1
|
Dewhirst MW. A translational review of hyperthermia biology. Int J Hyperthermia 2025; 42:2447952. [PMID: 39799944 DOI: 10.1080/02656736.2024.2447952] [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: 09/14/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/15/2025] Open
Abstract
This review was written to be included in the Special Collection 'Therapy Ultrasound: Medicine's Swiss Army Knife?' The purpose of this review is to provide basic presentation and interpretation of the fundamentals of hyperthermia biology, as it pertains to uses of therapeutic ultrasound. The fundamentals are presented but in the setting of a translational interpretation and a view toward the future. Subjects that require future research and development are highlighted. The effects of hyperthermia are time and temperature dependent. Because intra-tumoral temperatures are non-uniform in tumors, one has to account for differential biologic effects in different parts of a tumor that occur simultaneously during and after hyperthermia.
Collapse
Affiliation(s)
- Mark W Dewhirst
- Gustavo S. Montana Distinguished Professor Emeritus of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
2
|
Li Q, Liu H. Investigating the Prognostic Role of Telomerase-Related Cellular Senescence Gene Signatures in Breast Cancer Using Machine Learning. Biomedicines 2025; 13:826. [PMID: 40299459 PMCID: PMC12024799 DOI: 10.3390/biomedicines13040826] [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: 01/26/2025] [Revised: 03/24/2025] [Accepted: 03/29/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Telomeres and cellular senescence are critical biological processes implicated in cancer development and progression, including breast cancer, through their influence on genomic stability and modulation of the tumor microenvironment. Methods: This study integrated bulk RNA sequencing and single-cell RNA sequencing (scRNA-seq) data to establish a gene signature associated with telomere maintenance and cellular senescence for prognostic prediction in breast cancer. Telomere-related genes (TEGs) and senescence-associated genes were curated from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. A comprehensive machine learning framework incorporating 101 algorithmic combinations across 10 survival modeling approaches, including random survival forests and ridge regression, was employed to develop a robust prognostic model. Results: A set of 19 key telomere- and senescence-related genes was identified as the optimal prognostic signature. The model demonstrated strong predictive accuracy and was successfully validated in multiple independent cohorts. Functional enrichment analyses indicated significant associations with immune responses and aging-related pathways. Single-cell transcriptomic analysis revealed marked cellular heterogeneity, identifying distinct subpopulations (fibroblasts and immune cells) with divergent risk scores and biological pathway activity. Additionally, pseudo-time trajectory analysis and intercellular communication mapping provided insights into the dynamic evolution of the tumor microenvironment. Immunohistochemical (IHC) validation using data from the Human Protein Atlas confirmed differential protein expression between normal and tumor tissues for several of the selected genes, reinforcing their biological relevance and clinical utility. Conclusions: This study presents a novel 19-gene telomere- and senescence-associated signature with strong prognostic value in breast cancer. These findings enhance our understanding of tumor heterogeneity and may inform precision oncology approaches and future therapeutic strategies.
Collapse
Affiliation(s)
| | - Hongde Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China;
| |
Collapse
|
3
|
Singh P, Jay DG. The Role of eHsp90 in Extracellular Matrix Remodeling, Tumor Invasiveness, and Metastasis. Cancers (Basel) 2024; 16:3873. [PMID: 39594828 PMCID: PMC11592750 DOI: 10.3390/cancers16223873] [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: 10/03/2024] [Revised: 11/01/2024] [Accepted: 11/01/2024] [Indexed: 11/28/2024] Open
Abstract
Identifying proteins that act in tumor invasiveness and metastasis remains a critical unmet need in our search for effective cancer therapy. Hsp90, an abundant intracellular chaperone protein, plays a key role in maintaining cell homeostasis, and its elevated activity is pivotal in cancer progression. Due to the reliance of cancer cells on Hsp90's chaperone function to sustain tumor growth and spread, Hsp90 inhibitors have been the subject of numerous clinical trials over the past two decades. However, these efforts have largely been unsuccessful, primarily due to the cellular toxicity caused by pan-Hsp90 inhibitors at doses required for anticancer efficacy. Therefore, novel approaches to target Hsp90 are necessary. An identified subpopulation of Hsp90 located outside cells (eHsp90) may offer a promising alternative as a therapeutic target against cancer. Studies including our own have shown that eHsp90 is released specifically by cancer cells, and eHsp90 has unique interactors and functions extracellularly to promote tumor invasiveness, the initial step in metastasis. Inhibition of eHsp90 has been shown to suppress metastasis in animal models, indicating its therapeutic potential, although the underlying mechanisms remain incompletely understood. Cancer cells modulate the tumor microenvironment (TME) during the invasion, especially the ECM proteins and the state of the ECM is a strong predictor of invasive and metastatic cancer. Given that most of the known eHsp90 clients are ECM proteins or are proteins involved in ECM modulation, ECM remodelling could be the key mechanism through which eHsp90 enhances invasiveness. This review will focus on ECM modulation by eHsp90 as a driver of cancer invasion and metastasis. We will also discuss the potency of inhibiting eHsp90 in inhibiting invasion and metastatic spread in preclinical models and the using circulating Hsp90 patient samples as a biomarker of cancer invasion and metastasis.
Collapse
Affiliation(s)
- Pragya Singh
- Graduate School of Biomedical Sciences, Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Daniel G. Jay
- Graduate School of Biomedical Sciences, Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
4
|
Wang R, Ekem L, Gallagher J, Factor RE, Hall A, Ramanujam N. A color-based tumor segmentation method for clinical ex vivo breast tissue assessment utilizing a multi-contrast brightfield imaging strategy. JOURNAL OF BIOPHOTONICS 2024; 17:e202300241. [PMID: 38348582 PMCID: PMC11065618 DOI: 10.1002/jbio.202300241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 01/20/2024] [Accepted: 01/21/2024] [Indexed: 03/21/2024]
Abstract
We demonstrate an automated two-step tumor segmentation method leveraging color information from brightfield images of fresh core needle biopsies of breast tissue. Three different color spaces (HSV, CIELAB, YCbCr) were explored for the segmentation task. By leveraging white-light and green-light images, we identified two different types of color transformations that could separate adipose from benign and tumor or cancerous tissue. We leveraged these two distinct color transformation methods in a two-step process where adipose tissue segmentation was followed by benign tissue segmentation thereby isolating the malignant region of the biopsy. Our tumor segmentation algorithm and imaging probe could highlight suspicious regions on unprocessed biopsy tissue to guide selection of areas most similar to malignant tissues for tissue pathology whether it be formalin fixed or frozen sections, expedite tissue selection for molecular testing, detect positive tumor margins, or serve an alternative to tissue pathology, in countries where these services are lacking.
Collapse
Affiliation(s)
- Roujia Wang
- Department of Biomedical Engineering, Duke University, 27710 Durham, NC, USA
| | - Lillian Ekem
- Department of Biomedical Engineering, Duke University, 27710 Durham, NC, USA
| | - Jennifer Gallagher
- Department of Surgery, Duke University School of Medicine, 27710 Durham, NC, USA
| | | | - Allison Hall
- Department of Pathology, Duke University, 27710 Durham, NC, USA
| | - Nimmi Ramanujam
- Department of Biomedical Engineering, Duke University, 27710 Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, 27710 Durham, NC, USA
| |
Collapse
|
5
|
Singh P, Ramanathan V, Zhang Y, Georgakoudi I, Jay DG. Extracellular Hsp90 Binds to and Aligns Collagen-1 to Enhance Breast Cancer Cell Invasiveness. Cancers (Basel) 2023; 15:5237. [PMID: 37958410 PMCID: PMC10648158 DOI: 10.3390/cancers15215237] [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: 08/30/2023] [Revised: 10/09/2023] [Accepted: 10/21/2023] [Indexed: 11/15/2023] Open
Abstract
Cancer cell-secreted eHsp90 binds and activates proteins in the tumor microenvironment crucial in cancer invasion. Therefore, targeting eHsp90 could inhibit invasion, preventing metastasis-the leading cause of cancer-related mortality. Previous eHsp90 studies have solely focused on its role in cancer invasion through the 2D basement membrane (BM), a form of extracellular matrix (ECM) that lines the epithelial compartment. However, its role in cancer invasion through the 3D Interstitial Matrix (IM), an ECM beyond the BM, remains unexplored. Using a Collagen-1 binding assay and second harmonic generation (SHG) imaging, we demonstrate that eHsp90 directly binds and aligns Collagen-1 fibers, the primary component of IM. Furthermore, we show that eHsp90 enhances Collagen-1 invasion of breast cancer cells in the Transwell assay. Using Hsp90 conformation mutants and inhibitors, we established that the Hsp90 dimer binds to Collagen-1 via its N-domain. We also demonstrated that while Collagen-1 binding and alignment are not influenced by Hsp90's ATPase activity attributed to the N-domain, its open conformation is crucial for increasing Collagen-1 alignment and promoting breast cancer cell invasion. These findings unveil a novel role for eHsp90 in invasion through the IM and offer valuable mechanistic insights into potential therapeutic approaches for inhibiting Hsp90 to suppress invasion and metastasis.
Collapse
Affiliation(s)
- Pragya Singh
- Department of Developmental, Molecular and Chemical Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA; (P.S.); (I.G.)
| | - Varshini Ramanathan
- Department of Biomedical Engineering, Tufts University School of Engineering, Medford, MA 02155, USA; (V.R.); (Y.Z.)
| | - Yang Zhang
- Department of Biomedical Engineering, Tufts University School of Engineering, Medford, MA 02155, USA; (V.R.); (Y.Z.)
| | - Irene Georgakoudi
- Department of Developmental, Molecular and Chemical Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA; (P.S.); (I.G.)
- Department of Biomedical Engineering, Tufts University School of Engineering, Medford, MA 02155, USA; (V.R.); (Y.Z.)
| | - Daniel G. Jay
- Department of Developmental, Molecular and Chemical Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA; (P.S.); (I.G.)
| |
Collapse
|
6
|
Wang R, Deutsch RJ, Sunassee ED, Crouch BT, Ramanujam N. Adaptive Design of Fluorescence Imaging Systems for Custom Resolution, Fields of View, and Geometries. BME FRONTIERS 2023; 4:0005. [PMID: 37849673 PMCID: PMC10521686 DOI: 10.34133/bmef.0005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 11/27/2022] [Indexed: 10/19/2023] Open
Abstract
Objective and Impact Statement: We developed a generalized computational approach to design uniform, high-intensity excitation light for low-cost, quantitative fluorescence imaging of in vitro, ex vivo, and in vivo samples with a single device. Introduction: Fluorescence imaging is a ubiquitous tool for biomedical applications. Researchers extensively modify existing systems for tissue imaging, increasing the time and effort needed for translational research and thick tissue imaging. These modifications are application-specific, requiring new designs to scale across sample types. Methods: We implemented a computational model to simulate light propagation from multiple sources. Using a global optimization algorithm and a custom cost function, we determined the spatial positioning of optical fibers to generate 2 illumination profiles. These results were implemented to image core needle biopsies, preclinical mammary tumors, or tumor-derived organoids. Samples were stained with molecular probes and imaged with uniform and nonuniform illumination. Results: Simulation results were faithfully translated to benchtop systems. We demonstrated that uniform illumination increased the reliability of intraimage analysis compared to nonuniform illumination and was concordant with traditional histological findings. The computational approach was used to optimize the illumination geometry for the purposes of imaging 3 different fluorophores through a mammary window chamber model. Illumination specifically designed for intravital tumor imaging generated higher image contrast compared to the case in which illumination originally optimized for biopsy images was used. Conclusion: We demonstrate the significance of using a computationally designed illumination for in vitro, ex vivo, and in vivo fluorescence imaging. Application-specific illumination increased the reliability of intraimage analysis and enhanced the local contrast of biological features. This approach is generalizable across light sources, biological applications, and detectors.
Collapse
Affiliation(s)
- Roujia Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Riley J. Deutsch
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Brian T. Crouch
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nirmala Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
7
|
Dewi C, Fristiohady A, Amalia R, Khairul Ikram NK, Ibrahim S, Muchtaridi M. Signaling Pathways and Natural Compounds in Triple-Negative Breast Cancer Cell Line. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123661. [PMID: 35744786 PMCID: PMC9227697 DOI: 10.3390/molecules27123661] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 11/16/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, having a poor prognosis and rapid metastases. TNBC is characterized by the absence of estrogen, progesterone, and human epidermal growth receptor-2 (HER2) expressions and has a five-year survival rate. Compared to other breast cancer subtypes, TNBC patients only respond to conventional chemotherapies, and even then, with limited success. Shortages of chemotherapeutic medication can lead to resistance, pressured index therapy, non-selectivity, and severe adverse effects. Finding targeted treatments for TNBC is difficult owing to the various features of cancer. Hence, identifying the most effective molecular targets in TNBC pathogenesis is essential for predicting response to targeted therapies and preventing TNBC cell metastases. Nowadays, natural compounds have gained attention as TNBC treatments, and have offered new strategies for solving drug resistance. Here, we report a systematic review using the database from Pubmed, Science Direct, MDPI, BioScince, Springer, and Nature for articles screening from 2003 to 2022. This review analyzes relevant signaling pathways and the prospect of utilizing natural compounds as a therapeutic agent to improve TNBC treatments in the future.
Collapse
Affiliation(s)
- Citra Dewi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
- Pharmacy Department, Faculty of Science and Technology, Mandala Waluya University, Kendari 93561, Indonesia
| | - Adryan Fristiohady
- Faculty of Pharmacy, Halu Oleo University, Kampus Hijau Bumi Tridharma, Kendari 93232, Indonesia;
| | - Riezki Amalia
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
| | - Nur Kusaira Khairul Ikram
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
| | - Sugeng Ibrahim
- Department of Molecular Biology, Faculty of Medicine, Universitas Katolik Soegijapranata, Semarang 50234, Indonesia;
| | - Muchtaridi Muchtaridi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
- Correspondence:
| |
Collapse
|
8
|
Zhou W, Yang Y, Wang Z, Liu Y, Lari Najafi M. Impact of HSP90 α, CEA, NSE, SCC, and CYFRA21-1 on Lung Cancer Patients. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:6929971. [PMID: 34721827 PMCID: PMC8550848 DOI: 10.1155/2021/6929971] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/08/2021] [Accepted: 09/14/2021] [Indexed: 12/24/2022]
Abstract
Lung cancer is a lethal disease, and early diagnosis with the aid of biomarkers such as HSP90α protein can certainly assist the doctors to start treatment of patient at the earliest and can save their lives. To analyse the diagnostic value of HSP90α expression in lung cancer patients by collecting data of patients through IoT devices to avoid delay in treatments, a study has been presented in this paper where the significance of HSP90α biomarker is highlighted in early diagnosis of patients suffering from lung cancer. The second objective of the research study is to examine the correlation between the appearance level of HSP90α biomarker and the clinicopathological features of lung cancer. It is also evaluated whether the changes in HSP90α index are indicative or noteworthy before and after surgery of lung cancer patients. An observatory study of 78 patients with lung cancer in Qinhuangdao Hospital is presented in this paper where the samples were collected from June 2018 to March 2020. Their data were collected through IoT devices used in the latest healthcare facilities of the hospital. The ELISA method was utilized to identify the level of plasma HSP90 and to analyse HSP90 levels between the lung cancer group and healthy group of people. The relationship between HSP90 and the clinical pathological features of 78 patients suffering from lung cancer was analysed. An electrochemical luminescence method was used to detect CEA, NSE, SCC, and CYFRA21-1 levels. ROC curve and box plots were used to determine the analytic value of HSP90 and other biomarkers used in lung cancer diagnosis. Forty-two patients with moderate to early stage lung cancer with surgical correction were selected, and paired sample T test was used to analyse HSP90 levels before and after surgery. The plasma HSP90 level of lung cancer patients was quite higher as compared to the group of healthy people as per the values depicted in the research study. Second, HSP90 levels are substantially higher in pathologic type, differentiation degree, stage, and the existence of the lung, liver, and bone metastases (P < 0.05). The level of HSP90 expression was largely impacted by a few factors such as sex, age, smoking, and tumour location (P > 0.05). The ROC value for HSP90 was 0.599, while the area under the curve of HSP90 combined with other four tumour markers was 0.915 in the presented case study, indicating the presence of lung cancer. Patients with lung cancer had statistically significant differences in HSP90 expression levels before and after surgery (P < 0.05). It is concluded that the expression level of plasma HSP90α in lung cancer patients increases remarkably; therefore, HSP90 can be used to monitor presence of lung cancer before and after surgery in the patients.
Collapse
Affiliation(s)
- Wenwen Zhou
- Department of Oncology, First Hospital of Qinhuangdao, Qinhuangdao 066000, China
| | - Yanhong Yang
- Department of Oncology, First Hospital of Qinhuangdao, Qinhuangdao 066000, China
| | - Zhenzhen Wang
- Department of Oncology, First Hospital of Qinhuangdao, Qinhuangdao 066000, China
| | - Yan Liu
- Graduate School, Hebei Chengde Medical College, Chengde 067000, China
| | - Moslem Lari Najafi
- Pharmaceutical Science and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
9
|
Liu H, Zhang Z, Huang Y, Wei W, Ning S, Li J, Liang X, Liu K, Zhang L. Plasma HSP90AA1 Predicts the Risk of Breast Cancer Onset and Distant Metastasis. Front Cell Dev Biol 2021; 9:639596. [PMID: 34109171 PMCID: PMC8181396 DOI: 10.3389/fcell.2021.639596] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 05/03/2021] [Indexed: 12/24/2022] Open
Abstract
Aim We aimed to develop and validate a comprehensive nomogram containing pre-treatment plasma HSP90AA1 to predict the risk of breast cancer onset and metastasis. Methods We assessed the expression of HSP90s in breast cancer patients using an online database. To verify the results, 677 patients diagnosed with breast cancer and 146 patients with benign breast disease between 2014 and 2019 were selected from our hospital and were divided into cancer risk and metastasis risk cohorts. We focused on HSP90AA1 to elucidate the risks of onset and metastasis in the cohorts. Results Expression levels of HSP90AA1, HSP90AA2, HSP90AB1, HSP90B1, and TRAP1 were linked to disease progression. Survival analysis using the GEPIA and OncoLnc databases indicated that the upregulation of HSP90AA1 and HSP90AB1 was related to poor overall survival. In the cancer risk cohort, carcinoembryonic antigen (CEA), carbohydrate antigen 153 (CA153), HSP90AA1, T cells%, natural killer cells%, B cells%, neutrophil count, monocyte count, and d-dimer were incorporated into the nomogram. A high Harrell's concordance index (C-index) value of 0.771 [95% confidence interval (CI), 0.725-0.817] could still be reached in the interval validation. In the metastasis risk cohort, predictors contained in the prediction nomogram included the use of CEA, CA153, HSP90AA1, carbohydrate antigen 125 (CA125), natural killer cells%, B cells%, platelet count, monocyte count, and d-dimer. The C-index was 0.844 (95% CI, 0.801-0.887) and it was well-calibrated. HSP90AA1 raised net clinical benefit of breast cancer onset and metastasis risk prediction nomogram in a range of risk thresholds (5-92%) and (1-90%). Conclusion Our study revealed that pretreatment plasma HSP90AA1 combined with other markers could conveniently predict the risk of breast cancer onset and metastasis.
Collapse
Affiliation(s)
- Haizhou Liu
- Department of Research, Affiliated Tumor Hospital, Guangxi Medical University, Nanning, China
| | - Zihan Zhang
- Department of Research, Affiliated Tumor Hospital, Guangxi Medical University, Nanning, China
| | - Yi Huang
- Department of Research, Affiliated Tumor Hospital, Guangxi Medical University, Nanning, China
| | - Wene Wei
- Department of Research, Affiliated Tumor Hospital, Guangxi Medical University, Nanning, China
| | - Shufang Ning
- Department of Research, Affiliated Tumor Hospital, Guangxi Medical University, Nanning, China
| | - Jilin Li
- Department of Research, Affiliated Tumor Hospital, Guangxi Medical University, Nanning, China
| | - Xinqiang Liang
- Department of Research, Affiliated Tumor Hospital, Guangxi Medical University, Nanning, China
| | - Kaisheng Liu
- Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Litu Zhang
- Department of Research, Affiliated Tumor Hospital, Guangxi Medical University, Nanning, China
| |
Collapse
|
10
|
Wang R, Alvarez DA, Crouch BT, Pilani A, Lam C, Zhu C, Hughes P, Katz D, Haystead T, Ramanujam N. Understanding the sources of errors in ex vivo Hsp90 molecular imaging for rapid-on-site breast cancer diagnosis. BIOMEDICAL OPTICS EXPRESS 2021; 12:2299-2311. [PMID: 33996230 PMCID: PMC8086448 DOI: 10.1364/boe.418818] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/26/2021] [Accepted: 03/16/2021] [Indexed: 05/12/2023]
Abstract
Overexpression of heat shock protein 90 (Hsp90) on the surface of breast cancer cells makes it an attractive molecular biomarker for breast cancer diagnosis. Before a ubiquitous diagnostic method can be established, an understanding of the systematic errors in Hsp90-based imaging is essential. In this study, we investigated three factors that may influence the sensitivity of ex vivo Hsp90 molecular imaging: time-dependent tissue viability, nonspecific diffusion of an Hsp90 specific probe (HS-27), and contact-based imaging. These three factors will be important considerations when designing any diagnostic imaging strategy based on fluorescence imaging of a molecular target on tissue samples.
Collapse
Affiliation(s)
- Roujia Wang
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Daniel A. Alvarez
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Brian T. Crouch
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Aditi Pilani
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Christopher Lam
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Caigang Zhu
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
- Currently at Department of Biomedical Engineering, University of Kentucky, Lexington, Kentucky, 40506, USA
| | - Philip Hughes
- Department of Pharmacology and Cancer Biology, School of Medicine, Duke University, Durham, North Carolina 27708, USA
| | - David Katz
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Timothy Haystead
- Department of Pharmacology and Cancer Biology, School of Medicine, Duke University, Durham, North Carolina 27708, USA
| | - Nirmala Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
- Department of Pharmacology and Cancer Biology, School of Medicine, Duke University, Durham, North Carolina 27708, USA
| |
Collapse
|
11
|
Barth CW, Gibbs SL. Fluorescence Image-Guided Surgery - a Perspective on Contrast Agent Development. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2020; 11222:112220J. [PMID: 32255887 PMCID: PMC7115043 DOI: 10.1117/12.2545292] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In the past several decades, a number of novel fluorescence image-guided surgery (FGS) contrast agents have been under development, with many in clinical translation and undergoing clinical trials. In this review, we have identified and summarized the contrast agents currently undergoing clinical translation. In total, 39 novel FGS contrast agents are being studied in 85 clinical trials. Four FGS contrast agents are currently being studied in phase III clinical trials and are poised to reach FDA approval within the next two to three years. Among all novel FGS contrast agents, a wide variety of probe types, targeting mechanisms, and fluorescence properties exists. Clinically available FGS imaging systems have been developed for FDA approved FGS contrast agents, and thus further clinical development is required to yield FGS imaging systems tuned for the variety of contrast agents in the clinical pipeline. Additionally, study of current FGS contrast agents for additional disease types and development of anatomy specific contrast agents is required to provide surgeons FGS tools for all surgical specialties and associated comorbidities. The work reviewed here represents a significant effort from many groups and further development of this promising technology will have an enormous impact on surgical outcomes across all specialties.
Collapse
Affiliation(s)
- Connor W Barth
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201
| | - Summer L Gibbs
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201
- OHSU Center for Spatial Systems Biomedicine, Oregon Health & Science University, Portland, OR 97201
| |
Collapse
|