1
|
Wu Z, Wu W, Zhang C, Zhang W, Li Y, Ding T, Fang Z, Jing J, He X, Huang F. Enhanced diabetic foot ulcer treatment with a chitosan-based thermosensitive hydrogel loaded self-assembled multi-functional nanoparticles for antibacterial and angiogenic effects. Carbohydr Polym 2025; 347:122740. [PMID: 39486969 DOI: 10.1016/j.carbpol.2024.122740] [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: 07/17/2024] [Revised: 08/31/2024] [Accepted: 09/10/2024] [Indexed: 11/04/2024]
Abstract
Inhibiting bacterial growth and promoting angiogenesis are essential for enhancing wound healing in diabetic patients. Excessive oxidative stress at the wound site can also lead to an accumulation of reactive oxygen species. To address these challenges, a smart thermosensitive hydrogel loaded with therapeutic agents was developed. This formulation features self-assembled nanoparticles named CIZ, consisting of chlorogenic acid (CA), indocyanine green (ICG), and zinc ions (Zn2+). These nanoparticles are loaded into a chitosan-β-glycerophosphate hydrogel, named CIZ@G, which enables rapid gel formation under photothermal effects. The hydrogel demonstrates good biocompatibility and effectively releases drugs into diabetic foot ulcers (DFU) wound. Benefiting from the dual actions of CA and zinc ions, the hydrogel exhibits potent antioxidative and anti-inflammatory effects, enhances the expression of vascular endothelial growth factor (VEGF) and Platelet endothelial cell adhesion molecule-1 (CD31), and promotes angiogenesis. Both in vitro and in vivo experiments confirm that CIZ@G can effectively inhibit the growth of Staphylococcus aureus post-laser irradiation and accelerate wound remodeling within 14 days. This approach offers a new strategy for the treatment of diabetic foot ulcers (DFU), potentially transforming patient care in this challenging clinical area.
Collapse
Affiliation(s)
- Zhiwei Wu
- Department of orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Weiwei Wu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; Anhui Public Health Clinical Center, Hefei 230022, China
| | - Chi Zhang
- Anhui Public Health Clinical Center, Hefei 230022, China; Department of orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Wenbiao Zhang
- Department of orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yang Li
- Department of orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Tao Ding
- Department of orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Zhennan Fang
- Department of orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Juehua Jing
- Department of orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China..
| | - Xiaoyan He
- School of Life Sciences Anhui Medical University, Hefei 230032, China.
| | - Fei Huang
- Department of orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China..
| |
Collapse
|
2
|
Zhao J, Yu B, Li L, Guo S, Sha X, Ru W, Du GQ, Xue JY. Outer Membrane Vesicle-Cancer Hybrid Membrane Coating Indocyanine Green Nanoparticles for Enhancing Photothermal Therapy Efficacy in Tumors. ACS Biomater Sci Eng 2024; 10:7619-7631. [PMID: 39585170 DOI: 10.1021/acsbiomaterials.4c01251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Cell membrane-coated nanomaterials are increasingly recognized as effective in cancer treatment due to their unique benefits. This study introduces a novel hybrid membrane coating nanoparticle, termed cancer cell membrane (CCM)-outer membrane vesicle (OMV)@Lip-indocyanine green (ICG), which combines CCMs with bacterial OMV to encapsulate ICG-loaded liposomes. Comprehensive analyses were conducted to assess its physical and chemical properties as well as its functionality. Demonstrating targeted delivery capabilities and good biocompatibility, CCM-OMV@Lip-ICG nanoparticles showed promising photothermal and immunotherapeutic effects in tumor models. By inducing hyperthermia-induced tumor therapy and bolstering antitumor immunity, CCM-OMV@Lip-ICG nanoparticles exhibit a synergistic therapeutic effect, providing a new perspective for the management of cancer.
Collapse
Affiliation(s)
- Jing Zhao
- Guangdong Cardiovascular Institution, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Bo Yu
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province, Harbin 150028, China
| | - Lujing Li
- Department of Ultrasound, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Sihua Guo
- Guangdong Cardiovascular Institution, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Xuan Sha
- Guangdong Cardiovascular Institution, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Waner Ru
- Guangdong Cardiovascular Institution, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- College of Medicine, Shantou University, Shantou 515041, China
| | - Guo-Qing Du
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jing-Yi Xue
- Guangdong Cardiovascular Institution, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Science), Southern Medical University, Guangzhou 510080, China
| |
Collapse
|
3
|
Jiao Y, Liu Y, Jin M. Exploring the dark side of diagnostic dyes with a focus on Indocyanine green's adverse reactions. Sci Rep 2024; 14:30155. [PMID: 39627439 PMCID: PMC11614906 DOI: 10.1038/s41598-024-81903-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: 09/03/2024] [Accepted: 11/29/2024] [Indexed: 12/06/2024] Open
Abstract
Indocyanine green (ICG) is a diagnostic dye commonly used in medical imaging and liver function monitoring. Given its widespread use, there is a need for detailed evaluations of its adverse drug reactions in real-world settings. As the comprehensive overview of its safety profile is very limited, this study aimed to analyze the adverse events (AEs) associated with ICG using data from the Food and Drug Administration Adverse Event Reporting System (FAERS). Data between 2004Q1 and 2023Q4 were extracted from the FAERS database. Signal detection was performed using various disproportionality analysis algorithms, including reporting odds ratio (ROR), proportional reporting ratio, Bayesian confidence propagation neural network, and multiitem gamma Poisson shrinker. During the study period, a total of 62 ICG-related AEs were reported in the FAERS database. Significant clinical adverse reactions included anaphylactic shock (ROR: 92.10, 95% confidence interval (CI): 37.71-224.96), procedural hypotension (ROR: 1397.27, 95% CI: 443.31-4404.08), and urticaria (ROR: 10.88, 95% CI: 4.02-29.42). This study provides valuable insights into the safety profile of ICG, highlighting the need for further monitoring to ensure its safe clinical use in clinical practice. Ongoing pharmacovigilance and large-scale studies are warranted to fully understand the potential risks associated with ICG.
Collapse
Affiliation(s)
- Yan Jiao
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yahui Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Meng Jin
- Department of Radiation Oncology, Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China.
| |
Collapse
|
4
|
Wu X, Wu Q, Hou M, Jiang Y, Li M, Jia G, Yang H, Zhang C. Regenerating Chemotherapeutics through Copper-Based Nanomedicine: Disrupting Protein Homeostasis for Enhanced Tumor Therapy. Adv Healthc Mater 2024; 13:e2401954. [PMID: 39039985 DOI: 10.1002/adhm.202401954] [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: 05/27/2024] [Revised: 07/03/2024] [Indexed: 07/24/2024]
Abstract
The bis-(diethyldithiocarbamate)-copper (CuET), the disulfiram (DSF)-Cu complex, has exhibited noteworthy anti-tumor property. However, its efficacy is compromised due to the inadequate oxidative conditions and the limitation of bioavailable copper. Because CuET can inactivate valosin-containing protein (VCP), a bioinformatic pan-cancer analysis of VCP is first conducted in this study to identify CuET as a promising anticancer drug for diverse cancer types. Then, based on the drug action mechanism, a nanocomposite of CuET and copper oxide (CuO) is designed and fabricated utilizing bovine serum albumin (BSA) as the template (denoted as CuET-CuO@BSA, CCB). CCB manifests peroxidase (POD)-mimicking activity to oxidize the tumor endogenous H2O2 to generate reactive oxygen species (ROS), enhancing the chemotherapy effect of CuET. Furthermore, the cupric ions released after enzymatic reaction can regenerate CuET, which markedly perturbs intracellular protein homeostasis and induces apoptosis of tumor cells. Meanwhile, CCB triggers cuproptosis by inducing the aggregation of lipoylated proteins. The multifaceted action of CCB effectively inhibits tumor progression. Therefore, this study presents an innovative CuET therapeutic strategy that creates an oxidative microenvironment in situ and simultaneously self-supply copper source for CuET regeneration through the combination of CuO nanozyme with CuET, which holds promise for application of CuET for effective tumor therapy.
Collapse
Affiliation(s)
- Xubo Wu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Qinghe Wu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Mengfei Hou
- School of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, China
| | - Yifei Jiang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Meng Li
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Guoping Jia
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Huizhen Yang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Chunfu Zhang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| |
Collapse
|
5
|
Roberts R, Pachl M. Intraparenchymal Indocyanine Green Use Improves Nodal Yield During Minimally Invasive Tumor Nephrectomy in Children. J Laparoendosc Adv Surg Tech A 2024; 34:1039-1043. [PMID: 38967048 DOI: 10.1089/lap.2024.0114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024] Open
Abstract
Introduction: Correctly identifying positive lymph nodes associated with pediatric renal tumors is key to guiding management. Recommended targets for lymph node sampling are commonly missed during tumor nephrectomy, particularly if minimally invasive surgery (MIS) is performed. Indocyanine green (ICG) is used for lymph node mapping in adult oncology with excellent efficacy and safety profile. Materials and Methods: A prospective study was undertaken at a single-quaternary-level pediatric surgery center. All patients undergoing MIS radical or partial nephrectomy for renal tumors 2016-2023 were included. Patients managed from 2020 onwards received intra-parenchymal ICG prior to lymph node sampling. Main Results: Twenty-five patients underwent MIS nephrectomy at mean age 2 years 10 months. Eighteen patients were pre-ICG and 7 received ICG. ICG administration successfully demonstrated fluorescent nodes in all patients. Median number of nodes sampled was three pre-ICG and seven with ICG (P = 0.009). Forty-six nodes were sampled across 7 ICG patients-33 fluorescent, 10 non-fluorescent, and 3 identified histologically. Three nodes overall contained active disease, two pre-ICG and one fluorescent node with ICG. Neither operative time (180 pre-ICG versus 161 minutes ICG, P = 0.7) nor length of stay (72 versus 84 hours, P = 0.3) were significantly affected by ICG administration. There were no adverse events associated with ICG use. Conclusions: ICG is safe and effective at identifying nodes in MIS resection of pediatric renal tumors with the potential to increase the number of nodes sampled. Further research is needed, specifically a randomized control trial with extended follow-up.
Collapse
Affiliation(s)
- Rebecca Roberts
- Department of Paediatric Surgery and Urology, Birmingham Children's hospital, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, United Kingdom
| | - Max Pachl
- Department of Paediatric Surgery and Urology, Birmingham Children's hospital, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, United Kingdom
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
6
|
Jaiswal S, Cox KE, Amirfakhri S, Din Parast Saleh A, Kobayashi K, Lwin TM, Talib S, Aithal A, Mallya K, Jain M, Mohs AM, Hoffman RM, Batra SK, Bouvet M. Targeting Human Pancreatic Cancer with a Fluorophore-Conjugated Mucin 4 (MUC4) Antibody: Initial Characterization in Orthotopic Cell Line Mouse Models. J Clin Med 2024; 13:6211. [PMID: 39458160 PMCID: PMC11508345 DOI: 10.3390/jcm13206211] [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: 09/18/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Pancreatic cancer is the third leading cause of death related to cancer. The only possible cure presently is complete surgical resection; however, this is limited by difficulty in clearly defining tumor margins. Enhancement of the visualization of pancreatic ductal adenocarcinoma (PDAC) tumor margins using near-infrared dye-conjugated tumor-specific antibodies was pioneered by using anti-CEA, anti-CA19.9, and anti-MUC5AC in orthotopic mouse models of pancreatic cancer. Recently, an antibody to Mucin 4 (MUC4) conjugated to a fluorescent probe has shown promise in targeting colon tumors in orthotopic mouse models. Methods: In the present study, we targeted pancreatic cancer using an anti-MUC4 antibody conjugated to IRDye800 (anti-MUC4-IR800) in orthotopic mouse models. Two pancreatic cancer human cell lines were used, SW1990 and CD18/HPAF. Results: Anti-MUC4-IR800 targeted the two pancreatic cancer cell line tumors in orthotopic mouse models with high tumor-to-pancreas ratios and high tumor-to-liver ratios, with greater targeting seen in SW1990. Conclusions: The present results suggest anti-MUC4-IR800's potential to be used in fluorescence-guided surgical resection of pancreatic cancer.
Collapse
Affiliation(s)
- Sunidhi Jaiswal
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA; (S.J.); (S.A.); (A.D.P.S.); (K.K.); (R.M.H.)
- VA San Diego Healthcare System, La Jolla, CA 92161, USA
| | - Kristin E. Cox
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA; (S.J.); (S.A.); (A.D.P.S.); (K.K.); (R.M.H.)
- VA San Diego Healthcare System, La Jolla, CA 92161, USA
| | - Siamak Amirfakhri
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA; (S.J.); (S.A.); (A.D.P.S.); (K.K.); (R.M.H.)
- VA San Diego Healthcare System, La Jolla, CA 92161, USA
| | - Aylin Din Parast Saleh
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA; (S.J.); (S.A.); (A.D.P.S.); (K.K.); (R.M.H.)
| | - Keita Kobayashi
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA; (S.J.); (S.A.); (A.D.P.S.); (K.K.); (R.M.H.)
| | - Thinzar M. Lwin
- Department of Surgical Oncology, City of Hope, Duarte, CA 91010, USA;
| | - Sumbal Talib
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; (S.T.); (A.M.M.)
| | - Abhijit Aithal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; (A.A.); (K.M.); (M.J.); (S.K.B.)
| | - Kavita Mallya
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; (A.A.); (K.M.); (M.J.); (S.K.B.)
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; (A.A.); (K.M.); (M.J.); (S.K.B.)
| | - Aaron M. Mohs
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; (S.T.); (A.M.M.)
| | - Robert M. Hoffman
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA; (S.J.); (S.A.); (A.D.P.S.); (K.K.); (R.M.H.)
- VA San Diego Healthcare System, La Jolla, CA 92161, USA
- AntiCancer Inc., San Diego, CA 92111, USA
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; (A.A.); (K.M.); (M.J.); (S.K.B.)
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA; (S.J.); (S.A.); (A.D.P.S.); (K.K.); (R.M.H.)
- VA San Diego Healthcare System, La Jolla, CA 92161, USA
- UCSD Moores UCSD Cancer Center, 3855 Health Sciences Drive #0987, La Jolla, CA 92093-0987, USA
| |
Collapse
|
7
|
Kim SH, Chae Y, Kang BT, Lee S. Use of 18-Fluorodeoxyglucose Positron Emission Tomography and Near-Infrared Fluorescence-Guided Imaging Surgery in the Treatment of a Gastric Tumor in a Dog. Animals (Basel) 2024; 14:2917. [PMID: 39457847 PMCID: PMC11503833 DOI: 10.3390/ani14202917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
A 13-year-old Maltese dog with an abdominal mass underwent 18F-FDG PET/computed tomography (CT) for tumor localization and metastatic evaluation. PET/CT scans revealed a gastric mass near the esophagogastric junction and demonstrated mean and maximum standardized uptake values (SUVs) of 4.596 and 6.234, respectively, for the abdominal mass. Subsequent surgery incorporated ICG for NIR fluorescence-guided imaging, aiding in precise tumor localization and margin assessment. The excised mass was identified as a low-grade leiomyosarcoma on histopathology. The dog underwent PET/CT imaging six months postoperatively following the excision of the mass, which confirmed the absence of recurrence or residual lesions during follow-up. NIR fluorescence imaging using ICG demonstrated efficacy in real-time tumor visualization and margin assessment, a technique not previously reported in veterinary literature. The PET/CT findings complemented the diagnosis and provided valuable insights into metastasis. The absence of recurrence or complications in postoperative follow-up underscores the potential of these imaging modalities in enhancing surgical precision and improving prognosis in canine gastric tumors.
Collapse
Affiliation(s)
- Su-Hyeon Kim
- Department of Veterinary Surgery, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea;
- Department of Veterinary Surgery, Heamaru Referral Hospital, Seongnam 13590, Republic of Korea
| | - Yeon Chae
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (Y.C.); (B.-T.K.)
| | - Byeong-Teck Kang
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea; (Y.C.); (B.-T.K.)
| | - Sungin Lee
- Department of Veterinary Surgery, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea;
| |
Collapse
|
8
|
Vázquez-Villar V, Das C, Swift T, Elies J, Tolosa J, García-Martínez JC, Ruiz A. Oligo(styryl)benzenes liposomal AIE-dots for bioimaging and phototherapy in an in vitro model of prostate cancer. J Colloid Interface Sci 2024; 670:585-598. [PMID: 38776693 DOI: 10.1016/j.jcis.2024.05.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/27/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024]
Abstract
Whilst the development of advanced organic dots with aggregation-induced emission characteristics (AIE-dots) is being intensively studied, their clinical translation in efficient biotherapeutic devices has yet to be tackled. This study explores the synergistic interplay of oligo(styryl)benzenes (OSBs), potent fluorogens with an increased emission in the aggregate state, and Indocyanine green (ICG) as dual Near Infrared (NIR)-visible fluorescent nanovesicles with efficient reactive oxygen species (ROS) generation capacity for cancer treatment using photodynamic therapy (PDT). The co-loading of OSBs and ICG in different nanovesicles has been thoroughly investigated. The nanovesicles' physicochemical properties were manipulated via molecular engineering by modifying the structural properties of the lipid bilayer and the number of oligo(ethyleneoxide) chains in the OSB structure. Diffusion Ordered Spectroscopy (DOSY) NMR and spectrofluorometric studies revealed key differences in the structure of the vesicles and the arrangement of the OSB and ICG in the bilayer. The in vitro assessment of these OSB-ICG nanovesicles revealed that the formulations can increase the temperature and generate ROS after photoirradiation, showing for the first time their potential as dual photothermal/photodynamic (PTT/PDT) agents in the treatment of prostate cancer. Our study provides an exciting opportunity to extend the range of applications of OSB derivates to potentiate the toxicity of phototherapy in prostate and other types of cancer.
Collapse
Affiliation(s)
- Víctor Vázquez-Villar
- Universidad de Castilla-La Mancha, Departamento de Química Inorgánica, Orgánica y Bioquímica, Facultad de Farmacia, C/ José María Sánchez Ibáñez s/n, 02008 Albacete, Spain; Universidad de Castilla-La Mancha, Regional Center for Biomedical Research (CRIB), C/ Almansa 13, 02008 Albacete, Spain
| | - Chandrima Das
- Institute of Cancer Therapeutics, University of Bradford, Bradford, Richmond Rd, Bradford BD7 1DP, United Kingdom
| | - Thomas Swift
- School of Chemistry and Biosciences, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, United Kingdom
| | - Jacobo Elies
- Institute of Cancer Therapeutics, University of Bradford, Bradford, Richmond Rd, Bradford BD7 1DP, United Kingdom
| | - Juan Tolosa
- Universidad de Castilla-La Mancha, Departamento de Química Inorgánica, Orgánica y Bioquímica, Facultad de Farmacia, C/ José María Sánchez Ibáñez s/n, 02008 Albacete, Spain; Universidad de Castilla-La Mancha, Regional Center for Biomedical Research (CRIB), C/ Almansa 13, 02008 Albacete, Spain.
| | - Joaquín C García-Martínez
- Universidad de Castilla-La Mancha, Departamento de Química Inorgánica, Orgánica y Bioquímica, Facultad de Farmacia, C/ José María Sánchez Ibáñez s/n, 02008 Albacete, Spain; Universidad de Castilla-La Mancha, Regional Center for Biomedical Research (CRIB), C/ Almansa 13, 02008 Albacete, Spain.
| | - Amalia Ruiz
- Institute of Cancer Therapeutics, University of Bradford, Bradford, Richmond Rd, Bradford BD7 1DP, United Kingdom.
| |
Collapse
|
9
|
Yang T, Zhang Q, Miao Y, Lyu Y, Xu Y, Yang M, Mao C. Tumor-Homing Phage Nanofibers for Nanozyme-Enhanced Targeted Breast Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2403756. [PMID: 39233557 DOI: 10.1002/adma.202403756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/24/2024] [Indexed: 09/06/2024]
Abstract
Photodynamic therapy (PDT) eliminates cancer cells by converting endogenous oxygen into reactive oxygen species (ROS). However, its efficacy is significantly hindered by hypoxia in solid tumors. Hence, to engineer filamentous fd phage, a human-friendly bacteria-specific virus is proposed, into a nanozyme-nucleating photosensitizer-loaded tumor-homing nanofiber for enhanced production of ROS in a hypoxic tumor. Specifically, Pt-binding and tumor-homing peptides are genetically displayed on the sidewall and tip of the fd phage, respectively. The Pt-binding peptides induced nucleation and orientation of Pt nanozymes (PtNEs) on the sidewall of the phage. The resultant PtNE-coated tumor-homing phage exhibits significantly enhanced sustained catalytic conversion of hydrogen peroxide in hypoxic tumors into O2 for producing ROS needed for PDT, compared to non-phage-templated PtNE. Density functional theory (DFT) calculations verify the catalytic mechanism of the phage-templated PtNE. After intravenous injection of the PtNE-coated indocyanine green (ICG)-loaded tumor-homing phages into breast tumor-bearing mice, the nanofibers home to the tumors and effectively inhibit tumor growth by the PtNE-enhanced PDT. The nanofibers can also serve as a tumor-homing imaging probe due to the fluorescence of ICG. This work demonstrates that filamentous phage, engineered to become tumor-homing nanozyme-nucleating tumor-hypoxia-relieving nanofibers, can act as cancer-targeting nanozymes with improved catalytic performance for effective targeted PDT.
Collapse
Affiliation(s)
- Tao Yang
- School of Materials Science & Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, P. R. China
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, P. R. China
| | - Qinglei Zhang
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | - Yao Miao
- School of Materials Science & Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, P. R. China
| | - Yang Lyu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, P. R. China
| | - Yajing Xu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, P. R. China
| | - Mingying Yang
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | - Chuanbin Mao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, P. R. China
| |
Collapse
|
10
|
An J, Zhang Z, Zhang J, Zhang L, Liang G. Research progress in tumor therapy of carrier-free nanodrug. Biomed Pharmacother 2024; 178:117258. [PMID: 39111083 DOI: 10.1016/j.biopha.2024.117258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/24/2024] [Accepted: 08/02/2024] [Indexed: 08/25/2024] Open
Abstract
Carrier-free nanodrugs are a novel type of drug constructed by the self-assembly of drug molecules without carrier involvement. They have the characteristics of small particle size, easy penetration of various barriers, targeting tumors, and efficient release. In recent years, carrier-free nanodrugs have become a hot topic in tumor therapy as they solve the problems of low drug loading, poor biocompatibility, and low uptake efficiency of carrier nanodrugs. A series of recent studies have shown that carrier-free nanodrugs play a vital role in the treatment of various tumors, with similar or better effects than carrier nanodrugs. Based on the literature published in the past decades, this paper first summarizes the recent progress in the assembly modes of carrier-free nanodrugs, then describes common therapeutic modalities of carrier-free nanodrugs in tumor therapy, and finally depicts the existing challenges along with future trends of carrier-free nanodrugs. We hope that this review can guide the design and application of carrier-free nanodrugs in the future.
Collapse
Affiliation(s)
- Junling An
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, Henan, People's Republic of China.
| | - Zequn Zhang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, Henan, People's Republic of China.
| | - Jinrui Zhang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, Henan, People's Republic of China.
| | - Lingyang Zhang
- Institute of Biomedical Research, Henan Academy of Sciences, Zhengzhou, Henan, People's Republic of China.
| | - Gaofeng Liang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, Henan, People's Republic of China; Institute of Biomedical Research, Henan Academy of Sciences, Zhengzhou, Henan, People's Republic of China.
| |
Collapse
|
11
|
Zhang B, Lu J, Lin X, Wang J, Li Q, Jin T, Shi Q, Lu Y, Zhang J, Deng J, Zhang Y, Guo Y, Gao J, Chen H, Yan Y, Wu J, Gao J, Che J, Dong X, Gu Z, Lin N. Injectable and Sprayable Fluorescent Nanoprobe for Rapid Real-Time Detection of Human Colorectal Tumors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405275. [PMID: 38897213 DOI: 10.1002/adma.202405275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/08/2024] [Indexed: 06/21/2024]
Abstract
The development of minimally invasive surgery has greatly advanced precision tumor surgery, but sometime suffers from restricted visualization of the surgical field, especially during the removal of abdominal tumors. A 3-D inspection of tumors could be achieved by intravenously injecting tumor-selective fluorescent probes, whereas most of which are unable to instantly distinguish tumors via in situ spraying, which is urgently needed in the process of surgery in a convenient manner. In this study, this work has designed an injectable and sprayable fluorescent nanoprobe, termed Poly-g-BAT, to realize rapid tumor imaging in freshly dissected human colorectal tumors and animal models. Mechanistically, the incorporation of γ-glutamyl group facilitates the rapid internalization of Poly-g-BAT, and these internalized nanoprobes can be subsequently activated by intracellular NAD(P)H: quinone oxidoreductase-1 to release near-infrared fluorophores. As a result, Poly-g-BAT can achieve a superior tumor-to-normal ratio (TNR) up to 12.3 and enable a fast visualization (3 min after in situ spraying) of tumor boundaries in the xenograft tumor models, Apcmin/+ mice models and fresh human tumor tissues. In addition, Poly-g-BAT is capable of identifying minimal premalignant lesions via intravenous injection.
Collapse
Affiliation(s)
- Bo Zhang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Jialiang Lu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xu Lin
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jinqiang Wang
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qi Li
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Tingting Jin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Qiuqiu Shi
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yang Lu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jingyu Zhang
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jun Deng
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Yinqiong Zhang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Yu Guo
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jian Gao
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Haifeng Chen
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Youyou Yan
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Cancer Center of Zhejiang University, Hangzhou, 310006, China
- Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Hangzhou, 310024, China
| | - Jiahe Wu
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Cancer Center of Zhejiang University, Hangzhou, 310006, China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jianqing Gao
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jinxin Che
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Zhen Gu
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Nengming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Cancer Center of Zhejiang University, Hangzhou, 310006, China
- Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Hangzhou, 310024, China
| |
Collapse
|
12
|
Nguyen HN, Pertzborn D, Ziadat R, Ernst G, Guntinas-Lichius O, Von Eggeling F, Hoffmann F. Indocyanine green uptake by human tumor and non‑tumor cell lines and tissue. Biomed Rep 2024; 21:136. [PMID: 39114300 PMCID: PMC11304512 DOI: 10.3892/br.2024.1824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024] Open
Abstract
Indocyanine green (ICG) is a potential promising dye for a better intraoperative tumor border definition and an improved patient outcome by potentially improving tumor border visualization compared with traditional white light guided surgery. Here, the cellular uptake of ICG in human squamous cell carcinoma (SCC026) and immortalized non-cancer skin (HaCaT) cell lines was evaluated to study the tumor-specific cellular uptake of ICG. The spatial distribution of ICG inside tumor tissue was investigated in tissue sections of head and neck squamous cell carcinoma at a microscopic level. ICG uptake and internalization was observed in living cells after 2.5 h and in the nucleus after 24 h. In dead cells, higher and faster uptake was observed. In the tissue sections, higher ICG signal intensity could be detected in connective tissue and surrounding clusters and blood vessels. In conclusion, no distinct ICG uptake by tumor cells was detected in cancer cell lines and tumor tissue. ICG localization in certain regions of tumor tissue appears to be a result of enhanced tissue permeability and retention, but not specific to tumor cells.
Collapse
Affiliation(s)
- Hoang-Ngan Nguyen
- Working Group Innovative Biophotonics, Department of Otorhinolaryngology, Jena University Hospital, D-07747 Jena, Germany
| | - David Pertzborn
- Working Group Innovative Biophotonics, Department of Otorhinolaryngology, Jena University Hospital, D-07747 Jena, Germany
| | - Rafat Ziadat
- Working Group Innovative Biophotonics, Department of Otorhinolaryngology, Jena University Hospital, D-07747 Jena, Germany
| | - Günther Ernst
- Working Group Innovative Biophotonics, Department of Otorhinolaryngology, Jena University Hospital, D-07747 Jena, Germany
| | - Orlando Guntinas-Lichius
- Working Group Innovative Biophotonics, Department of Otorhinolaryngology, Jena University Hospital, D-07747 Jena, Germany
| | - Ferdinand Von Eggeling
- Working Group Innovative Biophotonics, Department of Otorhinolaryngology, Jena University Hospital, D-07747 Jena, Germany
| | - Franziska Hoffmann
- Working Group Innovative Biophotonics, Department of Otorhinolaryngology, Jena University Hospital, D-07747 Jena, Germany
| |
Collapse
|
13
|
Liu Z, Ali M, Sun Q, Zhang Q, Wei C, Wang Y, Tang D, Li X. Current status and future trends of real-time imaging in gastric cancer surgery: A literature review. Heliyon 2024; 10:e36143. [PMID: 39253259 PMCID: PMC11381608 DOI: 10.1016/j.heliyon.2024.e36143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/23/2024] [Accepted: 08/09/2024] [Indexed: 09/11/2024] Open
Abstract
Technological advances are crucial for the optimization of gastric cancer surgery, and the success of any gastric cancer surgery is based on the correct and precise anatomical determination of the primary tumour and tissue structures. Real-time imaging-guided surgery is showing increasing potential and utility, mainly because it helps to aid intraoperative decision-making. However, intraoperative imaging faces many challenges in the field of gastric cancer. This article summarizes and discusses the following clinical applications of real-time optical imaging and fluorescence-guided surgery for gastric cancer: (1) the potential of quantitative fluorescence imaging in assessing tissue perfusion, (2) vascular navigation and determination of tumour margins, (3) the advantages and limitations of lymph node drainage assessment, and (4) identification of peritoneal metastases. In addition, preclinical study of tumour-targeted fluorescence imaging are discussed.
Collapse
Affiliation(s)
- Zhu Liu
- The Yangzhou Clinical Medical College of Nanjing Medical University, Yangzhou, 225001, China
- Northern Jiangsu People's Hospital, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, China
| | - Muhammad Ali
- Clinical Medical College, Yangzhou University, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, China
| | - Qiannan Sun
- Northern Jiangsu People's Hospital, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, China
| | - Qi Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, 225001, China
- Northern Jiangsu People's Hospital, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, China
| | - Chen Wei
- Clinical Medical College, Yangzhou University, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, China
| | - Yong Wang
- Northern Jiangsu People's Hospital, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, China
| | - Dong Tang
- Clinical Medical College, Yangzhou University, Yangzhou, 225001, China
- Northern Jiangsu People's Hospital, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, China
| | - Xin Li
- Northern Jiangsu People's Hospital, Yangzhou, 225001, China
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, China
| |
Collapse
|
14
|
Wen Y, Wang J, Zheng W, Zhu J, Song X, Chen T, Zhang M, Huang Z, Li J. A supramolecular colloidal system based on folate-conjugated β-cyclodextrin polymer and indocyanine green for enhanced tumor-targeted cell imaging in 2D culture and 3D tumor spheroids. J Colloid Interface Sci 2024; 667:259-268. [PMID: 38636227 DOI: 10.1016/j.jcis.2024.04.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 04/20/2024]
Abstract
Indocyanine green (ICG) is an FDA-approved medical diagnostic agent that is widely used as a near-infrared (NIR) fluorescent imaging molecular probe. However, ICG tends to aggregate to form dimers or H-aggregates in water and lacks physical and optical stability, which greatly decreases its absorbance and fluorescence intensity in various applications. Additionally, ICG has no tissue- or tumor-targeting properties, and its structure is not easy to modify, which has further limited its application in cancer diagnosis. In this study, we addressed these challenges by developing a supramolecular colloidal carrier system that targets tumor cells. To this end, we synthesized a water-soluble β-cyclodextrin (β-CD) polymer conjugated with folate (FA), denoted PCD-FA, which is capable of forming inclusion complexes with ICG in water through host-guest interactions between the β-CD moieties and ICG molecules. The inclusion complexes formed by PCD-FA and ICG, called ICG@PCD-FA, dispersed stably in solution as colloidal nanoparticles, greatly improving the physical and optical properties of ICG by preventing ICG dimer formation, where ICG appeared as monomers and even J-aggregates. This resulted in stronger and more stable absorption at a longer wavelength of 900 nm, which may allow for deeper tissue penetration and imaging with reduced interference from biological tissues' autofluorescence. Moreover, ICG@PCD-FA showed a targeting effect on folate receptor-positive (FR+) tumor cells, which specifically highlighted FR+ cells via NIR endoscopic imaging. Notably, ICG@PCD-FA further improved permeation and accumulation in FR+ 3D tumor spheroids. Therefore, this ICG@PCD-FA supramolecular colloidal system may have a great potential for use in tumor NIR imaging and diagnostic applications.
Collapse
Affiliation(s)
- Yuting Wen
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore; National University of Singapore (Suzhou) Research Institute, Suzhou, Jiangsu 215000, China; National University of Singapore (Chongqing) Research Institute, Yubei District, Chongqing 401120, China
| | - Jianfeng Wang
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Wei Zheng
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Jingling Zhu
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore; NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Xia Song
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Taili Chen
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Miao Zhang
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Zhiwei Huang
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore; NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore.
| | - Jun Li
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore; National University of Singapore (Suzhou) Research Institute, Suzhou, Jiangsu 215000, China; National University of Singapore (Chongqing) Research Institute, Yubei District, Chongqing 401120, China; NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore.
| |
Collapse
|
15
|
Ram AS, Matuszewska K, McKenna C, Petrik J, Oblak ML. Validation of a semi-quantitative scoring system and workflow for analysis of fluorescence quantification in companion animals. Front Vet Sci 2024; 11:1392504. [PMID: 39144083 PMCID: PMC11322124 DOI: 10.3389/fvets.2024.1392504] [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: 02/27/2024] [Accepted: 07/11/2024] [Indexed: 08/16/2024] Open
Abstract
Significance Many commercially available near-infrared (NIR) fluorescence imaging systems lack algorithms for real-time quantifiable fluorescence data. Creation of a workflow for clinical assessment and post hoc analysis may provide clinical researchers with a method for intraoperative fluorescence quantification to improve objective outcome measures. Aim Scoring systems and verified image analysis are employed to determine the amount and intensity of fluorescence within surgical specimens both intra and postoperatively. Approach Lymph nodes from canine cancer patients were obtained during lymph node extirpation following peritumoral injection of indocyanine green (ICG). First, a semi-quantitative assessment of surface fluorescence was evaluated. Images obtained with a NIR exoscope were analysed to determine fluorescence thresholds and measure fluorescence amount and intensity. Results Post hoc fluorescence quantification (threshold of Hue = 165-180, Intensity = 30-255) displayed strong agreement with semi-quantitative scoring (k = 0.9734, p < 0.0001). Fluorescence intensity with either threshold of 35-255 or 45-255 were significant predictors of fluorescence and had high sensitivity and specificity (p < 0.05). Fluorescence intensity and quantification had a strong association (p < 0.001). Conclusion The validation of the semi-quantitative scoring system by image analysis provides a method for objective in situ observation of tissue fluorescence. The utilization of thresholding for ICG fluorescence intensity allows post hoc quantification of fluorescence when not built into the imaging system.
Collapse
Affiliation(s)
- Ann S. Ram
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - Kathy Matuszewska
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - Charly McKenna
- Department of Clinical Studies, University of Guelph, Guelph, ON, Canada
| | - Jim Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - Michelle L. Oblak
- Department of Clinical Studies, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
16
|
Xu H, Ye Z, Gao X, Dai Y, Luo Y, Han Z, Gu Y. Repurposing GnRH-A as a Near-Infrared Fluorescent Probe for Diagnosis and Surgical Navigation of Breast Cancer Tumors and Metastases. J Med Chem 2024; 67:12386-12398. [PMID: 38995618 DOI: 10.1021/acs.jmedchem.4c01142] [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: 07/13/2024]
Abstract
Breast cancer, globally the most common cancer in women, presents significant challenges in treatment. Breast-conserving surgery (BCS), a less traumatic and painful alternative to radical mastectomy, not only preserves the breast's appearance but also supports postsurgical functional recovery. However, accurately identifying tumors, precisely delineating margins, and thoroughly removing metastases remain complex surgical challenges, exacerbated by the limitations of current imaging techniques, including poor tumor uptake and low signal contrast. Addressing these challenges, our study developed a series of GnRHR-targeted probes (YQGN-n) for fluorescence imaging and surgical navigation of breast cancer through a drug repositioning strategy. Notably, YQGN-7, with its high cellular affinity (Kd of 217.8 nM), demonstrates exceptional selectivity and specificity for breast cancer tumors, surpassing traditional imaging agents like ICG in tumor uptake and pharmacokinetic properties. Furthermore, YQGN-7's effectiveness in surgical navigation, both for primary breast tumors and metastases, highlights its potential as a revolutionary tool in BCS.
Collapse
Affiliation(s)
- Haoran Xu
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Zhuoyi Ye
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Xin Gao
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Yue Dai
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Yang Luo
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Zhihao Han
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Yueqing Gu
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| |
Collapse
|
17
|
Thomson C. Near-infrared-guided Thoracoscopic Surgery and Future Near-infrared Targets. Vet Clin North Am Small Anim Pract 2024; 54:685-695. [PMID: 38508966 DOI: 10.1016/j.cvsm.2024.02.011] [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] [Indexed: 03/22/2024]
Abstract
Intraoperative near-infrared fluorescence imaging allows for real time, noninvasive visualization of anatomic structures (blood vessels, lymphatic vessels) or diseased states (cancer, inflammation). This technique is easily adapted to thoracoscopy and has allowed for improved detection of lung tumors and other various cancers, thoracic lymphatics, and cardiothoracic vasculature.
Collapse
Affiliation(s)
- Chris Thomson
- Surgical Oncology, Veterinary Specialty Hospital - North County, by Ethos Veterinary Health, 2055 Montiel Road. #104, San Marcos, CA 92069, USA.
| |
Collapse
|
18
|
Watanabe K, Takahashi H, Uehara S, Kato A, Fujii Y, Yanagita T, Suzuki T, Ushigome H, Maeda Y, Ogawa R, Matsuo Y, Mitsui A, Takiguchi S. Visualization of cecal tumor by near-infrared laparoscopy and intraoperative colonoscopy. Surg Case Rep 2024; 10:164. [PMID: 38951358 PMCID: PMC11217229 DOI: 10.1186/s40792-024-01964-0] [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: 04/21/2024] [Accepted: 06/24/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND In laparoscopic colorectal surgery, accurate localization of a tumor is essential for ensuring an adequate ablative margin. Therefore, a new method, near-infrared laparoscopy combined with intraoperative colonoscopy, was developed for visualizing the contour of a cecal tumor from outside of the bowel. The method was used after it was verified on a model that employed a silicone tube. CASE PRESENTATION The patient was a 77-year-old man with a cecal tumor near the appendiceal orifice. Laparoscopy was used to clamp of the terminal ileum, and a colonoscope was then inserted through the anus to the cecum. The laparoscope in the normal light mode could not be used to identify the cecal tumor. However, a laparoscope in the near-infrared ray mode could clearly visualize the contour of the cecal tumor from outside of the bowel, and the tumor could be safely resected by a stapler. The histopathological diagnosis of the resected specimen was adenocarcinoma with an invasion depth of M and a clear negative margin. CONCLUSIONS This is the first report of the laparoscopic detection of the contour of a cecal tumor from outside the bowel. This technique is useful and safe for contouring tumors in laparoscopic colorectal surgery and can be used in various surgeries that combine endoscopy and laparoscopy.
Collapse
Affiliation(s)
- Kaori Watanabe
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Hiroki Takahashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.
| | - Shuhei Uehara
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Akira Kato
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Yoshiaki Fujii
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Takeshi Yanagita
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Takuya Suzuki
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Hajime Ushigome
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Yuzo Maeda
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Ryo Ogawa
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Yoichi Matsuo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Akira Mitsui
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Kawasumi 1, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| |
Collapse
|
19
|
Yu H, Yao Y, Zhu T, Sun Y, Zhang M, Zhang Y, Cao M, Zhang W, Yao Y. The potential of indocyanine green fluorescence detection in surgical cut margin of breast conserving surgery. Gland Surg 2024; 13:1031-1044. [PMID: 39015719 PMCID: PMC11247585 DOI: 10.21037/gs-24-195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 06/24/2024] [Indexed: 07/18/2024]
Abstract
Background Fluorescence-guided surgery (FGS) is a cutting-edge technology that uses near-infrared (NIR) fluorescence imaging to guide surgeons in surgery. Indocyanine green (ICG) is a fluorescent dye, which can be used for in vivo imaging of tumor cells. We aimed to explore the use of ICG fluorescence-guided technology as a rapid intraoperative margin assessment method for breast cancer surgery. In addition, we also compared the dose selection of ICG. Methods This was a non-randomized prospective cohort study. Data were collected between August 2021 and October 2022 in the Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University. Upon specimen removal, tumor margins were immediately analyzed by ICG fluorescence detection and then sent to the pathology department for intraoperative frozen section analysis and subsequent routine pathological examination. Abnormal margin rates were calculated and compared using intraoperative frozen section analysis and under the guidance of ICG fluorescence. Results The study included 69 cases of breast cancer patients who underwent tumor resection assisted by ICG fluorescence-guided technology, including 18 patients with a 0.5 mg/kg dose and 51 patients with a 1.0 mg/kg dose. According to the study findings, the ICG test achieved a sensitivity of 81.82% and a specificity of 75.82%. At a dose of 0.5 mg/kg, the sensitivity was 66.67% whereas the specificity was 93.33%. At the dose of 1 mg/kg, the sensitivity was 87.5%, and the specificity was 74.42%. Similarly, for intraoperative frozen section analysis, the sensitivity was 81.82%, but the specificity was enhanced to 94.83%. Positive surgical cut margin was not identified in 2/69 by ICG fluorescence and frozen section analysis respectively. Conclusions The sensitivity of ICG fluorescence detection is comparable to that of frozen section analysis, but the specificity is poor. The sensitivity increased and the specificity decreased at 1 mg/kg compared to the 0.5 mg/kg dose. ICG fluorescence can be used as a supplementary tool for frozen section analysis. These findings support further development and clinical performance assessment of ICG fluorescence.
Collapse
Affiliation(s)
- Hao Yu
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College, Nanjing Medical University, Nanjing, China
| | - Yuhao Yao
- Department of Computer Science, Westcliff University, Irvine, CA, USA
| | - Tingting Zhu
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yulu Sun
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Meng Zhang
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yin Zhang
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Meng Cao
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Weijie Zhang
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yongzhong Yao
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College, Nanjing Medical University, Nanjing, China
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, School of Medicine, Southeast University, Nanjing, China
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
20
|
Jia G, Wang J, Wang H, Hu X, Long F, Yuan C, Liang C, Wang F. New insights into red blood cells in tumor precision diagnosis and treatment. NANOSCALE 2024; 16:11863-11878. [PMID: 38841898 DOI: 10.1039/d4nr01454e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Red blood cells (RBCs), which function as material transporters in organisms, are rich in materials that are exchanged with metabolically active tumor cells. Recent studies have demonstrated that tumor cells can regulate biological changes in RBCs, including influencing differentiation, maturation, and morphology. RBCs play an important role in tumor development and immune regulation. Notably, the novel scientific finding that RBCs absorb fragments of tumor-carrying DNA overturns the conventional wisdom that RBCs do not contain nucleic acids. RBC membranes are excellent biomimetic materials with significant advantages in terms of their biocompatibility, non-immunogenicity, non-specific adsorption resistance, and biodegradability. Therefore, RBCs provide a new research perspective for the development of tumor liquid biopsies, molecular imaging, drug delivery, and other tumor precision diagnosis and treatment technologies.
Collapse
Affiliation(s)
- Gaihua Jia
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Jun Wang
- Department of Laboratory Medicine, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430016, China.
| | - Hu Wang
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Xin Hu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Fei Long
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Chunhui Yuan
- Department of Laboratory Medicine, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430016, China.
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Chen Liang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Fubing Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
21
|
Hou Y, Zhu C, Ban G, Shen Z, Liang Y, Chen K, Wang C, Shi H. Advancements and Challenges in the Application of Metal-Organic Framework (MOF) Nanocomposites for Tumor Diagnosis and Treatment. Int J Nanomedicine 2024; 19:6295-6317. [PMID: 38919774 PMCID: PMC11198007 DOI: 10.2147/ijn.s463144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Nanoscale metal-organic frameworks (MOFs) offer high biocompatibility, nanomaterial permeability, substantial specific surface area, and well-defined pores. These properties make MOFs valuable in biomedical applications, including biological targeting and drug delivery. They also play a critical role in tumor diagnosis and treatment, including tumor cell targeting, identification, imaging, and therapeutic methods such as drug delivery, photothermal effects, photodynamic therapy, and immunogenic cell death. The diversity of MOFs with different metal centers, organics, and surface modifications underscores their multifaceted contributions to tumor research and treatment. This review is a summary of these roles and mechanisms. The final section of this review summarizes the current state of the field and discusses prospects that may bring MOFs closer to pharmaceutical applications.
Collapse
Affiliation(s)
- Yingze Hou
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
- Clinical Medical College, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
| | - Can Zhu
- Department of Urology, The Second Clinical Medical College of Anhui Medical University, Hefei, 230032, People’s Republic of China
| | - Ge Ban
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
| | - Zhean Shen
- Heart Center, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310000, People’s Republic of China
| | - Yingbing Liang
- Department of Chemistry and Biotechnology, Graduate School of Engineering Tottori University Koyama-Minami 4-101, Tottori, 680-8552, Japan
| | - Kun Chen
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
| | - Chenbo Wang
- School of Intelligent Medical Engineering, Sanquan College of Xinxiang Medical University, Xinxiang, 453003, People’s Republic of China
| | - Heng Shi
- Heart Center, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310000, People’s Republic of China
| |
Collapse
|
22
|
Lu K, Pan X, Zheng J, Cheng D, Zheng L, Zhang X. Theranostic nanoparticles ZIF-8@ICG for pH/NIR-responsive drug-release and NIR-guided chemo-phototherapy against non-small-cell lung cancer. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2024; 35:32. [PMID: 38896160 PMCID: PMC11186913 DOI: 10.1007/s10856-024-06802-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 05/17/2024] [Indexed: 06/21/2024]
Abstract
This study leverages nanotechnology by encapsulating indocyanine green (ICG) and paclitaxel (Tax) using zeolitic imidazolate frameworks-8 (ZIF-8) as a scaffold. This study aims to investigate the chemo-photothermal therapeutic potential of ZIF-8@ICG@Tax nanoparticles (NPs) in the treatment of non-small cell lung cancer (NSCLC). An "all-in-one" theranostic ZIF-8@ICG@Tax NPs was conducted by self-assembly based on electrostatic interaction. First, the photothermal effect, stability, pH responsiveness, drug release, and blood compatibility of ZIF-8@ICG@Tax were evaluated through in vitro testing. Furthermore, the hepatic and renal toxicity of ZIF-8@ICG@Tax were assessed through in vivo testing. Additionally, the anticancer effects of these nanoparticles were investigated both in vitro and in vivo. Uniform and stable chemo-photothermal ZIF-8@ICG@Tax NPs had been successfully synthesized and had outstanding drug releasing capacities. Moreover, ZIF-8@ICG@Tax NPs showed remarkable responsiveness dependent both on pH in the tumor microenvironment and NIR irradiation, allowing for targeted drug delivery and controlled drug release. NIR irradiation can enhance the tumor cell response to ZIF-8@ICG@Tax uptake, thereby promoting the anti-tumor growth in vitro and in vivo. ZIF-8@ICG@Tax and NIR irradiation have demonstrated remarkable synergistic anti-tumor growth properties compared to their individual components. This novel theranostic chemo-photothermal NPs hold great potential as a viable treatment option for NSCLC.
Collapse
Affiliation(s)
- Kaiming Lu
- Department of Operating Room, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, PR China
| | - Xiongfeng Pan
- Department of Thoracic Surgery, The Affiliated Cangnan Hospital of Wenzhou Medical University, Wenzhou, 325800, PR China
| | - Jinyu Zheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, PR China
| | - Dezhi Cheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, PR China
| | - Liangcheng Zheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, PR China
| | - Xinbo Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, PR China.
| |
Collapse
|
23
|
Cressoni C, Malandra S, Milan E, Boschi F, Nicolato E, Negri A, Veccia A, Bontempi P, Mangiameli D, Pietrobono S, Melisi D, Marzola P, Antonelli A, Speghini A. Injectable Thermogelling Nanostructured Ink as Simultaneous Optical and Magnetic Resonance Imaging Contrast Agent for Image-Guided Surgery. Biomacromolecules 2024; 25:3741-3755. [PMID: 38783486 DOI: 10.1021/acs.biomac.4c00312] [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: 05/25/2024]
Abstract
The development of efficient and biocompatible contrast agents is particularly urgent for modern clinical surgery. Nanostructured materials raised great interest as contrast agents for different imaging techniques, for which essential features are high contrasts, and in the case of precise clinical surgery, minimization of the signal spatial dispersion when embedded in biological tissues. This study deals with the development of a multimodal contrast agent based on an injectable hydrogel nanocomposite containing a lanthanide-activated layered double hydroxide coupled to a biocompatible dye (indocyanine green), emitting in the first biological window. This novel nanostructured thermogelling hydrogel behaves as an efficient tissue marker for optical and magnetic resonance imaging because the particular formulation strongly limits its spatial diffusion in biological tissue by exploiting a simple injection. The synergistic combination of these properties permits to employ the hydrogel ink simultaneously for both optical and magnetic resonance imaging, easy monitoring of the biological target, and, at the same time, increasing the spatial resolution during a clinical surgery. The biocompatibility and excellent performance as contrast agents are very promising for possible use in image-guided surgery, which is currently one of the most challenging topics in clinical research.
Collapse
Affiliation(s)
- Chiara Cressoni
- Nanomaterials Research Group, Department of Biotechnology and INSTM, RU of Verona, University of Verona, Strada le Grazie 15, 37134 Verona, Italy
| | - Sarah Malandra
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Urology Unit, University of Verona, Azienda Ospedaliera Universitaria Integrata Verona (AOUI), P.le A. Stefani 1, 37126 Verona, Italy
| | - Emil Milan
- Nanomaterials Research Group, Department of Biotechnology and INSTM, RU of Verona, University of Verona, Strada le Grazie 15, 37134 Verona, Italy
| | - Federico Boschi
- Department of Engineering for Innovation Medicine, University of Verona, Strada le Grazie 15, 37134 Verona, Italy
| | - Elena Nicolato
- Centre of Tecnological Platforms, University of Verona, Strada le Grazie 8, 37134 Verona, Italy
| | - Alessandro Negri
- Department of Engineering for Innovation Medicine, University of Verona, Strada le Grazie 15, 37134 Verona, Italy
| | - Alessandro Veccia
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Urology Unit, University of Verona, Azienda Ospedaliera Universitaria Integrata Verona (AOUI), P.le A. Stefani 1, 37126 Verona, Italy
| | - Pietro Bontempi
- Department of Engineering for Innovation Medicine, University of Verona, Strada le Grazie 15, 37134 Verona, Italy
| | - Domenico Mangiameli
- Department of Medicine, University of Verona, Piazzale Ludovico Antonio Scuro 10, 37124 Verona, Italy
| | - Silvia Pietrobono
- Department of Medicine, University of Verona, Piazzale Ludovico Antonio Scuro 10, 37124 Verona, Italy
| | - Davide Melisi
- Department of Medicine, University of Verona, Piazzale Ludovico Antonio Scuro 10, 37124 Verona, Italy
| | - Pasquina Marzola
- Department of Engineering for Innovation Medicine, University of Verona, Strada le Grazie 15, 37134 Verona, Italy
| | - Alessandro Antonelli
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Urology Unit, University of Verona, Azienda Ospedaliera Universitaria Integrata Verona (AOUI), P.le A. Stefani 1, 37126 Verona, Italy
| | - Adolfo Speghini
- Nanomaterials Research Group, Department of Biotechnology and INSTM, RU of Verona, University of Verona, Strada le Grazie 15, 37134 Verona, Italy
| |
Collapse
|
24
|
Loverro M, Bizzarri N, Capomacchia FM, Watrowski R, Querleu D, Gioè A, Naldini A, Santullo F, Foschi N, Fagotti A, Scambia G, Fanfani F. Indocyanine green fluorescence applied to gynecologic oncology: beyond sentinel lymph node. Int J Surg 2024; 110:3641-3653. [PMID: 38489558 PMCID: PMC11175818 DOI: 10.1097/js9.0000000000001318] [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: 09/16/2023] [Accepted: 02/26/2024] [Indexed: 03/17/2024]
Abstract
Indocyanine green (ICG), a well-known molecule employed in medicine for over five decades, has emerged as a versatile dye widely embraced across various surgical disciplines. In gynecologic oncology, its prevalent use revolves around the detection of sentinel lymph nodes. However, the true potential of ICG extends beyond this singular application, owing to its pragmatic utility, cost-effectiveness, and safety profile. Furthermore, ICG has been introduced in the theranostic landscape, marking a significant juncture in the evolution of its clinical utility. This narrative review aims to describe the expanding horizons of ICG fluorescence in gynecologic oncology, beyond the sentinel lymph node biopsy. The manifold applications reported within this manuscript include: 1) lymphography; 2) angiography; 3) nerve visualization; 4) ICG-driven resections; and 5) theranostic. The extensive exploration across these numerous applications, some of which are still in the preclinical phase, serves as a hypothesis generator, aiming to stimulate the development of clinical studies capable of expanding the use of this drug in our field, enhancing the care of gynecological cancer patients.
Collapse
Affiliation(s)
- Matteo Loverro
- UOC Ginecologia Oncologica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS
| | - Nicolò Bizzarri
- UOC Ginecologia Oncologica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS
| | | | - Rafał Watrowski
- Department of Gynecology and Obsterics, Helios Hospital Müllheim, Teaching Hospital of the University of Freiburg, 79379 Müllheim
- Faculty of Medicine, Medical Center - University of Freiburg, 79106 Freiburg, Germany
| | - Denis Querleu
- UOC Ginecologia Oncologica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS
| | - Alessandro Gioè
- UOC Ginecologia Oncologica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS
| | - Angelica Naldini
- UOC Ginecologia Oncologica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS
| | - Francesco Santullo
- Operational Unit of Peritoneum and Retroperitoneum Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli
| | - Nazario Foschi
- Urology Division, Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| | - Anna Fagotti
- UOC Ginecologia Oncologica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS
- Università Cattolica del Sacro Cuore
| | - Giovanni Scambia
- UOC Ginecologia Oncologica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS
- Università Cattolica del Sacro Cuore
| | - Francesco Fanfani
- UOC Ginecologia Oncologica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS
- Università Cattolica del Sacro Cuore
| |
Collapse
|
25
|
Gan YX, Yang ZL, Pan YX, Ou-Yang LY, Tang YH, Zhang YJ, Chen MS, Xu L. Change of indocyanine green clearance ability and liver function after transcatheter intra-arterial therapies and its impact on outcomes of resectable hepatocellular carcinoma: a retrospective cohort study. Int J Surg 2024; 110:2832-2844. [PMID: 38363991 DOI: 10.1097/js9.0000000000001156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/26/2024] [Indexed: 02/18/2024]
Abstract
BACKGROUND Indocyanine green (ICG) clearance test is a classical measurement of hepatic reserve, which involves surgical safety and patient recovery of hepatocellular carcinoma (HCC). The authors aim to compare effects of hepatic arterial infusion chemotherapy (HAIC) and transcatheter arterial chemoembolization (TACE) on liver function and outcomes of subsequent hepatectomy. MATERIAL AND METHODS HCC patients receiving HAIC/TACE in SYSUCC with repeated ICG clearance tests were retrospectively enrolled. ICG eliminating rate (ICG-K), ICG retention rate at 15 min (ICG-R15) and ordinary laboratory tests were collected. Peri-therapeutic changes of values were compared between the groups. Propensity score matching (PSM) and inverse probability of treatment weighing (IPTW) were employed to validate findings. Post-hepatectomy liver failure (PHLF), overall survival (OS) and recurrence-free survival (RFS) were analyzed in patients with subsequent curative hepatectomy. RESULTS Two hundred and four patients treated with HAIC ( n =130) and TACE ( n =74) were included. ΔICG-R15 was greater in the HAIC arm before matching (mean, 3.8% vs. 0.7%, P <0.001), after PSM (mean, 4.7% vs. 1.1%, P =0.014) and IPTW (mean, 2.0% vs. -3.6%, P <0.001). No difference was found for ΔALB, ΔALBI, ΔTBIL, ΔALT, ΔAST and ΔPT-INR. Multivariable analyses revealed elder age, cirrhosis, HAIC, greater ΔTBIL and ΔALBI were associated with deteriorating ICG-R15. Among those (105 for HAIC and 48 for TACE) receiving hepatectomy, occurrence of grade B/C PHLF (4.8% vs. 8.3%, P =0.616), OS (median, unreached vs. unreached, P =0.94) and RFS (median, 26.7 vs. 17.1 months, P =0.096) were comparable between the two arms. In subgroup analyses, preoperative HAIC yield superior RFS (median, 26.7 vs. 16.2 months, P =0.042) in patients with baseline ICG-R15 less than or equal to 10%. CONCLUSION Preoperative FOLFOX-HAIC caused apparent impairment of ICG clearance ability than TACE yet comparable impact on liver function and post-hepatectomy outcomes.
Collapse
Affiliation(s)
| | | | | | - Li-Ying Ou-Yang
- Intensive Care Unit, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center
| | - Yu-Hao Tang
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | | | - Li Xu
- Department of Liver Surgery
| |
Collapse
|
26
|
Li L, Jing J, Wang J, Zhang L, Zheng L, Chen L. [Preliminary application of indocyanine green fluorescence imaging technology in nasal endoscopic tumor surgery]. LIN CHUANG ER BI YAN HOU TOU JING WAI KE ZA ZHI = JOURNAL OF CLINICAL OTORHINOLARYNGOLOGY, HEAD, AND NECK SURGERY 2024; 38:284-287. [PMID: 38563169 PMCID: PMC11387301 DOI: 10.13201/j.issn.2096-7993.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Indexed: 04/04/2024]
Abstract
Objective:To preliminarily study the practical value of Indocyanine green(ICG) molecular fluorescence imaging technology in nasal endoscopic tumor surgery. Methods:Five patients with tumors related to nasal sinuses, orbital wall and skull base in the Department of Otolaryngology head and Neck Surgery, General Hospital of Xinjiang Military Command from December 2022 to April 2023 were enrolled. Among them, 3 were benign tumors and 2 were malignant tumors. All patients underwent surgery under the guidance of ICG molecular fluorescence imaging. ICG was administered intravenously through cubital vein at a dose of 0.5 mg/kg 12 to 24 h before surgery. Tumors were labeled by fluorescence imaging during the operation. surgeons cleared the tumor tissue strictly according to the labeled range and depth, malignant tumors were further expanded and cleaned according to pathology results. Results:All 5 patients achieved accurate tumor localization with the aid of fluorescence imaging technology. Resections were performed with reference to fluorescent labeling boundaries, all patients achieved complete tumor cleanup or negative margins. Conclusion:For tumor-related surgery under nasal endoscopy, ICG molecular fluorescence imaging technology can not only achieve accurate real-time positioning, but also provide evidence for surgeons to judge tumor boundaries. Therefore, we believe that the technology should have certain practical value in nasal endoscopic tumor surgery.
Collapse
Affiliation(s)
- Liang Li
- Department of Otolaryngology Head and Neck Surgery,General Hospital of Xinjiang Military Command,Urumqi,830000,China
| | - Jianjun Jing
- Department of Otolaryngology Head and Neck Surgery,General Hospital of Xinjiang Military Command,Urumqi,830000,China
| | - Jin Wang
- Department of Otolaryngology Head and Neck Surgery,General Hospital of PLA
| | - Longfang Zhang
- Department of Otolaryngology Head and Neck Surgery,General Hospital of Xinjiang Military Command,Urumqi,830000,China
| | - Liang Zheng
- Department of Otolaryngology Head and Neck Surgery,General Hospital of Xinjiang Military Command,Urumqi,830000,China
| | - Lei Chen
- Department of Otolaryngology Head and Neck Surgery,General Hospital of PLA
| |
Collapse
|
27
|
Sun S, Jiang G, Dong J, Xie X, Liao J, Tian Y. Photothermal hydrogels for infection control and tissue regeneration. Front Bioeng Biotechnol 2024; 12:1389327. [PMID: 38605983 PMCID: PMC11007110 DOI: 10.3389/fbioe.2024.1389327] [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: 02/21/2024] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
In this review, we report investigating photothermal hydrogels, innovative biomedical materials designed for infection control and tissue regeneration. These hydrogels exhibit responsiveness to near-infrared (NIR) stimulation, altering their structure and properties, which is pivotal for medical applications. Photothermal hydrogels have emerged as a significant advancement in medical materials, harnessing photothermal agents (PTAs) to respond to NIR light. This responsiveness is crucial for controlling infections and promoting tissue healing. We discuss three construction methods for preparing photothermal hydrogels, emphasizing their design and synthesis, which incorporate PTAs to achieve the desired photothermal effects. The application of these hydrogels demonstrates enhanced infection control and tissue regeneration, supported by their unique photothermal properties. Although research progress in photothermal hydrogels is promising, challenges remain. We address these issues and explore future directions to enhance their therapeutic potential.
Collapse
Affiliation(s)
- Siyu Sun
- College of Biomass Science and Engineering, Sichuan University, Chengdu, China
| | - Guangyang Jiang
- College of Biomass Science and Engineering, Sichuan University, Chengdu, China
| | - Jianru Dong
- College of Biomass Science and Engineering, Sichuan University, Chengdu, China
| | - Xi Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jinfeng Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yongqiang Tian
- College of Biomass Science and Engineering, Sichuan University, Chengdu, China
| |
Collapse
|
28
|
Zhang Y, Zhang P, Xu J, Zhao J, Yan R, Zhang A, Luo Y, Liao W, Huang C, Deng W, Nie Y. Novel indocyanine green-loaded photothermal nanoparticles targeting TRPV1 for thermal ablation treatment of severe murine asthma induced by ovalbumin and lipopolysaccharide. Int J Pharm 2024; 651:123778. [PMID: 38181990 DOI: 10.1016/j.ijpharm.2024.123778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/01/2024] [Accepted: 01/02/2024] [Indexed: 01/07/2024]
Abstract
To identify a replacement strategy for bronchial thermoplasty (BT) with non-invasive and free-of-severe side effect is urgently needed in the clinic for severe asthma treatment. In this study, PLGA-PEG@ICG@TRPV1 pAb (PIT) photothermal nanoparticles targeting bronchial TRPV1 were designed for photothermal therapy (PTT) against severe murine asthma induced by ovalbumin and lipopolysaccharide. PIT was formulated with a polyethylene glycol (PEG)-grafted poly (lactic-co-glycolic) acid (PLGA) coating as a skeleton structure to encapsulate indocyanine green (ICG) and was conjugated to the polyclonal antibody against transient receptor potential vanilloid 1 (TRPV1 pAb). The results revealed that PIT held good druggability due to its electronegativity and small diameter. PIT demonstrated great photothermal effects both in vivo and in vitro and exhibited good ability to target TRPV1 in vitro because of its selective cell uptake and specific cell toxicity toward TRPV1-overexpressing cells. The PIT treatment effectively reduced asthma symptoms in mice. This is evident from improvements in expiratory airflow limitation, significant decreases in inflammatory cell infiltration in the airways, and increases in goblet cell and columnar epithelial cell proliferation. In conclusion, PIT alleviates severe murine asthma symptoms through a combination of TRPV1 targeting and photothermal effects.
Collapse
Affiliation(s)
- Yidi Zhang
- Translational Medicine Research Institute, the First People's Hospital of Foshan, Foshan, 528000, PR China; School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Peifang Zhang
- Pulmonary and Critical Care Medicine, the First People's Hospital of Foshan, Foshan, 528000, PR China
| | - Jian Xu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Jingxin Zhao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Rong Yan
- Translational Medicine Research Institute, the First People's Hospital of Foshan, Foshan, 528000, PR China
| | - Aili Zhang
- Translational Medicine Research Institute, the First People's Hospital of Foshan, Foshan, 528000, PR China
| | - Yulong Luo
- Innovation Centre for Advanced Interdisciplinary Medicine, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510275, PR China
| | - Weiping Liao
- Foshan Fourth People's Hospital, Foshan, 528000, PR China.
| | - Chuqin Huang
- State Key Laboratory of Respiratory Diseases, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510120, PR China.
| | - Wenbin Deng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Guangzhou, 510275, PR China.
| | - Yichu Nie
- Translational Medicine Research Institute, the First People's Hospital of Foshan, Foshan, 528000, PR China.
| |
Collapse
|
29
|
Shevchenko I, Serban D, Dascalu AM, Tribus L, Alius C, Cristea BM, Suceveanu AI, Voiculescu D, Dumitrescu D, Bobirca F, Suceveanu AP, Georgescu DE, Serboiu CS. Factors Affecting the Efficiency of Near-Infrared Indocyanine Green (NIR/ICG) in Lymphatic Mapping for Colorectal Cancer: A Systematic Review. Cureus 2024; 16:e55290. [PMID: 38558607 PMCID: PMC10981778 DOI: 10.7759/cureus.55290] [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] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
As laparoscopy gained global popularity in oncologic surgery, the challenge of detecting lymph nodes spurred researchers to explore innovative techniques and approach the situation from a fresh perspective. While many proposed methods have faded into obscurity, the utilization of indocyanine green (ICG) in the surgical treatment of oncologic patients has continued to advance. The immense potential of this dye is widely acknowledged, yet its full extent and limitations in lymphatic mapping for colorectal cancer remain to be precisely determined. This article aims to assess the magnitude of its potential and explore the constraints based on insights from clinical studies published by pioneering researchers. A systematic review of the existing literature, comprising articles in English, was conducted using the Scopus, PubMed, and Springer Link databases. The search employed keywords such as "colorectal cancer" AND/OR "indocyanine green," "fluorescence" AND/OR "lymphatic mapping" AND/OR "lymph nodes." Initially identifying 129 articles, the application of selection criteria narrowed down the pool to 10 articles, which served as the primary sources of data for our review. Despite the absence of a standardized protocol for the application of ICG in colorectal cancer, particularly in the context of lymphatic mapping, the detection rates have exhibited considerable variation across studies. Nevertheless, all authors unanimously regarded this technique as beneficial and promising. Additionally, it is advocated as an adjunctive tool to enhance the accuracy of cancer staging. Near-infrared (NIR)-enhanced surgery holds the promise of transforming the landscape of oncologic surgery, emerging as a valuable tool for surgeons. However, the absence of a standardized technique and the subjective nature of result assessment impose limitations on the potential of this method. Consequently, it can be inferred that the establishment of a universally accepted protocol, encompassing parameters such as dose, concentration, technique, and site of administration of ICG, along with the optimal time needed for fluorescence visualization, would enhance the outcomes. Emphasizing the accurate selection of patients is crucial to prevent the occurrence of false-negative results.
Collapse
Affiliation(s)
- Irina Shevchenko
- Surgery, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
- General Surgery, Emergency University Hospital Bucharest, Bucharest, ROU
| | - Dragos Serban
- Surgery, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
- General Surgery, Emergency University Hospital Bucharest, Bucharest, ROU
| | - Ana Maria Dascalu
- Ophthalmology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Laura Tribus
- Gastroenterology, Faculty of Oral Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Catalin Alius
- Surgery, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
- General Surgery, Emergency University Hospital Bucharest, Bucharest, ROU
| | - Bogdan Mihai Cristea
- Anatomy, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Andra Iulia Suceveanu
- Gastroenterology, Faculty of Medicine, Ovidius University of Constanta, Constanta, ROU
- Gastroenterology, Clinical Emergency Hospital St Apostle Andrew, Constanta, ROU
| | - Daniel Voiculescu
- Surgery, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
- General Surgery, Emergency University Hospital Bucharest, Bucharest, ROU
| | - Dan Dumitrescu
- Surgery, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
- General Surgery, Emergency University Hospital Bucharest, Bucharest, ROU
| | - Florin Bobirca
- Surgery, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
- General Surgery, Clinic Hospital "Dr. Ioan Cantacuzino" Bucharest, Bucharest, ROU
| | - Adrian Paul Suceveanu
- Medicine, Faculty of Medicine, Ovidius University of Constanta, Constanta, ROU
- Gastroenterology, Clinical Emergency Hospital St Apostle Andrew, Constanta, ROU
| | - Dragos Eugen Georgescu
- Surgery, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Crenguta Sorina Serboiu
- Radiology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| |
Collapse
|
30
|
Zhao X, Li S, Song Y, Fan L. Construction of a near infrared fluorescence system for imaging of biological tissues. Sci Rep 2024; 14:1626. [PMID: 38238385 PMCID: PMC10796378 DOI: 10.1038/s41598-024-51583-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 01/07/2024] [Indexed: 01/22/2024] Open
Abstract
Surgical procedures often rely on unaided visual observation or endoscopic assistance, which may pose challenges in cases involving intricate anatomical relationships. Real-time imaging technologies capable of intraoperative visualization of target organs have the potential to enhance the precision of surgical procedures by facilitating accurate identification, separation, and protection of vital tissues or organs. Despite these advantages, the widespread adoption of such technologies has been hindered by factors such as the prohibitive cost of equipment. This study aims to optimize and develop a device based on Indocyanine Green (ICG) for fluorescence imaging. The objective is to monitor changes in the average fluorescence intensity of ICG in the bladder, offering valuable guidance for surgeries involving the bladder. 1. Male rabbits were administered 0.01 mg/ml ICG via the renal pelvis and ear vein to obtain fluorescence images of the ureter, bladder, and small intestine. 2. After ligating the bilateral ureters of male rabbits, a retrograde bladder perfusion of 5 ml 0.01 mg/ml ICG was conducted to capture fluorescence images of the bladder over time. The average fluorescence intensity was computed using Image Pro Plus 6.0, and the corresponding curve was generated using Prism 8.0. Using a similar methodology, the average fluorescence intensity of male rabbits without ureteral ligation was measured and plotted over time. 1. The developed device facilitated imaging of the ureter, bladder, and small intestine. 2. The bladder's average fluorescence intensity exhibited changes over time in response to urine production and ureteral ligation, contrasting with observations without ureteral ligation. We have successfully constructed and optimized a modular fluorescence imaging system for organs and tissues. This system proves effective in imaging experiments involving hollow organs in animals and offers valuable insights for relevant surgical procedures.
Collapse
Affiliation(s)
- Xu Zhao
- Department of Urology, General Hospital of Northern Theater Command, Shenhe District, No. 83, Wenhua Rd, Shenyang, 110000, China
- Department of Graduate School, China Medical University, Shenyang, China
| | - Shilin Li
- Department of Urology, General Hospital of Northern Theater Command, Shenhe District, No. 83, Wenhua Rd, Shenyang, 110000, China
- Department of Graduate School, China Medical University, Shenyang, China
| | - Yue Song
- Department of Urology, General Hospital of Northern Theater Command, Shenhe District, No. 83, Wenhua Rd, Shenyang, 110000, China.
| | - Lianhui Fan
- Department of Urology, General Hospital of Northern Theater Command, Shenhe District, No. 83, Wenhua Rd, Shenyang, 110000, China.
| |
Collapse
|
31
|
Yao S, Wu D, Hu X, Chen Y, Fan W, Mou X, Cai Y, Yang X. Platelet membrane-coated bio-nanoparticles of indocyanine green/elamipretide for NIR diagnosis and antioxidant therapy in acute kidney injury. Acta Biomater 2024; 173:482-494. [PMID: 37981044 DOI: 10.1016/j.actbio.2023.11.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/03/2023] [Accepted: 11/08/2023] [Indexed: 11/21/2023]
Abstract
Acute kidney injury (AKI) is a prevalent condition in critically ill patients that is often associated with significant morbidity and mortality. As the lack of effective early diagnosis methods often delays AKI treatment, there is currently no definitive clinical intervention available. In this study, we aimed to address these challenges by developing a nano-system called Platelet membranes-ICG-SS31-PLGA (PISP), which was designed to selectively target to the kidney site, taking advantage of the natural tendency of platelets to accumulate at sites of vascular injury. This approach allowed for the accumulation of PISP within the kidney as the disease progresses. By incorporating ICG, the in vivo distribution of PISP can be observed for NIR diagnosis of AKI. This non-invasive imaging technique holds great promise for early detection and monitoring of AKI. Furthermore, Elamipretide (SS31) acts as a mitochondria-targeted antioxidant that protects against mitochondrial damage and reduces oxidative stress, inflammation, and apoptosis. The combination of diagnostic and therapeutic capabilities within a single nano-system makes the PISP approach a valuable tool for addressing AKI. This intervention helps to prevent the deterioration of AKI and promotes the recovery. STATEMENT OF SIGNIFICANCE.
Collapse
Affiliation(s)
- Shijie Yao
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Danping Wu
- Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xiaojuan Hu
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Yang Chen
- Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Weijiao Fan
- Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xiaozhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China.
| | - Yu Cai
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China.
| | - Xianghong Yang
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China.
| |
Collapse
|
32
|
Shu WJ, Ma Z, Tian X, Wang F. Near-Infrared Fluorescence Imaging of miRNA Using a Transmembrane Polypeptide-Based Genetic Reporter. SMALL METHODS 2024; 8:e2300990. [PMID: 37882335 DOI: 10.1002/smtd.202300990] [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: 08/03/2023] [Revised: 09/29/2023] [Indexed: 10/27/2023]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that play important regulatory roles in multiple biological processes. Many miRNAs exhibit unique expression patterns and are considered as theranostic biomarkers in a variety of human diseases. A reporter system that is capable of imaging miRNA in vivo is crucial for investigating miRNA biology. In the present study, an organic anion-transporting polypeptide 1B3 (OATP1B3)-based genetic switch system is designed and optimized to achieve near-infrared fluorescent imaging of miRNA by the uptake of indocyanine green (ICG) dye. The reporter system, named miR-ON-OB3, is shown to be efficient to regulate the expression of OATP1B3 in mammalian cells. Notably, the results indicate that the system is of high sensitivity for near-infrared fluorescence imaging of both exogenous and endogenous miRNA in mammalian cells. Moreover, the system is proved to be functional for real-time near-infrared fluorescence imaging of miRNA in living mice. This study establishes a novel genetic encoded reporter for near-infrared fluorescence imaging of miRNA, which may provide a potential tool for in vivo imaging of miRNA in clinical applications due to the clinical availability of ICG.
Collapse
Affiliation(s)
- Wen-Jie Shu
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Zhe Ma
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xiaojie Tian
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Fu Wang
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Xianyang Key Laboratory of Molecular Imaging and Drug Synthesis, School of Pharmacy, Shaanxi Institute of International Trade & Commerce, Xianyang, Shaanxi, 712046, China
| |
Collapse
|
33
|
Chen J, Li J, Zhong C, Ling Y, Liu D, Li X, Xu J, Liu Q, Guo Y, Wang L. Nanobody-loaded nanobubbles targeting the G250 antigen with ultrasound/photoacoustic/fluorescence multimodal imaging capabilities for specifically enhanced imaging of RCC. NANOSCALE 2023; 16:343-359. [PMID: 38062769 DOI: 10.1039/d3nr04097f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
Clinicians have attempted to discover a noninvasive, easy-to-perform, and accurate method to distinguish benign and malignant renal masses. The targeted nanobubbles (NBs) we constructed that target the specific membrane antigen of renal cell carcinoma (RCC), G250, and contain indocyanine green (ICG) provide multimodal enhanced imaging capability in ultrasound/photoacoustic/fluorescence for RCC which may possibly solve this problem. In this study, we encapsulated ICG in the lipid shell of the NBs by mechanical oscillation, then anti-G250 nanobodies (AGN) were coupled to the surfaces by the biotin-streptavidin bridge method, and the nanobubble named AGN/ICG-NB was completely constructed. The average particle diameter of the prepared AGN/ICG-NBs was (427.2 ± 4.50) nm, and the zeta potential was (-13.33 ± 1.01) mV. Immunofluorescence and flow cytometry confirmed the specific binding capability of AGN/ICG-NBs to G250-positive cells. In vitro imaging experiments confirmed the multimodal imaging capability of AGN/ICG-NBs, and the in vivo imaging experiments demonstrated the specifically enhanced ability of AGN/ICG-NBs for ultrasound/photoacoustic/fluorescence imaging of human-derived RCC tumors. The biosafety of AGN/ICG-NB was verified by CCK-8 assay, organ H&E staining and blood biochemical indices. In conclusion, the targeted nanobubbles we prepared with ultrasound/photoacoustic/fluorescence multimodal imaging capabilities provide a potentially feasible approach to address the need for early diagnosis and differential diagnosis of renal masses.
Collapse
Affiliation(s)
- Jiajiu Chen
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China.
| | - Jingyi Li
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China.
| | - Chengjie Zhong
- The Second Clinical Medical College, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yi Ling
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing 400038, P.R. China.
| | - Deng Liu
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing 400038, P.R. China.
| | - Xin Li
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing 400038, P.R. China.
| | - Jing Xu
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China.
| | - Qiuli Liu
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China.
| | - Yanli Guo
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing 400038, P.R. China.
| | - Luofu Wang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, P.R. China.
| |
Collapse
|
34
|
Zhang X, He Z. Cell Membrane Coated pH-Responsive Intelligent Bionic Delivery Nanoplatform for Active Targeting in Photothermal Therapy. Int J Nanomedicine 2023; 18:7729-7744. [PMID: 38115989 PMCID: PMC10729683 DOI: 10.2147/ijn.s436940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/29/2023] [Indexed: 12/21/2023] Open
Abstract
Aim To produce pH-responsive bionic high photothermal conversion nanoparticles actively targeting tumors for sensitizing photothermal therapy (PTT). Materials and Methods The bionic nanoparticles (ICG-PEI@HM NPs) were prepared by electrostatic adsorption of indocyanine green (ICG) coupled to polyethyleneimine (PEI) and modified with tumor cell membranes. In vitro and in vivo experiments were conducted to investigate the efficacy of ICG-PEI@HM-mediated PTT. Results The intelligent responsiveness of ICG-PEI@HM to pH promoted the accumulation of ICG and enhanced the PTT performance of ICG-PEI@HM NPs. Compared with free ICG, NPs exhibited great photothermal stability, cellular uptake, and active tumor targeting for PTT. Conclusion ICG-PEI@HM NPs can enhance the efficacy of PTT and can be used as a new strategy for the construction of photothermal agents.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Pathology, Jining No.1 People’s Hospital, Jining, Shandong, 272000, People’s Republic of China
| | - Zelai He
- Department of Radiation Oncology, the First Affiliated Hospital of Bengbu Medical College & Tumor Hospital Affiliated to Bengbu Medical College, Bengbu, Anhui, 233004, People’s Republic of China
| |
Collapse
|
35
|
Xiang C, Fu Y, Hao T, Wei L, Liu Y, Fan ZC, Guo N, Yu P, Teng YO. Podophyllotoxin-loaded PEGylated E-selectin peptide conjugate targeted cancer site to enhance tumor inhibition and reduce side effect. Eur J Med Chem 2023; 260:115780. [PMID: 37666045 DOI: 10.1016/j.ejmech.2023.115780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/19/2023] [Accepted: 08/29/2023] [Indexed: 09/06/2023]
Abstract
E-selectin, which is highly expressed in vascular endothelial cells near tumor and get involved in the all tumor growth steps: occurrence, proliferation and metastasis, is considered as a promise targeted protein for antitumor drug discovery. Herein, we would like to report the design, preparation and the anticancer evaluation of the peptide-PEG-podophyllotoxin conjugate(PEG-Pep-PODO), in which the short peptide (CIELLQAR) was used as the E-selectin ligand for the targeting purpose and the PEG portion the molecule got the conjugate self-assembled to form a water soluble nanoparticle. In vitro release study showed that the conjugated and entrapped PODO could be released simultaneously in the presence of GSH (highly expressed in tumor environmental conditions) and the GSH would catalyze the break of the disufur bond which linked of the PODO and the peptide-PEG portion of the conjugate. Cell adhesion test of the PEG-Pep-PODO indicated that E-selectin ligand peptide CIELLQAR could get specifically and efficiently binding to the E-selectin expressing human umbilical vein endothelial cells (HUVEC). In vitro cytotoxicity assay further revealed that PEG-Pep-PODO significantly improved the selectivity of PEG-Pep-PODO for killing the tumor cells and normal cells compared with PODO solution formulation. More importantly, the in vivo experiment demonstrated that the conjugate would accumulate of the PODO payload in tumor through targeting endothelial cells in the tumor microenvironment, which resulted in the much improved in vivo inhibition of tumor growth, intratumoral microvessel density, and decreased systemic toxicity of this nanoparticle over the free PODO. Furthermore, this water soluble conjugate greatly improved the pharmacokinetic properties of the mother molecule.
Collapse
Affiliation(s)
- Cen Xiang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin, 300457, China; College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Ying Fu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Tiantian Hao
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin, 300457, China; Medicinal Chemistry Department, Shouyao Holdings (Beijing) Co., Ltd., Beijing, China
| | - Linlin Wei
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Yuning Liu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Zhen-Chuan Fan
- College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China.
| | - Na Guo
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Peng Yu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Yu-Ou Teng
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin, 300457, China.
| |
Collapse
|
36
|
Zeting Y, Shuli M, Yue L, Haowei F, Jing S, Yueping Z, Jie W, Teng C, Wanli D, Zhang K, Peihao Y. Tissue adhesive indocyanine green-locking granular gel-mediated photothermal therapy combined with checkpoint inhibitor for preventing postsurgical recurrence and metastasis of colorectal cancer. Bioeng Transl Med 2023; 8:e10576. [PMID: 38023716 PMCID: PMC10658503 DOI: 10.1002/btm2.10576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 06/23/2023] [Accepted: 06/30/2023] [Indexed: 12/01/2023] Open
Abstract
Developing effective therapy to inhibit postoperative recurrence and metastasis of colorectal cancer (CRC) is challenging and significant to reduce mortality and morbidity. Here, a granular hydrogel, assembled from gelatin microgels by dialdehyde starch and interpenetrated with in situ polymerized poly(sulfobetaine methacrylate-co-N-isopropylacrylamide) (P(SBMA-co-NIPAM)), is prepared to load and lock Food and Drug Administration (FDA)-approved indocyanine green (ICG) with definite photothermal function and biosafety for photothermal therapy (PTT) combining with checkpoint inhibitor. The presence of P(SBMA-co-NIPAM) endows granular hydrogel with high retention to water-soluble ICG, preventing easy diffusion and rapid scavenging of ICG. The ICG-locking granular hydrogel can be spread and adhered onto the surgery site at wet state in vivo, exerting a persistent and stable PTT effect. Combined with αPD-L1 treatment, ICG-locking granular hydrogel-mediated PTT can eradicate postsurgery residual and metastatic tumors, and prevent long-term tumor recurrence. Further mechanistic studies indicate that combination treatment effectively promotes dendritic cells maturation in lymph nodes, enhances the number and infiltration of CD8+ T and CD4+ T cells in tumor tissue, and improves memory T cell number in spleen, thus activating the antitumor immune response. Overall, ICG-locking gel-mediated PTT is expected to exhibit broad clinical applications in postoperative treatment of cancers, like CRC.
Collapse
Affiliation(s)
- Yuan Zeting
- Interventional Cancer Institute of Chinese Integrative Medicine & Putuo HospitalShanghai University of Traditional Chinese MedicineShanghaiP. R. China
- Central Laboratory, Putuo HospitalShanghai University of Traditional Chinese MedicineShanghaiP. R. China
- Department of Pharmaceutics, School of PharmacyEast China University of Science and TechnologyShanghaiChina
- Shanghai Putuo Central School of Clinical MedicineAnhui Medical UniversityHefeiP. R. China
| | - Ma Shuli
- Interventional Cancer Institute of Chinese Integrative Medicine & Putuo HospitalShanghai University of Traditional Chinese MedicineShanghaiP. R. China
- Central Laboratory, Putuo HospitalShanghai University of Traditional Chinese MedicineShanghaiP. R. China
- Department of Pharmaceutics, School of PharmacyEast China University of Science and TechnologyShanghaiChina
| | - Li Yue
- Interventional Cancer Institute of Chinese Integrative Medicine & Putuo HospitalShanghai University of Traditional Chinese MedicineShanghaiP. R. China
- Central Laboratory, Putuo HospitalShanghai University of Traditional Chinese MedicineShanghaiP. R. China
| | - Fang Haowei
- Department of Polymer Materials, School of Materials Science and EngineeringShanghai UniversityShanghaiP. R. China
| | - Shang Jing
- Interventional Cancer Institute of Chinese Integrative Medicine & Putuo HospitalShanghai University of Traditional Chinese MedicineShanghaiP. R. China
- Central Laboratory, Putuo HospitalShanghai University of Traditional Chinese MedicineShanghaiP. R. China
- Shanghai Putuo Central School of Clinical MedicineAnhui Medical UniversityHefeiP. R. China
| | - Zhan Yueping
- Interventional Cancer Institute of Chinese Integrative Medicine & Putuo HospitalShanghai University of Traditional Chinese MedicineShanghaiP. R. China
- Central Laboratory, Putuo HospitalShanghai University of Traditional Chinese MedicineShanghaiP. R. China
| | - Wang Jie
- Department of General Surgery, Putuo HospitalShanghai University of Traditional Chinese MedicineShanghaiP. R. China
| | - Chen Teng
- Department of General Surgery, Putuo HospitalShanghai University of Traditional Chinese MedicineShanghaiP. R. China
| | - Deng Wanli
- Department of Oncology, Putuo HospitalShanghai University of Traditional Chinese MedicineShanghaiP. R. China
| | - Kunxi Zhang
- Interventional Cancer Institute of Chinese Integrative Medicine & Putuo HospitalShanghai University of Traditional Chinese MedicineShanghaiP. R. China
- Department of Polymer Materials, School of Materials Science and EngineeringShanghai UniversityShanghaiP. R. China
| | - Yin Peihao
- Interventional Cancer Institute of Chinese Integrative Medicine & Putuo HospitalShanghai University of Traditional Chinese MedicineShanghaiP. R. China
- Department of Pharmaceutics, School of PharmacyEast China University of Science and TechnologyShanghaiChina
- Department of General Surgery, Putuo HospitalShanghai University of Traditional Chinese MedicineShanghaiP. R. China
| |
Collapse
|
37
|
Lu Z, Li J, Chen B, Feng J, Hu Q, Jin Y, Fu Z. Mitochondria Targeted Nanoparticles Potentiate Tumor Chemo-Phototherapy by Toxic Oxidative Stress Mediated Oxeiptosis. Macromol Biosci 2023; 23:e2300151. [PMID: 37295777 DOI: 10.1002/mabi.202300151] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/06/2023] [Indexed: 06/12/2023]
Abstract
Insufficient accumulation of drug at the tumor site and the low drug response are the main reason for the unsatisfactory effect of cancer therapy. Delivery drugs exquisitely to subcellular level can be employed to reduce side effects, and expand the therapeutic window. Herein, a triphenylphosphine (TPP) modified lipid nanoparticles is designed which are loaded with the photosensitizer indocyanine green (ICG) and chemotherapeutic paclitaxel (PTX) for mitochondria-targeted chemo-phototherapy. Owing to the movement of majority mitochondria along microtubules in cytoplasm, mitochondrial targeting may enable PTX to act more effectively. Meanwhile, the existence of chemo-drug potentiates the phototherapy to achieve synergistic anti-tumor activity. As expected, mitochondria targeting nanomedicine (M-ICG-PTX NPs) showed improved mitochondria targeted cellular distribution and enhanced cell cytotoxicity in vitro. Also, M-ICG-PTX NPs exhibited higher tumor growth inhibition ability by promoting cell apoptosis and oxeiptosis pathway, and high effective inhibition of primary tumor growth and tumor metastasis. Taken together, M-ICG-PTX NPs may be promising nanoplatforms to achieve potent therapeutic effect for the combination of chemo- and photo-therapy (PTT).
Collapse
Affiliation(s)
- Zhuoting Lu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Jingyu Li
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Baohong Chen
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Jiayu Feng
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Qinglian Hu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| |
Collapse
|
38
|
Millard M, Bernhard Y, Canilho N, Grandemange S, Parant S, Mourer M, Lassalle HP, Pasc A. Enhanced stability and photothermal efficiency of Indocyanine Green J-aggregates by nanoformulation with Calix[4]arene for photothermal therapy of cancers. Colloids Surf B Biointerfaces 2023; 230:113516. [PMID: 37660516 DOI: 10.1016/j.colsurfb.2023.113516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 07/17/2023] [Accepted: 08/13/2023] [Indexed: 09/05/2023]
Abstract
Photothermal therapy (PTT) is a method of growing attention, owing to its controllable process, high efficiency and minimal side effect. Indocyanine Green (ICG) is as Food and Drug Administration (FDA) approved agent that stands on the frontline of further developments of PTT toward clinics. However, the applicability of ICG-mediated PTT is limited by the rapid in vivo clearance and photo-degradation of ICG. To improve those parameters, nanosized ICG-loaded nanoparticles (ICG-J/CX) were fabricated in this study by co-assembly of anionic ICG J-aggregates (ICG-J) with cationic tetraguanidinium calix[4]arene (CX). This very simple approach produces ICG-J/CX with a well-defined nanometer range size and a close to neutral charge. The nanoparticles demonstrate high photothermal conversion efficiency (PCE) and dramatically improved photostability, as compared with ICG. The in vitro cellular uptake and cytotoxicity studies further demonstrated that the ICG-J/CX nanoparticles enhance uptake and photothermal efficiency in comparison with ICG or non-formulated ICG-J, overall demonstrating that ICG-J/CX mediated photothermal therapy have significant potential for attaining cancer treatment.
Collapse
Affiliation(s)
- Marie Millard
- Université de Lorraine, CNRS, L2CM UMR 7053, F-54506 Vandoeuvre-lès-Nancy, France; Université de Lorraine, CNRS UMR 7039, CRAN, F-54000 Nancy, France
| | - Yann Bernhard
- Université de Lorraine, CNRS, L2CM UMR 7053, F-54506 Vandoeuvre-lès-Nancy, France
| | - Nadia Canilho
- Université de Lorraine, CNRS, L2CM UMR 7053, F-54506 Vandoeuvre-lès-Nancy, France
| | | | - Stéphane Parant
- Université de Lorraine, CNRS, L2CM UMR 7053, F-54506 Vandoeuvre-lès-Nancy, France
| | - Maxime Mourer
- Université de Lorraine, CNRS, L2CM UMR 7053, F-54506 Vandoeuvre-lès-Nancy, France
| | - Henri-Pierre Lassalle
- Université de Lorraine, CNRS UMR 7039, CRAN, F-54000 Nancy, France; Institut de Cancérologie de Lorraine, Unité de Recherche Translationnelle F-54000 Nancy, France.
| | - Andreea Pasc
- Université de Lorraine, CNRS, L2CM UMR 7053, F-54506 Vandoeuvre-lès-Nancy, France.
| |
Collapse
|
39
|
Shi NQ, Cui XY, Zhou C, Tang N, Cui DX. Application of near-infrared fluorescence imaging in theranostics of gastrointestinal tumors. Gastroenterol Rep (Oxf) 2023; 11:goad055. [PMID: 37781571 PMCID: PMC10533422 DOI: 10.1093/gastro/goad055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/06/2023] [Accepted: 08/28/2023] [Indexed: 10/03/2023] Open
Abstract
Gastrointestinal cancers have become an important cause of cancer-related death in humans. Improving the early diagnosis rate of gastrointestinal tumors and improving the effect of surgical treatment can significantly improve the survival rate of patients. The conventional diagnostic method is high-definition white-light endoscopy, which often leads to missed diagnosis. For surgical treatment, intraoperative tumor localization and post-operative anastomotic state evaluation play important roles in the effect of surgical treatment. As a new imaging method, near-infrared fluorescence imaging (NIRFI) has its unique advantages in the diagnosis and auxiliary surgical treatment of gastrointestinal tumors due to its high sensitivity and the ability to image deep tissues. In this review, we focus on the latest advances of NIRFI technology applied in early diagnosis of gastrointestinal tumors, identification of tumor margins, identification of lymph nodes, and assessment of anastomotic leakage. In addition, we summarize the advances of NIRFI systems such as macro imaging and micro imaging systems, and also clearly describe the application process of NIRFI from system to clinical application, and look into the prospect of NIRFI applied in the theranostics of gastrointestinal tumors.
Collapse
Affiliation(s)
- Nan-Qing Shi
- Department of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Institute of Nano Biomedicine and Engineering, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Xin-Yuan Cui
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Cheng Zhou
- Department of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Institute of Nano Biomedicine and Engineering, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Ning Tang
- Department of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Institute of Nano Biomedicine and Engineering, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Da-Xiang Cui
- Department of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Institute of Nano Biomedicine and Engineering, Shanghai Jiao Tong University, Shanghai, P. R. China
- National Engineering Center for Nanotechnology, Shanghai, P. R. China
| |
Collapse
|
40
|
Wang J, Liao H, Ban J, Li S, Xiong X, He Q, Shi X, Shen H, Yang S, Sun C, Liu L. Multifunctional Near-Infrared Dye IR-817 Encapsulated in Albumin Nanoparticles for Enhanced Imaging and Photothermal Therapy in Melanoma. Int J Nanomedicine 2023; 18:4949-4967. [PMID: 37693889 PMCID: PMC10488832 DOI: 10.2147/ijn.s425013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/21/2023] [Indexed: 09/12/2023] Open
Abstract
Background Near-infrared cyanine dyes have high sensitivity and spatial resolution imaging capabilities, but they also have unavoidable drawbacks such as photobleaching, low water solubility, fluorescence quenching, and toxic side effects. As an effective biologic drug carrier, albumin combines with cyanine dyes to form albumin@dye nanoparticles. These nanoparticles can alleviate the aforementioned issues and are widely used in tumor imaging and photothermal therapy. Methods Herein, a newly synthesized near-infrared dye IR-817 was combined with bovine serum albumin (BSA) to create BSA@IR-817 nanoparticles. Through the detection of fluorescence emission and absorption, the optimal concentration and ratio of BSA and IR-817 were determined. Subsequently, dynamic light scattering (DLS) measurements and scanning electron microscopy (SEM) were used for the physical characterization of the BSA@IR-817 nanoparticles. Finally, in vitro and in vivo experiments were conducted to assess the fluorescence imaging and photothermal therapeutic potential of BSA@IR-817 nanoparticles. Results IR-817 was adsorbed onto the BSA carrier by covalent conjugation and supramolecular encapsulation, resulting in the formation of dispersed, homogeneous, and stable nanoparticles with a particle size range of 120-220 nm. BSA@IR-817 not only improved the poor water solubility, fluorescence quenching, and toxic side effects of IR-817 but also enhanced the absorption and fluorescence emission peaks in the near-infrared region, as well as the fluorescence in the visible spectrum. In addition, BSA@IR-817 combined with laser 808 irradiation was able to convert light energy into heat energy with temperatures exceeding 50 °C. By creating a mouse model of subcutaneous melanoma, it was discovered that the tumor inhibition rate of BSA@IR-817 was greater than 99% after laser irradiation and that it achieved nearly complete tumor ablation without causing significant toxicity. Conclusion Our research, therefore, proposes the use of safe and effective photothermal nanoparticles for the imaging, diagnosis, and treatment of melanoma, and offers a promising strategy for future biomedical applications.
Collapse
Affiliation(s)
- Jianv Wang
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Hongye Liao
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Jieming Ban
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, National Traditional Chinese Medicine Clinical Research Base, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Sen Li
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, National Traditional Chinese Medicine Clinical Research Base, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Xia Xiong
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Qingqing He
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Xinyu Shi
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, National Traditional Chinese Medicine Clinical Research Base, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Hongping Shen
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, National Traditional Chinese Medicine Clinical Research Base, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Sijin Yang
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, National Traditional Chinese Medicine Clinical Research Base, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Changzhen Sun
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, National Traditional Chinese Medicine Clinical Research Base, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Li Liu
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| |
Collapse
|
41
|
Yue NN, Xu HM, Xu J, Zhu MZ, Zhang Y, Tian CM, Nie YQ, Yao J, Liang YJ, Li DF, Wang LS. Application of Nanoparticles in the Diagnosis of Gastrointestinal Diseases: A Complete Future Perspective. Int J Nanomedicine 2023; 18:4143-4170. [PMID: 37525691 PMCID: PMC10387254 DOI: 10.2147/ijn.s413141] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/02/2023] [Indexed: 08/02/2023] Open
Abstract
The diagnosis of gastrointestinal (GI) diseases currently relies primarily on invasive procedures like digestive endoscopy. However, these procedures can cause discomfort, respiratory issues, and bacterial infections in patients, both during and after the examination. In recent years, nanomedicine has emerged as a promising field, providing significant advancements in diagnostic techniques. Nanoprobes, in particular, offer distinct advantages, such as high specificity and sensitivity in detecting GI diseases. Integration of nanoprobes with advanced imaging techniques, such as nuclear magnetic resonance, optical fluorescence imaging, tomography, and optical correlation tomography, has significantly enhanced the detection capabilities for GI tumors and inflammatory bowel disease (IBD). This synergy enables early diagnosis and precise staging of GI disorders. Among the nanoparticles investigated for clinical applications, superparamagnetic iron oxide, quantum dots, single carbon nanotubes, and nanocages have emerged as extensively studied and utilized agents. This review aimed to provide insights into the potential applications of nanoparticles in modern imaging techniques, with a specific focus on their role in facilitating early and specific diagnosis of a range of GI disorders, including IBD and colorectal cancer (CRC). Additionally, we discussed the challenges associated with the implementation of nanotechnology-based GI diagnostics and explored future prospects for translation in this promising field.
Collapse
Affiliation(s)
- Ning-ning Yue
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Hao-ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, People’s Republic of China
| | - Jing Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, People’s Republic of China
| | - Min-zheng Zhu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, Guangdong, People’s Republic of China
| | - Cheng-Mei Tian
- Department of Emergency, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Yu-qiang Nie
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, People’s Republic of China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Yu-jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - De-feng Li
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Li-sheng Wang
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
42
|
Yuan Y, Li X, Bao X, Huangfu M, Zhang H. The magic mirror: a novel intraoperative monitoring method for parathyroid glands. Front Endocrinol (Lausanne) 2023; 14:1160902. [PMID: 37284221 PMCID: PMC10239973 DOI: 10.3389/fendo.2023.1160902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/01/2023] [Indexed: 06/08/2023] Open
Abstract
The accurate detection of parathyroid glands (PGs) during surgery is of great significance in thyroidectomy and parathyroidectomy, which protects the function of normal PGs to prevent postoperative hypoparathyroidism and the thorough removal of parathyroid lesions. Existing conventional imaging techniques have certain limitations in the real-time exploration of PGs. In recent years, a new, real-time, and non-invasive imaging system known as the near-infrared autofluorescence (NIRAF) imaging system has been developed to detect PGs. Several studies have confirmed that this system has a high parathyroid recognition rate and can reduce the occurrence of transient hypoparathyroidism after surgery. The NIRAF imaging system, like a magic mirror, can monitor the PGs during surgery in real time, thus providing great support for surgeries. In addition, the NIRAF imaging system can evaluate the blood supply of PGs by utilizing indocyanine green (ICG) to guide surgical strategies. The NIRAF imaging system and ICG complement each other to protect normal parathyroid function and reduce postoperative complications. This article reviews the effectiveness of the NIRAF imaging system in thyroidectomies and parathyroidectomies and briefly discusses some existing problems and prospects for the future.
Collapse
|
43
|
Li C, Huang J, Yuan L, Xie W, Ying Y, Li C, Yu Y, Pan Y, Qu W, Hao H, Algharib SA, Chen D, Xie S. Recent progress of emitting long-wavelength carbon dots and their merits for visualization tracking, target delivery and theranostics. Theranostics 2023; 13:3064-3102. [PMID: 37284447 PMCID: PMC10240821 DOI: 10.7150/thno.80579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/07/2023] [Indexed: 06/08/2023] Open
Abstract
As a novel strategy for in vivo visualization tracking and monitoring, carbon dots (CDs) emitting long wavelengths (LW, 600-950 nm) have received tremendous attention due to their deep tissue penetration, low photon scattering, satisfactory contrast resolution and high signal-to-background ratios. Although, the mechanism of CDs emitting LW remains controversial and what properties are best for in vivo visualization have not been specifically elucidated, it is more conducive to the in vivo application of LW-CDs through rational design and ingenious synthesis based on the appreciation of the luminescence mechanism. Therefore, this review analyzes the current tracer technologies applied in vivo and their advantages and disadvantages, with emphasis on the physical mechanism of emitting LW fluorescence for in vivo imaging. Subsequently, the general properties and merits of LW-CDs for tracking and imaging are summarized. More importantly, the factors affecting the synthesis of LW-CDs and its luminescence mechanism are highlighted. Simultaneously, the application of LW-CDs for disease diagnosis, integration of diagnosis and therapy are summarized. Finally, the bottlenecks and possible future directions of LW-CDs in visualization tracking and imaging in vivo are detailly discussed.
Collapse
Affiliation(s)
- Chao Li
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jiamin Huang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Liwen Yuan
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Wenqing Xie
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yupeng Ying
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Chengzhe Li
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yahang Yu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yuanhu Pan
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Wei Qu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Haihong Hao
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Samah Attia Algharib
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh 13736, QG, Egypt
| | - Dongmei Chen
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Shuyu Xie
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
- Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| |
Collapse
|
44
|
Ma Z, Deng J, Ma B, Chen H. Real-time Indocyanine Green Fluorescence Technique Reduces Anastomotic Leakage in Bilioenteric Anastomosis: A Case Report and literature review. Photodiagnosis Photodyn Ther 2023:103609. [PMID: 37187271 DOI: 10.1016/j.pdpdt.2023.103609] [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: 03/03/2023] [Revised: 04/25/2023] [Accepted: 05/09/2023] [Indexed: 05/17/2023]
Abstract
BACKGROUND Anastomotic leakage is a serious complication that can occur in bilioenteric anastomosis surgery, leading to significant morbidity and mortality. Currently, practitioners rely on subjective measures to determine anastomotic perfusion and mechanical integrity, which have limitations. The use of indocyanine green fluorescence technology has become increasingly widespread in clinical practice, especially in gastrointestinal-related surgery. This technique has a unique role in evaluating the blood perfusion of anastomoses and reducing the incidence of anastomotic leakage. However, there have been no reports of its use in bilioenteric anastomosis surgery. Further research is needed to investigate the potential benefits of indocyanine green fluorescence technology in improving outcomes and reducing complications in this type of surgery. CASE SUMMARY a 50-year-old female patient underwent total laparoscopic radical resection of cholangiocarcinoma. During the surgery, indocyanine green fluorescence technology was used to complete the biliary intestinal anastomosis under full visual and dynamic monitoring. The patient recovered well after the operation without experiencing biliary leakage or other complications. CONCLUSION The present case study underscores the potential advantages associated with the incorporation of intraoperative real-time indocyanine green (ICG) technology in bilioenteric anastomosis surgery. By facilitating enhanced visualization and assessment of anastomotic perfusion and mechanical stability, this state-of-the-art technique may help mitigate the occurrence of anastomotic leaks while simultaneously improving patient outcomes. Notably, intravenous administration of ICG at a dose of 2.5 mg/kg, administered 24 hours prior to surgery, has been found to yield optimal visualization outcomes.
Collapse
Affiliation(s)
- Zhen Ma
- Lanzhou University Second Hospital, Lanzhou 730030, China; Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou 730030, China; Key Laboratory of the Digestive System tumours of Gansu Province
| | - Junge Deng
- Lanzhou University Second Hospital, Lanzhou 730030, China; Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Bin Ma
- Lanzhou University Second Hospital, Lanzhou 730030, China; Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou 730030, China; Key Laboratory of the Digestive System tumours of Gansu Province
| | - Hao Chen
- Lanzhou University Second Hospital, Lanzhou 730030, China; Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou 730030, China; Key Laboratory of the Digestive System tumours of Gansu Province.
| |
Collapse
|
45
|
Srivastava I, Lew B, Wang Y, Blair S, George MB, Hajek BS, Bangru S, Pandit S, Wang Z, Ludwig J, Flatt K, Gruebele M, Nie S, Gruev V. Cell-Membrane Coated Nanoparticles for Tumor Delineation and Qualitative Estimation of Cancer Biomarkers at Single Wavelength Excitation in Murine and Phantom Models. ACS NANO 2023; 17:8465-8482. [PMID: 37126072 DOI: 10.1021/acsnano.3c00578] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Real-time guidance through fluorescence imaging improves the surgical outcomes of tumor resections, reducing the chances of leaving positive margins behind. As tumors are heterogeneous, it is imperative to interrogate multiple overexpressed cancer biomarkers with high sensitivity and specificity to improve surgical outcomes. However, for accurate tumor delineation and ratiometric detection of tumor biomarkers, current methods require multiple excitation wavelengths to image multiple biomarkers, which is impractical in a clinical setting. Here, we have developed a biomimetic platform comprising near-infrared fluorescent semiconducting polymer nanoparticles (SPNs) with red blood cell membrane (RBC) coating, capable of targeting two representative cell-surface biomarkers (folate, αυβ3 integrins) using a single excitation wavelength for tumor delineation during surgical interventions. We evaluate our single excitation ratiometric nanoparticles in in vitro tumor cells, ex vivo tumor-mimicking phantoms, and in vivo mouse xenograft tumor models. Favorable biological properties (improved biocompatibility, prolonged blood circulation, reduced liver uptake) are complemented by superior spectral features: (i) specific fluorescence enhancement in tumor regions with high tumor-to-normal tissue (T/NT) ratios in ex vivo samples and (ii) estimation of cell-surface tumor biomarkers with single wavelength excitation providing insights about cancer progression (metastases). Our single excitation, dual output approach has the potential to differentiate between the tumor and healthy regions and simultaneously provide a qualitative indicator of cancer progression, thereby guiding surgeons in the operating room with the resection process.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sushant Bangru
- Department of Cell Biology, Duke University, Durham, North Carolina 27705, United States
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Wang J, Xu Y, Zhang Y, Tian H. Safety and effectiveness of fluorescence laparoscopy in precise hepatectomy: A meta-analysis. Photodiagnosis Photodyn Ther 2023; 42:103599. [PMID: 37156455 DOI: 10.1016/j.pdpdt.2023.103599] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/17/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND To perform a systematic review of the safety and effectiveness of fluorescence laparoscopy-guided precise hepatectomy. METHODS We searched the PubMed, Embase, Web of Science, and Cochrane Library databases from inception to December 1, 2022, using the search terms "indocyanine green," "ICG," "infracyanine green," "laparoscopy," "liver resection," and "hepatectomy." After performing a methodological quality assessment of the included studies, the overall results were subjected to meta-analysis using Review Manager 5.3. RESULTS After screening, the meta-analysis included a total of 13 articles. The studies included 1,115 patients who were grouped into the fluorescence laparoscopy (490 patients) and conventional laparoscopy (625 patients) groups. All articles included in the meta-analysis were of high quality. The results of the meta-analysis revealed that compared to the conventional laparoscopy group, the fluorescence laparoscopy group had a higher R0 resection rate (odds ratio=4.03, 95% confidence interval [1.50, 10.83], P=0.006), lower blood transfusion rate (odds ratio=0.46, 95% confidence interval [0.21, 0.97], P=0.04) and lower blood loss (mean difference=-36.58; 95% confidence interval [-59.75, -13.41], P=0.002). However, the length of hospital stay, operative time, and incidence of postoperative complications did not differ significantly between both groups (P>0.05). CONCLUSION Compared to conventional laparoscopy, fluorescence laparoscopy provides better application effects in hepatectomy. The surgical procedure has demonstrated good safety and feasibility, which make it worthy of popularization.
Collapse
Affiliation(s)
- Junqiang Wang
- Shandong University of Traditional Chinese Medicine, Shandong Jinan 250355,China
| | - Ying Xu
- Shandong First Medical University, Shandong Taian 271016, China
| | - Yuhua Zhang
- Shandong University of Traditional Chinese Medicine, Shandong Jinan 250355,China
| | - Hu Tian
- Shandong Provincial Qianfoshan Hospital, Shandong Jinan 250014, China.
| |
Collapse
|
47
|
Su H, Xu Z, Bao M, Luo S, Liang J, Pei W, Guan X, Liu Z, Jiang Z, Zhang M, Zhao Z, Jin W, Zhou H. Lateral pelvic sentinel lymph node biopsy using indocyanine green fluorescence navigation: can it be a powerful supplement tool for predicting the status of lateral pelvic lymph nodes in advanced lower rectal cancer. Surg Endosc 2023; 37:4088-4096. [PMID: 36997652 DOI: 10.1007/s00464-023-10033-w] [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: 06/20/2022] [Accepted: 03/12/2023] [Indexed: 05/05/2023]
Abstract
BACKGROUND An innovative instrument for laparoscopy using indocyanine green (ICG) allows easy detection of sentinel lymph nodes (SLNs) in lateral pelvic lymph nodes (LPLNs). Here, we investigated the safety and efficacy of lateral pelvic SLN biopsy (SLNB) using ICG fluorescence navigation in advanced lower rectal cancer and evaluated the sensitivity and specificity of this technique to predict the status of LPLN. METHODS From April 1, 2017 to December 1, 2020, we conducted lateral pelvic SLNB using ICG fluorescence navigation during laparoscopic total mesorectal excision and lateral pelvic lymph node dissection (LLND) in 23 patients with advanced low rectal cancer who presented with LPLN but without LPLN enlargement. Data regarding clinical characteristics, surgical and pathological outcomes, lymph node findings, and postoperative complications were collected and analyzed. RESULTS We successfully performed the surgery using fluorescence navigation. One patient underwent bilateral LLND and 22 patients underwent unilateral LLND. The lateral pelvic SLN were clearly fluorescent before dissection in 21 patients. Lateral pelvic SLN metastasis was diagnosed in 3 patients and negative in 18 patients by frozen pathological examination. Among the 21 patients in whom lateral pelvic SLN was detected, the dissected lateral pelvic non-SLNs were all negative. All dissected LPLNs were negative in two patients without fluorescent lateral pelvic SLN. CONCLUSION This study indicated that lateral pelvic SLNB using ICG fluorescence navigation shows promise as a safe and feasible procedure for advanced lower rectal cancer with good accuracy, and no false-negative cases were found. No metastasis in SLNB seemed to reflect all negative LPLN metastases, and this technique can replace preventive LLND for advanced lower rectal cancer.
Collapse
Affiliation(s)
- Hao Su
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Zheng Xu
- Department of Colorectal Surgery, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100021, China
| | - Mandula Bao
- Department of Pancreatic and Gastric Surgery, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100021, China
| | - Shou Luo
- Department of Colorectal Surgery, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100021, China
| | - Jianwei Liang
- Department of Colorectal Surgery, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100021, China
| | - Wei Pei
- Department of Colorectal Surgery, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100021, China
| | - Xu Guan
- Department of Colorectal Surgery, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100021, China
| | - Zheng Liu
- Department of Colorectal Surgery, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100021, China
| | - Zheng Jiang
- Department of Colorectal Surgery, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100021, China
| | - Mingguang Zhang
- Department of Colorectal Surgery, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100021, China
| | - Zhixun Zhao
- Department of Colorectal Surgery, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100021, China
| | - Weisen Jin
- Department of Anorectal Diseases, Third Medical Center of Chinese PLA General Hospital, Haidian District, Beijing, 100039, People's Republic of China.
| | - Haitao Zhou
- Department of Colorectal Surgery, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
48
|
Lu WL, Kuang H, Gu J, Hu X, Chen B, Fan Y. GAP-43 targeted indocyanine green-loaded near-infrared fluorescent probe for real-time mapping of perineural invasion lesions in pancreatic cancer in vivo. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 50:102671. [PMID: 37054805 DOI: 10.1016/j.nano.2023.102671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/22/2023] [Accepted: 03/26/2023] [Indexed: 04/15/2023]
Abstract
OBJECTIVE Perineural invasion (PNI) is associated with local recurrence, distant metastasis, and a poor prognosis in pancreatic cancer. However, rare attempt was made to identified the PNI intraoperative. To facilitate precise R0 excision of the tumor, we planned to develop a fluorescent probe for intraoperative imaging of the PNI using GAP-43 as the target and indocyanine green (ICG) as the carrier. METHODS The probe was created by binding peptide antibody and ICG. Its targeting was tested in vitro and in vivo using a co-culture model of PC12 and tumor cells to create an in vitro neural invasion model and a mouse sciatic nerve invasion model. The small animal imaging system and surgical navigation system confirmed the probe's potential clinical applicability. The sciatic nerve damage model was created to confirm the probe's targeting. RESULTS We used the pancreatic cancer samples and the public database to confirm that GAP-43 was preferentially overexpressed in pancreatic cancer, particularly in PNI. PC12 cells showed high GAP-43RA-PEG-ICG probe-specific absorption after being co-cultured with tumor cells in vitro. In the sciatic nerve invasion experiment, animals in probe group displayed a significantly stronger fluorescence signal at the PNI compared to ICG-NP and the contralateral normal nerves groups. Although only 60 % of mice appeared to have R0 resections by the naked eye, small animal imaging systems and surgical fluorescence navigation systems could remove the tumor with R0 precision. The injury model used in the probe imaging experimental trials demonstrated that the probe was specifically targeted to the injured nerve, regardless of whether the injury was infiltrated by a tumor or physical. CONCLUSION We developed the GAP-43Ra-ICG-PEG, an active-targeting near-infrared fluorescent (NIF) probe, that specifically binds to GAP-43-positive neural cells in an in vitro model of PNI. The probe efficiently visualized PNI lesions in pancreatic cancer in preclinical models, opening up new possibilities for NIRF-guided pancreatic surgery, particularly for PNI patients.
Collapse
Affiliation(s)
- Wen Liang Lu
- The Department of General Surgery & Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Department of Thyroid and breast surgery, Maternal and Child Health Hospital of Hubei Province, Wuhan 430070, China
| | - Houfang Kuang
- Department of General Surgery, Wuhan Children(,) hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430016, China
| | - Jianyou Gu
- The Department of General Surgery & Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiaojun Hu
- The Department of General Surgery & Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Department of Hepatobiliary Surgery, The Fifth Affifiliated Hospital of Southern Medical University, Guangzhou 510920, China
| | - Bo Chen
- Department of Thyroid and breast surgery, Maternal and Child Health Hospital of Hubei Province, Wuhan 430070, China
| | - Yingfang Fan
- The Department of General Surgery & Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| |
Collapse
|
49
|
Chauhan N, Cabrera M, Chowdhury P, Nagesh PK, Dhasmana A, Pranav, Jaggi M, Chauhan SC, Yallapu MM. Indocyanine Green-based Glow Nanoparticles Probe for Cancer Imaging. Nanotheranostics 2023; 7:353-367. [PMID: 37151801 PMCID: PMC10161388 DOI: 10.7150/ntno.78405] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 03/22/2023] [Indexed: 08/31/2023] Open
Abstract
Indocyanine green (ICG) is one of the FDA-approved near infra-red fluorescent (NIRF) probes for cancer imaging and image-guided surgery in the clinical setting. However, the limitations of ICG include poor photostability, high concentration toxicity, short circulation time, and poor cancer cell specificity. To overcome these hurdles, we engineered a nanoconstruct composed of poly (vinyl pyrrolidone) (PVP)-indocyanine green that is cloaked self-assembled with tannic acid (termed as indocyanine green-based glow nanoparticles probe, ICG-Glow NPs) for the cancer cell/tissue-specific targeting. The self-assembled ICG-Glow NPs were confirmed by spherical nanoparticles formation (DLS and TEM) and spectral analyses. The NIRF imaging characteristic of ICG-Glow NPs was established by superior fluorescence counts on filter paper and chicken tissue. The ICG-Glow NPs exhibited excellent hemo and cellular compatibility with human red blood cells, kidney normal, pancreatic normal, and other cancer cell lines. An enhanced cancer-specific NIRF binding and imaging capability of ICG-Glow NPs was confirmed using different human cancer cell lines and human tumor tissues. Additionally, tumor-specific binding/accumulation of ICG-Glow NPs was confirmed in MDA-MB-231 xenograft mouse model. Collectively, these findings suggest that ICG-Glow NPs have great potential as a novel and safe NIRF imaging probe for cancer cell/tumor imaging. This can lead to a quicker cancer diagnosis facilitating precise disease detection and management.
Collapse
Affiliation(s)
- Neeraj Chauhan
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| | - Marco Cabrera
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| | - Pallabita Chowdhury
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Prashanth K.B. Nagesh
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Laboratory of Signal Transduction, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| | - Pranav
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| | - Subhash C. Chauhan
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| | - Murali M. Yallapu
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| |
Collapse
|
50
|
Wu Y, Chen Z, Shen D, He Z, Lv J, Li H, Yang M, Tan J, Yuan J, Gao J, Yuan Z. A Lysosome-Targeted Near-Infrared Fluorescent Probe with Excellent Water Solubility for Surgery Navigation in Breast Cancer. ACS OMEGA 2023; 8:12481-12488. [PMID: 37033849 PMCID: PMC10077528 DOI: 10.1021/acsomega.3c00601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/13/2023] [Indexed: 06/19/2023]
Abstract
To get a tumor-targeted contrast agent for imaging guide resection of tumors, we designed a novel fluorescent probe based on the heptamethine cyanine core, Cy7-MO, which has excellent water solubility and near-infrared photophysical and lysosomal targeting properties. The chemical structure of Cy7-MO was characterized by nuclear magnetic resonance spectroscopy and high-resolution mass spectrometry. The toxicity of Cy7-MO was evaluated by cell counting kit-8. Then, a cellular-level study was conducted to evaluate the suborganelle localization in 4T1-Luc1 cells, and it was also used for surgical navigation in orthotopic breast tumor resection in vivo. The results showed that Cy7-MO was well targeted to lysosomes. Importantly, the Cy7-MO probe was found to be well tolerable and exhibited excellent biocompatibility. Moreover, the orthotopic breast tumor margin was clearly visualized through fluorescence guiding of Cy7-MO. Finally, the correct tumor tissues were completely removed, and a negative margin was obtained successfully, which demonstrated an enhanced precision of surgery.
Collapse
Affiliation(s)
- Yumei Wu
- Key
Laboratory of Basic Pharmacology of Ministry of Education and Joint
International Research Laboratory of Ethnomedicine of Ministry of
Education, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
- Key
Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou
Province, School of Pharmacy, Zunyi Medical
University, Zunyi, Guizhou Province 563000, China
- Guizhou
International Scientific and Technological Cooperation Base for Medical
Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
| | - Zhengjun Chen
- Key
Laboratory of Basic Pharmacology of Ministry of Education and Joint
International Research Laboratory of Ethnomedicine of Ministry of
Education, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
- Key
Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou
Province, School of Pharmacy, Zunyi Medical
University, Zunyi, Guizhou Province 563000, China
- Guizhou
International Scientific and Technological Cooperation Base for Medical
Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
| | - Dan Shen
- Key
Laboratory of Basic Pharmacology of Ministry of Education and Joint
International Research Laboratory of Ethnomedicine of Ministry of
Education, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
- Key
Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou
Province, School of Pharmacy, Zunyi Medical
University, Zunyi, Guizhou Province 563000, China
- Guizhou
International Scientific and Technological Cooperation Base for Medical
Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
| | - Zhiquan He
- Morphological
Laboratory, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
| | - Jiajia Lv
- Key
Laboratory of Basic Pharmacology of Ministry of Education and Joint
International Research Laboratory of Ethnomedicine of Ministry of
Education, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
- Key
Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou
Province, School of Pharmacy, Zunyi Medical
University, Zunyi, Guizhou Province 563000, China
- Guizhou
International Scientific and Technological Cooperation Base for Medical
Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
| | - Hongyu Li
- Key
Laboratory of Basic Pharmacology of Ministry of Education and Joint
International Research Laboratory of Ethnomedicine of Ministry of
Education, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
- Key
Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou
Province, School of Pharmacy, Zunyi Medical
University, Zunyi, Guizhou Province 563000, China
- Guizhou
International Scientific and Technological Cooperation Base for Medical
Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
| | - Mingyan Yang
- Key
Laboratory of Basic Pharmacology of Ministry of Education and Joint
International Research Laboratory of Ethnomedicine of Ministry of
Education, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
- Key
Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou
Province, School of Pharmacy, Zunyi Medical
University, Zunyi, Guizhou Province 563000, China
- Guizhou
International Scientific and Technological Cooperation Base for Medical
Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
| | - Jun Tan
- Department
of Histology and Embryology, Zunyi Medical
University, Zunyi, Guizhou Province 563000, China
| | - Jianrong Yuan
- Key
Laboratory of Basic Pharmacology of Ministry of Education and Joint
International Research Laboratory of Ethnomedicine of Ministry of
Education, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
- Key
Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou
Province, School of Pharmacy, Zunyi Medical
University, Zunyi, Guizhou Province 563000, China
- Guizhou
International Scientific and Technological Cooperation Base for Medical
Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
| | - Jie Gao
- Key
Laboratory of Basic Pharmacology of Ministry of Education and Joint
International Research Laboratory of Ethnomedicine of Ministry of
Education, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
- Key
Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou
Province, School of Pharmacy, Zunyi Medical
University, Zunyi, Guizhou Province 563000, China
- Guizhou
International Scientific and Technological Cooperation Base for Medical
Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
| | - Zeli Yuan
- Key
Laboratory of Basic Pharmacology of Ministry of Education and Joint
International Research Laboratory of Ethnomedicine of Ministry of
Education, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
- Key
Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou
Province, School of Pharmacy, Zunyi Medical
University, Zunyi, Guizhou Province 563000, China
- Guizhou
International Scientific and Technological Cooperation Base for Medical
Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, Zunyi, Guizhou Province 563000, China
| |
Collapse
|