1
|
Saadan N, Ahmed WU, Kadi AA, Al-Mutairi MS, Al-Wabli RI, Rahman AFMM. Synthesis and Evaluation of Thiazolyl-indole-2-carboxamide Derivatives as Potent Multitarget Anticancer Agents. ACS OMEGA 2024; 9:41944-41967. [PMID: 39398118 PMCID: PMC11465279 DOI: 10.1021/acsomega.4c06889] [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: 07/26/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 10/15/2024]
Abstract
Cancer is a complex disease driven by the dysregulation of multiple signaling pathways and cellular processes. The development of compounds capable of exerting multitarget actions against these key pathways involved in cancer progression is a promising therapeutic approach. Here, a series of novel (E/Z)-N-(4-(2-(2-(substituted)hydrazineyl)-2-oxoethyl)thiazol-2-yl)-1H-indole-2-carboxamide derivatives (6a-6z) were designed, synthesized, and evaluated for their biological activity. Compounds 6e, 6i, 6q, 6v, 7a, and 7b exhibited exceptional cytotoxicity against various cancer cell lines, particularly 6i (IC50 = 6.10 ± 0.4 μM against MCF-7 cell lines) and 6v (IC50 = 6.49 ± 0.3 μM against MCF-7 cell lines). These potent compounds inhibited key protein kinases like EGFR, HER2, VEGFR-2, and CDK2, induced cell cycle arrest at the G2/M phase, and promoted apoptosis. Docking studies revealed improved binding affinity of 6i and 6v with target proteins compared to reference drugs. These findings highlight the promising potential of 6i and 6v as multitarget cancer therapeutics deserving further development.
Collapse
Affiliation(s)
- Njood
M. Saadan
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Wahid U. Ahmed
- School
of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Adnan A. Kadi
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Maha S. Al-Mutairi
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Reem I. Al-Wabli
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - A. F. M. Motiur Rahman
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
2
|
Um‐e‐Kalsoom, Wang S, Qu J, Liu L. Innovative optical imaging strategies for monitoring immunotherapy in the tumor microenvironments. Cancer Med 2024; 13:e70155. [PMID: 39387259 PMCID: PMC11465031 DOI: 10.1002/cam4.70155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 08/01/2024] [Accepted: 08/16/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND The tumor microenvironment (TME) plays a critical role in cancer progression and response to immunotherapy. Immunotherapy targeting the immune system has emerged as a promising treatment modality, but challenges in understanding the TME limit its efficacy. Optical imaging strategies offer noninvasive, real-time insights into the interactions between immune cells and the TME. OBJECTIVE This review assesses the progress of optical imaging technologies in monitoring immunotherapy within the TME and explores their potential applications in clinical trials and personalized cancer treatment. METHODS This is a comprehensive literature review based on the advances in optical imaging modalities including fluorescence imaging (FLI), bioluminescence imaging (BLI), and photoacoustic imaging (PAI). These modalities were analyzed for their capacity to provide high-resolution, real-time imaging of immune cell dynamics, tumor vasculature, and other critical components of the TME. RESULTS Optical imaging techniques have shown significant potential in tracking immune cell infiltration, assessing immune checkpoint inhibitors, and visualizing drug delivery within the TME. Technologies like FLI and BLI are pivotal in tracking immune responses in preclinical models, while PAI provides functional imaging with deeper tissue penetration. The integration of these modalities with immunotherapy holds promise for improving treatment monitoring and outcomes. CONCLUSION Optical imaging is a powerful tool for understanding the complexities of the TME and optimizing immunotherapy. Further advancements in imaging technologies, combined with nanomaterial-based approaches, could pave the way for enhanced diagnostic accuracy and therapeutic efficacy in cancer treatment.
Collapse
Affiliation(s)
- Um‐e‐Kalsoom
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Shiqi Wang
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Liwei Liu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| |
Collapse
|
3
|
Shao W, Lin X, Huang Y, Qu L, Zhuo W, Liu H. Rapid patient-specific organ dose estimation in computed tomography scans via integration of radiomics features and neural networks. Quant Imaging Med Surg 2024; 14:7379-7391. [PMID: 39429608 PMCID: PMC11485356 DOI: 10.21037/qims-24-645] [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: 03/28/2024] [Accepted: 08/22/2024] [Indexed: 10/22/2024]
Abstract
Background Computed tomography (CT) offers detailed cross-sectional images of internal anatomy for disease detection but carries a risk of solid cancer or blood malignancies due to exposure to X-ray radiation. This study aimed to develop a new method to quickly predict patient-specific organ doses from CT examinations by training neural networks (NNs) based on radiomics features. Methods CT Digital Imaging and Communications in Medicine (DICOM) image data were exported to DeepViewer, a clinical autosegmentation software, to segment the regions of interest (ROIs) for patient organs. Radiomics feature extraction was performed based on the selected CT data and ROIs. Reference organ doses were computed using Monte Carlo (MC) simulations. Patient-specific organ doses were predicted by training a NN model based on radiomics features and reference doses. For the dose prediction performance, the relative root mean squared error (RRMSE), mean absolute percentage error (MAPE), and coefficient of determination (R2) were evaluated on the test sets. The robustness of the NN model was evaluated via the random rearrangement of patient samples in the training and test sets. Results The maximal difference between the reference and predicted doses was less than 1 mGy for all investigated organs. The range of MAPE was 1.68% to 5.2% for head organs, 11.42% to 15.2% for chest organs, and 5.0% to 8.0% for abdominal organs; the maximal R2 values were 0.93, 0.86, and 0.89 for the head, chest, and abdominal organs, respectively. Conclusions The radiomics feature-based NN model can achieve accurate prediction of patient-specific organ doses at a high speed of less than 1 second using a single central processing unit, which supports its use as a user-friendly online clinical application.
Collapse
Affiliation(s)
- Wencheng Shao
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Xin Lin
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Ying Huang
- Institute of Modern Physics, Fudan University, Shanghai, China
| | - Liangyong Qu
- Department of Radiology, Shanghai Zhongye Hospital, Shanghai, China
| | - Weihai Zhuo
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Haikuan Liu
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Lancia A, Ingrosso G, Detti B, Festa E, Bonzano E, Linguanti F, Camilli F, Bertini N, La Mattina S, Orsatti C, Francolini G, Abenavoli EM, Livi L, Aristei C, de Jong D, Al Feghali KA, Siva S, Becherini C. Biology-guided radiotherapy in metastatic prostate cancer: time to push the envelope? Front Oncol 2024; 14:1455428. [PMID: 39314633 PMCID: PMC11417306 DOI: 10.3389/fonc.2024.1455428] [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: 06/26/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
The therapeutic landscape of metastatic prostate cancer has undergone a profound revolution in recent years. In addition to the introduction of novel molecules in the clinics, the field has witnessed a tremendous development of functional imaging modalities adding new biological insights which can ultimately inform tailored treatment strategies, including local therapies. The evolution and rise of Stereotactic Body Radiotherapy (SBRT) have been particularly notable in patients with oligometastatic disease, where it has been demonstrated to be a safe and effective treatment strategy yielding favorable results in terms of disease control and improved oncological outcomes. The possibility of debulking all sites of disease, matched with the ambition of potentially extending this treatment paradigm to polymetastatic patients in the not-too-distant future, makes Biology-guided Radiotherapy (BgRT) an attractive paradigm which can be used in conjunction with systemic therapy in the management of patients with metastatic prostate cancer.
Collapse
Affiliation(s)
- Andrea Lancia
- Department of Radiation Oncology, San Matteo Hospital Foundation Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Pavia, Italy
| | | | - Beatrice Detti
- Radiotherapy Unit Prato, Usl Centro Toscana, Presidio Villa Fiorita, Prato, Italy
| | - Eleonora Festa
- Radiation Oncology Section, University of Perugia, Perugia, Italy
| | - Elisabetta Bonzano
- Department of Radiation Oncology, San Matteo Hospital Foundation Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Pavia, Italy
| | | | - Federico Camilli
- Radiation Oncology Section, University of Perugia, Perugia, Italy
| | - Niccolò Bertini
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Salvatore La Mattina
- Department of Radiation Oncology, San Matteo Hospital Foundation Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Pavia, Italy
| | - Carolina Orsatti
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Giulio Francolini
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | | | - Lorenzo Livi
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Cynthia Aristei
- Radiation Oncology Section, University of Perugia, Perugia, Italy
| | - Dorine de Jong
- Medical Affairs, RefleXion Medical, Inc., Hayward, CA, United States
| | | | - Shankar Siva
- Department of Radiation Oncology, Sir Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Carlotta Becherini
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| |
Collapse
|
5
|
Nguyen TNH, Horowitz LF, Krilov T, Lockhart E, Kenerson HL, Gujral TS, Yeung RS, Arroyo-Currás N, Folch A. Label-free, real-time monitoring of cytochrome C drug responses in microdissected tumor biopsies with a multi-well aptasensor platform. SCIENCE ADVANCES 2024; 10:eadn5875. [PMID: 39241078 PMCID: PMC11378948 DOI: 10.1126/sciadv.adn5875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 07/31/2024] [Indexed: 09/08/2024]
Abstract
Functional assays on intact tumor biopsies can complement genomics-based approaches for precision oncology, drug testing, and organs-on-chips cancer disease models by capturing key therapeutic response determinants, such as tissue architecture, tumor heterogeneity, and the tumor microenvironment. Most of these assays rely on fluorescent labeling, a semiquantitative method best suited for single-time-point assays or labor-intensive immunostaining analysis. Here, we report integrated aptamer electrochemical sensors for on-chip, real-time monitoring of cytochrome C, a cell death indicator, from intact microdissected tissues with high affinity and specificity. The platform features a multi-well sensor layout and a multiplexed electronic setup. The aptasensors measure increases in cytochrome C in the supernatant of mouse or human microdissected tumors after exposure to various drug treatments. Because of the sensor's high affinity, it primarily tracks rising concentrations of cytochrome C, capturing dynamic changes during apoptosis. This approach could help develop more advanced cancer disease models and apply to other complex in vitro disease models, such as organs-on-chips and organoids.
Collapse
Affiliation(s)
- Tran N H Nguyen
- Department of Bioengineering, University of Washington, Seattle, WA 98105, USA
| | - Lisa F Horowitz
- Department of Bioengineering, University of Washington, Seattle, WA 98105, USA
| | - Timothy Krilov
- Department of Bioengineering, University of Washington, Seattle, WA 98105, USA
| | - Ethan Lockhart
- Department of Bioengineering, University of Washington, Seattle, WA 98105, USA
| | - Heidi L Kenerson
- Department of Surgery, University of Washington, Seattle, WA 98105, USA
| | - Taranjit S Gujral
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98105, USA
| | - Raymond S Yeung
- Department of Surgery, University of Washington, Seattle, WA 98105, USA
| | | | - Albert Folch
- Department of Bioengineering, University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
6
|
Kaur J, Kaur P. A systematic literature analysis of multi-organ cancer diagnosis using deep learning techniques. Comput Biol Med 2024; 179:108910. [PMID: 39032244 DOI: 10.1016/j.compbiomed.2024.108910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
Cancer is becoming the most toxic ailment identified among individuals worldwide. The mortality rate has been increasing rapidly every year, which causes progression in the various diagnostic technologies to handle this illness. The manual procedure for segmentation and classification with a large set of data modalities can be a challenging task. Therefore, a crucial requirement is to significantly develop the computer-assisted diagnostic system intended for the initial cancer identification. This article offers a systematic review of Deep Learning approaches using various image modalities to detect multi-organ cancers from 2012 to 2023. It emphasizes the detection of five supreme predominant tumors, i.e., breast, brain, lung, skin, and liver. Extensive review has been carried out by collecting research and conference articles and book chapters from reputed international databases, i.e., Springer Link, IEEE Xplore, Science Direct, PubMed, and Wiley that fulfill the criteria for quality evaluation. This systematic review summarizes the overview of convolutional neural network model architectures and datasets used for identifying and classifying the diverse categories of cancer. This study accomplishes an inclusive idea of ensemble deep learning models that have achieved better evaluation results for classifying the different images into cancer or healthy cases. This paper will provide a broad understanding to the research scientists within the domain of medical imaging procedures of which deep learning technique perform best over which type of dataset, extraction of features, different confrontations, and their anticipated solutions for the complex problems. Lastly, some challenges and issues which control the health emergency have been discussed.
Collapse
Affiliation(s)
- Jaspreet Kaur
- Department of Computer Engineering & Technology, Guru Nanak Dev University, Amritsar, Punjab, India.
| | - Prabhpreet Kaur
- Department of Computer Engineering & Technology, Guru Nanak Dev University, Amritsar, Punjab, India.
| |
Collapse
|
7
|
Soares Dos Santos MP, Bernardo RMC, Vidal J, Moreira A, Torres DFM, Herdeiro CAR, Santos HA, Gonçalves G. Next-generation chemotherapy treatments based on black hole algorithms: From cancer remission to chronic disease management. Comput Biol Med 2024; 180:108961. [PMID: 39106673 DOI: 10.1016/j.compbiomed.2024.108961] [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: 03/15/2024] [Revised: 07/10/2024] [Accepted: 07/26/2024] [Indexed: 08/09/2024]
Abstract
PROBLEM Therapeutic planning strategies have been developed to enhance the effectiveness of cancer drugs. Nevertheless, their performance is highly limited by the inefficient biological representativeness of predictive tumor growth models, which hinders their translation to clinical practice. OBJECTIVE This study proposes a disruptive approach to oncology based on nature-inspired control using realistic Black Hole physical laws, in which tumor masses are trapped to experience attraction dynamics on their path to complete remission or to become a chronic disease. This control method is designed to operate independently of individual patient idiosyncrasies, including high tumor heterogeneities and highly uncertain tumor dynamics, making it a promising avenue for advancing beyond the limitations of the traditional survival probabilistic paradigm. DESIGN Here, we provide a multifaceted study of chemotherapy therapeutic planning that includes: (1) the design of a pioneering controller algorithm based on physical laws found in the Black Holes; (2) investigation of the ability of this controller algorithm to ensure stable equilibrium treatments; and (3) simulation tests concerning tumor volume dynamics using drugs with significantly different pharmacokinetics (Cyclophosphamide and Atezolizumab), tumor volumes (200 mm3 and 12 732 mm3) and modeling characterizations (Gompertzian and Logistic tumor growth models). RESULTS Our results highlight the ability of this new astrophysical-inspired control algorithm to perform effective chemotherapy treatments for multiple tumor-treatment scenarios, including tumor resistance to chemotherapy, clinical scenarios modelled by time-dependent parameters, and highly uncertain tumor dynamics. CONCLUSIONS Our findings provide strong evidence that cancer therapy inspired by phenomena found in black holes can emerge as a disruptive paradigm. This opens new high-impacting research directions, exploring synergies between astrophysical-inspired control algorithms and Artificial Intelligence applied to advanced personalized cancer therapeutics.
Collapse
Affiliation(s)
- Marco P Soares Dos Santos
- Center for Mechanical Technology & Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal; Intelligent Systems Associate Laboratory (LASI), Guimarães, Portugal.
| | - Rodrigo M C Bernardo
- Center for Mechanical Technology & Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal
| | - JoãoV Vidal
- Department of Physics and Aveiro Institute of Materials (CICECO), University of Aveiro, Aveiro, Portugal; Department of Physics and Institute for Nanostructures, Nanomodelling and Nanofabrication (I3N), University of Aveiro, Aveiro, Portugal
| | - Ana Moreira
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193, Aveiro, Portugal
| | - Delfim F M Torres
- Center for Research and Development in Mathematics and Applications (CIDMA), Department of Mathematics, University of Aveiro, Aveiro, Portugal
| | - Carlos A R Herdeiro
- Center for Research and Development in Mathematics and Applications (CIDMA), Department of Mathematics, University of Aveiro, Aveiro, Portugal
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Gil Gonçalves
- Center for Mechanical Technology & Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal; Intelligent Systems Associate Laboratory (LASI), Guimarães, Portugal
| |
Collapse
|
8
|
Jha CB, Singh C, Randhawa JK, Kaul A, Varshney R, Singh S, Kaushik A, Manna K, Mathur R. Synthesis and evaluation of curcumin reduced and capped gold nanoparticles as a green diagnostic probe with therapeutic potential. Colloids Surf B Biointerfaces 2024; 241:114050. [PMID: 38936032 DOI: 10.1016/j.colsurfb.2024.114050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/04/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
Curcumin, a compound in turmeric, shows promise for its anti-cancer properties. In this study, we successfully synthesised curcumin-reduced and capped gold nanoparticles. Most evaluations have been limited to in-vitro studies for these nanoparticles; our study takes a step further by highlighting the in-vivo assessment of these curcumin-reduced and capped gold nanoparticles (GNPCs) using non-invasive imaging (SPECT and optical) and possible therapeutic potential. The GNPCs showed an average hydrodynamic diameter of 58 nm and a PDI of 0.336. The synthesised and fully characterised GNPCs showed ex-vivo hemolysis value of ≤ 1.74 % and serum stability of ≥ 95 % over 24 h. Using in-vivo non-invasive (SPECT and optical Imaging), prolonged circulation and enhanced bioavailability of GNPCs were seen. The biodistribution studies after radiolabelling GNPCs with 99 mTc complemented the optical imaging. The SPECT images showed higher uptake of the GNPCs at the tumour site, viz the contralateral muscle and the native Curcumin, resulting in a high target-to-non-target ratio that differentiated the tumour sufficiently and enhanced the diagnostics. Other organs also accumulate radiolabeled GNPCs in systemic circulation; bio dosimetry is performed. It was found that the dose received by the different organs was safe for use, and the in-vivo toxicity studies in rats indicated negligible toxicity over 30 days. The tumour growth was also reduced in mice models treated with GNPCs compared to the control. These significant findings demonstrate that GNPC shows synergistic activity in vivo, indicating its ability as a green diagnostic probe that has the potential for therapy.
Collapse
Affiliation(s)
- Chandan Bhogendra Jha
- Division of Radiological, Nuclear and Imaging Sciences, Institute of Nuclear Medicine and Allied Sciences, DRDO, Delhi 110054, India; Department of Chemistry, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Chitrangda Singh
- Division of Radiological, Nuclear and Imaging Sciences, Institute of Nuclear Medicine and Allied Sciences, DRDO, Delhi 110054, India
| | | | - Ankur Kaul
- Division of Radiological, Nuclear and Imaging Sciences, Institute of Nuclear Medicine and Allied Sciences, DRDO, Delhi 110054, India
| | - Raunak Varshney
- Division of Radiological, Nuclear and Imaging Sciences, Institute of Nuclear Medicine and Allied Sciences, DRDO, Delhi 110054, India
| | - Sweta Singh
- Division of Radiological, Nuclear and Imaging Sciences, Institute of Nuclear Medicine and Allied Sciences, DRDO, Delhi 110054, India
| | - Aruna Kaushik
- Division of Radiological, Nuclear and Imaging Sciences, Institute of Nuclear Medicine and Allied Sciences, DRDO, Delhi 110054, India
| | - Kuntal Manna
- Department of Chemistry, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Rashi Mathur
- Division of Radiological, Nuclear and Imaging Sciences, Institute of Nuclear Medicine and Allied Sciences, DRDO, Delhi 110054, India.
| |
Collapse
|
9
|
Suri S, Boora GS, Kaur R, Chauhan A, Ghoshal S, Pal A. Recent advances in minimally invasive biomarkers of OSCC: from generalized to personalized approach. FRONTIERS IN ORAL HEALTH 2024; 5:1426507. [PMID: 39157206 PMCID: PMC11327221 DOI: 10.3389/froh.2024.1426507] [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: 05/01/2024] [Accepted: 07/16/2024] [Indexed: 08/20/2024] Open
Abstract
Oral cancer is the 6th most common type of cancer worldwide, and oral squamous cell carcinoma (OSCC) accounts for >90% of oral cancers. It is a major health problem, particularly in low- and middle-income countries (LMICs), due to both its high incidence and significant mortality and morbidity. Despite being a global burden, and even with the significant advancement in the management of OSCC, the overall outcome of the disease is still abysmal. With the advent of time, advanced diagnostic and treatment approaches have come into practice, but the burden of the disease has not improved significantly. Major reasons attributed to the poor outcome are delay in diagnosis, locoregional recurrence and resistance to the currently available treatment regimen. In this review, we have highlighted the existing challenges in the diagnosis and have emphasized the advancements in minimally invasive biomarkers. Additionally, the importance of collaborative multidimensional approaches involving clinicians and researchers has been discussed, as well as the need to redefine and establish better utility and management of existing diagnostic and treatment protocols along with the minimally invasive/non-invasive biomarkers.
Collapse
Affiliation(s)
- Smriti Suri
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh,India
| | - Geeta S. Boora
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh,India
| | - Rajandeep Kaur
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh,India
| | - Anshika Chauhan
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh,India
| | - Sushmita Ghoshal
- Department of Radiotherapy, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arnab Pal
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh,India
| |
Collapse
|
10
|
Russo L, Charles-Davies D, Bottazzi S, Sala E, Boldrini L. Radiomics for clinical decision support in radiation oncology. Clin Oncol (R Coll Radiol) 2024; 36:e269-e281. [PMID: 38548581 DOI: 10.1016/j.clon.2024.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 02/14/2024] [Accepted: 03/08/2024] [Indexed: 07/09/2024]
Abstract
Radiomics is a promising tool for the development of quantitative biomarkers to support clinical decision-making. It has been shown to improve the prediction of response to treatment and outcome in different settings, particularly in the field of radiation oncology by optimising the dose delivery solutions and reducing the rate of radiation-induced side effects, leading to a fully personalised approach. Despite the promising results offered by radiomics at each of these stages, standardised methodologies, reproducibility and interpretability of results are still lacking, limiting the potential clinical impact of these tools. In this review, we briefly describe the principles of radiomics and the most relevant applications of radiomics at each stage of cancer management in the framework of radiation oncology. Furthermore, the integration of radiomics into clinical decision support systems is analysed, defining the challenges and offering possible solutions for translating radiomics into a clinically applicable tool.
Collapse
Affiliation(s)
- L Russo
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Dipartimento di Scienze Radiologiche ed Ematologiche. Università Cattolica Del Sacro Cuore, Rome, Italy.
| | - D Charles-Davies
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - S Bottazzi
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - E Sala
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Dipartimento di Scienze Radiologiche ed Ematologiche. Università Cattolica Del Sacro Cuore, Rome, Italy
| | - L Boldrini
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
11
|
Chinnappan R, Makhzoum T, Arai M, Hajja A, Abul Rub F, Alodhaibi I, Alfuwais M, Elahi MA, Alshehri EA, Ramachandran L, Mani NK, Abrahim S, Mir MS, Al-Kattan K, Mir TA, Yaqinuddin A. Recent Advances in Biosensor Technology for Early-Stage Detection of Hepatocellular Carcinoma-Specific Biomarkers: An Overview. Diagnostics (Basel) 2024; 14:1519. [PMID: 39061656 PMCID: PMC11276200 DOI: 10.3390/diagnostics14141519] [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: 06/06/2024] [Revised: 07/06/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Hepatocellular carcinoma is currently the most common malignancy of the liver. It typically occurs due to a series of oncogenic mutations that lead to aberrant cell replication. Most commonly, hepatocellular carcinoma (HCC) occurs as a result of pre-occurring liver diseases, such as hepatitis and cirrhosis. Given its aggressive nature and poor prognosis, the early screening and diagnosis of HCC are crucial. However, due to its plethora of underlying risk factors and pathophysiologies, patient presentation often varies in the early stages, with many patients presenting with few, if any, specific symptoms in the early stages. Conventionally, screening and diagnosis are performed through radiological examination, with diagnosis confirmed by biopsy. Imaging modalities tend to be limited by their requirement of large, expensive equipment; time-consuming operation; and a lack of accurate diagnosis, whereas a biopsy's invasive nature makes it unappealing for repetitive use. Recently, biosensors have gained attention for their potential to detect numerous conditions rapidly, cheaply, accurately, and without complex equipment and training. Through their sensing platforms, they aim to detect various biomarkers, such as nucleic acids, proteins, and even whole cells extracted by a liquid biopsy. Numerous biosensors have been developed that may detect HCC in its early stages. We discuss the recent updates in biosensing technology, highlighting its competitive potential compared to conventional methodology and its prospects as a tool for screening and diagnosis.
Collapse
Affiliation(s)
- Raja Chinnappan
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.M.); (M.A.); (A.H.); (F.A.R.); (I.A.); (M.A.); (M.A.E.); (K.A.-K.); (T.A.M.)
- Tissue/Organ Bioengineering & BioMEMS Laboratory, Organ Transplant Centre of Excellence (TR&I-Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia;
| | - Tariq Makhzoum
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.M.); (M.A.); (A.H.); (F.A.R.); (I.A.); (M.A.); (M.A.E.); (K.A.-K.); (T.A.M.)
| | - Momo Arai
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.M.); (M.A.); (A.H.); (F.A.R.); (I.A.); (M.A.); (M.A.E.); (K.A.-K.); (T.A.M.)
| | - Amro Hajja
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.M.); (M.A.); (A.H.); (F.A.R.); (I.A.); (M.A.); (M.A.E.); (K.A.-K.); (T.A.M.)
| | - Farah Abul Rub
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.M.); (M.A.); (A.H.); (F.A.R.); (I.A.); (M.A.); (M.A.E.); (K.A.-K.); (T.A.M.)
| | - Ibrahim Alodhaibi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.M.); (M.A.); (A.H.); (F.A.R.); (I.A.); (M.A.); (M.A.E.); (K.A.-K.); (T.A.M.)
| | - Mohammed Alfuwais
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.M.); (M.A.); (A.H.); (F.A.R.); (I.A.); (M.A.); (M.A.E.); (K.A.-K.); (T.A.M.)
| | - Muhammad Affan Elahi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.M.); (M.A.); (A.H.); (F.A.R.); (I.A.); (M.A.); (M.A.E.); (K.A.-K.); (T.A.M.)
| | - Eman Abdullah Alshehri
- Tissue/Organ Bioengineering & BioMEMS Laboratory, Organ Transplant Centre of Excellence (TR&I-Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia;
| | - Lohit Ramachandran
- Microfluidics, Sensors & Diagnostics (μSenD) Laboratory, Centre for Microfluidics, Biomarkers, Photoceutics and Sensors (μBioPS), Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India; (L.R.); (N.K.M.)
| | - Naresh Kumar Mani
- Microfluidics, Sensors & Diagnostics (μSenD) Laboratory, Centre for Microfluidics, Biomarkers, Photoceutics and Sensors (μBioPS), Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India; (L.R.); (N.K.M.)
| | - Shugufta Abrahim
- Graduate School of Science and Engineering for Education, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan;
| | - Mohammad Shabab Mir
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh 147301, Punjab, India;
| | - Khaled Al-Kattan
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.M.); (M.A.); (A.H.); (F.A.R.); (I.A.); (M.A.); (M.A.E.); (K.A.-K.); (T.A.M.)
- Lung Health Centre Department, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Tanveer Ahmad Mir
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.M.); (M.A.); (A.H.); (F.A.R.); (I.A.); (M.A.); (M.A.E.); (K.A.-K.); (T.A.M.)
- Tissue/Organ Bioengineering & BioMEMS Laboratory, Organ Transplant Centre of Excellence (TR&I-Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia;
| | - Ahmed Yaqinuddin
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.M.); (M.A.); (A.H.); (F.A.R.); (I.A.); (M.A.); (M.A.E.); (K.A.-K.); (T.A.M.)
| |
Collapse
|
12
|
Zhang K, Abdoli N, Gilley P, Sadri Y, Chen X, Thai TC, Dockery L, Moore K, Mannel RS, Qiu Y. Developing a Novel Image Marker to Predict the Clinical Outcome of Neoadjuvant Chemotherapy (NACT) for Ovarian Cancer Patients. ARXIV 2024:arXiv:2309.07087v2. [PMID: 37744460 PMCID: PMC10516116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Objective Neoadjuvant chemotherapy (NACT) is one kind of treatment for advanced stage ovarian cancer patients. However, due to the nature of tumor heterogeneity, the clinical outcome to NACT vary significantly among different subgroups. The patients with partial responses to NACT may lead to suboptimal debulking surgery, which will result in adverse prognosis. To address this clinical challenge, the purpose of this study is to develop a novel image marker to achieve high accuracy prognosis prediction of the NACT at an early stage. Methods For this purpose, we first computed a total of 1373 radiomics features to quantify the tumor characteristics, which can be grouped into three categories: geometric, intensity, and texture features. Second, all these features were optimized by principal component analysis algorithm to generate a compact and informative feature cluster. This cluster was used as input for developing and optimizing support vector machine (SVM) based classifiers, which indicated the likelihood of the patient receiving suboptimal cytoreduction after the NACT treatment. Two different kernels for SVM algorithm were explored and compared. To validate this scheme, a total of 42 ovarian cancer patients were retrospectively collected. A nested leave-one-out cross-validation framework was adopted for model performance assessment. Results The results demonstrated that the model with a Gaussian radial basis function kernel SVM yielded an AUC (area under the ROC [receiver characteristic operation] curve) of 0.806 ± 0.078. Meanwhile, this model achieved overall accuracy (ACC) of 83.3%, positive predictive value (PPV) of 81.8%, and negative predictive value (NPV) of 83.9%. Conclusion This study provides meaningful information for the development of radiomics based image markers in NACT treatment outcome prediction.
Collapse
Affiliation(s)
- Ke Zhang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA 73019
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK, USA 73019
| | - Neman Abdoli
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK, USA 73019
| | - Patrik Gilley
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK, USA 73019
| | - Youkabed Sadri
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK, USA 73019
| | - Xuxin Chen
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK, USA 73019
| | - Theresa C. Thai
- Department of Radiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA 73104
| | - Lauren Dockery
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA 73104
| | - Kathleen Moore
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA 73104
| | - Robert S. Mannel
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA 73104
| | - Yuchen Qiu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA 73019
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK, USA 73019
| |
Collapse
|
13
|
Xie Z, Zhao H, Song L, Ye Q, Zhong L, Li S, Zhang R, Wang M, Chen X, Lu Z, Yang W, Zhao Y. A radiograph-based deep learning model improves radiologists' performance for classification of histological types of primary bone tumors: A multicenter study. Eur J Radiol 2024; 176:111496. [PMID: 38733705 DOI: 10.1016/j.ejrad.2024.111496] [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: 09/25/2023] [Revised: 04/03/2024] [Accepted: 05/02/2024] [Indexed: 05/13/2024]
Abstract
PURPOSE To develop a deep learning (DL) model for classifying histological types of primary bone tumors (PBTs) using radiographs and evaluate its clinical utility in assisting radiologists. METHODS This retrospective study included 878 patients with pathologically confirmed PBTs from two centers (638, 77, 80, and 83 for the training, validation, internal test, and external test sets, respectively). We classified PBTs into five categories by histological types: chondrogenic tumors, osteogenic tumors, osteoclastic giant cell-rich tumors, other mesenchymal tumors of bone, or other histological types of PBTs. A DL model combining radiographs and clinical features based on the EfficientNet-B3 was developed for five-category classification. The area under the receiver operating characteristic curve (AUC), accuracy, sensitivity, and specificity were calculated to evaluate model performance. The clinical utility of the model was evaluated in an observer study with four radiologists. RESULTS The combined model achieved a macro average AUC of 0.904/0.873, with an accuracy of 67.5 %/68.7 %, a macro average sensitivity of 66.9 %/57.2 %, and a macro average specificity of 92.1 %/91.6 % on the internal/external test set, respectively. Model-assisted analysis improved accuracy, interpretation time, and confidence for junior (50.6 % vs. 72.3 %, 53.07[s] vs. 18.55[s] and 3.10 vs. 3.73 on a 5-point Likert scale [P < 0.05 for each], respectively) and senior radiologists (68.7 % vs. 75.3 %, 32.50[s] vs. 21.42[s] and 4.19 vs. 4.37 [P < 0.05 for each], respectively). CONCLUSION The combined DL model effectively classified histological types of PBTs and assisted radiologists in achieving better classification results than their independent visual assessment.
Collapse
Affiliation(s)
- Zhuoyao Xie
- Department of Radiology, The Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics, Guangdong Province), 183 Zhongshan Da Dao Xi, Guangzhou, Guangdong, 510630, China.
| | - Huanmiao Zhao
- Guangdong Provincial Key Laboratory of Medical Image Processing, School of Biomedical Engineering, Southern Medical University, 1023 Shatai Road, Baiyun District, Guangzhou, 510515, Guangdong, China.
| | - Liwen Song
- Department of Radiology, The Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics, Guangdong Province), 183 Zhongshan Da Dao Xi, Guangzhou, Guangdong, 510630, China.
| | - Qiang Ye
- Department of Radiology, The Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics, Guangdong Province), 183 Zhongshan Da Dao Xi, Guangzhou, Guangdong, 510630, China.
| | - Liming Zhong
- Guangdong Provincial Key Laboratory of Medical Image Processing, School of Biomedical Engineering, Southern Medical University, 1023 Shatai Road, Baiyun District, Guangzhou, 510515, Guangdong, China.
| | - Shisi Li
- Department of Radiology, The Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics, Guangdong Province), 183 Zhongshan Da Dao Xi, Guangzhou, Guangdong, 510630, China.
| | - Rui Zhang
- Department of Radiology, The Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics, Guangdong Province), 183 Zhongshan Da Dao Xi, Guangzhou, Guangdong, 510630, China.
| | - Menghong Wang
- Department of Radiology, The Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics, Guangdong Province), 183 Zhongshan Da Dao Xi, Guangzhou, Guangdong, 510630, China.
| | - Xiaqing Chen
- Department of Radiology, The Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics, Guangdong Province), 183 Zhongshan Da Dao Xi, Guangzhou, Guangdong, 510630, China.
| | - Zixiao Lu
- Department of Radiology, The Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics, Guangdong Province), 183 Zhongshan Da Dao Xi, Guangzhou, Guangdong, 510630, China.
| | - Wei Yang
- Guangdong Provincial Key Laboratory of Medical Image Processing, School of Biomedical Engineering, Southern Medical University, 1023 Shatai Road, Baiyun District, Guangzhou, 510515, Guangdong, China.
| | - Yinghua Zhao
- Department of Radiology, The Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics, Guangdong Province), 183 Zhongshan Da Dao Xi, Guangzhou, Guangdong, 510630, China.
| |
Collapse
|
14
|
Sun X, Li Y, Liu X, Cui D, Shi Y, Huang G. Tumor-specific enhanced NIR-II photoacoustic imaging via photothermal and low-pH coactivated AuNR@PNIPAM-VAA nanogel. J Nanobiotechnology 2024; 22:326. [PMID: 38858673 PMCID: PMC11163807 DOI: 10.1186/s12951-024-02617-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Properly designed second near-infrared (NIR-II) nanoplatform that is responsive tumor microenvironment can intelligently distinguish between normal and cancerous tissues to achieve better targeting efficiency. Conventional photoacoustic nanoprobes are always "on", and tumor microenvironment-responsive nanoprobe can minimize the influence of endogenous chromophore background signals. Therefore, the development of nanoprobe that can respond to internal tumor microenvironment and external stimulus shows great application potential for the photoacoustic diagnosis of tumor. RESULTS In this work, a low-pH-triggered thermal-responsive volume phase transition nanogel gold nanorod@poly(n-isopropylacrylamide)-vinyl acetic acid (AuNR@PNIPAM-VAA) was constructed for photoacoustic detection of tumor. Via an external near-infrared photothermal switch, the absorption of AuNR@PNIPAM-VAA nanogel in the tumor microenvironment can be dynamically regulated, so that AuNR@PNIPAM-VAA nanogel produces switchable photoacoustic signals in the NIR-II window for tumor-specific enhanced photoacoustic imaging. In vitro results show that at pH 5.8, the absorption and photoacoustic signal amplitude of AuNR@PNIPAM-VAA nanogel in NIR-II increases up obviously after photothermal modulating, while they remain slightly change at pH 7.4. Quantitative calculation presents that photoacoustic signal amplitude of AuNR@PNIPAM-VAA nanogel at 1064 nm has ~ 1.6 folds enhancement as temperature increases from 37.5 °C to 45 °C in simulative tumor microenvironment. In vivo results show that the prepared AuNR@PNIPAM-VAA nanogel can achieve enhanced NIR-II photoacoustic imaging for selective tumor detection through dynamically responding to thermal field, which can be precisely controlled by external light. CONCLUSIONS This work will offer a viable strategy for the tumor-specific photoacoustic imaging using NIR light to regulate the thermal field and target the low pH tumor microenvironment, which is expected to realize accurate and dynamic monitoring of tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Xiaodong Sun
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Yujie Li
- Reproductive Medicine Research Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
| | - Xiaowan Liu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Dandan Cui
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Yujiao Shi
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.
| | - Guojia Huang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| |
Collapse
|
15
|
Cai Y, Luo M, Yang W, Xu C, Wang P, Xue G, Jin X, Cheng R, Que J, Zhou W, Pang B, Xu S, Li Y, Jiang Q, Xu Z. The Deep Learning Framework iCanTCR Enables Early Cancer Detection Using the T-cell Receptor Repertoire in Peripheral Blood. Cancer Res 2024; 84:1915-1928. [PMID: 38536129 DOI: 10.1158/0008-5472.can-23-0860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 07/20/2023] [Accepted: 03/19/2024] [Indexed: 06/05/2024]
Abstract
T cells recognize tumor antigens and initiate an anticancer immune response in the very early stages of tumor development, and the antigen specificity of T cells is determined by the T-cell receptor (TCR). Therefore, monitoring changes in the TCR repertoire in peripheral blood may offer a strategy to detect various cancers at a relatively early stage. Here, we developed the deep learning framework iCanTCR to identify patients with cancer based on the TCR repertoire. The iCanTCR framework uses TCRβ sequences from an individual as an input and outputs the predicted cancer probability. The model was trained on over 2,000 publicly available TCR repertoires from 11 types of cancer and healthy controls. Analysis of several additional publicly available datasets validated the ability of iCanTCR to distinguish patients with cancer from noncancer individuals and demonstrated the capability of iCanTCR for the accurate classification of multiple cancers. Importantly, iCanTCR precisely identified individuals with early-stage cancer with an AUC of 86%. Altogether, this work provides a liquid biopsy approach to capture immune signals from peripheral blood for noninvasive cancer diagnosis. SIGNIFICANCE Development of a deep learning-based method for multicancer detection using the TCR repertoire in the peripheral blood establishes the potential of evaluating circulating immune signals for noninvasive early cancer detection.
Collapse
Affiliation(s)
- Yideng Cai
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Meng Luo
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Wenyi Yang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Chang Xu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Pingping Wang
- School for Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, China
| | - Guangfu Xue
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xiyun Jin
- School for Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, China
| | - Rui Cheng
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Jinhao Que
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Wenyang Zhou
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Boran Pang
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shouping Xu
- Department of Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yu Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Qinghua Jiang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
- School for Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, China
| | - Zhaochun Xu
- School for Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, China
| |
Collapse
|
16
|
Wegierak D, Cooley MB, Perera R, Wulftange WJ, Gurkan UA, Kolios MC, Exner AA. Decorrelation Time Mapping as an Analysis Tool for Nanobubble-Based Contrast Enhanced Ultrasound Imaging. IEEE TRANSACTIONS ON MEDICAL IMAGING 2024; 43:2370-2380. [PMID: 38329864 PMCID: PMC11234354 DOI: 10.1109/tmi.2024.3364076] [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] [Indexed: 02/10/2024]
Abstract
Nanobubbles (NBs; ~100-500 nm diameter) are preclinical ultrasound (US) contrast agents that expand applications of contrast enhanced US (CEUS). Due to their sub-micron size, high particle density, and deformable shell, NBs in pathological states of heightened vascular permeability (e.g. in tumors) extravasate, enabling applications not possible with microbubbles (~1000-10,000 nm diameter). A method that can separate intravascular versus extravascular NB signal is needed as an imaging biomarker for improved tumor detection. We present a demonstration of decorrelation time (DT) mapping for enhanced tumor NB-CEUS imaging. In vitro models validated the sensitivity of DT to agent motion. Prostate cancer mouse models validated in vivo imaging potential and sensitivity to cancerous tissue. Our findings show that DT is inversely related to NB motion, offering enhanced detail of NB dynamics in tumors, and highlighting the heterogeneity of the tumor environment. Average DT was high in tumor regions (~9 s) compared to surrounding normal tissue (~1 s) with higher sensitivity to tumor tissue compared to other mapping techniques. Molecular NB targeting to tumors further extended DT (11 s) over non-targeted NBs (6 s), demonstrating sensitivity to NB adherence. From DT mapping of in vivo NB dynamics we demonstrate the heterogeneity of tumor tissue while quantifying extravascular NB kinetics and delineating intra-tumoral vasculature. This new NB-CEUS-based biomarker can be powerful in molecular US imaging, with improved sensitivity and specificity to diseased tissue and potential for use as an estimator of vascular permeability and the enhanced permeability and retention (EPR) effect in tumors.
Collapse
|
17
|
Someya Y, Iima M, Imai H, Isoda H, Ohno T, Kataoka M, Bihan DL, Nakamoto Y. In Vivo and Post-mortem Comparisons of IVIM/Time-dependent Diffusion MR Imaging Parameters in Melanoma and Breast Cancer Xenograft Models. Magn Reson Med Sci 2024:mp.2023-0078. [PMID: 38797683 DOI: 10.2463/mrms.mp.2023-0078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024] Open
Abstract
PURPOSE We aimed to investigate the changes in intravoxel incoherent motion (IVIM) and diffusion parameters between in vivo and post-mortem conditions and the time dependency of these parameters using two different mouse tumor models with different vessel lumen sizes. METHODS Six B16 and six MDA-MB-231 xenograft mice were scanned using 7 Tesla MRI under both in vivo/post-mortem conditions. Diffusion weighted imaging with 17 b-values (0-3000 s/mm2) were obtained at two diffusion times (9 and 27.6 ms). The shifted apparent diffusion coefficient (sADC) using 2 b-values (200 and 1500 s/mm2), non-Gaussian diffusion and IVIM parameters (ADC0, K, fIVIM) were estimated at each of the diffusion times. The results were evaluated by repeated measures two-way analysis of variance and post hoc Bonferroni test. RESULTS In B16 tumors, fIVIM significantly decreased with post-mortem conditions (from 12.6 ± 6.5% to 5.2 ± 1.9%, P < 0.05 at long diffusion time; from 11.0 ± 2.4% to 4.6 ± 2.7%, P < 0.05 at short diffusion time). In MDA-MB-231 tumors, fIVIM also significantly decreased (from 8.8 ± 3.8% to 2.6 ± 1.1%, P < 0.05 at long; from 7.9 ± 5.4% to 2.9 ± 1.1%, P < 0.05 at short). No diffusion time dependency was observed (P = 0.59 in B16 and P = 0.77 in MDA-MB-231). The sADC and ADC0 values tended to decrease and the K value tended to increase after sacrificing and when increasing the diffusion time. CONCLUSION The fIVIM values dropped after sacrificing, confirming that IVIM MRI is a promising quantitative parameter to evaluate blood microcirculation. The presence of residual post-mortem fIVIM values suggested that the influence of water molecule diffusion in the blood lumen may contribute to the IVIM effect. Diffusion MRI parameter's time dependency and those changes after sacrificing could possibly provide additional insights into diffusion hindrance mechanisms.
Collapse
Affiliation(s)
- Yuko Someya
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
- Department of Diagnostic Radiology, Kobe City Medical Center General Hospital, Kobe, Hyogo, Japan
| | - Mami Iima
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
- Department of Clinical Innovative Medicine, Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Kyoto, Japan
| | - Hirohiko Imai
- Department of Systems Science, Graduate School of Informatics, Kyoto University, Kyoto, Kyoto, Japan
| | - Hiroyoshi Isoda
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Tsuyoshi Ohno
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Masako Kataoka
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Denis Le Bihan
- NeuroSpin/Joliot, CEA-Saclay Center, Paris-Saclay University, Gif-sur-Yvette, France
- Human Brain Research Center, Kyoto University Graduate School of Medicine, Kyoto, Kyoto, Japan
- National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| |
Collapse
|
18
|
Urbano-Gámez JD, Guzzi C, Bernal M, Solivera J, Martínez-Zubiaurre I, Caro C, García-Martín ML. Tumor versus Tumor Cell Targeting in Metal-Based Nanoparticles for Cancer Theranostics. Int J Mol Sci 2024; 25:5213. [PMID: 38791253 PMCID: PMC11121233 DOI: 10.3390/ijms25105213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/05/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
The application of metal-based nanoparticles (mNPs) in cancer therapy and diagnostics (theranostics) has been a hot research topic since the early days of nanotechnology, becoming even more relevant in recent years. However, the clinical translation of this technology has been notably poor, with one of the main reasons being a lack of understanding of the disease and conceptual errors in the design of mNPs. Strikingly, throughout the reported studies to date on in vivo experiments, the concepts of "tumor targeting" and "tumor cell targeting" are often intertwined, particularly in the context of active targeting. These misconceptions may lead to design flaws, resulting in failed theranostic strategies. In the context of mNPs, tumor targeting can be described as the process by which mNPs reach the tumor mass (as a tissue), while tumor cell targeting refers to the specific interaction of mNPs with tumor cells once they have reached the tumor tissue. In this review, we conduct a critical analysis of key challenges that must be addressed for the successful targeting of either tumor tissue or cancer cells within the tumor tissue. Additionally, we explore essential features necessary for the smart design of theranostic mNPs, where 'smart design' refers to the process involving advanced consideration of the physicochemical features of the mNPs, targeting motifs, and physiological barriers that must be overcome for successful tumor targeting and/or tumor cell targeting.
Collapse
Affiliation(s)
- Jesús David Urbano-Gámez
- Biomedical Magnetic Resonance Laboratory—BMRL, Andalusian Public Foundation Progress and Health—FPS, 41092 Seville, Spain; (J.D.U.-G.); (C.G.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
| | - Cinzia Guzzi
- Biomedical Magnetic Resonance Laboratory—BMRL, Andalusian Public Foundation Progress and Health—FPS, 41092 Seville, Spain; (J.D.U.-G.); (C.G.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
| | - Manuel Bernal
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Andalucía Tech, 29071 Malaga, Spain
| | - Juan Solivera
- Department of Neurosurgery, Reina Sofia University Hospital, 14004 Cordoba, Spain;
| | - Iñigo Martínez-Zubiaurre
- Department of Clinical Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, P.O. Box 6050, Langnes, 9037 Tromsö, Norway;
| | - Carlos Caro
- Biomedical Magnetic Resonance Laboratory—BMRL, Andalusian Public Foundation Progress and Health—FPS, 41092 Seville, Spain; (J.D.U.-G.); (C.G.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
| | - María Luisa García-Martín
- Biomedical Magnetic Resonance Laboratory—BMRL, Andalusian Public Foundation Progress and Health—FPS, 41092 Seville, Spain; (J.D.U.-G.); (C.G.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
- Biomedical Research Networking Center in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| |
Collapse
|
19
|
Camli B, Andrus L, Roy A, Mishra B, Xu C, Georgakoudi I, Tkaczyk T, Ben-Yakar A. Two photon imaging probe with highly efficient autofluorescence collection at high scattering and deep imaging conditions. BIOMEDICAL OPTICS EXPRESS 2024; 15:3163-3182. [PMID: 38855663 PMCID: PMC11161376 DOI: 10.1364/boe.520729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/05/2024] [Accepted: 04/06/2024] [Indexed: 06/11/2024]
Abstract
In this paper, we present a 2-photon imaging probe system featuring a novel fluorescence collection method with improved and reliable efficiency. The system aims to miniaturize the potential of 2-photon imaging in the metabolic and morphological characterization of cervical tissue at sub-micron resolution over large imaging depths into a flexible and clinically viable platform towards the early detection of cancers. Clinical implementation of such a probe system is challenging due to inherently low levels of autofluorescence, particularly when imaging deep in highly scattering tissues. For an efficient collection of fluorescence signals, our probe employs 12 0.5 NA collection fibers arranged around a miniaturized excitation objective. By bending and terminating a multitude of collection fibers at a specific angle, we increase collection area and directivity significantly. Positioning of these fibers allows the collection of fluorescence photons scattered away from their ballistic trajectory multiple times, which offers a system collection efficiency of 4%, which is 55% of what our bench-top microscope with 0.75 NA objective achieves. We demonstrate that the collection efficiency is largely maintained even at high scattering conditions and high imaging depths. Radial symmetry of arrangement maintains uniformity of collection efficiency across the whole FOV. Additionally, our probe can image at different tissue depths via axial actuation by a dc servo motor, allowing depth dependent tissue characterization. We designed our probe to perform imaging at 775 nm, targeting 2-photon autofluorescence from NAD(P)H and FAD molecules, which are often used in metabolic tissue characterization. An air core photonic bandgap fiber delivers laser pulses of 100 fs duration to the sample. A miniaturized objective designed with commercially available lenses of 3 mm diameter focuses the laser beam on tissue, attaining lateral and axial imaging resolutions of 0.66 µm and 4.65 µm, respectively. Characterization results verify that our probe achieves collection efficiency comparable to our optimized bench-top 2-photon imaging microscope, minimally affected by imaging depth and radial positioning. We validate autofluorescence imaging capability with excised porcine vocal fold tissue samples. Images with 120 µm FOV and 0.33 µm pixel sizes collected at 2 fps confirm that the 300 µm imaging depth was achieved.
Collapse
Affiliation(s)
- Berk Camli
- Department of Mechanical Engineering, UT Austin, Austin, Texas, USA
| | - Liam Andrus
- Department of Biomedical Engineering, UT Austin, Austin, Texas, USA
| | - Aditya Roy
- Department of Mechanical Engineering, UT Austin, Austin, Texas, USA
| | - Biswajit Mishra
- Department of Mechanical Engineering, UT Austin, Austin, Texas, USA
| | - Chris Xu
- School of Applied and Engineering Physics, Cornell University, Ithaca, New York, USA
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Tomasz Tkaczyk
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Adela Ben-Yakar
- Department of Mechanical Engineering, UT Austin, Austin, Texas, USA
- Department of Biomedical Engineering, UT Austin, Austin, Texas, USA
- Department of Electrical and Computer Engineering, UT Austin, Austin, Texas, USA
| |
Collapse
|
20
|
Wang M, Liu Z, Liu C, He W, Qin D, You M. DNAzyme-based ultrasensitive immunoassay: Recent advances and emerging trends. Biosens Bioelectron 2024; 251:116122. [PMID: 38382271 DOI: 10.1016/j.bios.2024.116122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/03/2024] [Accepted: 02/08/2024] [Indexed: 02/23/2024]
Abstract
Immunoassay, as the most commonly used method for protein detection, is simple to operate and highly specific. Sensitivity improvement is always the thrust of immunoassays, especially for the detection of trace quantities. The emergence of artificial enzyme, i.e., DNAzyme, provides a novel approach to improve the detection sensitivity of immunoassay. Simultaneously, its advantages of simple synthesis and high stability enable low cost, broad applicability and long shelf life for immunoassay. In this review, we summarized the recent advances in DNAzyme-based immunoassay. First, we summarized the existing different DNAzymes based on their catalytic activities. Next, the common signal amplification strategies used for DNAzyme-based immunoassays were reviewed to cater to diverse detection requirements. Following, the wide applications in disease diagnosis, environmental monitoring and food safety were discussed. Finally, the current challenges and perspectives on the future development of DNAzyme-based immunoassays were also provided.
Collapse
Affiliation(s)
- Meng Wang
- Department of Biomedical Engineering, School of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing, 400065, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Zhe Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China; Department of Rehabilitation Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Chang Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Wanghong He
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China; Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, 100050, PR China
| | - Dui Qin
- Department of Biomedical Engineering, School of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing, 400065, PR China.
| | - Minli You
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China.
| |
Collapse
|
21
|
Fujita K, Urano Y. Activity-Based Fluorescence Diagnostics for Cancer. Chem Rev 2024; 124:4021-4078. [PMID: 38518254 DOI: 10.1021/acs.chemrev.3c00612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024]
Abstract
Fluorescence imaging is one of the most promising approaches to achieve intraoperative assessment of the tumor/normal tissue margins during cancer surgery. This is critical to improve the patients' prognosis, and therefore various molecular fluorescence imaging probes have been developed for the identification of cancer lesions during surgery. Among them, "activatable" fluorescence probes that react with cancer-specific biomarker enzymes to generate fluorescence signals have great potential for high-contrast cancer imaging due to their low background fluorescence and high signal amplification by enzymatic turnover. Over the past two decades, activatable fluorescence probes employing various fluorescence control mechanisms have been developed worldwide for this purpose. Furthermore, new biomarker enzymatic activities for specific types of cancers have been identified, enabling visualization of various types of cancers with high sensitivity and specificity. This Review focuses on recent advances in the design, function and characteristics of activatable fluorescence probes that target cancer-specific enzymatic activities for cancer imaging and also discusses future prospects in the field of activity-based diagnostics for cancer.
Collapse
|
22
|
Takahashi-Yamashiro K, Miyazono K. Tissue clearing method in visualization of cancer progression and metastasis. Ups J Med Sci 2024; 129:10634. [PMID: 38716075 PMCID: PMC11075440 DOI: 10.48101/ujms.v129.10634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 03/07/2024] [Indexed: 05/24/2024] Open
Abstract
Since various imaging modalities have been developed, cancer metastasis can be detected from an early stage. However, limitations still exist, especially in terms of spatial resolution. Tissue-clearing technology has emerged as a new imaging modality in cancer research, which has been developed and utilized for a long time mainly in neuroscience field. This method enables us to detect cancer metastatic foci with single-cell resolution at whole mouse body/organ level. On top of that, 3D images of cancer metastasis of whole mouse organs make it easy to understand their characteristics. Recently, further applications of tissue clearing methods were reported in combination with reporter systems, labeling, and machine learning. In this review, we would like to provide an overview of this technique and current applications in cancer research and discuss their potentials and limitations.
Collapse
Affiliation(s)
- Kei Takahashi-Yamashiro
- Department of Chemistry, Faculty of Science, University of Alberta, Edmonton, Alberta, Canada
| | - Kohei Miyazono
- Department of Applied Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Laboratory for Cancer Invasion and Metastasis, Institute for Medical Sciences, RIKEN, Yokohama City, Kanagawa, Japan
| |
Collapse
|
23
|
Zhang K, Abdoli N, Gilley P, Sadri Y, Chen X, Thai TC, Dockery L, Moore K, Mannel RS, Qiu Y. Developing a novel image marker to predict the clinical outcome of neoadjuvant chemotherapy (NACT) for ovarian cancer patients. Comput Biol Med 2024; 172:108240. [PMID: 38460312 DOI: 10.1016/j.compbiomed.2024.108240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 02/13/2024] [Accepted: 02/26/2024] [Indexed: 03/11/2024]
Abstract
OBJECTIVE Neoadjuvant chemotherapy (NACT) is one kind of treatment for advanced stage ovarian cancer patients. However, due to the nature of tumor heterogeneity, the clinical outcomes to NACT vary significantly among different subgroups. Partial responses to NACT may lead to suboptimal debulking surgery, which will result in adverse prognosis. To address this clinical challenge, the purpose of this study is to develop a novel image marker to achieve high accuracy prognosis prediction of NACT at an early stage. METHODS For this purpose, we first computed a total of 1373 radiomics features to quantify the tumor characteristics, which can be grouped into three categories: geometric, intensity, and texture features. Second, all these features were optimized by principal component analysis algorithm to generate a compact and informative feature cluster. This cluster was used as input for developing and optimizing support vector machine (SVM) based classifiers, which indicated the likelihood of receiving suboptimal cytoreduction after the NACT treatment. Two different kernels for SVM algorithm were explored and compared. A total of 42 ovarian cancer cases were retrospectively collected to validate the scheme. A nested leave-one-out cross-validation framework was adopted for model performance assessment. RESULTS The results demonstrated that the model with a Gaussian radial basis function kernel SVM yielded an AUC (area under the ROC [receiver characteristic operation] curve) of 0.806 ± 0.078. Meanwhile, this model achieved overall accuracy (ACC) of 83.3%, positive predictive value (PPV) of 81.8%, and negative predictive value (NPV) of 83.9%. CONCLUSION This study provides meaningful information for the development of radiomics based image markers in NACT treatment outcome prediction.
Collapse
Affiliation(s)
- Ke Zhang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA, 73019; School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK, USA, 73019
| | - Neman Abdoli
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK, USA, 73019
| | - Patrik Gilley
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK, USA, 73019
| | - Youkabed Sadri
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK, USA, 73019
| | - Xuxin Chen
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK, USA, 73019
| | - Theresa C Thai
- Department of Radiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA, 73104
| | - Lauren Dockery
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA, 73104
| | - Kathleen Moore
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA, 73104
| | - Robert S Mannel
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA, 73104
| | - Yuchen Qiu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA, 73019; School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK, USA, 73019.
| |
Collapse
|
24
|
Xu Y, Liang H, Zeng Q, He F, Liu C, Gai S, Ding H, Yang P. A bubble-enhanced lanthanide-doped up/down-conversion platform with tumor microenvironment response for dual-modal photoacoustic and near-infrared-II fluorescence imaging. J Colloid Interface Sci 2024; 659:149-159. [PMID: 38159491 DOI: 10.1016/j.jcis.2023.12.088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024]
Abstract
As an important tumor diagnosis strategy in precision medicine, multimodal imaging has been widely studied. However, the weak imaging signal with low spatial resolution and the constant signal of lack of specific activation severely limit its disease diagnosis. Herein, a bubble-enhanced lanthanide-based up/down-conversion platform with tumor microenvironment response for dual-mode imaging, LDNP@DMSN-Au@CaCO3 nanoparticles (named as LDAC NPs) were successfully developed. Combining the advantages of photoacoustic imaging (PAI) and the second near-infrared window (NIR-II) fluorescence imaging (FI), significantly improved the accuracy of diseases diagnosis. LDAC NPs with flower-like structure were synthesized through the encapsulation of uniform lanthanide-doped nanoparticles (NaYbF4:Ce,Er@NaYF4 named LDNPs) with dendritic mesoporous silica (DMSN). The gold nanoparticles (Au NPs) were then in situ grown on the surface of DMSN and the surface were finally coated with a layer of calcium carbonate (CaCO3). Under the excitation of the 980 nm laser, LDNPs showed strong emission of NIR-II at 1550 nm due to the doping of Ce and Er ions, showcasing excellent spatial resolution and deep tissue penetration characteristics, while the resulting visible light emission (540 nm) enables Au NPs to generate PAI signals with the aid of LDNPs via the fluorescence resonance energy transfer effect. In acidic tumoral environment, CaCO3 layer could produce CO2 microbubbles, and the PAI signals of LDAC NPs could be further enhanced with the generation of CO2 bubbles due to the bubble cavitation effect. Simultaneously, the NIR-II FI of LDAC NPs was self-enhanced with the degradation of the CaCO3. This intelligent nanoparticle with stimulus-activated dual-mode imaging capability holds great promise in future precision diagnostics.
Collapse
Affiliation(s)
- Yuening Xu
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin 150001, PR China
| | - Haoran Liang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin 150001, PR China
| | - Qingtan Zeng
- Changhai Hospital Affiliated to Navy Military Medical University, Shanghai, PR China
| | - Fei He
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin 150001, PR China.
| | - Changlin Liu
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin 150001, PR China
| | - Shili Gai
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin 150001, PR China
| | - He Ding
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin 150001, PR China.
| | - Piaoping Yang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin 150001, PR China.
| |
Collapse
|
25
|
Saladino GM, Brodin B, Kakadiya R, Toprak MS, Hertz HM. Iterative nanoparticle bioengineering enabled by x-ray fluorescence imaging. SCIENCE ADVANCES 2024; 10:eadl2267. [PMID: 38517973 DOI: 10.1126/sciadv.adl2267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 02/16/2024] [Indexed: 03/24/2024]
Abstract
Nanoparticles (NPs) are currently developed for drug delivery and molecular imaging. However, they often get intercepted before reaching their target, leading to low targeting efficacy and signal-to-noise ratio. They tend to accumulate in organs like lungs, liver, kidneys, and spleen. The remedy is to iteratively engineer NP surface properties and administration strategies, presently a time-consuming process that includes organ dissection at different time points. To improve this, we propose a rapid iterative approach using whole-animal x-ray fluorescence (XRF) imaging to systematically evaluate NP distribution in vivo. We applied this method to molybdenum-based NPs and clodronate liposomes for tumor targeting with transient macrophage depletion, leading to reduced accumulations in lungs and liver and eventual tumor detection. XRF computed tomography (XFCT) provided 3D insight into NP distribution within the tumor. We validated the results using a multiscale imaging approach with dye-doped NPs and gene expression analysis for nanotoxicological profiling. XRF imaging holds potential for advancing therapeutics and diagnostics in preclinical pharmacokinetic studies.
Collapse
Affiliation(s)
- Giovanni M Saladino
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691, Stockholm, Sweden
| | - Bertha Brodin
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691, Stockholm, Sweden
| | - Ronak Kakadiya
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691, Stockholm, Sweden
| | - Muhammet S Toprak
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691, Stockholm, Sweden
| | - Hans M Hertz
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691, Stockholm, Sweden
| |
Collapse
|
26
|
Tandon R, Agrawal S, Rathore NPS, Mishra AK, Jain SK. A systematic review on deep learning-based automated cancer diagnosis models. J Cell Mol Med 2024; 28:e18144. [PMID: 38426930 PMCID: PMC10906380 DOI: 10.1111/jcmm.18144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 12/08/2023] [Accepted: 01/16/2024] [Indexed: 03/02/2024] Open
Abstract
Deep learning is gaining importance due to its wide range of applications. Many researchers have utilized deep learning (DL) models for the automated diagnosis of cancer patients. This paper provides a systematic review of DL models for automated diagnosis of cancer patients. Initially, various DL models for cancer diagnosis are presented. Five major categories of cancers such as breast, lung, liver, brain and cervical cancer are considered. As these categories of cancers have a very high percentage of occurrences with high mortality rate. The comparative analysis of different types of DL models is drawn for the diagnosis of cancer at early stages by considering the latest research articles from 2016 to 2022. After comprehensive comparative analysis, it is found that most of the researchers achieved appreciable accuracy with implementation of the convolutional neural network model. These utilized the pretrained models for automated diagnosis of cancer patients. Various shortcomings with the existing DL-based automated cancer diagnosis models are also been presented. Finally, future directions are discussed to facilitate further research for automated diagnosis of cancer patients.
Collapse
Affiliation(s)
| | | | | | - Abhinava K. Mishra
- Molecular, Cellular and Developmental Biology DepartmentUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| | | |
Collapse
|
27
|
Nguyen TNH, Horowitz L, Krilov T, Lockhart E, Kenerson HL, Yeung RS, Arroyo-Currás N, Folch A. Label-Free, Real-Time Monitoring of Cytochrome C Responses to Drugs in Microdissected Tumor Biopsies with a Multi-Well Aptasensor Platform. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578278. [PMID: 38352494 PMCID: PMC10862797 DOI: 10.1101/2024.01.31.578278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Functional assays on intact tumor biopsies can potentially complement and extend genomics-based approaches for precision oncology, drug testing, and organs-on-chips cancer disease models by capturing key determinants of therapeutic response, such as tissue architecture, tumor heterogeneity, and the tumor microenvironment. Currently, most of these assays rely on fluorescent labeling, a semi-quantitative method best suited to be a single-time-point terminal assay or labor-intensive terminal immunostaining analysis. Here, we report integrated aptamer electrochemical sensors for on-chip, real-time monitoring of increases of cytochrome C, a cell death indicator, from intact microdissected tissues with high affinity and specificity. The platform features a multi-well sensor layout and a multiplexed electronic setup. The aptasensors measure increases in cytochrome C in the supernatant of mouse or human microdissected tumors after exposure to various drug treatments. Since the aptamer probe can be easily exchanged to recognize different targets, the platform could be adapted for multiplexed monitoring of various biomarkers, providing critical information on the tumor and its microenvironment. This approach could not only help develop more advanced cancer disease models but also apply to other complex in vitro disease models, such as organs-on-chips and organoids.
Collapse
|
28
|
Zhang DG, Pan YJ, Chen BQ, Lu XC, Xu QX, Wang P, Kankala RK, Jiang NN, Wang SB, Chen AZ. Protein-guided biomimetic nanomaterials: a versatile theranostic nanoplatform for biomedical applications. NANOSCALE 2024; 16:1633-1649. [PMID: 38168813 DOI: 10.1039/d3nr05495k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Over the years, bioinspired mineralization-based approaches have been applied to synthesize multifunctional organic-inorganic nanocomposites. These nanocomposites can address the growing demands of modern biomedical applications. Proteins, serving as vital biological templates, play a pivotal role in the nucleation and growth processes of various organic-inorganic nanocomposites. Protein-mineralized nanomaterials (PMNMs) have attracted significant interest from researchers due to their facile and convenient preparation, strong physiological activity, stability, impressive biocompatibility, and biodegradability. Nevertheless, few comprehensive reviews have expounded on the progress of these nanomaterials in biomedicine. This article systematically reviews the principles and strategies for constructing nanomaterials using protein-directed biomineralization and biomimetic mineralization techniques. Subsequently, we focus on their recent applications in the biomedical field, encompassing areas such as bioimaging, as well as anti-tumor, anti-bacterial, and anti-inflammatory therapies. Furthermore, we discuss the challenges encountered in practical applications of these materials and explore their potential in future applications. This review aspired to catalyze the continued development of these bioinspired nanomaterials in drug development and clinical diagnosis, ultimately contributing to the fields of precision medicine and translational medicine.
Collapse
Affiliation(s)
- Da-Gui Zhang
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Yu-Jing Pan
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Biao-Qi Chen
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Xiao-Chang Lu
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Qin-Xi Xu
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Pei Wang
- Jiangxi Provincial Key Laboratory of Oral Biomedicine, Jiangxi Province Clinical Research Center for Oral Diseases, School of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Ranjith Kumar Kankala
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Ni-Na Jiang
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Shi-Bin Wang
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Ai-Zheng Chen
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| |
Collapse
|
29
|
Feng P, Xue X, Bukhari I, Qiu C, Li Y, Zheng P, Mi Y. Gut microbiota and its therapeutic implications in tumor microenvironment interactions. Front Microbiol 2024; 15:1287077. [PMID: 38322318 PMCID: PMC10844568 DOI: 10.3389/fmicb.2024.1287077] [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: 09/01/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024] Open
Abstract
The development of cancer is not just the growth and proliferation of a single transformed cell, but its tumor microenvironment (TME) also coevolves with it, which is primarily involved in tumor initiation, development, metastasis, and therapeutic responses. Recent years, TME has been emerged as a potential target for cancer diagnosis and treatment. However, the clinical efficacy of treatments targeting the TME, especially its specific components, remains insufficient. In parallel, the gut microbiome is an essential TME component that is crucial in cancer immunotherapy. Thus, assessing and constructing frameworks between the gut microbiota and the TME can significantly enhance the exploration of effective treatment strategies for various tumors. In this review the role of the gut microbiota in human cancers, including its function and relationship with various tumors was summarized. In addition, the interaction between the gut microbiota and the TME as well as its potential applications in cancer therapeutics was described. Furthermore, it was summarized that fecal microbiota transplantation, dietary adjustments, and synthetic biology to introduce gut microbiota-based medical technologies for cancer treatment. This review provides a comprehensive summary for uncovering the mechanism underlying the effects of the gut microbiota on the TME and lays a foundation for the development of personalized medicine in further studies.
Collapse
Affiliation(s)
- Pengya Feng
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Children Rehabilitation Medicine, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xia Xue
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ihtisham Bukhari
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunjing Qiu
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingying Li
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengyuan Zheng
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Mi
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
30
|
Skrodzki D, Molinaro M, Brown R, Moitra P, Pan D. Synthesis and Bioapplication of Emerging Nanomaterials of Hafnium. ACS NANO 2024; 18:1289-1324. [PMID: 38166377 DOI: 10.1021/acsnano.3c08917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
A significant amount of progress in nanotechnology has been made due to the development of engineered nanoparticles. The use of metallic nanoparticles for various biomedical applications has been extensively investigated. Biomedical research is highly focused on them because of their inert nature, nanoscale structure, and similar size to many biological molecules. The intrinsic characteristics of these particles, including electronic, optical, physicochemical, and surface plasmon resonance, that can be altered by altering their size, shape, environment, aspect ratio, ease of synthesis, and functionalization properties, have led to numerous biomedical applications. Targeted drug delivery, sensing, photothermal and photodynamic therapy, and imaging are some of these. The promising clinical results of NBTXR3, a high-Z radiosensitizing nanomaterial derived from hafnium, have demonstrated translational potential of this metal. This radiosensitization approach leverages the dependence of energy attenuation on atomic number to enhance energy-matter interactions conducive to radiation therapy. High-Z nanoparticle localization in tumor issue differentially increases the effect of ionizing radiation on cancer cells versus nearby healthy ones and mitigates adverse effects by reducing the overall radiation burden. This principle enables material multifunctionality as contrast agents in X-ray-based imaging. The physiochemical properties of hafnium (Z = 72) are particularly advantageous for these applications. A well-placed K-edge absorption energy and high mass attenuation coefficient compared to elements in human tissue across clinical energy ranges leads to significant attenuation. Chemical reactivity allows for variety in nanoparticle synthesis, composition, and functionalization. Nanoparticles such as hafnium oxide exhibit excellent biocompatibility due to physiochemical inertness prior to incidence with ionizing radiation. Additionally, the optical and electronic properties are applicable in biosensing, optical component coatings, and semiconductors. The wide interest has prompted extensive research in design and synthesis to facilitate property fine-tuning. This review summarizes synthetic methods for hafnium-based nanomaterials and applications in therapy, imaging, and biosensing with a mechanistic focus. A discussion and future perspective section highlights clinical progress and elaborates on current challenges. By focusing on factors impacting applicational effectiveness and examining limitations this review aims to support researchers and expedite clinical translation of future hafnium-based nanomedicine.
Collapse
Affiliation(s)
- David Skrodzki
- Department of Materials Science and Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Matthew Molinaro
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Richard Brown
- Department of Nuclear Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Parikshit Moitra
- Department of Nuclear Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Dipanjan Pan
- Department of Materials Science and Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Department of Nuclear Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Huck Institutes of the Life Sciences, 101 Huck Life Sciences Building, University Park, Pennsylvania 16802, United States
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
31
|
Evans SM, Ivanova K, Rome R, Cossio D, Pilgrim C, Zalcberg J, Antill Y, Blake L, Du Guesclin A, Garrett A, Giffard D, Golobic N, Moir D, Parikh S, Parisi A, Sanday K, Shadbolt C, Smith M, Te Marvelde L, Williams K. Registry-derived stage (RD-Stage) for capturing cancer stage at diagnosis for endometrial cancer. BMC Cancer 2023; 23:1222. [PMID: 38087227 PMCID: PMC10714535 DOI: 10.1186/s12885-023-11615-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Capture of cancer stage at diagnosis is important yet poorly reported by health services to population-based cancer registries. In this paper we describe current completeness of stage information for endometrial cancer available in Australian cancer registries; and develop and validate a set of rules to enable cancer registry medical coders to calculate stage using data available to them (registry-derived stage or 'RD-Stage'). METHODOLOGY Rules for deriving RD-stage (Endometrial carcinoma) were developed using the American Joint Commission on Cancer (AJCC) TNM (tumour, nodes, metastasis) Staging System (8th Edition). An expert working group comprising cancer specialists responsible for delivering cancer care, epidemiologists and medical coders reviewed and endorsed the rules. Baseline completeness of data fields required to calculate RD-Stage, and calculation of the proportion of cases for whom an RD stage could be assigned, was assessed across each Australian jurisdiction. RD-Stage (Endometrial cancer) was calculated by Victorian Cancer Registry (VCR) medical coders and compared with clinical stage recorded by the patient's treating clinician and captured in the National Gynae-Oncology Registry (NGOR). RESULTS The necessary data completeness level for calculating RD-Stage (Endometrial carcinoma) across various Australian jurisdictions varied from 0 to 89%. Three jurisdictions captured degree of spread of cancer, rendering RD-Stage unable to be calculated. RD-Stage (Endometrial carcinoma) could not be derived for 64/485 (13%) cases and was not captured for 44/485 (9%) cases in NGOR. At stage category level (I, II, III, IV), there was concordance between RD-Stage and NGOR captured stage in 393/410 (96%) of cases (95.8%, Kendall's coefficient = 0.95). CONCLUSION A lack of consistency in data captured by, and data sources reporting to, population-based cancer registries meant that it was not possible to provide national endometrial carcinoma stage data at diagnosis. In a sample of Victorian cases, where surgical pathology was available, there was very good concordance between RD-Stage (Endometrial carcinoma) and clinician-recorded stage data available from NGOR. RD-Stage offers promise in capturing endometrial cancer stage at diagnosis for population epidemiological purposes when it is not provided by health services, but requires more extensive validation.
Collapse
Affiliation(s)
- S M Evans
- Cancer Council Victoria, Melbourne, Australia.
| | - K Ivanova
- Cancer Council Victoria, Melbourne, Australia
| | - R Rome
- Epworth Health Care, Melbourne, Australia
| | - D Cossio
- Cancer Alliance Queensland, Woolloongabba, Australia
| | - Chc Pilgrim
- Central Clinical School, Department of Surgery, The Alfred, Monash University, Melbourne, Australia
| | - J Zalcberg
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Y Antill
- Monash University, Melbourne, Australia
| | - L Blake
- Cancer Council Victoria, Melbourne, Australia
| | - A Du Guesclin
- Department of Anatomical Pathology, The Alfred, Melbourne, Australia
| | - A Garrett
- Queensland Centre for Gynaecological Cancer, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - D Giffard
- Cancer Alliance Queensland, Woolloongabba, Australia
| | - N Golobic
- Cancer Alliance Queensland, Woolloongabba, Australia
| | - D Moir
- Department of Anatomical Pathology, The Alfred, Melbourne, Australia
| | - S Parikh
- Cancer Council Victoria, Melbourne, Australia
| | - A Parisi
- ACT Cancer Registry Australian Capital Territory Health, Deakin, Australia
| | - K Sanday
- Queensland Centre for Gynaecological Cancer, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - C Shadbolt
- Royal Women's Hospital, Melbourne, Australia
| | - M Smith
- ACT Cancer Registry Australian Capital Territory Health, Deakin, Australia
| | | | - K Williams
- Cancer Council Victoria, Melbourne, Australia
| |
Collapse
|
32
|
Pandey S, Choudhary P, Gajbhiye V, Jadhav S, Bodas D. In vivo imaging of prostate tumor-targeted folic acid conjugated quantum dots. Cancer Nanotechnol 2023. [DOI: 10.1186/s12645-023-00162-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
Abstract
AbstractCancer is a major threat to human health; thus, early detection is imperative for successful management. Rapid diagnosis can be achieved by imaging primary (subcutaneous) tumors using fluorophores conjugated with tumor markers. Here, the application of biocompatible, quantum efficient, monodisperse, and photostable polymer-coated quantum dots (PQDs) is demonstrated for targeted prostate tumor imaging in living SCID mice. Briefly, PQDs (blue) are conjugated to folic acid (FA-PQDs) using DCC-NHS chemistry. Initially, in vitro targeted imaging via FA-PQDs is evaluated in LNCaP cells. The confocal microscopic evaluation demonstrates the uptake of FA-PQDs. To understand the dispersion of PQDs in vivo, the biodistribution of PQDs is assessed at different time intervals (1- 180 min) using whole-body fluorescence imaging and computed tomography (CT) scan. PQDs are seen to accumulate in organs like the liver, kidneys, spleen, lungs, and urinary bladder within 60 min, however, PQDs are not observed at 180 min indicating renal clearance. Further, to target the prostate tumor (~ 200 mm3) in mice, FA-PQDs are injected intravenously, and whole-body fluorescence imaging along with a CT scan is recorded. FA-PQDs are seen at the tumor site as compared to PQDs. The results confirm that the FA-PQDs function as excellent nanoprobes for targeted tumor imaging in vivo.
Collapse
|
33
|
Hagos G, Hammad N, Stanway S, Yusuf A, Hailemariam T. Cancer care in Needle Hospital, Hargeisa, Somaliland. Ecancermedicalscience 2023; 17:1642. [PMID: 38414938 PMCID: PMC10898905 DOI: 10.3332/ecancer.2023.1642] [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: 07/18/2023] [Indexed: 02/29/2024] Open
Abstract
Somaliland is an autonomous region in the northern part of Somalia that declared its independence in 1991. It is a low-income country (LIC) with a population size of 5.7 million with a gross domestic product per capita of $775. Health services are delivered by public, private and non-governmental organisations. The public health care system in Somaliland is facing huge challenges. Seven percent of the population suffers from non-communicable diseases, but data on cancer incidence and mortality are not available. Much of the emphasis in public health has been placed on primary care and maternal and child health. There is still a large gap in cancer prevention, early detection and screening in the country. Additionally, there is no cancer registry or published data on cancer. Currently, there are a few private hospitals that provide chemotherapy services in Somaliland of which Needle Hospital is one. Services provided in this hospital include medical oncology for all solid tumours, palliative care, follow-up and cancer health education. The hospital provides services for patients from Somaliland and neighbouring countries including Djibouti, Somalia and Ethiopia. As a new oncology clinic in an LIC, the clinic is facing many challenges, like the absence of a multidisciplinary tumour board, presentation of patients at the advanced stage of tumours and poor cancer awareness in the general population.
Collapse
Affiliation(s)
| | - Nazik Hammad
- Saint Michael’s Hospital, University of Toronto, Toronto M5B 1W8, Canada
| | | | | | | |
Collapse
|
34
|
Fu D, Yang F, Zhang J, Xiang Z, Wang Y. Near-Infrared Rechargeable Persistent Luminescence Nanoparticles for Biomedical Implants In Vivo Noninvasive Bioimaging. ACS APPLIED MATERIALS & INTERFACES 2023; 15:53310-53317. [PMID: 37947316 DOI: 10.1021/acsami.3c12947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Luminescent imaging has garnered significant attention for in vivo tracking of biomedical implants during and after surgery due to its human friendliness, affordability, and high sensitivity. However, conventional fluorescent probes are susceptible to background autofluorescence interference from living tissues, often resulting in poor signal-to-noise ratios. Herein, we report a background interference-free persistent luminescent implant (PLI) with excellent persistent luminescence (PL) performance, which can be clearly and long-term detected by an optical imaging system after implantation. Rechargeable near-infrared persistent luminescence nanoparticles (PLNPs) were prepared first via a simple hydrothermal approach and then modified by PEGylation to improve their hydrophilicity, biocompatibility, and compatibility with polymer substrates. The PEGylated PLNPs were facilely complexed into a polymer matrix to fabricate the PLI. The obtained PLIs can well inherit the PL properties of PLNPs, exhibiting good PL optical imaging performance without tissue autofluorescence interference. Furthermore, both PLNPs and PLIs possess good biocompatibility, and the addition of PLNPs has no negative impact on the biocompatibility of the polymer matrix. This work fully utilizes the luminescent properties of PLNPs and adapts this PL to the field of biomedical implant imaging, which provides new insight for designing biomedical imaging systems.
Collapse
Affiliation(s)
- Daihua Fu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Fan Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Jiayi Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Zhen Xiang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| |
Collapse
|
35
|
Shams M, Abdallah S, Alsadoun L, Hamid YH, Gasim R, Hassan A. Oncological Horizons: The Synergy of Medical and Surgical Innovations in Cancer Treatment. Cureus 2023; 15:e49249. [PMID: 38143618 PMCID: PMC10743204 DOI: 10.7759/cureus.49249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2023] [Indexed: 12/26/2023] Open
Abstract
The landscape of cancer treatment has witnessed a remarkable transformation in recent years, marked by the convergence of medical and surgical innovations. Historically, cancer therapy faced challenges, including limited efficacy and severe side effects. This narrative review explores the historical progression of cancer treatments, shedding light on significant breakthroughs in both medical and surgical oncology. It comprehensively addresses the medical domain, covering chemotherapy, targeted therapies, immunotherapy, hormonal treatments, and radiological procedures. Simultaneously, it delves into the surgical realm, discussing the evolution of surgical techniques, minimally invasive procedures, and the role of surgery across various stages of cancer. The article emphasizes the fusion of medical and surgical approaches, highlighting neoadjuvant and adjuvant therapies and the significance of multidisciplinary tumor boards. It also addresses innovations, challenges, and the pivotal role of patient-centered care. Furthermore, it offers insights into the future directions and forecasts in the constantly evolving field of integrated oncological care. This review provides a comprehensive understanding of the dynamic and transformative nature of cancer treatment, reflecting the unwavering commitment of the medical and surgical communities in the ongoing fight against cancer.
Collapse
Affiliation(s)
| | | | - Lara Alsadoun
- Trauma and Orthopaedics, Chelsea and Westminster Hospital, London, GBR
| | - Yusra H Hamid
- Community Medicine, Faculty of Medicine, University of Khartoum, Khartoum, SDN
| | - Rayan Gasim
- Internal Medicine, University of Khartoum, Khartoum, SDN
| | | |
Collapse
|
36
|
Saborido-Moral JD, Fernández-Patón M, Tejedor-Aguilar N, Cristian-Marín A, Torres-Espallardo I, Campayo-Esteban JM, Pérez-Calatayud J, Baltas D, Martí-Bonmatí L, Carles M. Free automatic software for quality assurance of computed tomography calibration, edges and radiomics metrics reproducibility. Phys Med 2023; 114:103153. [PMID: 37778209 DOI: 10.1016/j.ejmp.2023.103153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/16/2023] [Accepted: 09/22/2023] [Indexed: 10/03/2023] Open
Abstract
PURPOSE To develop a QA procedure, easy to use, reproducible and based on open-source code, to automatically evaluate the stability of different metrics extracted from CT images: Hounsfield Unit (HU) calibration, edge characterization metrics (contrast and drop range) and radiomic features. METHODS The QA protocol was based on electron density phantom imaging. Home-made open-source Python code was developed for the automatic computation of the metrics and their reproducibility analysis. The impact on reproducibility was evaluated for different radiation therapy protocols, and phantom positions within the field of view and systems, in terms of variability (Shapiro-Wilk test for 15 repeated measurements carried out over three days) and comparability (Bland-Altman analysis and Wilcoxon Rank Sum Test or Kendall Rank Correlation Coefficient). RESULTS Regarding intrinsic variability, most metrics followed a normal distribution (88% of HU, 63% of edge parameters and 82% of radiomic features). Regarding comparability, HU and contrast were comparable in all conditions, and drop range only in the same CT scanner and phantom position. The percentages of comparable radiomic features independent of protocol, position and system were 59%, 78% and 54%, respectively. The non-significantly differences in HU calibration curves obtained for two different institutions (7%) translated in comparable Gamma Index G (1 mm, 1%, >99%). CONCLUSIONS An automated software to assess the reproducibility of different CT metrics was successfully created and validated. A QA routine proposal is suggested.
Collapse
Affiliation(s)
- Juan D Saborido-Moral
- La Fe Health Research Institute, Biomedical Imaging Research Group (GIBI230-PREBI) and Imaging La Fe Node at Distributed Network for Biomedical Imaging (ReDIB) Unique Scientific and Technical Infrastructures (ICTS), 46026 Valencia, Spain.
| | - Matías Fernández-Patón
- La Fe Health Research Institute, Biomedical Imaging Research Group (GIBI230-PREBI) and Imaging La Fe Node at Distributed Network for Biomedical Imaging (ReDIB) Unique Scientific and Technical Infrastructures (ICTS), 46026 Valencia, Spain
| | - Natalia Tejedor-Aguilar
- Department of Radiation Oncology, La Fe Polytechnic and University Hospital, Valencia, Spain
| | - Andrei Cristian-Marín
- Department of Radiation Protection, La Fe Polytechnic and University Hospital, Valencia, Spain
| | | | - Juan M Campayo-Esteban
- Department of Radiation Protection, La Fe Polytechnic and University Hospital, Valencia, Spain
| | - José Pérez-Calatayud
- Department of Radiation Oncology, La Fe Polytechnic and University Hospital, Valencia, Spain
| | - Dimos Baltas
- Division of Medical Physics, Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg, Heidelberg, Germany
| | - Luis Martí-Bonmatí
- La Fe Health Research Institute, Biomedical Imaging Research Group (GIBI230-PREBI) and Imaging La Fe Node at Distributed Network for Biomedical Imaging (ReDIB) Unique Scientific and Technical Infrastructures (ICTS), 46026 Valencia, Spain
| | - Montserrat Carles
- La Fe Health Research Institute, Biomedical Imaging Research Group (GIBI230-PREBI) and Imaging La Fe Node at Distributed Network for Biomedical Imaging (ReDIB) Unique Scientific and Technical Infrastructures (ICTS), 46026 Valencia, Spain
| |
Collapse
|
37
|
Chen X, Li Y, Zhou Z, Zhang Y, Chang L, Gao X, Li Q, Luo H, Westover KD, Zhu J, Wei X. Dynamic ultrasound molecular-targeted imaging of senescence in evaluation of lapatinib resistance in HER2-positive breast cancer. Cancer Med 2023; 12:19904-19920. [PMID: 37792675 PMCID: PMC10587953 DOI: 10.1002/cam4.6607] [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: 03/24/2023] [Revised: 07/21/2023] [Accepted: 09/22/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Prolonged treatment of HER2+ breast cancer with lapatinib (LAP) causes cellular senescence and acquired drug resistance, which often associating with poor prognosis for patients. We aim to explore the correlation between cellular senescence and LAP resistance in HER2+ breast cancer, screen for molecular marker of reversible senescence, and construct targeted nanobubbles for ultrasound molecular imaging to dynamically evaluate LAP resistance. METHODS AND RESULTS In this study, we established a new cellular model of reversible cellular senescence using LAP and HER2+ breast cancer cells and found that reversible senescence contributed to LAP resistance in HER2+ breast cancer. Then, we identified ecto-5'-nucleotidase (NT5E) as a marker of reversible senescence in HER2+ breast cancer. Based on this, we constructed NT5E-targeted nanobubbles (NT5E-FITC-NBs) as a new molecular imaging modality which could both target reversible senescent cells and be used for ultrasound imaging. NT5E-FITC-NBs showed excellent physical and imaging characteristics. As an ultrasound contrast agent, NT5E-FITC-NBs could accurately identify reversible senescent cells both in vitro and in vivo. CONCLUSIONS Our data demonstrate that cellular senescence-based ultrasound-targeted imaging can identify reversible senescence and evaluate LAP resistance effectively in HER2+ breast cancer cells, which has the potential to improve cancer treatment outcomes by altering therapeutic strategies ahead of aggressive recurrences.
Collapse
Affiliation(s)
- Xiaoyu Chen
- Department of Diagnostic and Therapeutic UltrasonographyTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for CancerTianjinChina
- Department of UltrasoundTianjin HospitalTianjinChina
| | - Ying Li
- Breast Cancer CenterTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Zhiwei Zhou
- Department of Radiation Oncology and BiochemistryUniversity of Texas Southwestern Medical CenterTexasDallasUSA
| | - Yanqiu Zhang
- Department of Diagnostic and Therapeutic UltrasonographyTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Luchen Chang
- Department of Diagnostic and Therapeutic UltrasonographyTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Xiujun Gao
- School of Biomedical Engineering and Technology, Tianjin Medical UniversityTianjinChina
| | - Qing Li
- Cancer CenterDaping Hospital, Third Military Medical UniversityChongqingChina
| | - Hao Luo
- Cancer CenterDaping Hospital, Third Military Medical UniversityChongqingChina
| | - Kenneth D. Westover
- Department of Radiation Oncology and BiochemistryUniversity of Texas Southwestern Medical CenterTexasDallasUSA
| | - Jialin Zhu
- Department of Diagnostic and Therapeutic UltrasonographyTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Xi Wei
- Department of Diagnostic and Therapeutic UltrasonographyTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for CancerTianjinChina
| |
Collapse
|
38
|
Albahli S, Nazir T. A Circular Box-Based Deep Learning Model for the Identification of Signet Ring Cells from Histopathological Images. Bioengineering (Basel) 2023; 10:1147. [PMID: 37892876 PMCID: PMC10604551 DOI: 10.3390/bioengineering10101147] [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: 08/15/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 10/29/2023] Open
Abstract
Signet ring cell (SRC) carcinoma is a particularly serious type of cancer that is a leading cause of death all over the world. SRC carcinoma has a more deceptive onset than other carcinomas and is mostly encountered in its later stages. Thus, the recognition of SRCs at their initial stages is a challenge because of different variants and sizes and illumination changes. The recognition process of SRCs at their early stages is costly because of the requirement for medical experts. A timely diagnosis is important because the level of the disease determines the severity, cure, and survival rate of victims. To tackle the current challenges, a deep learning (DL)-based methodology is proposed in this paper, i.e., custom CircleNet with ResNet-34 for SRC recognition and classification. We chose this method because of the circular shapes of SRCs and achieved better performance due to the CircleNet method. We utilized a challenging dataset for experimentation and performed augmentation to increase the dataset samples. The experiments were conducted using 35,000 images and attained 96.40% accuracy. We performed a comparative analysis and confirmed that our method outperforms the other methods.
Collapse
Affiliation(s)
- Saleh Albahli
- Department of Information Technology, College of Computer, Qassim University, Buraydah 51452, Saudi Arabia;
| | - Tahira Nazir
- Faculty of Computing, Riphah International University, Islamabad 44600, Pakistan
| |
Collapse
|
39
|
Najafi S, Mortezaee K. Advances in dendritic cell vaccination therapy of cancer. Biomed Pharmacother 2023; 164:114954. [PMID: 37257227 DOI: 10.1016/j.biopha.2023.114954] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/16/2023] [Accepted: 05/27/2023] [Indexed: 06/02/2023] Open
Abstract
Traditionally, vaccines have helped eradication of several infectious diseases and also saved millions of lives in the human history. Those prophylactic vaccines have acted through inducing immune responses against a live attenuated, killed organism or antigenic subunits to protect the recipient against a real infection caused by the pathogenic microorganism. Nevertheless, development of anticancer vaccines as valuable targets in human health has faced challenges and requires further optimizations. Dendritic cells (DCs) are the most potent antigen presenting cells (APCs) that play essential roles in tumor immunotherapies through induction of CD8+ T cell immunity. Accordingly, various strategies have been tested to employ DCs as therapeutic vaccines for exploiting their activity against tumor cells. Application of whole tumor cells or purified/recombinant antigen peptides are the most common approaches for pulsing DCs, which then are injected back into the patients. Although some hopeful results are reported for a number of DC vaccines tested in animal and clinical trials of cancer patients, such approaches are still inefficient and require optimization. Failure of DC vaccination is postulated due to immunosuppressive tumor microenvironment (TME), overexpression of checkpoint proteins, suboptimal avidity of tumor-associated antigen (TAA)-specific T lymphocytes, and lack of appropriate adjuvants. In this review, we have an overview of the current experiments and trials evaluated the anticancer efficacy of DC vaccination as well as focusing on strategies to improve their potential including combination therapy with immune checkpoint inhibitors (ICIs).
Collapse
Affiliation(s)
- Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
40
|
Qin J, Guo N, Yang J, Chen Y. Recent Advances of Metal-Polyphenol Coordination Polymers for Biomedical Applications. BIOSENSORS 2023; 13:776. [PMID: 37622862 PMCID: PMC10452320 DOI: 10.3390/bios13080776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/24/2023] [Accepted: 07/28/2023] [Indexed: 08/26/2023]
Abstract
Nanomedicine has provided cutting-edge technologies and innovative methods for modern biomedical research, offering unprecedented opportunities to tackle crucial biomedical issues. Nanomaterials with unique structures and properties can integrate multiple functions to achieve more precise diagnosis and treatment, making up for the shortcomings of traditional treatment methods. Among them, metal-polyphenol coordination polymers (MPCPs), composed of metal ions and phenolic ligands, are considered as ideal nanoplatforms for disease diagnosis and treatment. Recently, MPCPs have been extensively investigated in the field of biomedicine due to their facile synthesis, adjustable structures, and excellent biocompatibility, as well as pH-responsiveness. In this review, the classification of various MPCPs and their fabrication strategies are firstly summarized. Then, their significant achievements in the biomedical field such as biosensing, drug delivery, bioimaging, tumor therapy, and antibacterial applications are highlighted. Finally, the main limitations and outlooks regarding MPCPs are discussed.
Collapse
Affiliation(s)
- Jing Qin
- College of Advanced Materials Engineering, Jiaxing Nanhu University, Jiaxing 314001, China; (N.G.); (J.Y.); (Y.C.)
| | | | | | | |
Collapse
|
41
|
Papachristou N, Kotronoulas G, Dikaios N, Allison SJ, Eleftherochorinou H, Rai T, Kunz H, Barnaghi P, Miaskowski C, Bamidis PD. Digital Transformation of Cancer Care in the Era of Big Data, Artificial Intelligence and Data-Driven Interventions: Navigating the Field. Semin Oncol Nurs 2023; 39:151433. [PMID: 37137770 DOI: 10.1016/j.soncn.2023.151433] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 05/05/2023]
Abstract
OBJECTIVES To navigate the field of digital cancer care and define and discuss key aspects and applications of big data analytics, artificial intelligence (AI), and data-driven interventions. DATA SOURCES Peer-reviewed scientific publications and expert opinion. CONCLUSION The digital transformation of cancer care, enabled by big data analytics, AI, and data-driven interventions, presents a significant opportunity to revolutionize the field. An increased understanding of the lifecycle and ethics of data-driven interventions will enhance development of innovative and applicable products to advance digital cancer care services. IMPLICATIONS FOR NURSING PRACTICE As digital technologies become integrated into cancer care, nurse practitioners and scientists will be required to increase their knowledge and skills to effectively use these tools to the patient's benefit. An enhanced understanding of the core concepts of AI and big data, confident use of digital health platforms, and ability to interpret the outputs of data-driven interventions are key competencies. Nurses in oncology will play a crucial role in patient education around big data and AI, with a focus on addressing any arising questions, concerns, or misconceptions to foster trust in these technologies. Successful integration of data-driven innovations into oncology nursing practice will empower practitioners to deliver more personalized, effective, and evidence-based care.
Collapse
Affiliation(s)
- Nikolaos Papachristou
- Medical Physics and Digital Innovation Laboratory, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | | | - Nikolaos Dikaios
- Centre for Vision Speech and Signal Processing, University of Surrey, Guildford, UK; Mathematics Research Centre, Academy of Athens, Athens, Greece
| | - Sarah J Allison
- Department of Sport, Exercise and Rehabilitation, Faculty of Health and Life Sciences, Northumbria University, Newcastle, UK; School of Bioscience and Medicine, Faculty of Health & Medical Sciences, University of Surrey, Guildford, UK
| | | | - Taranpreet Rai
- Centre for Vision Speech and Signal Processing, University of Surrey, Guildford, UK; Datalab, The Veterinary Health Innovation Engine (vHive), Guildford, UK
| | - Holger Kunz
- Institute of Health Informatics, University College London, London, UK
| | - Payam Barnaghi
- UK Dementia Research Institute Care Research and Technology Centre, Imperial College London, London, UK
| | - Christine Miaskowski
- School of Nursing, University California San Francisco, San Francisco, California, USA
| | - Panagiotis D Bamidis
- Medical Physics and Digital Innovation Laboratory, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
42
|
Qin W, Chandra J, Abourehab MAS, Gupta N, Chen ZS, Kesharwani P, Cao HL. New opportunities for RGD-engineered metal nanoparticles in cancer. Mol Cancer 2023; 22:87. [PMID: 37226188 DOI: 10.1186/s12943-023-01784-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/26/2023] [Indexed: 05/26/2023] Open
Abstract
The advent of nanotechnology has opened new possibilities for bioimaging. Metal nanoparticles (such as gold, silver, iron, copper, etc.) hold tremendous potential and offer enormous opportunities for imaging and diagnostics due to their broad optical characteristics, ease of manufacturing technique, and simple surface modification. The arginine-glycine-aspartate (RGD) peptide is a three-amino acid sequence that seems to have a considerably greater ability to adhere to integrin adhesion molecules that exclusively express on tumour cells. RGD peptides act as the efficient tailoring ligand with a variety of benefits including non-toxicity, greater precision, rapid clearance, etc. This review focuses on the possibility of non-invasive cancer imaging using metal nanoparticles with RGD assistance.
Collapse
Affiliation(s)
- Wei Qin
- Xi'an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, College of Pharmacy, Xi'an Medical University, Xi'an, 710021, China
| | - Jyoti Chandra
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Neelima Gupta
- Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Zhe-Sheng Chen
- Institute for Biotechnology, College of Pharmacy and Health Sciences, St. John's University, Queens, New York, 11439, USA
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical science, Chennai, India.
| | - Hui-Ling Cao
- Xi'an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, College of Pharmacy, Xi'an Medical University, Xi'an, 710021, China.
| |
Collapse
|
43
|
Crintea A, Motofelea AC, Șovrea AS, Constantin AM, Crivii CB, Carpa R, Duțu AG. Dendrimers: Advancements and Potential Applications in Cancer Diagnosis and Treatment-An Overview. Pharmaceutics 2023; 15:pharmaceutics15051406. [PMID: 37242648 DOI: 10.3390/pharmaceutics15051406] [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: 03/05/2023] [Revised: 04/17/2023] [Accepted: 04/29/2023] [Indexed: 05/28/2023] Open
Abstract
Cancer is a leading cause of death worldwide, and the main treatment methods for this condition are surgery, chemotherapy, and radiotherapy. These treatment methods are invasive and can cause severe adverse reactions among organisms, so nanomaterials are increasingly used as structures for anticancer therapies. Dendrimers are a type of nanomaterial with unique properties, and their production can be controlled to obtain compounds with the desired characteristics. These polymeric molecules are used in cancer diagnosis and treatment through the targeted distribution of some pharmacological substances. Dendrimers have the ability to fulfill several objectives in anticancer therapy simultaneously, such as targeting tumor cells so that healthy tissue is not affected, controlling the release of anticancer agents in the tumor microenvironment, and combining anticancer strategies based on the administration of anticancer molecules to potentiate their effect through photothermal therapy or photodynamic therapy. The purpose of this review is to summarize and highlight the possible uses of dendrimers regarding the diagnosis and treatment of oncological conditions.
Collapse
Affiliation(s)
- Andreea Crintea
- Department of Molecular Sciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Alexandru Cătălin Motofelea
- Department of Internal Medicine, Faculty of Medicine, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Alina Simona Șovrea
- Department of Morphological Sciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400000 Cluj-Napoca, Romania
| | - Anne-Marie Constantin
- Department of Morphological Sciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400000 Cluj-Napoca, Romania
| | - Carmen-Bianca Crivii
- Department of Morphological Sciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400000 Cluj-Napoca, Romania
| | - Rahela Carpa
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Institute for Research-Development-Innovation in Applied Natural Sciences, Babeș-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Alina Gabriela Duțu
- Department of Molecular Sciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| |
Collapse
|
44
|
Xu X, Liu A, Liu S, Ma Y, Zhang X, Zhang M, Zhao J, Sun S, Sun X. Application of molecular dynamics simulation in self-assembled cancer nanomedicine. Biomater Res 2023; 27:39. [PMID: 37143168 PMCID: PMC10161522 DOI: 10.1186/s40824-023-00386-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 04/21/2023] [Indexed: 05/06/2023] Open
Abstract
Self-assembled nanomedicine holds great potential in cancer theragnostic. The structures and dynamics of nanomedicine can be affected by a variety of non-covalent interactions, so it is essential to ensure the self-assembly process at atomic level. Molecular dynamics (MD) simulation is a key technology to link microcosm and macroscale. Along with the rapid development of computational power and simulation methods, scientists could simulate the specific process of intermolecular interactions. Thus, some experimental observations could be explained at microscopic level and the nanomedicine synthesis process would have traces to follow. This review not only outlines the concept, basic principle, and the parameter setting of MD simulation, but also highlights the recent progress in MD simulation for self-assembled cancer nanomedicine. In addition, the physicochemical parameters of self-assembly structure and interaction between various assembled molecules under MD simulation are also discussed. Therefore, this review will help advanced and novice researchers to quickly zoom in on fundamental information and gather some thought-provoking ideas to advance this subfield of self-assembled cancer nanomedicine.
Collapse
Affiliation(s)
- Xueli Xu
- School of Science, Shandong Jianzhu University, Jinan, 250101, China
| | - Ao Liu
- School of Science, Shandong Jianzhu University, Jinan, 250101, China
| | - Shuangqing Liu
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Yanling Ma
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Xinyu Zhang
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Meng Zhang
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Jinhua Zhao
- School of Science, Shandong Jianzhu University, Jinan, 250101, China
| | - Shuo Sun
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, 02115, USA
| | - Xiao Sun
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China.
| |
Collapse
|
45
|
Yan SY, Yang YW, Jiang XY, Hu S, Su YY, Yao H, Hu CH. Fat quantification: Imaging methods and clinical applications in cancer. Eur J Radiol 2023; 164:110851. [PMID: 37148843 DOI: 10.1016/j.ejrad.2023.110851] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 05/08/2023]
Abstract
Recently, the study of the relationship between lipid metabolism and cancer has evolved. The characteristics of intratumoral and peritumoral fat are distinct and changeable during cancer development. Subcutaneous and visceral adipose tissue are also associated with cancer prognosis. In non-invasive imaging, fat quantification parameters such as controlled attenuation parameter, fat volume fraction, and proton density fat fraction from different imaging methods complement conventional images by providing concrete fat information. Therefore, measuring the changes of fat content for further understanding of cancer characteristics has been applied in both research and clinical settings. In this review, the authors summarize imaging advances in fat quantification and highlight their clinical applications in cancer precaution, auxiliary diagnosis and classification, therapy response monitoring, and prognosis.
Collapse
Affiliation(s)
- Suo Yu Yan
- Department of Radiology, The First Affiliated Hospital to Soochow University, Suzhou 215006, PR China
| | - Yi Wen Yang
- Department of Radiology, The First Affiliated Hospital to Soochow University, Suzhou 215006, PR China
| | - Xin Yu Jiang
- Department of Radiology, The First Affiliated Hospital to Soochow University, Suzhou 215006, PR China
| | - Su Hu
- Department of Radiology, The First Affiliated Hospital to Soochow University, Suzhou 215006, PR China
| | - Yun Yan Su
- Department of Radiology, The First Affiliated Hospital to Soochow University, Suzhou 215006, PR China.
| | - Hui Yao
- Department of Radiology, The First Affiliated Hospital to Soochow University, Suzhou 215006, PR China; Department of General Surgery, The First Affiliated Hospital to Soochow University, Suzhou 215006, PR China.
| | - Chun Hong Hu
- Department of Radiology, The First Affiliated Hospital to Soochow University, Suzhou 215006, PR China.
| |
Collapse
|
46
|
Steyaert S, Pizurica M, Nagaraj D, Khandelwal P, Hernandez-Boussard T, Gentles AJ, Gevaert O. Multimodal data fusion for cancer biomarker discovery with deep learning. NAT MACH INTELL 2023; 5:351-362. [PMID: 37693852 PMCID: PMC10484010 DOI: 10.1038/s42256-023-00633-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 02/17/2023] [Indexed: 09/12/2023]
Abstract
Technological advances now make it possible to study a patient from multiple angles with high-dimensional, high-throughput multi-scale biomedical data. In oncology, massive amounts of data are being generated ranging from molecular, histopathology, radiology to clinical records. The introduction of deep learning has significantly advanced the analysis of biomedical data. However, most approaches focus on single data modalities leading to slow progress in methods to integrate complementary data types. Development of effective multimodal fusion approaches is becoming increasingly important as a single modality might not be consistent and sufficient to capture the heterogeneity of complex diseases to tailor medical care and improve personalised medicine. Many initiatives now focus on integrating these disparate modalities to unravel the biological processes involved in multifactorial diseases such as cancer. However, many obstacles remain, including lack of usable data as well as methods for clinical validation and interpretation. Here, we cover these current challenges and reflect on opportunities through deep learning to tackle data sparsity and scarcity, multimodal interpretability, and standardisation of datasets.
Collapse
Affiliation(s)
- Sandra Steyaert
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
| | - Marija Pizurica
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
| | | | | | - Tina Hernandez-Boussard
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
- Department of Biomedical Data Science, Stanford University
| | - Andrew J Gentles
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
- Department of Biomedical Data Science, Stanford University
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University
- Department of Biomedical Data Science, Stanford University
| |
Collapse
|
47
|
German-Cortés J, Vilar-Hernández M, Rafael D, Abasolo I, Andrade F. Solid Lipid Nanoparticles: Multitasking Nano-Carriers for Cancer Treatment. Pharmaceutics 2023; 15:pharmaceutics15030831. [PMID: 36986692 PMCID: PMC10056426 DOI: 10.3390/pharmaceutics15030831] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/25/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Despite all the advances seen in recent years, the severe adverse effects and low specificity of conventional chemotherapy are still challenging problems regarding cancer treatment. Nanotechnology has helped to address these questions, making important contributions in the oncological field. The use of nanoparticles has allowed the improvement of the therapeutic index of several conventional drugs and facilitates the tumoral accumulation and intracellular delivery of complex biomolecules, such as genetic material. Among the wide range of nanotechnology-based drug delivery systems (nanoDDS), solid lipid nanoparticles (SLNs) have emerged as promising systems for delivering different types of cargo. Their solid lipid core, at room and body temperature, provides SLNs with higher stability than other formulations. Moreover, SLNs offer other important features, namely the possibility to perform active targeting, sustained and controlled release, and multifunctional therapy. Furthermore, with the possibility to use biocompatible and physiologic materials and easy scale-up and low-cost production methods, SLNs meet the principal requirements of an ideal nanoDDS. The present work aims to summarize the main aspects related to SLNs, including composition, production methods, and administration routes, as well as to show the most recent studies about the use of SLNs for cancer treatment.
Collapse
Affiliation(s)
- Júlia German-Cortés
- Drug Delivery & Targeting Group, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
| | - Mireia Vilar-Hernández
- Drug Delivery & Targeting Group, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
| | - Diana Rafael
- Drug Delivery & Targeting Group, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Functional Validation & Preclinical Research (FVPR), U20 ICTS Nanbiosis, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
- Correspondence: (D.R.); (I.A.); (F.A.)
| | - Ibane Abasolo
- Drug Delivery & Targeting Group, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Functional Validation & Preclinical Research (FVPR), U20 ICTS Nanbiosis, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
- Servei de Bioquímica, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain
- Correspondence: (D.R.); (I.A.); (F.A.)
| | - Fernanda Andrade
- Drug Delivery & Targeting Group, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departament de Farmàcia i Tecnologia Farmacèutica i Fisicoquímica, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain
- Correspondence: (D.R.); (I.A.); (F.A.)
| |
Collapse
|
48
|
Pourmadadi M, Rajabzadeh-Khosroshahi M, Eshaghi MM, Rahmani E, Motasadizadeh H, Arshad R, Rahdar A, Pandey S. TiO2-based nanocomposites for cancer diagnosis and therapy: A comprehensive review. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
49
|
Goel M, Mackeyev Y, Krishnan S. Radiolabeled nanomaterial for cancer diagnostics and therapeutics: principles and concepts. Cancer Nanotechnol 2023; 14:15. [PMID: 36865684 PMCID: PMC9968708 DOI: 10.1186/s12645-023-00165-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/13/2023] [Indexed: 03/01/2023] Open
Abstract
In the last three decades, radiopharmaceuticals have proven their effectiveness for cancer diagnosis and therapy. In parallel, the advances in nanotechnology have fueled a plethora of applications in biology and medicine. A convergence of these disciplines has emerged more recently with the advent of nanotechnology-aided radiopharmaceuticals. Capitalizing on the unique physical and functional properties of nanoparticles, radiolabeled nanomaterials or nano-radiopharmaceuticals have the potential to enhance imaging and therapy of human diseases. This article provides an overview of various radionuclides used in diagnostic, therapeutic, and theranostic applications, radionuclide production through different techniques, conventional radionuclide delivery systems, and advancements in the delivery systems for nanomaterials. The review also provides insights into fundamental concepts necessary to improve currently available radionuclide agents and formulate new nano-radiopharmaceuticals.
Collapse
Affiliation(s)
- Muskan Goel
- Amity School of Applied Sciences, Amity University, Gurugram, Haryana 122413 India
| | - Yuri Mackeyev
- Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center, Houston, TX 77030 USA
| | - Sunil Krishnan
- Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center, Houston, TX 77030 USA
| |
Collapse
|
50
|
Vladimirov N, Perlman O. Molecular MRI-Based Monitoring of Cancer Immunotherapy Treatment Response. Int J Mol Sci 2023; 24:3151. [PMID: 36834563 PMCID: PMC9959624 DOI: 10.3390/ijms24043151] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Immunotherapy constitutes a paradigm shift in cancer treatment. Its FDA approval for several indications has yielded improved prognosis for cases where traditional therapy has shown limited efficiency. However, many patients still fail to benefit from this treatment modality, and the exact mechanisms responsible for tumor response are unknown. Noninvasive treatment monitoring is crucial for longitudinal tumor characterization and the early detection of non-responders. While various medical imaging techniques can provide a morphological picture of the lesion and its surrounding tissue, a molecular-oriented imaging approach holds the key to unraveling biological effects that occur much earlier in the immunotherapy timeline. Magnetic resonance imaging (MRI) is a highly versatile imaging modality, where the image contrast can be tailored to emphasize a particular biophysical property of interest using advanced engineering of the imaging pipeline. In this review, recent advances in molecular-MRI based cancer immunotherapy monitoring are described. Next, the presentation of the underlying physics, computational, and biological features are complemented by a critical analysis of the results obtained in preclinical and clinical studies. Finally, emerging artificial intelligence (AI)-based strategies to further distill, quantify, and interpret the image-based molecular MRI information are discussed in terms of perspectives for the future.
Collapse
Affiliation(s)
- Nikita Vladimirov
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Or Perlman
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|