FI, Italy; (A.A.); (C.T.S.)
| | - Valentina Onnis
- Department of Life and Environmental Sciences, Unit of Pharmaceuitical, Pharmacological and Nutraceutical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, I-09042 Monserrato, CA, Italy; (D.M.); (A.D.)
| |
Collapse
|
24
|
Yang L, Guo W, Ding H, Gao X, Xu Y, Wang M, Yang X, Zhao Y, Wang W, Liu W, Jia F, Hou D, Nanding A, Cheng L, Meng H, Wang K. Evaluation of the safety, biodistribution, dosimetry of [ 18F]AlF-NYM005 and initial experience in clear cell renal cell carcinoma: an interim analysis of a prospective trial. Eur J Nucl Med Mol Imaging 2025; 52:1354-1369. [PMID: 39676103 DOI: 10.1007/s00259-024-07007-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 11/24/2024] [Indexed: 12/17/2024]
Abstract
PURPOSE This first-in-human study aimed to evaluate the radiation dosimetry and whole-body biodistribution of [18F]AlF-NYM005, a novel small-molecule carbonic anhydrase IX (CAIX) targeting agent, and to investigate its ability to detect CAIX-positive tumors using PET scans in a cohort of clear cell renal cell carcinoma (ccRCC) patients. METHODS [18F]AlF-NYM005 was synthesized using a fully automatic cassette module Mortenon M1 (Nuoyu, China). Thirty-five patients with a suspicious lesion considered primary renal malignancy or a history of ccRCC were prospectively recruited and studied. All patients underwent [18F]AlF-NYM005 PET/CT examinations and the maximum standardized uptake value (SUVmax) was measured on conventional [18F]AlF-NYM005 PET/CT images. Among these patients, five patients underwent dynamic [18F]AlF-NYM005 PET/CT scanning (120 min) of the lower abdomen. Another subset of five ccRCC patients underwent sequential whole-body PET scans at 30, 60, 90, and 120 min (one of the five patients underwent additional 150 min and 180 min scans) after [18F]AlF-NYM005 injection to assess biodistribution and dosimetry. The influx constant (Ki) was calculated from the dynamic [18F]AlF-NYM005 PET/CT data using the Patlak model. Whole-body biodistribution was calculated as time-activity curves (TACs) describing dynamic uptake patterns in the patients' major organs, followed by calculation of tracer kinetics and cumulative organ activity. Effective doses of [18F]AlF-NYM005 and individual organ doses were also calculated. RESULTS [18F]AlF-NYM005 was successfully synthesized with a radiochemical purity of > 95% and an average labeling yield of 36.5 ± 8.3%. All patients tolerated the PET examinations well, and no adverse side effects were observed. The total body effective dose was 7.6E-03 mSv/MBq. The highest agent uptake was observed in the kidneys, stomach, and liver, contributing to an effective dose of 0.0126 ± 0.0029 mSv/MBq. The TACs showed optimal normal organ uptake with high tumor uptake and long retention of up to 2 h post-injection. Notably, a rapid increase of the tracer followed by a rapid decrease in the blood pool, kidney, liver, and tumor lesions was observed, indicating that [18F]AlF-NYM005 was rapidly eliminated from blood and urine. For the kinetic data analysis, the Ki for the primary kidney lesions had a mean of 0.082 ± 0.057 ml/g/min. The CAIX-positive tumors displayed rapid uptake, and all lesions were detectable within 30 min, with no additional lesions observed in the subsequent multi-time point scans. The patient-level sensitivity, specificity, and accuracy of [18F]AlF-NYM005 PET/CT were 93.8%, 75.0%, and 90% for group 1 and 92.3%, 100%, and 93.3% for group 2, respectively. For per-lymph node analysis, [18F]AlF-NYM005 PET/CT demonstrated 92.9% sensitivity, 90.5% specificity, and 91.8% accuracy in diagnosing metastatic lymph nodes. For per-distant metastasis analysis, it showed 90.5% sensitivity, 91.3% specificity, and 90.6% accuracy. The SUVmax of [18F]AlF-NYM005 PET/CT for primary ccRCC lesions was 15.5 ± 7.35. Tumor uptake was positive correlated with immunohistochemical staining findings. CONCLUSION This pilot study in ccRCC patients has demonstrated the safety, acceptable radiation dosimetry, favorable biodistribution, and exceptional tumor uptake of [18F]AlF-NYM005. The preliminary diagnostic study indicated the potential utility of [18F]AlF-NYM005 PET/CT, showing promising results in the diagnosis of primary or metastatic ccRCC. TRIAL REGISTRATION This study was registered at ClinicalTrial.gov (ChiCTR2200058108) as NYPILOT on 29 March, 2022.
Collapse
Affiliation(s)
- Liping Yang
- Department of PET-CT, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Wei Guo
- Department of PET-CT, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Hongchao Ding
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xing Gao
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuchao Xu
- School of Nuclear Science and Technology, University of South China, Hunan, China
| | - Menglu Wang
- Department of PET-CT, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Xinyue Yang
- Department of PET-CT, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Yue Zhao
- Department of PET-CT, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Wenzhi Wang
- Department of PET-CT, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Wei Liu
- Department of PET-CT, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Fan Jia
- Department of PET-CT, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Dayong Hou
- Department of PET-CT, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Abiyasi Nanding
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Liang Cheng
- Department of PET-CT, Harbin Medical University Cancer Hospital, Harbin, 150001, China.
| | - Hongxue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, 150001, China.
| | - Kezheng Wang
- Department of PET-CT, Harbin Medical University Cancer Hospital, Harbin, 150001, China.
| |
Collapse
|
25
|
Amiranda S, Succoio M, Anzilotti S, Cuomo O, Petrozziello T, Tedeschi V, Finizio A, Mele G, Parkkila S, Annunziato L, De Simone G, Pignataro G, Secondo A, Zambrano N. Pharmacological inhibition of carbonic anhydrases with a positively charged pyridinium sulfonamide phenocopies the neuroprotective effects of Car9 genetic ablation in a murine setting of oxygen/glucose deprivation followed by re-oxygenation and is associated with improved neuronal function in ischemic rats. Heliyon 2025; 11:e42457. [PMID: 40028587 PMCID: PMC11868941 DOI: 10.1016/j.heliyon.2025.e42457] [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: 06/03/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 03/05/2025] Open
Abstract
Carbonic anhydrases constitute a family of metalloenzymes vital for maintaining acid-base balance and regulating pH in physio-pathological processes. These findings suggest carbonic anhydrases as potential therapeutic targets for treating pH-associated disorders, including cerebral ischemia, to mitigate hypoxia- and reoxygenation-induced neuronal damage. A focus on carbonic anhydrase IX showed that ischemic stress altered subcellular distributions of this enzyme in rodent neuronal populations. Given the enzyme's canonical membrane localization, we implemented pharmacological inhibition using a membrane-impermeant sulfonamide inhibitor in neuronal models of brain ischemia. The treatments exerted neuroprotective effects on neurons from Car9 knockout mice. Moreover, administration of the sulfonamide inhibitor to rats subjected to transient middle cerebral artery occlusion decreased infarct volumes and improved neurological deficits. Our results support the involvement of carbonic anhydrase IX in postischemic damage and pave the way for possible pharmacological interventions with selective inhibitors in the management of brain ischemia.
Collapse
Affiliation(s)
- Sara Amiranda
- Dipartimento di Medicina molecolare e Biotecnologie mediche, Università degli Studi di Napoli Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore S.C.aR.L., Napoli, Italy
| | - Mariangela Succoio
- Dipartimento di Medicina molecolare e Biotecnologie mediche, Università degli Studi di Napoli Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore S.C.aR.L., Napoli, Italy
| | - Serenella Anzilotti
- Department of Human Sciences and Quality of Life Promotion, Università San Raffaele, Rome, Italy
| | - Ornella Cuomo
- Division of Pharmacology, Dipartimento di Neuroscienze e Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Tiziana Petrozziello
- Division of Pharmacology, Dipartimento di Neuroscienze e Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Valentina Tedeschi
- Division of Pharmacology, Dipartimento di Neuroscienze e Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Arianna Finizio
- Dipartimento di Medicina molecolare e Biotecnologie mediche, Università degli Studi di Napoli Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore S.C.aR.L., Napoli, Italy
| | - Giorgia Mele
- Dipartimento di Medicina molecolare e Biotecnologie mediche, Università degli Studi di Napoli Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore S.C.aR.L., Napoli, Italy
| | - Seppo Parkkila
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Fimlab Ltd, Tampere University Hospital, Tampere, Finland
| | | | - Giuseppina De Simone
- Istituto di Biostrutture e Bioimmagini, CNR, Via Pietro Castellino 111, 80131, Napoli, Italy
| | - Giuseppe Pignataro
- Division of Pharmacology, Dipartimento di Neuroscienze e Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Agnese Secondo
- Division of Pharmacology, Dipartimento di Neuroscienze e Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli Federico II, Napoli, Italy
| | - Nicola Zambrano
- Dipartimento di Medicina molecolare e Biotecnologie mediche, Università degli Studi di Napoli Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore S.C.aR.L., Napoli, Italy
| |
Collapse
|
26
|
Nencetti S, Cuffaro D, Ciccone L, Nocentini A, Di Stefano M, Poli G, Macchia M, Tuccinardi T, Nuti E, Supuran CT, Rossello A, Orlandini E. A series of benzensulfonamide derivatives as new potent carbonic anhydrase IX and XII inhibitors. Future Med Chem 2025; 17:271-285. [PMID: 39878534 PMCID: PMC11792798 DOI: 10.1080/17568919.2025.2453420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025] Open
Abstract
AIM Human carbonic anhydrases (hCAs) are involved in many physiological processes including respiration, pH control, ion transport, bone resorption, and gastric fluid secretion. Recently, CA IX and CA XII have been studied for their role in cancer diseases, motivating the design of inhibitors of these isoforms. MATERIAL AND METHOD Here, we used the tail approach to design a new series of monoaryl (1a-i) and bicyclic (1j-n) benzensulfonamide derivatives CA IX and CA XII inhibitors. All synthesized compounds were investigated toward a panel of hCAs, and most of them exhibited potent CA inhibitory activity for CA II, CA IX and CA XII with Ki values. In silico studies were performed to investigate the binding mode between inhibitors and CA. RESULTS AND CONCLUSION The best compound was 1i that showed a low nanomolar range of Ki value as CA inhibitor (Ki = 9.4, 5.6 and 6.3 nM hCA II, IX and XII, respectively).
Collapse
Affiliation(s)
| | | | - Lidia Ciccone
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Alessio Nocentini
- Department of Neurofarba, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Sesto Fiorentino, Italy
| | | | - Giulio Poli
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Marco Macchia
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | - Elisa Nuti
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Claudiu T. Supuran
- Department of Neurofarba, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Sesto Fiorentino, Italy
| | - Armando Rossello
- Department of Pharmacy, University of Pisa, Pisa, Italy
- Research Center “E. Piaggio” Università di, Pisa, Italy
| | - Elisabetta Orlandini
- Research Center “E. Piaggio” Università di, Pisa, Italy
- Department of Earth Sciences, University of Pisa, Pisa, Italy
| |
Collapse
|
27
|
Chen S, Shcherbina A, Schafer ST, Mattingly ZA, Ramesh J, Narayanan C, Banerjee S, Heath B, Regester M, Chen I, Thakurela S, Hallmayer J, O'Hara R, Solomon M, Nordahl CW, Amaral DG, Chetty S. Cellular mechanisms of early brain overgrowth in autistic children: elevated levels of GPX4 and resistance to ferroptosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635706. [PMID: 39975145 PMCID: PMC11838294 DOI: 10.1101/2025.01.30.635706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Autistic individuals with disproportionate megalencephaly (ASD-DM), characterized by enlarged brains relative to body height, have higher rates of intellectual disability and face more severe cognitive challenges than autistic children with average brain sizes. The cellular and molecular mechanisms underlying this neurophenotype remain poorly understood. To investigate these mechanisms, we generated human induced pluripotent stem cells from non-autistic typically developing children and autistic children with and without disproportionate megalencephaly. We assessed these children longitudinally from ages two to twelve years using magnetic resonance imaging and comprehensive cognitive and medical evaluations. We show that neural progenitor cells (NPCs) derived from ASD-DM children exhibit increased rates of cell survival and suppressed cell death, accompanied by heightened oxidative stress and ferrous iron accumulation. Despite these stressors, ASD-DM NPCs actively suppress apoptosis and ferroptosis by regulating proteins such as caspase-3 (CASP3), poly(ADP-ribose) polymerase 1 (PARP1), and glutathione peroxidase 4 (GPX4). Cellular ferroptotic signatures are further supported by elevated expression of selenocysteine genes, including GPX4 , in the blood of ASD-DM children and their mothers, suggesting potential hereditary or environmental influences. Furthermore, we show that peripheral expression of GPX4 and other selenocysteine genes correlate with cognitive outcomes (IQ). These findings underscore the role of ferroptosis in autism, pointing to potential diagnostic biomarkers and targets for intervention.
Collapse
|
28
|
Mehrotra S, Kaur N, Kaur S, Matharoo K, Pandey RK. From antibodies to nanobodies: The next frontier in cancer theranostics for solid tumors. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 144:287-329. [PMID: 39978969 DOI: 10.1016/bs.apcsb.2024.10.014] [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: 02/22/2025]
Abstract
The field of cancer therapeutics has witnessed significant advancements over the past decades, particularly with the emergence of immunotherapy. This chapter traces the transformative journey from traditional antibody-based therapies to the innovative use of nanobodies in the treatment and diagnosis of solid tumors. Nanobodies are the smallest fragments of antibodies derived from camelid immunoglobulins and have redefined the possibilities in cancer theranostics due to their unique structural and functional properties. We provide an overview of the biochemical characteristics of nanobodies that make them particularly suitable for theranostic applications, such as their small size, high stability, enhanced infiltration into the complex tumor microenvironment (TME) and ability to bind with high affinity to epitopes that are inaccessible to conventional antibodies. Further, their ease of modification and functionalization has enabled the development of nanobody-based drug conjugates/toxins and radiolabeled compounds for precise imaging and targeted radiotherapy. We elucidate how nanobodies are being served as valuable tools for prognostic assessment, enabling clinicians to predict disease aggressiveness, monitor treatment response, and stratify patients for personalized therapeutic interventions.
Collapse
Affiliation(s)
- Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India.
| | - Navdeep Kaur
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Sukhpreet Kaur
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Kawaljit Matharoo
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | | |
Collapse
|
29
|
Gong Q, Song X, Tong Y, Huo L, Zhao X, Han Y, Shen W, Ru J, Shen X, Liang C. Recent advances of anti-tumor nano-strategies via overturning pH gradient: alkalization and acidification. J Nanobiotechnology 2025; 23:42. [PMID: 39849540 PMCID: PMC11761731 DOI: 10.1186/s12951-025-03134-2] [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: 10/17/2024] [Accepted: 01/19/2025] [Indexed: 01/25/2025] Open
Abstract
The acidic tumor microenvironment, a hallmark of many solid tumors, is primarily induced by the high glycolytic rate of tumor cells. To avoid acidosis, tumor cells ingeniously maintain an acidic extracellular pH while keeping a relatively alkaline intracellular pH. Overturning the unique pH gradient of tumor cells has exhibited to be a viable approach for cancer therapy. In this review, the formation and regulatory mechanisms of the acidic microenvironment in solid tumors will be firstly outlined. Subsequently, we will comprehensively summarize the latest advancements in anti-tumor therapy via using nanomedicines to manipulate the tumor pH gradient, including acidifying intracellular environment and alkalizing extracellular environment. Following this, we will discuss the future potential of strategies employing nanomedicines to reverse tumor pH gradient. This review aims to foster research on therapeutic approaches targeting the pH regulation of solid tumors and holds an optimistic outlook for the future development of this field.
Collapse
Affiliation(s)
- Qiufang Gong
- Institute for Advanced Research, Cixi Biomedical Research Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xuejiao Song
- School of Physical and Mathematical Sciences, Nanjing Tech University (Nanjing Tech), Nanjing, 211816, China.
| | - Yao Tong
- Institute for Advanced Research, Cixi Biomedical Research Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Lixuan Huo
- Institute for Advanced Research, Cixi Biomedical Research Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xuefen Zhao
- Institute for Advanced Research, Cixi Biomedical Research Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yingying Han
- Institute for Advanced Research, Cixi Biomedical Research Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Wei Shen
- Institute for Advanced Research, Cixi Biomedical Research Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jiaxi Ru
- Institute for Advanced Research, Cixi Biomedical Research Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xian Shen
- Institute for Advanced Research, Cixi Biomedical Research Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Chao Liang
- Institute for Advanced Research, Cixi Biomedical Research Institute, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
30
|
Talani C, Olsson H, Roberg K, Wiechec E, Almangush A, Mäkitie AA, Farnebo L. Predicting Early Death in Head and Neck Cancer-A Pilot Study. Cancers (Basel) 2025; 17:302. [PMID: 39858084 PMCID: PMC11763563 DOI: 10.3390/cancers17020302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Background: The aim of this study was to evaluate biomarkers and biological characteristics of tumor biopsies from patients with head and neck cancer (HNC) to assess the risk of early death. Furthermore, we analyzed whether any combination of markers could be used for the prognostication of death within six months after cancer diagnosis. Materials and Methods: Patients diagnosed with HNC, receiving curative treatment decision at a multidisciplinary tumor board meeting, and who died within six months of diagnosis were included in this study. Nine patients who died within six months from diagnosis were identified and matched according to the tumor site and stage to seventeen patients who survived for at least two years. Results: The expression of markers was compared between the early-death patients and survivors. There was significantly higher Ki-67 expression in patients who died within six months than in those surviving for two years, with a mean difference of 21% (p = 0.038). A significant difference in cytoplasmic survivin expression was noted where early-death patients had increased expression compared to the survivors (p = 0.021). Furthermore, the intensity of survivin staining differed between the groups (p = 0.006). Conclusions: The results of this pilot study indicate that Ki67 and survivin could be potential prognostic biomarkers for early death in patients with HNC and possibly included in a panel of prognostic markers of value for treatment decision making.
Collapse
Affiliation(s)
- Charbél Talani
- Division of Sensory Organs and Communication, Department of Biomedical and Clinical Sciences, Linköping University, 581 83 Linköping, Sweden;
- Region Östergötland Anaesthetics, Operations and Specialty Surgery Center, Department of Otorhinolaryngology, 582 25 Linköping, Sweden; (K.R.)
| | - Hans Olsson
- Department of Pathology, Clinical and Experimental Medicine, Medical Faculty, Linköping University, 581 83 Linköping, Sweden;
| | - Karin Roberg
- Region Östergötland Anaesthetics, Operations and Specialty Surgery Center, Department of Otorhinolaryngology, 582 25 Linköping, Sweden; (K.R.)
- Division of Cell Biology, Department of Biomedical and Clinical Sciences, Linköping University, 581 83 Linköping, Sweden
| | - Emilia Wiechec
- Region Östergötland Anaesthetics, Operations and Specialty Surgery Center, Department of Otorhinolaryngology, 582 25 Linköping, Sweden; (K.R.)
- Division of Cell Biology, Department of Biomedical and Clinical Sciences, Linköping University, 581 83 Linköping, Sweden
| | - Alhadi Almangush
- Institute of Biomedicine, Pathology, University of Turku, 20014 Turku, Finland
- Department of Pathology, University of Helsinki, 00014 Helsinki, Finland
| | - Antti A. Mäkitie
- Division of Ear, Nose and Throat Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, 171 76 Stockholm, Sweden;
- Department of Otorhinolaryngology, Head and Neck Surgery, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Lovisa Farnebo
- Division of Sensory Organs and Communication, Department of Biomedical and Clinical Sciences, Linköping University, 581 83 Linköping, Sweden;
- Region Östergötland Anaesthetics, Operations and Specialty Surgery Center, Department of Otorhinolaryngology, 582 25 Linköping, Sweden; (K.R.)
| |
Collapse
|
31
|
Solmaz ÖA, Kutluer N, Bozan MB. Carbonic Anhydrase IX Enzyme in Triple Negative Breast Carcinoma: Relationship With Prognostic Factors and Response to Neoadjuvant Chemotherapy. Eur J Breast Health 2025; 21:57-62. [PMID: 39744916 PMCID: PMC11706115 DOI: 10.4274/ejbh.galenos.2024.2024-6-1] [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: 06/15/2024] [Accepted: 10/15/2024] [Indexed: 01/11/2025]
Abstract
Objective Triple negative breast carcinoma (TNBC) is characterized by the absence of estrogen receptor, progesterone receptor and human epidermal growth factor receptor-2 receptor expression. Carbonic anhydrase IX (CA IX) is a tumor-associated cell surface glycoprotein that is involved in adaptation to hypoxia-induced acidosis and plays a role in cancer progression. The aim of this study was to investigate CA IX expression in TNBC and its relationship with treatment effect. Materials and Methods Immunohistochemical staining was performed on tru-cut biopsy materials with CA IX antibody. Positive staining was graded as low (<10%) and high (>10%). In addition, the relationship between tumor diameter, histological grade and the treatment effect on mastectomy materials performed after neoadjuvant treatment was evaluated. Results TNBCs with positive staining for CA IX exhibited higher histological grade, and higher Ki-67 index compared to TNBCs with negative staining (p < 0.05). The response to treatment decreased as the degree of CA IX staining increased. There was no significant difference between the high staining group and low staining group in terms of patient age, tumor diameter and breast localisation. Conclusion CA IX enzyme is a poor prognostic marker in TNBC cases. However, overexpression of CA IX was associated with reduced response to treatment.
Collapse
Affiliation(s)
- Özgen Arslan Solmaz
- Department of Pathology, Elazığ Fethi Sekin City Hospital, University of Health Sciences Turkey, Elazığ, Turkey
| | - Nizamettin Kutluer
- Department of General Surgery, Elazığ Fethi Sekin City Hospital, University of Health Sciences Turkey, Elazığ, Turkey
| | - Mehmet Buğra Bozan
- Department of General Surgery, Elazığ Fethi Sekin City Hospital, University of Health Sciences Turkey, Elazığ, Turkey
| |
Collapse
|
32
|
Petrosiute A, Musvicaitė J, Petroška D, Ščerbavičienė A, Arnold S, Matulienė J, Žvirblienė A, Matulis D, Lučiūnaitė A. CCL2-CCR2 Axis Inhibition in Osteosarcoma Cell Model: The Impact of Oxygen Level on Cell Phenotype. J Cell Physiol 2025; 240:e31489. [PMID: 39587819 PMCID: PMC11747949 DOI: 10.1002/jcp.31489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/14/2024] [Accepted: 11/06/2024] [Indexed: 11/27/2024]
Abstract
Treatment of osteosarcoma is hampered by tumor hypoxia and requires alternative approaches. Although the CCL2-CCR2 axis is indispensable in tumor-induced inflammation and angiogenesis, its blockade has not been effective to date. This study aimed to characterize how CCR2 inhibition affects the crosstalk of osteosarcoma cells with immune cells to better delineate tumor resistance mechanisms that help withstand such treatment. In this study, 143B cells were exposed to healthy donor PBMC supernatants in a transwell assay lacking direct cell-to-cell contact and subjected to different oxygen concentrations. In addition, mice bearing orthotopic 143B tumors were subjected to CCR2 antagonist treatment. Our findings show that hypoxic conditions alter cytokine and cancer- related protein expression on cells and impair CCR2 antagonist effects in the experimental osteosarcoma model. CCL2-CCR2 axis blockade in the 143B xenografts, which are positive for hypoxia marker CAIX, did not slow 143B tumor growth or metastasis but altered tumor microenvironment by VEGFR downregulation and shift in the CD44-positive cell population towards high CD44 expression. This study highlights differential responses of tumor cells to CCR2 antagonists in the presence of different oxygen saturations and expands our knowledge of compensatory mechanisms leading to CCL2-CCR2 treatment resistance.
Collapse
Affiliation(s)
- Agne Petrosiute
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences CenterVilnius UniversityVilniusLithuania
| | - Justina Musvicaitė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences CenterVilnius UniversityVilniusLithuania
| | - Donatas Petroška
- National Center of PathologyAffiliate of Vilnius University Hospital Santaros KlinikosVilniusLithuania
| | - Alvilė Ščerbavičienė
- Department of Biological Models, Institute of Biochemistry, Life Sciences CenterVilnius UniversityVilniusLithuania
| | - Sascha Arnold
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences CenterVilnius UniversityVilniusLithuania
| | - Jurgita Matulienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences CenterVilnius UniversityVilniusLithuania
| | - Aurelija Žvirblienė
- Department of Immunology, Institute of Biotechnology, Life Sciences CenterVilnius UniversityVilniusLithuania
| | - Daumantas Matulis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences CenterVilnius UniversityVilniusLithuania
| | - Asta Lučiūnaitė
- Department of Immunology, Institute of Biotechnology, Life Sciences CenterVilnius UniversityVilniusLithuania
| |
Collapse
|
33
|
Abha Mishra KM, Kumari N, Carta F, Renzi G, Supuran CT, Sethi KK. Design, Synthesis, and In Vitro Evaluation of Aromatic Sulfonamides as Human Carbonic Anhydrase I, II, IX, and XII Inhibitors and Their Antioxidant Activity. J Biochem Mol Toxicol 2025; 39:e70135. [PMID: 39812110 DOI: 10.1002/jbt.70135] [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: 09/11/2024] [Revised: 12/09/2024] [Accepted: 12/29/2024] [Indexed: 01/30/2025]
Abstract
This study is focused on the design, synthesis, and evaluation of some sulfonamide derivatives for their inhibitory effects on human carbonic anhydrase (hCA) enzymes I, II, IX, and XII as well as for their antioxidant activity. The purity of the synthesized molecules was confirmed by the HPLC purity analysis and was found in the range of 93%-100%. The inhibition constant (Ki) against hCA I ranged from 0.75 nM to 1972 nM. The sulfonamides inhibited isoform hCA II significantly, with a Ki ranging from 0.09 to 56 nM. Similarly, the inhibitory effects on hCA IX and XII were found with Ki spanning from 27.8 to 2099 nM and 9.43 to 509 nM, respectively. Most of the synthesized compounds showed significant inhibition in comparison to standard drugs such as acetazolamide, ethoxzolamide, zonisamide, methazolamide, dorzolamide, and SLC-0111. Antioxidant activity was assessed using the DPPH assay, with compound 13 showing better antioxidant activity with an IC50 of 54.8 µg/mL, as compared to the standard ascorbic acid (IC50 64.7 µg/mL). The molecular docking studies provided insights into the binding modes of these compounds. The in silico physicochemical properties, pharmacokinetic/ADME, and toxicity properties evaluations confirmed favorable drug-likeness properties, complying with Lipinski's rule. These findings underscore the therapeutic potential of these compounds for the treatment of retinal/cerebral edema, glaucoma, edema, epilepsy management, high-altitude sickness, and cancer.
Collapse
Affiliation(s)
- K M Abha Mishra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Nutan Kumari
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Fabrizio Carta
- NEUROFARBA Department, Sezione di Farmaceutica e Nutraceutica, Università degli Studi di Firenze, Sesto Fiorentino, Italy
| | - Gioele Renzi
- NEUROFARBA Department, Sezione di Farmaceutica e Nutraceutica, Università degli Studi di Firenze, Sesto Fiorentino, Italy
| | - Claudiu T Supuran
- NEUROFARBA Department, Sezione di Farmaceutica e Nutraceutica, Università degli Studi di Firenze, Sesto Fiorentino, Italy
| | - Kalyan K Sethi
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Guwahati, India
| |
Collapse
|
34
|
Giovannuzzi S, Supuran CT. Lactonase activity of α-carbonic anhydrases allows identification of novel inhibitors. Arch Pharm (Weinheim) 2025; 358:e2400705. [PMID: 39651798 PMCID: PMC11704026 DOI: 10.1002/ardp.202400705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/11/2024]
Abstract
Lactones, a diverse and abundant class of molecules found in nature, exhibit a wide range of bioactivities, including anti-inflammatory, anticancer, and antibacterial effects. Among them, acyl homoserine lactones (AHSLs) play a crucial role in quorum sensing, influencing bacterial pathogenicity and biofilm formation in Gram-negative bacteria. Paraoxonases (PONs), calcium-containing enzymes known for their lactonase activity, have been shown to hydrolyze AHSLs and reduce the biofilm formation of several pathogenic bacteria. In this study, we explored the potential lactonase activity of a class of zinc(II) enzymes, the carbonic anhydrases (CAs), aiming to uncover new insights into their catalytic versatility. Using LC-MS and MS/MS analyses, we investigated the lactonase activity of CAs and assessed several lactones through a stopped-flow kinetic assay as substrates/inhibitors. Our findings reveal that lactones are novel "prodrug" inhibitors of CAs, with lactones DHC and 6 showing the most promising inhibition constants (KIs) in the low micromolar range against both human and bacterial isozymes.
Collapse
Affiliation(s)
- Simone Giovannuzzi
- NEUROFARBA Department, Pharmaceutical and Nutraceutical SectionUniversity of FlorenceFirenzeItaly
| | - Claudiu T. Supuran
- NEUROFARBA Department, Pharmaceutical and Nutraceutical SectionUniversity of FlorenceFirenzeItaly
| |
Collapse
|
35
|
Hoffmann S, Berger BT, Lucas LR, Schiele F, Park JE. Discovery of Carbonic Anhydrase 9 as a Novel CLEC2 Ligand in a Cellular Interactome Screen. Cells 2024; 13:2083. [PMID: 39768175 PMCID: PMC11674933 DOI: 10.3390/cells13242083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/13/2024] [Accepted: 12/13/2024] [Indexed: 01/30/2025] Open
Abstract
Membrane proteins, especially extracellular domains, are key therapeutic targets due to their role in cell communication and associations. Yet, their functions and interactions often remain unclear. This study presents a general method to discover interactions of membrane proteins with immune cells and subsequently to deorphanize their respective receptors. We developed a comprehensive recombinant protein library of extracellular domains of human transmembrane proteins and proteins found in the ER-Golgi-lysosomal systems. Using this library, we conducted a flow-cytometric screen that identified several cell surface binding events, including an interaction between carbonic anhydrase 9 (CAH9/CA9/CAIX) and CD14high cells. Further analysis revealed this interaction was indirect and mediated via platelets bound to the monocytes. CA9, best known for its diverse roles in cancer, is a promising therapeutic target. We utilized our library to develop an AlphaLISA high-throughput screening assay, identifying CLEC2 as one robust CA9 binding partner. A five-amino-acid sequence (EDLPT) in CA9, identical to a CLEC2 binding domain in Podoplanin (PDPN), was found to be essential for this interaction. Like PDPN, CA9-induced CLEC2 signaling is mediated via Syk. A Hodgkin's lymphoma cell line (HDLM-2) endogenously expressing CA9 can activate Syk-dependent CLEC2 signaling, providing enticing evidence for a novel function of CA9 in hematological cancers. In conclusion, we identified numerous interactions with monocytes and platelets and validated one, CA9, as an endogenous CLEC2 ligand. We provide a new list of other putative CA9 interaction partners and uncovered CA9-induced CLEC2 activation, providing new insights for CA9-based therapeutic strategies.
Collapse
Affiliation(s)
- Sebastian Hoffmann
- Division of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach, Germany (L.R.L.)
| | - Benedict-Tilman Berger
- Division of High-Throughput Biology, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach, Germany; (B.-T.B.); (F.S.)
| | - Liane Rosalie Lucas
- Division of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach, Germany (L.R.L.)
| | - Felix Schiele
- Division of High-Throughput Biology, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach, Germany; (B.-T.B.); (F.S.)
- Division of Biotherapeutics Discovery, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach, Germany
| | - John Edward Park
- Division of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach, Germany (L.R.L.)
| |
Collapse
|
36
|
Lou K, Wang J, He H, Wang Y, Mi Y, Li W, Chen L, Zhang Y, Mao Y, Lin J, Fu H, Yu C. Value of [ 68Ga]Ga-NYM046 PET/CT, in Comparison with 18F-FDG PET/CT, for Diagnosis of Clear Cell Renal Cell Carcinoma. J Nucl Med 2024; 65:1884-1890. [PMID: 39542699 PMCID: PMC11619588 DOI: 10.2967/jnumed.124.267527] [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: 01/31/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024] Open
Abstract
This study aimed to investigate the diagnostic efficacy of [68Ga]Ga-NYM046 PET/CT in animal models and patients with clear cell renal cell carcinoma (ccRCC) and to compare its performance with that of 18F-FDG PET/CT. Methods: The in vivo biodistribution of [68Ga]Ga-NYM046 was evaluated in mice bearing OS-RC-2 xenografts. Twelve patients with ccRCC were included in the study; all completed paired [68Ga]Ga-NYM046 PET/CT and 18F-FDG PET/CT. The diagnostic efficacies of these 2 PET tracers were compared. Moreover, the positive rate of carbonic anhydrase IX in the pathologic tissue sections was compared with the SUVmax obtained by PET/CT. Results: The tumor accumulation of [68Ga]Ga-NYM046 at 1 h after injection in OS-RC-2 xenograft tumor models was 7.21 ± 2.39 injected dose per gram of tissue. Apart from tumors, the kidney and stomach showed high-uptake distributions. In total, 9 primary tumors, 96 involved lymph nodes, and 147 distant metastases in 12 patients were evaluated using [68Ga]Ga-NYM046 and 18F-FDG PET/CT. Compared with 18F-FDG PET/CT, [68Ga]Ga-NYM046 PET/CT detected more primary tumors (9 vs. 1), involved lymph nodes (95 vs. 92), and distant metastases (137 vs. 127). In quantitative analysis, the primary tumors' SUVmax (median, 13.5 vs. 2.4; z = -2.668, P = 0.008) was significantly higher in [68Ga]Ga-NYM046 PET/CT. Conversely, the involved lymph nodes' SUVmax (median, 5.9 vs. 7.6; z = -3.236, P = 0.001) was higher in 18F-FDG PET/CT. No significant differences were found for distant metastases (median SUVmax, 5.0 vs. 5.0; z = -0.381, P = 0.703). Higher [68Ga]Ga-NYM046 uptake in primary tumors corresponded to higher expression of carbonic anhydrase IX, with an R 2 value of 0.8274. Conclusion: [68Ga]Ga-NYM046 PET/CT offers a viable strategy for detecting primary tumors, involved lymph nodes, and distant metastases in patients with ccRCC.
Collapse
Affiliation(s)
- Kequan Lou
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Jialiang Wang
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Huihui He
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yanjuan Wang
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yuanyuan Mi
- Department of Urological Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Wenjin Li
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Liping Chen
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yu Zhang
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yong Mao
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China; and
| | - Jianguo Lin
- Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Haitian Fu
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China;
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Chunjing Yu
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China;
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
37
|
Bunev AS, Shetnev AA, Shemchuk OS, Kozhukhov PK, Sharonova TV, Tyuryaeva II, Khotin MG, Ageev SV, Kholmurodova DK, Rizaev JA, Semenov KN, Sharoyko VV. Combination of Carbonic Anhydrase Isoform IX Inhibitors and Gefitinib Suppresses on the Invasive Potential of Non-Small Cell Lung Cancer Cells. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:2227-2237. [PMID: 39865035 DOI: 10.1134/s0006297924120113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/24/2024] [Accepted: 11/05/2024] [Indexed: 01/28/2025]
Abstract
Human carbonic anhydrase IX (CAIX) plays a key role in maintaining pH homeostasis of malignant neoplasms, thus creating a favorable microenvironment for the growth, invasion, and metastasis of tumor cells. Recent studies have established that inhibition of CAIX expressed on the surface of tumor cells significantly increases the efficacy of classical chemotherapeutic agents and makes it possible to suppress the resistance of tumor cells to chemotherapy, as well as to increase their sensitivity to drugs (in particular, to reduce the required dose of cytostatic agents). In this work, we studied the ability of new CAIX inhibitors based on substituted 1,2,4-oxadiazole-containing primary aromatic sulfonamides, to potentiate the cytostatic effect of gefitinib (selective inhibitor of epidermal growth factor receptor tyrosine kinase domain) under hypoxic conditions. We investigated a combined effect of gefitinib and CAIX inhibitors 4-(3-phenyl-1,2,4-oxadiazol-5-yl)thiophene-2-sulfonamide (1), 4-(5-(thiophene-3-yl)-1,2,4-oxadiazol-3-yl)benzenesulfonamide (2), 4-(3-(pyridin-2-yl)-1,2,4-oxadiazol-5-yl)thiophene-2-sulfonamide (3), and 4-(5-methyl-1,2,4-oxadiazol-3-yl)benzenesulfonamide (4) on gefitinib cytotoxicity, cell proliferation, activation of caspases-3/7, and cell cycle control in human lung adenocarcinoma A549 cells. It was found that the combinations of compounds 1 and 2 with gefitinib suppressed the invasive potential of A549 cells. Compound 1 had the greatest effect and can be considered as a promising candidate for further research.
Collapse
Affiliation(s)
- Alexander S Bunev
- Medicinal Chemistry Center, Togliatti State University, Togliatti, 445020, Russia
| | - Anton A Shetnev
- Institute of Biophysics of the Future, Dolgoprudny, Moscow Region, 141701, Russia
| | - Olga S Shemchuk
- Pavlov First St. Petersburg State Medical University, St. Petersburg, 197022, Russia
| | - Pavel K Kozhukhov
- Pavlov First St. Petersburg State Medical University, St. Petersburg, 197022, Russia
| | | | - Irina I Tyuryaeva
- St. Petersburg State University, St. Petersburg, 199034, Russia
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia
| | - Mikhail G Khotin
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia
| | - Sergey V Ageev
- Pavlov First St. Petersburg State Medical University, St. Petersburg, 197022, Russia
- St. Petersburg State University, St. Petersburg, 199034, Russia
| | - Dilafruz K Kholmurodova
- Scientific and Practice Center for Immunology, Allergology and Human Genomics, Samarkand State Medical University, Samarkand, 100400, Uzbekistan
| | - Jasur A Rizaev
- Scientific and Practice Center for Immunology, Allergology and Human Genomics, Samarkand State Medical University, Samarkand, 100400, Uzbekistan
| | - Konstantin N Semenov
- Pavlov First St. Petersburg State Medical University, St. Petersburg, 197022, Russia
- St. Petersburg State University, St. Petersburg, 199034, Russia
- Scientific and Practice Center for Immunology, Allergology and Human Genomics, Samarkand State Medical University, Samarkand, 100400, Uzbekistan
| | - Vladimir V Sharoyko
- Medicinal Chemistry Center, Togliatti State University, Togliatti, 445020, Russia.
- Pavlov First St. Petersburg State Medical University, St. Petersburg, 197022, Russia
- St. Petersburg State University, St. Petersburg, 199034, Russia
- Scientific and Practice Center for Immunology, Allergology and Human Genomics, Samarkand State Medical University, Samarkand, 100400, Uzbekistan
| |
Collapse
|
38
|
Onyeogaziri FC, Smith R, Arce M, Huang H, Erzar I, Rorsman C, Malinverno M, Orsenigo F, Sundell V, Fernando D, Daniel G, Niemelä M, Laakso A, Jahromi BR, Olsson AK, Magnusson PU. Pharmacological blocking of neutrophil extracellular traps attenuates immunothrombosis and neuroinflammation in cerebral cavernous malformation. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1549-1567. [PMID: 39632986 PMCID: PMC11634782 DOI: 10.1038/s44161-024-00577-y] [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: 01/18/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024]
Abstract
Cerebral cavernous malformation (CCM) is a neurovascular disease with symptoms such as strokes, hemorrhages and neurological deficits. With surgery being the only treatment strategy, understanding the molecular mechanisms of CCM is crucial in finding alternative therapeutic options for CCM. Neutrophil extracellular traps (NETs) were recently reported in CCM, and NETs were shown to have positive or negative effects in different disease contexts. In this study, we investigated the roles of NETs in CCM by pharmacologically inhibiting NET formation using Cl-amidine (a peptidyl arginine deiminase inhibitor). We show here that Cl-amidine treatment reduced lesion burden, coagulation and endothelial-to-mesenchymal transition. Furthermore, NETs promoted the activation of microglia and fibroblasts, leading to increased neuroinflammation and a chronic wound microenvironment in CCM. The inhibition of NET formation caused endothelial quiescence and promoted a healthier microenvironment. Our study suggests the inhibition of NETs as a potential therapeutic strategy in CCM.
Collapse
Affiliation(s)
- Favour C Onyeogaziri
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ross Smith
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Maximiliano Arce
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Hua Huang
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Iza Erzar
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Charlotte Rorsman
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Matteo Malinverno
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Fabrizio Orsenigo
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Veronica Sundell
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Dinesh Fernando
- Department of Biomaterials and Technology/Wood Science, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Geoffrey Daniel
- Department of Biomaterials and Technology/Wood Science, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Mika Niemelä
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Aki Laakso
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Behnam Rezai Jahromi
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Peetra U Magnusson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
39
|
Ullah MZ, Hussain Z, Shakir SA, Mahmood M, Ejaz SA, Aziz M, Fayyaz A, Iqbal J, Mumtaz A. Exploration of newly synthesized deferasirox derivatives as potential anti-cancer agents via in-vitro and in-silico approaches. Int J Biol Macromol 2024; 283:137971. [PMID: 39581395 DOI: 10.1016/j.ijbiomac.2024.137971] [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: 01/04/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Carbonic anhydrase IX (CA IX), upregulated by hypoxia-inducible factor (HIF), plays a crucial role in regulation of intracellular and extracellular pH, which is essential for the growth and spread of tumors. The overexpression of CA IX in breast cancer is linked to a low post-radiation patient survival rate. Under normoxic conditions, CA IX expression is relatively low, but hypoxia-inducible factors (HIFs) upregulate its expression when oxygen levels drop. This adaptation supports the tumor's acidic microenvironment, aiding processes like metastasis, immune evasion, and resistance to therapies. Due to these functions, CA IX is considered a promising target for cancer therapy, with inhibitors in development aimed at disrupting its activity and thus hindering tumor growth and survival. Thus, various derivatives of already reported anticancer drug i.e., deferasirox were synthesized and their effect on CA IX enzyme were assessed. Additionally, the binding affinities of deferasirox derivatives with three distinct receptor proteins i.e., Tumor Protein P53 (TP53), Nuclear factor kappa B (NF-κB) and caspase 3 (pdb: 3DCY, 1NFI, 3DEI) were also observed. Their anticancer effect was evaluated by using non-invasive human breast cancer cells i.e., MCF-7 and glioblastoma cells (U87). Among all derivatives, the four thioureas derivatives showed more anticancer potential. The 4-(3,5-bis(2-hydroxyphenyl)-1H-1,2,4-triazol-1-yl)-N-((3,4-dimethoxyphenyl)carbamothioyl) benzamide (6) derivative exhibited maximum anticancer potential (0.33 ± 0.02 μM) with greater binding affinity at different protein receptors. The MTT results further confirmed the enzyme inhibition results of deferasirox derivatives. In conclusion, targeting hypoxia-induced CA IX expression in breast cancer through the use of deferasirox-derived thiourea derivatives presents a promising therapeutic approach.
Collapse
Affiliation(s)
- Muhammad Zahid Ullah
- Department of Chemistry, COMSATS University Islamabad, Abbottabad 22060, Pakistan
| | - Zahid Hussain
- Department of Chemistry, COMSATS University Islamabad, Abbottabad 22060, Pakistan; Center for Advance Drug Research, COMSATS University Islamabad, Abbottabad 22060, Pakistan
| | - Syed Ahmad Shakir
- Department of Chemistry, COMSATS University Islamabad, Abbottabad 22060, Pakistan
| | - Mahnoor Mahmood
- Department of Chemistry, COMSATS University Islamabad, Abbottabad 22060, Pakistan
| | - Syeda Abida Ejaz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacv, The Islamia University of Bahawalpur, 63100, Pakistan
| | - Mubashir Aziz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacv, The Islamia University of Bahawalpur, 63100, Pakistan
| | - Ammara Fayyaz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacv, The Islamia University of Bahawalpur, 63100, Pakistan
| | - Jamshed Iqbal
- Department of Chemistry, COMSATS University Islamabad, Abbottabad 22060, Pakistan; Center for Advance Drug Research, COMSATS University Islamabad, Abbottabad 22060, Pakistan.
| | - Amara Mumtaz
- Department of Chemistry, COMSATS University Islamabad, Abbottabad 22060, Pakistan.
| |
Collapse
|
40
|
Jang JH, Kim JY, Lee TJ. Recent advances in anticancer mechanisms of molecular glue degraders: focus on RBM39-dgrading synthetic sulfonamide such as indisulam, E7820, tasisulam, and chloroquinoxaline sulfonamide. Genes Genomics 2024; 46:1345-1361. [PMID: 39271535 DOI: 10.1007/s13258-024-01565-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024]
Abstract
Synthetic sulfonamide anticancer drugs, including E7820, indisulam, tasisulam, and chloroquinoxaline sulfonamide, exhibit diverse mechanisms of action and therapeutic potential, functioning as molecular glue degraders. E7820 targets RBM39, affecting RNA splicing and angiogenesis by suppressing integrin α2. Phase I studies have demonstrated some stability in advanced solid malignancies; however, further efficacy studies are required. Indisulam causes G1 cell cycle arrest and delays the G1/S transition by modulating splicing through RBM39 degradation via DCAF15. Despite its limited initial efficacy, it shows promise in combination therapies, particularly for hematopoietic malignancies and gliomas. Tasisulam inhibits VEGF signaling, suppresses angiogenesis, and induces apoptosis. Although early trials indicated broad activity, safety concerns have halted its development. Chloroquinoxaline sulfonamide, initially investigated for cell cycle arrest and topoisomerase II inhibition, was discontinued owing to its limited efficacy and toxicity, despite promising initial results. Recent studies revealed the structural interaction of E7820 with DCAF15 and RBM39, although phase II trials on myeloid malignancies have shown limited efficacy. Indisulam is effective against glioblastoma and neuroblastoma, with potential synergy in combination therapies and metabolic disruption. Recent research on tasisulam reveals its potential in cancer therapy by targeting RBM39 degradation through DCAF15-mediated pathways. Understanding these mechanisms could lead to new treatments that affect alternative splicing and improve cancer therapies Overall, although these drugs exhibit promising mechanisms of action, further research is required to optimize their clinical efficacy and safety.
Collapse
Affiliation(s)
- Ji Hoon Jang
- Department of Anatomy, College of Medicine, Yeungnam University, 170 Hyeonchung-ro, Nam-gu, Daegu, 42415, Republic of Korea
| | - Joo-Young Kim
- Department of Anatomy, College of Medicine, Yeungnam University, 170 Hyeonchung-ro, Nam-gu, Daegu, 42415, Republic of Korea
| | - Tae-Jin Lee
- Department of Anatomy, College of Medicine, Yeungnam University, 170 Hyeonchung-ro, Nam-gu, Daegu, 42415, Republic of Korea.
| |
Collapse
|
41
|
Noor A, McGowan ER, Van Zuylekom JK, Cullinane C, Roselt PD, Hicks RJ, Wheatcroft MP, Donnelly PS. Preclinical evaluation and automated synthesis of [ 89Zr]ZrDFOSquaramide-girentuximab for diagnostic imaging of carbonic anhydrase IX positive tumours. EJNMMI Radiopharm Chem 2024; 9:80. [PMID: 39589414 PMCID: PMC11599670 DOI: 10.1186/s41181-024-00310-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: 09/17/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Carbonic Anhydrase IX (CAIX) is a zinc metalloenzyme that is over-expressed in many cancers making it a valid target for targeted diagnostic imaging with Positron Emission Tomography (PET). The monoclonal antibody girentuximab binds to CAIX and when radiolabelled with positron-emitting zirconium-89 can be used for diagnostic PET imaging of CAIX positive tumours. RESULTS Reaction of desferrioxamine squaramide ethyl ester with girentuximab allowed isolation of a conjugate with desferrioxamine squaramide (DFOSq) covalently attached to girentuximab through stable vinylogous amide linkages to give DFOSq-girentuximab. This conjugate was radiolabelled with zirconium-89 to give [89Zr]ZrDFOSq-girentuximab and the tumour uptake of the tracer was evaluated in CAIX positive HT29 tumour-bearing mice. Analysis of the PET images and biodistribution studies showed that the tracer displays high tumour uptake. An automated process for production of [89Zr]ZrDFOSq-girentuximab was developed, using [89Zr]ZrCl4 as a starting material that was also synthesized in an automated process. This automated process allows isolation of [89Zr]ZrDFOSq-girentuximab in radiochemical yields of 80-90% and in > 95% radiochemical purity. CONCLUSIONS [89Zr]ZrDFOSq-girentuximab has high uptake in CAIX positive tumours. An automated procedure for the synthesis of [89Zr]ZrDFOSq-girentuximab using [89Zr]ZrCl4 as a starting material has been developed. This automated process could be readily adapted to other antibodies.
Collapse
Affiliation(s)
- Asif Noor
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Emily R McGowan
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | | | - Carleen Cullinane
- Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Peter D Roselt
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
- Department of Radiopharmaceutical Sciences, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Rodney J Hicks
- Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
- Melbourne Theranostic Innovation Centre, Level 8/14-20 Blackwood St, North Melbourne VIC, Melbourne, 3051, Australia
| | - Michael P Wheatcroft
- Telix Pharmaceuticals Limited, Suite 401, 55 Flemington Road, North Melbourne, Melbourne, VIC, 3051, Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
42
|
Windsor P, Ouyang H, G da Costa JA, Rama Damodaran A, Chen Y, Bhagi-Damodaran A. Gas Tunnel Engineering of Prolyl Hydroxylase Reprograms Hypoxia Signaling in Cells. Angew Chem Int Ed Engl 2024; 63:e202409234. [PMID: 39168829 DOI: 10.1002/anie.202409234] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/02/2024] [Accepted: 08/20/2024] [Indexed: 08/23/2024]
Abstract
Cells have evolved intricate mechanisms for recognizing and responding to changes in oxygen (O2) concentrations. Here, we have reprogrammed cellular hypoxia (low O2) signaling via gas tunnel engineering of prolyl hydroxylase 2 (PHD2), a non-heme iron dependent O2 sensor. Using computational modeling and protein engineering techniques, we identify a gas tunnel and critical residues therein that limit the flow of O2 to PHD2's catalytic core. We show that systematic modification of these residues can open the constriction topology of PHD2's gas tunnel. Using kinetic stopped-flow measurements with NO as a surrogate diatomic gas, we demonstrate up to 3.5-fold enhancement in its association rate to the iron center of tunnel-engineered mutants. Our most effectively designed mutant displays 9-fold enhanced catalytic efficiency (kcat/KM=830±40 M-1 s-1) in hydroxylating a peptide mimic of hypoxia inducible transcription factor HIF-1α, as compared to WT PHD2 (kcat/KM=90±9 M-1 s-1). Furthermore, transfection of plasmids that express designed PHD2 mutants in HEK-293T mammalian cells reveal significant reduction of HIF-1α and downstream hypoxia response transcripts under hypoxic conditions of 1 % O2. Overall, these studies highlight activation of PHD2 as a new pathway to reprogram hypoxia responses and HIF signaling in cells.
Collapse
Affiliation(s)
- Peter Windsor
- Department of Chemistry, University of Minnesota, Twin Cities, Minneapolis, MN 55455, United States
| | - Haiping Ouyang
- Department of Biochemistry and Molecular Biology, University of Minnesota, Twin Cities, Minneapolis, MN 55455, United States
| | - Joseph A G da Costa
- Department of Chemistry, University of Minnesota, Twin Cities, Minneapolis, MN 55455, United States
| | - Anoop Rama Damodaran
- Department of Chemistry, University of Minnesota, Twin Cities, Minneapolis, MN 55455, United States
| | - Yue Chen
- Department of Biochemistry and Molecular Biology, University of Minnesota, Twin Cities, Minneapolis, MN 55455, United States
| | - Ambika Bhagi-Damodaran
- Department of Chemistry, University of Minnesota, Twin Cities, Minneapolis, MN 55455, United States
| |
Collapse
|
43
|
Głowacka K, Ibanez S, Renoult O, Vermonden P, Giolito MV, Özkan K, Degavre C, Aubert L, Guilbaud C, Laloux-Morris F, Richiardone E, Ambroise J, Bouzin C, Brusa D, Dehairs J, Swinnen J, Corbet C, Larondelle Y, Feron O. Acid-exposed and hypoxic cancer cells do not overlap but are interdependent for unsaturated fatty acid resources. Nat Commun 2024; 15:10107. [PMID: 39572570 PMCID: PMC11582563 DOI: 10.1038/s41467-024-54435-3] [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: 03/13/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
Cancer cells in acidic tumor regions are aggressive and a key therapeutic target, but distinguishing between acid-exposed and hypoxic cells is challenging. Here, we use carbonic anhydrase 9 (CA9) antibodies to mark acidic areas in both hypoxic and respiring tumor areas, along with an HRE-GFP reporter for hypoxia, to isolate distinct cell populations from 3D tumor spheroids. Transcriptomic analysis of CA9-positive, hypoxia-negative cells highlights enriched fatty acid desaturase activity. Inhibiting or silencing stearoyl-CoA desaturase-1 (SCD1) induces ferroptosis in CA9-positive acidic cancer cells and delays mouse tumor growth, an effect enhanced by omega-3 fatty acid supplementation. Using acid-exposed cancer cells and patient-derived tumor organoids, we show that SCD1 inhibition increases acidic cancer cell reliance on external mono-unsaturated fatty acids, depriving hypoxic cells of essential resources. This bystander effect provides unbiased evidence for a lack of full overlap between hypoxic and acidic tumor compartments, highlighting a rationale for targeting desaturase activity in cancer.
Collapse
Affiliation(s)
- Katarzyna Głowacka
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Sébastien Ibanez
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Ophélie Renoult
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Perrine Vermonden
- Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium
| | - Maria Virginia Giolito
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Kübra Özkan
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Charline Degavre
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Léo Aubert
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Céline Guilbaud
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Florine Laloux-Morris
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Elena Richiardone
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Jérôme Ambroise
- Centre des Technologies Moléculaires Appliquées (CTMA), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Caroline Bouzin
- Imaging Platform 2IP, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Davide Brusa
- CytoFlux-Flow Cytometry and Cell Sorting Platform, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Jonas Dehairs
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Johan Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Yvan Larondelle
- Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Louvain-la-Neuve, Belgium
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) Department, WEL Research Institute, Wavre, Belgium.
| |
Collapse
|
44
|
Combs JE, Murray AB, Lomelino CL, Mboge MY, Mietzsch M, Horenstein NA, Frost SC, McKenna R, Becker HM. Disruption of the Physical Interaction Between Carbonic Anhydrase IX and the Monocarboxylate Transporter 4 Impacts Lactate Transport in Breast Cancer Cells. Int J Mol Sci 2024; 25:11994. [PMID: 39596062 PMCID: PMC11593560 DOI: 10.3390/ijms252211994] [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: 10/10/2024] [Revised: 11/02/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
It has been previously established that breast cancer cells exhibit high expression of the monocarboxylate (lactate) transporters (MCT1 and/or MCT4) and carbonic anhydrase IX (CAIX) and form a functional metabolon for proton-coupled lactate export, thereby stabilizing intracellular pH. CD147 is the MCT accessory protein that facilitates the creation of the MCT/CAIX complex. This study describes how the small molecule Beta-Galactose 2C (BGal2C) blocks the physical and functional interaction between CAIX and either MCT1 or MCT4 in Xenopus oocytes, which reduces the rate of proton and lactate flux with an IC50 of ~90 nM. This value is similar to the Ki for inhibition of CAIX activity. Furthermore, it is shown that BGal2C blocks hypoxia-induced lactate transport in MDA-MB-231 and MCF-7 breast cancer cells, both of which express CAIX. As in oocytes, BGal2C interferes with the physical interaction between CAIX and MCTs in both cell types. Finally, X-ray crystallographic studies highlight unique interactions between BGal2C and a CAIX-mimic that are not observed within the CAII active site and which may underlie the strong specificity of BGal2C for CAIX. These studies demonstrate the utility of a novel sulfonamide in interfering with elevated proton and lactate flux, a hallmark of many solid tumors.
Collapse
Affiliation(s)
- Jacob E. Combs
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA (M.M.); (S.C.F.)
| | - Akilah B. Murray
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA (M.M.); (S.C.F.)
| | - Carrie L. Lomelino
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA (M.M.); (S.C.F.)
| | - Mam Y. Mboge
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA (M.M.); (S.C.F.)
| | - Mario Mietzsch
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA (M.M.); (S.C.F.)
| | | | - Susan C. Frost
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA (M.M.); (S.C.F.)
| | - Robert McKenna
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA (M.M.); (S.C.F.)
| | - Holger M. Becker
- Institute of Physiological Chemistry, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
45
|
Mishra KA, Sethi KK. Unveiling tomorrow: Carbonic anhydrase activators and inhibitors pioneering new frontiers in Alzheimer's disease. Arch Pharm (Weinheim) 2024:e2400748. [PMID: 39506506 DOI: 10.1002/ardp.202400748] [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: 09/16/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder and a principal basis of dementia in the elderly population globally. Recently, human carbonic anhydrases (hCAs, EC 4.2.1.1) were demonstrated as possible new targets for treating AD. hCAs are vital for maintaining pH balance and performing other physiological processes as they catalyze the reversible hydration of carbon dioxide to bicarbonate and a proton. Current research indicates that hCA plays a role in brain functions critical for transmitting neural signals. Activation of carbonic anhydrase (CA) has emerged as a promising avenue in addressing memory loss and cognitive issues. Conversely, the exploration of CA inhibition represents a novel frontier in this field. By enhancing glial fitness and cerebrovascular health and blocking amyloid-β (Aβ)-induced mitochondrial dysfunction pathways, cytochrome C (CytC) release, caspase 9 activation, and H2O2 generation in neurons, CA inhibitors improve cognition and lessen the pathology caused by Aβ. Recent research has pushed hCAs into the spotlight as critical players in AD pathogenesis and precise therapeutic targets. The captivating dilemma of choosing between hCA inhibitors and activators looms large, as inhibitors reduce Aβ aggregation and improve cerebral blood flow, while activators enhance cerebrovascular functions and restore pH balance. The current review sheds light on the clinical evidence for hCAs and the roles of inhibitors and activators in AD. Additionally, this review offers a fascinating outlook on the data that may aid medicinal chemists in designing and developing new leads that are more effective and selective for upcoming in vitro and in vivo studies, allowing for the discovery and introduction of novel drug candidates for the treatment of AD to the market and into the clinical pipeline.
Collapse
Affiliation(s)
- Km Abha Mishra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Guwahati, Guwahati, Assam, India
| | - Kalyan K Sethi
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Guwahati, Guwahati, Assam, India
| |
Collapse
|
46
|
Knudsen-Clark AM, Mwangi D, Cazarin J, Morris K, Baker C, Hablitz LM, McCall MN, Kim M, Altman BJ. Circadian rhythms of macrophages are altered by the acidic tumor microenvironment. EMBO Rep 2024; 25:5080-5112. [PMID: 39415049 PMCID: PMC11549407 DOI: 10.1038/s44319-024-00288-2] [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: 08/01/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are prime therapeutic targets due to their pro-tumorigenic functions, but varying efficacy of macrophage-targeting therapies highlights our incomplete understanding of how macrophages are regulated within the tumor microenvironment (TME). The circadian clock is a key regulator of macrophage function, but how circadian rhythms of macrophages are influenced by the TME remains unknown. Here, we show that conditions associated with the TME such as polarizing stimuli, acidic pH, and lactate can alter circadian rhythms in macrophages. While cyclic AMP (cAMP) has been reported to play a role in macrophage response to acidic pH, our results indicate pH-driven changes in circadian rhythms are not mediated solely by cAMP signaling. Remarkably, circadian disorder of TAMs was revealed by clock correlation distance analysis. Our data suggest that heterogeneity in circadian rhythms within the TAM population level may underlie this circadian disorder. Finally, we report that circadian regulation of macrophages suppresses tumor growth in a murine model of pancreatic cancer. Our work demonstrates a novel mechanism by which the TME influences macrophage biology through modulation of circadian rhythms.
Collapse
Affiliation(s)
- Amelia M Knudsen-Clark
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Daniel Mwangi
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Juliana Cazarin
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Kristina Morris
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Cameron Baker
- Genomics Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Lauren M Hablitz
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Matthew N McCall
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Brian J Altman
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA.
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
47
|
Han Mİ, Gündüz MG, Ammara A, Supuran CT, Doğan ŞD. Tail-approach based design, synthesis, and molecular modeling of benzenesulfonamides carrying thiadiazole and urea moieties as novel carbonic anhydrase inhibitors. Arch Pharm (Weinheim) 2024; 357:e2400439. [PMID: 39079940 DOI: 10.1002/ardp.202400439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 11/06/2024]
Abstract
We synthesized herein 16 compounds (SUT1-SUT16) as potential carbonic anhydrase (CA) inhibitors utilizing the tail-approach design. Based on this strategy, we connected benzenesulfonamide, the zinc-binding scaffold, to different urea moieties with the 1,3,4-thiadiazole ring as a linker. We obtained the target compounds by the reaction of 4-(5-amino-1,3,4-thiadiazol-2-yl)benzenesulfonamide with aryl isocyanates. Upon confirmation of their structures, the compounds were screened for their ability to inhibit the tumor-related human (h) isoforms human carbonic anhydrase (hCA) IX and XII, as well as the physiologically dominant hCA I and II. Most of the molecules demonstrated Ki values ≤ 10 nM with different selectivity profiles. The binding modes of SUT9, SUT10, and SUT5, the most effective inhibitors of hCA II, IX, and XII, respectively, were predicted by molecular docking. SUT16 (4-{5-[3-(naphthalen-1-yl)ureido]-1,3,4-thiadiazol-2-yl}benzenesulfonamide) was found to be the most selective inhibitor of the cancer-associated isoforms hCA IX and XII over the off-target isoforms, hCAI and II. The interaction dynamics and stability of SUT16 within hCA IX and XII were investigated by molecular dynamics simulations as well as dynophore analysis. Based on computational data, increased hydrophobic contacts and hydrogen bonds in the tail part of these molecules within hCA IX and XII were found as favorable interactions leading to effective inhibitors of cancer-related isoforms.
Collapse
Affiliation(s)
- M İhsan Han
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | - Miyase Gözde Gündüz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Andrea Ammara
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Firenze, Italy
| | - Claudiu T Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Firenze, Italy
| | - Şengül Dilem Doğan
- Department of Basic Sciences, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| |
Collapse
|
48
|
Mo X, Rao DP, Kaur K, Hassan R, Abdel-Samea AS, Farhan SM, Bräse S, Hashem H. Indole Derivatives: A Versatile Scaffold in Modern Drug Discovery-An Updated Review on Their Multifaceted Therapeutic Applications (2020-2024). Molecules 2024; 29:4770. [PMID: 39407697 PMCID: PMC11477627 DOI: 10.3390/molecules29194770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
Indole derivatives have become an important class of compounds in medicinal chemistry, recognized for their wide-ranging biological activities and therapeutic potential. This review provides a comprehensive overview of recent advances in the evaluation of indole-based compounds in the last five years, highlighting their roles in cancer treatment, infectious disease management, anti-inflammatory therapies, metabolic disorder interventions, and neurodegenerative disease management. Indole derivatives have shown significant efficacy in targeting diverse biological pathways, making them valuable scaffolds in designing new drugs. Notably, these compounds have demonstrated the ability to combat drug-resistant cancer cells and pathogens, a significant breakthrough in the field, and offer promising therapeutic options for chronic diseases such as diabetes and hypertension. By summarizing recent key findings and exploring the underlying biological mechanisms, this review underscores the potential of indole derivatives in addressing major healthcare challenges, thereby instilling hope and optimism in the field of modern medicine.
Collapse
Affiliation(s)
- Xingyou Mo
- School of Engineering, Guangzhou College of Technology and Business, Guangzhou 510850, China
| | - Devendra Pratap Rao
- Coordination Chemistry Laboratory, Department of Chemistry, Dayanand Anglo-Vedic (PG) College, Kanpur 208001, Uttar Pradesh, India
| | - Kirandeep Kaur
- Department of Chemistry, Maharaja Ranjit Singh Punjab Technical University, Bathinda 151001, Punjab, India
| | - Roket Hassan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sohag University, Sohag 82524, Egypt
| | - Ahmed S. Abdel-Samea
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Deraya University, New Minia 61768, Egypt
| | - Sara Mahmoud Farhan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Deraya University, New Minia 61768, Egypt
| | - Stefan Bräse
- Institute of Biological and Chemical Systems—Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Kaiserstrasse 12, 76131 Karlsruhe, Germany
| | - Hamada Hashem
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sohag University, Sohag 82524, Egypt
| |
Collapse
|
49
|
Zhou Y, Li C, Chen X, Zhao Y, Liao Y, Huang P, Wu W, Nieto NS, Li L, Tang W. Development of folate receptor targeting chimeras for cancer selective degradation of extracellular proteins. Nat Commun 2024; 15:8695. [PMID: 39379374 PMCID: PMC11461649 DOI: 10.1038/s41467-024-52685-9] [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: 12/15/2023] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
Targeted protein degradation has emerged as a novel therapeutic modality to treat human diseases by utilizing the cell's own disposal systems to remove protein target. Significant clinical benefits have been observed for degrading many intracellular proteins. Recently, the degradation of extracellular proteins in the lysosome has been developed. However, there have been limited successes in selectively degrading protein targets in disease-relevant cells or tissues, which would greatly enhance the development of precision medicine. Additionally, most degraders are not readily available due to their complexity. We report a class of easily accessible Folate Receptor TArgeting Chimeras (FRTACs) to recruit the folate receptor, primarily expressed on malignant cells, to degrade extracellular soluble and membrane cancer-related proteins in vitro and in vivo. Our results indicate that FRTAC is a general platform for developing more precise and effective chemical probes and therapeutics for the study and treatment of cancers.
Collapse
Affiliation(s)
- Yaxian Zhou
- Lachman Institute of Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Chunrong Li
- Lachman Institute of Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Xuankun Chen
- Lachman Institute of Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Yuan Zhao
- Lachman Institute of Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Yaxian Liao
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Penghsuan Huang
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Wenxin Wu
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Nicholas S Nieto
- Lachman Institute of Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Lingjun Li
- Lachman Institute of Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Weiping Tang
- Lachman Institute of Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| |
Collapse
|
50
|
Lim HY, Dolzhenko AV. 1,3,5-Triazine as a promising scaffold in the development of therapeutic agents against breast cancer. Eur J Med Chem 2024; 276:116680. [PMID: 39018924 DOI: 10.1016/j.ejmech.2024.116680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
1,3,5-Triazine scaffold has garnered considerable interest due to its wide-ranging pharmacological properties, particularly in the field of cancer research. Breast cancer is the most commonly diagnosed cancer among women. Approximately one in eight women will receive a diagnosis of invasive breast cancer during their lifetime. The five-year survival rate for invasive breast cancer is less than 30 %, indicating a need to develop a more effective therapeutic agent targeting breast cancer. This review discusses bioactive 1,3,5-triazines targeting breast cancer cells by the inhibition of different enzymes, which include PI3K, mTOR, EGFR, VEGFR, FAK, CDK, DHFR, DNA topoisomerase, ubiquitin-conjugating enzyme, carbonic anhydrase, and matrix metalloproteinase. The anticancer agent search in some drug discovery programs is based on compound screening for antiproliferative activity. Often, multiple targets contribute to the anticancer effect of 1,3,5-triazines and this approach allows identification of active molecules prior to identification of their targets.
Collapse
Affiliation(s)
- Han Yin Lim
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan, 47500, Malaysia.
| | - Anton V Dolzhenko
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan, 47500, Malaysia; Curtin Medical School, Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, GPO Box U1987 Perth, Western, Bentley, 6845, Australia
| |
Collapse
|