1
|
Alaei E, Hashemi F, Farahani N, Tahmasebi S, Nabavi N, Daneshi S, Mahmoodieh B, Rahimzadeh P, Taheriazam A, Hashemi M. Peptides in breast cancer therapy: From mechanisms to emerging drug delivery and immunotherapy strategies. Pathol Res Pract 2025; 269:155946. [PMID: 40174279 DOI: 10.1016/j.prp.2025.155946] [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: 01/15/2025] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
Breast cancer therapy can be improved by the application of multifunctional peptides and they have unique features, such as high specificity, minimized toxicity, and the capability to influence diverse processes. The role of peptides in breas cancer therapy is highlighted in the present review, examining their functions as therapeutic agents, diagnostic tools, and drug delivery application. Therapeutic peptides have displayed the ability to regulate key pathways in breast tumor, including HER2, VEGF, and EGFR, providing ideal alternatives to the conventional chemotherapy with reduced adverse effects. Additionally, peptide-based vaccines and immune-modulating peptides have demonstrated the capacity in enhancing anti-cancer immunity. The incorporation of peptides into nanoparticles has improved the delivery of drugs and genes, enhanced anti-cancer efficacy while minimizing side impacts. The progresses in the peptide engineering, including stapled peptides, peptide-drug conjugates, and cell-penetrating peptides, have remarkably increased their therapeutic efficacy and stability, elevating their applications in breast cancer therapy. Peptides can be developed using bioinformatics and high-throughput screening technologies to optimize pharmacokinetics and bioavailability. Despite their promise, peptides demonstrate challenges such as enzymatic degradation, limited stability, and high production costs. These obstacles can be addressed through strategies such as peptide cyclization, the employement of non-natural amino acids, and nanoparticle encapsulation. This review explores these recent advancements and strategies, providing ideal insights into the clinical potential of peptides in breast tumor therapy.
Collapse
Affiliation(s)
- Elmira Alaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Safa Tahmasebi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Behnaz Mahmoodieh
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
2
|
Samari M, Alamzadeh Z, Irajirad R, Sarikhani A, Mahabadi VP, Ghaznavi H, Khoei S. FROP-1 peptide-conjugated ultrasmall superparamagnetic nanoparticles as a targeted T1-weighted MR contrast agent for breast cancer: in vitro study. BMC Biomed Eng 2025; 7:5. [PMID: 40307948 PMCID: PMC12044754 DOI: 10.1186/s42490-025-00091-7] [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: 12/15/2024] [Accepted: 04/02/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND The aim of this study was to produce ultrasmall superparamagnetic iron oxide (USPIO) nanoparticles (NPs) conjugated to the FROP-1 peptide for targeted magnetic resonance imaging (MRI) of breast cancer cell lines and to evaluate its application as a specific and targeted T1-weighted MR imaging contrast agent in vitro. Sodium citrate-stabilized Fe3O4 NPs were conjugated with the FROP-1 peptide by 1-ethyl-3-(3-dimethylaminopropyl) carbide diamide hydrochloride (EDC) to form a novel Fe3O4@FROP-1 specific target contrast agent. The specificity and targeting of Fe3O4@FROP-1 to bind FROP-1 receptors were investigated in vitro by cellular uptake and cellular MR imaging. RESULTS In this study, the synthesis of water-soluble ultrasmall Fe3O4 NPs was performed by the co-precipitation method. XRD, TEM, and VSM analyses showed the formation of the Fe3O4 NPs with an average size of about 3.78 ± 0.2 nm. FT-IR spectroscopy approved the conjugation of the FROP-1 peptide with the Fe3O4 NPs. The synthesized Fe3O4@FROP-1 NPs showed good biocompatibility, and the high r1 relaxivity and r2/r1, respectively, were 2.608 mM- 1S- 1 and 1.18. The biocompatibility of the Fe3O4 and Fe3O4@FROP-1 NPs on the MCF-7, SKBR-3, MDA-MB-231, and MCF-10 cell lines was determined using cytotoxicity analysis. The specific targeting effect on the cells was verified by in vitro cellular uptake and cell MR imaging. CONCLUSION It was found that the contrast intensity of the Fe3O4@FROP-1 nanoprobe increases as Fe concentration increases. Cellular uptake of the Fe3O4 and Fe3O4@FROP-1 NPs was quantified using ICP-MS. The synthesized NPs had better imaging performance than Dotarem (gadoterate meglumine). The findings showed that Fe3O4@FROP-1 NPs have potential utility as a specific and targeted T1-weighted contrast agent in breast cancer MR imaging.
Collapse
Affiliation(s)
- Melika Samari
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Medical Physics Department, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Alamzadeh
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Irajirad
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Sarikhani
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Medical Physics Department, Iran University of Medical Sciences, Tehran, Iran
| | | | - Habib Ghaznavi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Samideh Khoei
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Medical Physics Department, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Tually P, Quinto VG, Omar Y, Novruzov F, Yudistiro R, Sathekge M, Currie G, Galette P, Patel N, Brown T, Bolland G, Lo Bue R, Cade D. Real world experience with [ 99mTc]Tc-HYNIC-iPSMA SPECT prostate cancer detection: interim results from the global NOBLE registry. EJNMMI REPORTS 2024; 8:43. [PMID: 39738799 DOI: 10.1186/s41824-024-00226-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/24/2024] [Indexed: 01/02/2025]
Abstract
PURPOSE [99mTc]Tc-HYNIC-iPSMA is a novel technetium-99m-labelled small molecule inhibitor of the prostate-specific membrane antigen (PSMA) for detecting prostate cancer (PC). The objective of this registry was to collect and evaluate [99mTc]Tc-HYNIC-iPSMA patient data and images to establish the safety and tolerability, and clinical utility of this agent in imaging at different stages of PC. METHODS Patients 18 to 80 years old with primary staging and metastatic PC were eligible. Patients unable to perform prescribed examinations, undergo a [99mTc]Tc-HYNIC-iPSMA planar and SPECT or SPECT/CT (when available), or sign a patient informed consent form were excluded from the registry. All eligible patients underwent a screening and baseline visit before imaging with [99mTc]Tc-HYNIC-iPSMA. The primary safety endpoint was assessed by collecting and grading all treatment-related adverse events using the Common Terminology Criteria for Adverse Events. Patients were followed until disease progression, death, serious or intolerable adverse events, registry termination by the sponsor, patient withdrawal, or lost to follow-up. Analysis was planned for when data was available from 40 enrolled patients. RESULTS 40 patients enrolled in 6 countries and received [99mTc]Tc-HYNIC-iPSMA tracer administration followed by planar and SPECT imaging. Of the 40 patients included, investigators reported a change in management due to the [99mTc]Tc-HYNIC-iPSMA imaging in 17/40 of patients (42.5%). No adverse events were reported. CONCLUSIONS [99mTc]Tc-HYNIC-iPSMA is a promising option to identify PSMA-positive prostate cancer on SPECT and could improve patient access to PSMA imaging worldwide.
Collapse
Affiliation(s)
- Pete Tually
- Department of Nuclear Medicine, Charles Sturt University, TeleMedVET, Perth, WA, Australia
| | | | | | - Fuad Novruzov
- Department of Nuclear Medicine, Azerbaijan National Centre of Oncology, M. Xiyabani Street No. 137, Baku, Azerbaijan
| | - Ryan Yudistiro
- Department of Nuclear Medicine, Siloam Hospital, Jakarta, Indonesia
| | - Mike Sathekge
- University of Pretoria Nuclear Medicine Department, Gauteng, South Africa
| | - Geoffrey Currie
- School of Dentistry and Medical Sciences, Charles Sturt University, Bathurst, Australia
| | | | - Neel Patel
- Telix Pharmaceuticals, Melbourne, Australia
| | | | | | | | - David Cade
- Telix Pharmaceuticals, Melbourne, Australia.
| |
Collapse
|
4
|
Yang Y, Xie T, Gao P, Han W, Liu Y, Wang Y. Hsa_Circ_002144 Promotes Glycolysis and Immune Escape of Breast Cancer Through miR-326/PKM Axis. Cancer Biother Radiopharm 2024; 39:755-769. [PMID: 38963787 DOI: 10.1089/cbr.2024.0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024] Open
Abstract
Background: Breast cancer is a leading cause of cancer-related deaths in women worldwide, posing a significant threat to female health. Therefore, it is crucial to search for new therapeutic targets and prognostic biomarkers for breast cancer patients. Method: Bioinformatics analysis, quantitative real-time PCR (qRT-PCR), and fluorescence in situ hybridization (FISH) were employed to investigate the expression of hsa_circ_002144 in breast cancer. Transwell assay, Western blotting, and cell viability assay were utilized to assess the impact of hsa_circ_002144 on the proliferation, migration, and invasion of breast cancer cells. Additionally, a mouse model was established to validate its functionality. Flow cytometry, WB analysis, enzyme-linked immunosorbent assay (ELISA), qRT-PCR, exosomes isolation, and co-culture system were employed to elucidate the molecular mechanism underlying macrophage polarization. Result: we have discovered for the first time that hsa_circ_002144 is highly expressed in breast cancer. It affected tumor growth and metastasis and could influence macrophage polarization through the glycolytic pathway. Conclusion: This finding provides a new direction for breast cancer treatment and prognosis assessment.
Collapse
Affiliation(s)
- Yong Yang
- College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang City, China
| | - Tianhao Xie
- General Surgery, The Affiliated Hospital of Hebei University, Baoding City, China
| | - Peng Gao
- Anesthesiology department, Affiliated hospital of Qingdao university, Qingdao City, China
| | - Weijun Han
- Third Surgery, Baoji traditional Chinese Medicine Hospital in Shaanxi Province, Baoji City, China
| | - Yuhong Liu
- Rheumatology and Immunology Department, The Affiliated Hospital of Yan 'an University, Yan 'an City, China
| | - Yanmei Wang
- School of Nursing and Health, Medical College of Yan 'an University, Yan 'an City, China
| |
Collapse
|
5
|
Ferreira AH, Real CC, Malafaia O. Heterobivalent Dual-Target Peptide for Integrin-α vβ 3 and Neuropeptide Y Receptors on Breast Tumor. Pharmaceuticals (Basel) 2024; 17:1328. [PMID: 39458969 PMCID: PMC11510292 DOI: 10.3390/ph17101328] [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: 08/23/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Heterodimer peptides targeting more than one receptor can be advantageous, as tumors can simultaneously express more than one receptor type. For human breast cancer, a promising biological target is tumor angiogenesis through αvβ3 integrin expression. Another promising target is Neuropeptide Y receptors, considering Y1R is overexpressed in 90% of human breast tumors. This article details the development and preclinical evaluation, both in vitro and in vivo, of a novel heterodimer peptide dual-receptor-targeting probe, [99mTc]HYNIC-cRGDfk-NPY, designed for imaging breast tumors. Methods: Female BALB/c healthy mice were used to perform biodistrubution studies and female SCID mice were subcutaneously injected with MCF-7 and MDA-MB-231 tumor cells. [99mTc]HYNIC-cRGDfk-NPY was intravenously administered to the mice, followed by ex vivo biodistribution studies and small-animal SPECT/CT imaging. Nonspecific tracer uptake in both models was determined by coinjecting an excess of unlabeled HYNIC-cRGDfk-NPY (100 µg) along with the radiolabeled tracer. Results: Imaging and biodistribution data demonstrate good uptake to estrogen receptor-positive (MCF-7) and triple-negative (MDA-MB-231) tumor models. The in vivo tumor uptakes of radiolabeled conjugate were 9.30 ± 3.25% and 4.93 ± 1.01% for MCF-7 and MDA-MB231, respectively. The tumor/muscle ratios were 5.65 ± 0.94 for the MCF-7 model and 7.78 ± 3.20 for MDA-MB231. Conclusions: [99mTc]HYNIC-cRGDfk-NPY demonstrated rapid blood clearance, renal excretion, and in vivo tumor uptake, highlighting its potential as a tumor imaging agent.
Collapse
Affiliation(s)
- Aryel H. Ferreira
- MackGraphe-Mackenzie Institute for Research in Graphene and Nanotechnologies, Mackenzie Presbyterian University, São Paulo 01302-907, Brazil
- Mackenzie Evangelical College of Paraná, Mackenzie Presbyterian University, Curitiba 80730-000, Brazil
- Nuclear and Energy Research Institute, Instituto de Pesquisas Energéticas e Nucleares da Comissão Nacional de Energia Nuclear—São Paulo (IPEN-CNEN/SP), São Paulo 05508-000, Brazil
| | - Caroline C. Real
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Osvaldo Malafaia
- Mackenzie Evangelical College of Paraná, Mackenzie Presbyterian University, Curitiba 80730-000, Brazil
| |
Collapse
|
6
|
Kolay S, Kumar N, Guleria M, Das T. [ 99mTc]Tc-labeled HYNIC conjugated chlorambucil as a tumor targeting Agent: Synthesis, characterization and ex-vivo evaluation. Bioorg Med Chem Lett 2024; 105:129730. [PMID: 38583784 DOI: 10.1016/j.bmcl.2024.129730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/22/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
Chlorambucil is an alkylating drug that finds application towards chemotherapy of different types of cancers. In order to explore the possibility of utilization of this drug as an imaging agent for early diagnosis of solid tumors, attempt was made to synthesize a 99mTc complex of chlorambucil and evaluate its potential in tumor bearing small animal model. HYNIC-chlorambucil was synthesized by conjugation of HYNIC with chlorambucil via an ethylenediamine linker. All the intermediates and final product were purified and characterized by standard spectroscopic techniques viz. FT-IR, 1H/13C-NMR as well as by mass spectrometry. HYNIC-chlorambucil conjugate was radiolabeled with [99mTc]Tc and found to be formed with > 95 % radiochemical purity via RP-HPLC studies. The partition coefficient (Log10Po/w) of the synthesized complex was found to be -0.78 ± 0.25 which indicated the moderate hydrophilic nature for the complex. Biological behaviour of [99mTc]Tc-HYNIC-chlorambucil, studied in fibrosarcoma bearing Swiss mice, revealed a tumor uptake of about 4.16 ± 1.52 %IA/g at 30 min post-administration, which declined to 1.91 ± 0.13 % IA/g and 1.42 ± 0.14 %IA/g at 1 h and 2 h post-administration, respectively. A comparison of different [99mTc]Tc-chlorambucil derivatives (reported in the contemporary literature) formulated using different methodologies revealed that tumor uptake and pharmacokinetics exhibited by these agents strongly depend on the lipophilicity/hydrophilicity of such agents, which in turn is dependent on the bifunctional chelators used for formulating the radiolabeled chlorambucils.
Collapse
Affiliation(s)
- Soumi Kolay
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Naveen Kumar
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Mohini Guleria
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Tapas Das
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India.
| |
Collapse
|
7
|
Mendonça FF, Sobral DV, Durante ACR, Miranda ACC, Mejia J, de Paula Faria D, Marques FLN, de Barboza MF, Fuscaldi LL, Malavolta L. Assessment of bioactive peptides derived from laminin-111 as prospective breast cancer-targeting agents. Amino Acids 2024; 56:1. [PMID: 38285098 PMCID: PMC10824877 DOI: 10.1007/s00726-023-03379-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/24/2023] [Indexed: 01/30/2024]
Abstract
Breast cancer remains a pressing public health issue primarily affecting women. Recent research has spotlighted bioactive peptides derived from laminin-111, implicated in breast tumor development. Remarkably, the sequences IKVAV, YIGSR, and KAFDITYVRLKF from the α1, β1, and γ1 chains, respectively, have garnered significant attention. This study aims to assess the potential of these radiolabeled peptides as targeting agents for breast cancer. The three peptides were synthesized using the Fmoc strategy, purified via reversed-phase high-performance liquid chromatography (RP-HPLC), and characterized through mass spectrometry. Iodine-131 (131I) radiolabeling was performed using the chloramine T method, exhibiting high radiochemical yield and stability for [131I]I-YIKVAV and [131I]I-YIGSR. Conversely, [131I]I-KAFDITYVRLKF demonstrated low radiochemical yield and stability and was excluded from the biological studies. The lipophilicity of the compounds ranged from - 2.12 to - 1.10. Serum protein binding assay for [131I]I-YIKVAV and [131I]I-YIGSR reached ≅ 48% and ≅ 25%, respectively. Affinity for breast cancer cells was evaluated using MDA-MB-231 and MCF-7 tumor cell lines, indicating the affinity of the radiopeptides with these tumor cells. Ex vivo biodistribution profiles of the radiopeptides were assessed in the MDA-MB-231 breast tumor animal model, revealing tumor tissue accumulation, supported by a high tumor-to-contralateral muscle ratio and autoradiography. These results signify the effective penetration of YIKVAV and YIGSR into tumor tissue. Therefore, the synthesized α1 and β1 peptide fragments exhibit favorable characteristics as potential breast cancer-targeting agents, promising future exploration as radiopharmaceuticals for breast cancer.
Collapse
Affiliation(s)
- Fernanda Ferreira Mendonça
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Rua Dr. Cesareo Motta Jr. 61, Sao Paulo, CEP 01221-020, Brazil
| | - Danielle Vieira Sobral
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Rua Dr. Cesareo Motta Jr. 61, Sao Paulo, CEP 01221-020, Brazil
| | - Ana Claudia Ranucci Durante
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Rua Dr. Cesareo Motta Jr. 61, Sao Paulo, CEP 01221-020, Brazil
| | | | - Jorge Mejia
- Hospital Israelita Albert Einstein, Sao Paulo, 05521-200, Brazil
| | - Daniele de Paula Faria
- Laboratory of Nuclear Medicine (LIM-43), Department of Radiology and Oncology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, 01246-903, Brazil
| | - Fabio Luiz Navarro Marques
- Laboratory of Nuclear Medicine (LIM-43), Department of Radiology and Oncology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, 01246-903, Brazil
| | | | - Leonardo Lima Fuscaldi
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Rua Dr. Cesareo Motta Jr. 61, Sao Paulo, CEP 01221-020, Brazil
| | - Luciana Malavolta
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Rua Dr. Cesareo Motta Jr. 61, Sao Paulo, CEP 01221-020, Brazil.
| |
Collapse
|
8
|
Li X, Lv X, Li H, Zhang G, Long Y, Li K, Fan Y, Jin D, Zhou F, Liu H. Undifferentially Expressed CXXC5 as a Transcriptionally Regulatory Biomarker of Breast Cancer. Adv Biol (Weinh) 2023; 7:e2300189. [PMID: 37423953 DOI: 10.1002/adbi.202300189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/17/2023] [Indexed: 07/11/2023]
Abstract
This work hypothesizes that some genes undergo radically changed transcription regulations (TRs) in breast cancer (BC), but don't show differential expressions for unknown reasons. The TR of a gene is quantitatively formulated by a regression model between the expression of this gene and multiple transcription factors (TFs). The difference between the predicted and real expression levels of a gene in a query sample is defined as the mqTrans value of this gene, which quantitatively reflects its regulatory changes. This work systematically screens the undifferentially expressed genes with differentially expressed mqTrans values in 1036 samples across five datasets and three ethnic groups. This study calls the 25 genes satisfying the above hypothesis in at least four datasets as dark biomarkers, and the strong dark biomarker gene CXXC5 (CXXC Finger Protein 5) is even supported by all the five independent BC datasets. Although CXXC5 does not show differential expressions in BC, its transcription regulations show quantitative associations with BCs in diversified cohorts. The overlapping long noncoding RNAs (lncRNAs) may have contributed their transcripts to the expression miscalculations of dark biomarkers. The mqTrans analysis serves as a complementary view of the transcriptome-based detections of biomarkers that are ignored by many existing studies.
Collapse
Affiliation(s)
- Xue Li
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
- School of Biology and Engineering, Guizhou Medical University, Guiyang, 550025, China
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Xiaoying Lv
- School of Biology and Engineering, Guizhou Medical University, Guiyang, 550025, China
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Haijun Li
- School of Biology and Engineering, Guizhou Medical University, Guiyang, 550025, China
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Gongyou Zhang
- School of Biology and Engineering, Guizhou Medical University, Guiyang, 550025, China
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Yaohang Long
- School of Biology and Engineering, Guizhou Medical University, Guiyang, 550025, China
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Kewei Li
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, 130012, China
- College of Computer Science and Technology, Jilin University, Changchun, 130012, China
| | - Yusi Fan
- College of Software, Jilin University, Changchun, 130012, China
| | - Dawei Jin
- Research Institute of Guizhou Huada Life Big Data, Guiyang, Guizhou, 550025, China
| | - Fengfeng Zhou
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, 130012, China
- College of Computer Science and Technology, Jilin University, Changchun, 130012, China
| | - Hongmei Liu
- School of Biology and Engineering, Guizhou Medical University, Guiyang, 550025, China
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang, Guizhou, 550025, China
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, 130012, China
| |
Collapse
|
9
|
Luo X, Zhang Z, Cheng C, Wang T, Fang D, Zuo C, Yuan G, Li R, Li X. SPECT Imaging with Tc-99m-Labeled HYNIC-FAPI-04 to Extend the Differential Time Window in Evaluating Tumor Fibrosis. Pharmaceuticals (Basel) 2023; 16:ph16030423. [PMID: 36986521 PMCID: PMC10051245 DOI: 10.3390/ph16030423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/16/2023] Open
Abstract
The so-far used Ga-68- or F-18-labelled tracers are of a relative short time window in differentiating tumor fibrosis. SPECT applicable imaging probe, 99mTc-HYNIC-FAPI-04, was synthesized and evaluated in tumor cells and animal models of FAP-positive glioma and FAP-negative hepatoma, and then compared with 18F-FDG or 68Ga-FAPI-04 PET/CT. The radio-labeling rate of 99mTc-HYNIC-FAPI-04 was greater than 90%, and the radiochemical purity was >99% after purification with sep-pak C18 column. In vitro cell uptake experiments of 99mTc-HYNIC-FAPI-04 showed good FAP binding specificity, and the cellular uptake significantly decreased when blocked by DOTA-FAPI-04, reflecting the similar targeting mechanism of HYNIC-FAPI-04 and DOTA-FAPI-04. SPECT/CT imaging showed that U87MG tumor was distinguishable and of a high uptake of 99mTc-HYNIC-FAPI-04 (2.67 ± 0.35 %ID/mL at 1.5 h post injection (h P.I.), while tumor signal of FAP-negative HUH-7 was as low as 0.34 ± 0.06 %ID/mL. At 5 h P.I., U87MG tumor was still distinguishable (1.81 ± 0.20 %ID/mL). In comparison, although U87MG tumor was of obvious 68Ga-FAPI-04 uptake and clearly visible at 1 h P.I., the tumorous radioactive signals were fuzzy at 1.5 h P.I. 99mTc-HYNIC-FAPI-04 specifically bound to FAP-positive tumors and qualified with the ability of evaluating tumor fibrosis over longer time windows.
Collapse
Affiliation(s)
- Xiu Luo
- Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai 200433, China (X.L.)
| | - Zhe Zhang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Chao Cheng
- Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai 200433, China (X.L.)
| | - Tao Wang
- Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai 200433, China (X.L.)
| | - Danzhou Fang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Changjing Zuo
- Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai 200433, China (X.L.)
| | - Gengbiao Yuan
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
- Correspondence: (G.Y.); (R.L.)
| | - Rou Li
- Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai 200433, China (X.L.)
- Correspondence: (G.Y.); (R.L.)
| | - Xiao Li
- Department of Nuclear Medicine, Shanghai Changhai Hospital, Shanghai 200433, China (X.L.)
| |
Collapse
|
10
|
Breast-Specific Gamma Imaging: An Added Value in the Diagnosis of Breast Cancer, a Systematic Review. Cancers (Basel) 2022; 14:cancers14194619. [PMID: 36230540 PMCID: PMC9559460 DOI: 10.3390/cancers14194619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Breast-specific gamma imaging represents an emergent instrument for breast cancer detection. We selected on Medline articles published from 1995 to 2022 that compare various imaging modalities with breast-specific gamma imaging. The aim of this paper was to assess if this imaging method is a more valuable choice in detecting breast malignant lesions compared to morphological counterparts such mammography, ultrasound, and magnetic resonance imaging in terms of specificity, sensibility and positive and negative predictive value. At the cost of a major radiology burden, breast-specific gamma imaging is more specific, with a sensibility comparable to magnetic resonance imaging and higher than ultrasonography and mammography. Abstract Purpose: Breast cancer is the most common solid tumor and the second highest cause of death in the United States. Detection and diagnosis of breast tumors includes various imaging modalities, such as mammography (MMG), ultrasound (US), and contrast-enhancement MRI. Breast-specific gamma imaging (BSGI) is an emerging tool, whereas morphological imaging has the disadvantage of a higher absorbed dose. Our aim was to assess if this imaging method is a more valuable choice in detecting breast malignant lesions compared to morphological counterparts. Methods: research on Medline from 1995 to June 2022 was conducted. Studies that compared at least one anatomical imaging modality with BSGI were screened and assessed through QUADAS2 for risk of bias and applicability concerns assessment. Sensitivity, specificity, positive and negative predictive value (PPV and NPV) were reported. Results: A total of 15 studies compared BSGI with MMG, US, and MRI. BSGI sensitivity was similar to MRI, but specificity was higher. Specificity was always higher than MMG and US. BSGI had higher PPV and NPV. When used for the evaluation of a suspected breast lesion, the overall sensitivity was better than the examined overall sensitivity when BSGI was excluded. Risk of bias and applicability concerns domain showed mainly low risk of bias. Conclusion: BSGI is a valuable imaging modality with similar sensitivity to MRI but higher specificity, although at the cost of higher radiation burden.
Collapse
|
11
|
Mat Isa SZ, Zainon R, Tamal M. State of the Art in Gold Nanoparticle Synthesisation via Pulsed Laser Ablation in Liquid and Its Characterisation for Molecular Imaging: A Review. MATERIALS (BASEL, SWITZERLAND) 2022; 15:875. [PMID: 35160822 PMCID: PMC8838486 DOI: 10.3390/ma15030875] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/11/2022]
Abstract
With recent advances in nanotechnology, various nanomaterials have been used as drug carriers in molecular imaging for the treatment of cancer. The unique physiochemical properties and biocompatibility of gold nanoparticles have developed a breakthrough in molecular imaging, which allows exploration of gold nanoparticles in drug delivery for diagnostic purpose. The conventional gold nanoparticles synthetisation methods have limitations with chemical contaminations during the synthesisation process and the use of higher energy. Thus, various innovative approaches in gold nanoparticles synthetisation are under development. Recently, studies have been focused on the development of eco-friendly, non-toxic, cost-effective and simple gold nanoparticle synthesisation. The pulsed laser ablation in liquid (PLAL) technique is a versatile synthetic and convincing technique due to its high efficiency, eco-friendly and facile method to produce gold nanoparticle. Therefore, this study aimed to review the eco-friendly gold nanoparticle synthesisation method via the PLAL method and to characterise the gold nanoparticles properties for molecular imaging. This review paper provides new insight to understand the PLAL technique in producing gold nanoparticles and the PLAL parameters that affect gold nanoparticle properties to meet the desired needs in molecular imaging.
Collapse
Affiliation(s)
- Siti Zaleha Mat Isa
- Department of Biomedical Imaging, Advanced Medical and Dental Institute, Universiti Sains Malaysia, SAINS@BERTAM, Kepala Batas 13200, Pulau Pinang, Malaysia;
| | - Rafidah Zainon
- Department of Biomedical Imaging, Advanced Medical and Dental Institute, Universiti Sains Malaysia, SAINS@BERTAM, Kepala Batas 13200, Pulau Pinang, Malaysia;
| | - Mahbubunnabi Tamal
- Department of Biomedical Engineering, College of Engineering, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia;
| |
Collapse
|
12
|
Zhang B, Tian T, Xiao D, Gao S, Cai X, Lin Y. Facilitating In Situ Tumor Imaging with a Tetrahedral DNA Framework‐Enhanced Hybridization Chain Reaction Probe. ADVANCED FUNCTIONAL MATERIALS 2022. [DOI: 10.1002/adfm.202109728] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Bowen Zhang
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Sichuan Chengdu 610041 China
| | - Taoran Tian
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Sichuan Chengdu 610041 China
| | - Dexuan Xiao
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Sichuan Chengdu 610041 China
| | - Shaojingya Gao
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Sichuan Chengdu 610041 China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Sichuan Chengdu 610041 China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases National Clinical Research Center for Oral Diseases West China Hospital of Stomatology Sichuan University Sichuan Chengdu 610041 China
- College of Biomedical Engineering Sichuan University Sichuan Chengdu 610041 China
| |
Collapse
|
13
|
Han L, Song X. Lutein induces an inhibitory effect on the malignant progression of pancreatic adenocarcinoma by targeting BAG3/cholesterol homeostasis. J Biochem Mol Toxicol 2021; 36:e22958. [PMID: 34783111 DOI: 10.1002/jbt.22958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/07/2021] [Accepted: 10/26/2021] [Indexed: 11/06/2022]
Abstract
Pancreatic adenocarcinoma (PDAC) is a fatal malignancy and patients with PDAC are mostly diagnosed at advanced stages. Lutein is a natural compound that belongs to the non-vitamin A carotenoids family and has presented antitumor effects on multiple cancer types. However, the function of lutein in PDAC and the mechanisms are not reported. Here, we explored the role of lutein in PDAC progression. Bioinformatic analysis identified that lutein is correlated with PDAC. Lutein suppressed the proliferation, migration, and invasion of PANC-1 cells. The upregulated genes in PDAC patients were identified and the overlap analysis predicted BAG3 as one target of lutein. Lutein repressed BAG3 expression and bioinformatics analysis predicted the interaction between lutein and BAG3. The inhibitory effects of lutein on PANC-1 cell proliferation, migration, and invasion are reversed by overexpression of BAG3. GSEA analysis identified that cholesterol homeostasis as one of the downstream signaling pathways of BAG3. In conclusion, lutein induced an inhibitory effect on the malignant progression of PDAC by targeting BAG3/cholesterol homeostasis. Lutein may be applied as a promising candidate for PDAC therapy.
Collapse
Affiliation(s)
- Lulu Han
- Department of Neurology Ward 3, The Fifth People's Hospital of Jinan, Jinan, Shandong, P.R. China
| | - Xiaoming Song
- Department of Gerontology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, P.R. China
| |
Collapse
|
14
|
Najafi M, Majidpoor J, Toolee H, Mortezaee K. The current knowledge concerning solid cancer and therapy. J Biochem Mol Toxicol 2021; 35:e22900. [PMID: 34462987 DOI: 10.1002/jbt.22900] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 07/29/2021] [Accepted: 08/20/2021] [Indexed: 12/25/2022]
Abstract
Solid cancers comprise a large number of new cases and deaths from cancer each year globally. There are a number of strategies for addressing tumors raised from solid organs including surgery, chemotherapy, radiotherapy, targeted therapy, immunotherapy, combinational therapy, and stem cell and extracellular vesicle (EV) therapy. Surgery, radiotherapy, and chemotherapy are the dominant cures, but are not always effective, in which even in a localized tumor there is a possibility of tumor relapse after surgical resection. Over half of the cancer patients will receive radiotherapy as a part of their therapeutic schedule. Radiotherapy can cause an abscopal response for boosting the activity of the immune system outside the local field of radiation, but it may also cause an unwanted bystander effect, predisposing nonradiated cells into carcinogenesis. In the context of immunotherapy, immune checkpoint inhibition is known as the standard-of-care, but the major concern is in regard with cold cancers that show low responses to such therapy. Stem-cell therapy can be used to send prodrugs toward the tumor area; this strategy, however, has its own predicaments, such as unwanted attraction toward the other sites including healthy tissues and its instability. A substitute to such therapy and quite a novel strategy is to use EVs, by virtue of their stability and potential to cross biological barriers and long-term storage of contents. Combination therapy is the current focus. Despite advances in the field, there are still unmet concerns in the area of effective cancer therapy, raising challenges and opportunities for future investigations.
Collapse
Affiliation(s)
- Masoud Najafi
- Medical Technology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Heidar Toolee
- Department of Anatomy, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Keywan Mortezaee
- Cancer and Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran.,Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
15
|
Kim SJ, Lee JH, Park WJ, Kim S. Bioinformatic Exploration for Prognostic Significance of Sphingolipid Metabolism-Related Genes in Invasive Ductal Carcinoma Using the Cancer Genome Atlas Cohort. Int J Gen Med 2021; 14:4423-4434. [PMID: 34413672 PMCID: PMC8370849 DOI: 10.2147/ijgm.s328376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 07/29/2021] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Sphingolipid metabolism is a highly controlled process that is involved in regulating bioactive lipid signaling pathways and serves important roles in several cellular processes in breast cancer. Invasive ductal carcinoma (IDC), which is characterized by the malignant proliferation of the ductal epithelium and stromal invasion, is the most common type of breast cancer. Recent advances in genetic research have accelerated the discovery of novel prognostic factors and therapeutic targets for the disease. The aim of the present study was to investigate the expression and prognostic significance of sphingolipid metabolism-related genes in female IDC. METHODS The present study used gene expression RNAseq data obtained from The Cancer Genome Atlas breast invasive carcinoma (TCGA BRCA) datasets. RESULTS Sphingolipid metabolism-related genes exhibited dysregulated mRNA expression levels in IDC. The Student's t-test revealed that SMPDL3B, B4GALNT1, LPAR2, and LASS2 were significantly upregulated, while LASS3, LPAR1, B4GALT6, GAL3ST1, HPGD, ST8SIA1, UGT8, and S1PR1 were significantly downregulated in female IDC tissues compared with normal solid tissues. Kaplan-Meier survival analyses revealed that high SMPDL3B mRNA expression levels were associated with good prognosis in female IDC, suggesting that SMPDL3B plays a tumor suppressor role. To the best of our knowledge, the present study was the first to report that dysregulated expressions of SMPDL3B are significantly associated with age, estrogen receptor status, progesterone receptor status, and histological subtype. CONCLUSION Taken together, our study indicated that SMPDL3B may have a pathophysiological role and serve as a novel prognostic biomarker in IDC.
Collapse
Affiliation(s)
- Su-Jeong Kim
- Department of Biochemistry, College of Medicine, Gachon University, Yeonsu-gu, Incheon, 21999, Republic of Korea
| | - Jae-Ho Lee
- Department of Anatomy, School of Medicine, Keimyung University, Dalseo-gu, Daegu, 42601, Republic of Korea
| | - Woo-Jae Park
- Department of Biochemistry, College of Medicine, Gachon University, Yeonsu-gu, Incheon, 21999, Republic of Korea
| | - Shin Kim
- Department of Immunology, School of Medicine, Keimyung University, Dalseo-gu, Daegu, 42601, Republic of Korea
- Institute of Medical Science, Keimyung University, Dalseo-gu, Daegu, 42601, Republic of Korea
- Institute for Cancer Research, Keimyung University Dongsan Medical Center, Dalseo-gu, Daegu, 42601, Republic of Korea
| |
Collapse
|
16
|
Hormati A, Shiran JA, Molazadeh M, Kaboudin B, Ahmadpour S. Synthesis of New Thioureas Derivatives and Evaluation of Their Efficacy as Proliferation Inhibitors in MCF-7 Breast Cancer Cells by Using 99mTc-MIBI Radiotracer. Med Chem 2021; 17:766-778. [PMID: 32334505 DOI: 10.2174/1573406416666200425224921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 03/26/2020] [Accepted: 03/26/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND OBJECTIVES Anti-tumor activity of some thioureas derivatives is well documented in literature and received considerable attention. The present study aims to synthesize and characterize some novel thioureas and carbonylthioureas as anti-tumor agents for MCF-7 breast cancer cells in vitro and in vivo. MATERIALS AND METHODS Several 1-allyl-3-(substituted phenyl), N,N'-(phenylene) bis(3- allyldithithiourea) and 1-cyclopropanecarbonyl-3-(substituted phenyl)-thioureas derivatives were synthesized and confirmed by FT-IR spectroscopy, NMR and 13C-NMR. Anti-tumor activity of these compounds was determined by various in vitro and in vivo assays including; MTT, tumor volume measurement as well as,99mTc-MIBI tumor uptake in MCF-7 tumor bearing nude mice. RESULTS Among all of the synthesized compounds, some thioureas derivatives [3i] and [4b] at 100 nM concentration exhibited significant inhibitory effects on the proliferation of MCF-7 cell in vitro. However, this inhibition was not observed in HUVEC human endothelial normal cells. In vivo anti-tumor effects of the synthesized compounds on MCF-7 xenograft mouse models demonstrated a reduction in the tumor volume for various concentrations between 2 to 10 mg/kg after 21 days. These effects were comparable with Tamoxifen as standard anti-estrogen drug. According to the 99mTc-MIBI biodistribution result, treatment of MCF-7 bearing nude mice with both [3i] and [4b] compounds at the maximum concentration (10 mg/kg) can lead to a significant decrease of 99mTc- MIBI tumor uptake. CONCLUSION Compounds [3i] and [4b] suppressed the growth of MCF-7 cells in the xenograft nude mice at the doses that were well-tolerated. Our study suggests that these new compounds with their significant anti-tumor effects, may serve as useful candidates for breast cancer therapy.
Collapse
Affiliation(s)
- Ahmad Hormati
- Gastroenterology and Hepatology Diseases Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Jafar Abbasi Shiran
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| | - Mikaeil Molazadeh
- Department of Medical Physics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Babak Kaboudin
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| | - Sajjad Ahmadpour
- Gastroenterology and Hepatology Diseases Research Center, Qom University of Medical Sciences, Qom, Iran
| |
Collapse
|
17
|
Kręcisz P, Czarnecka K, Królicki L, Mikiciuk-Olasik E, Szymański P. Radiolabeled Peptides and Antibodies in Medicine. Bioconjug Chem 2020; 32:25-42. [PMID: 33325685 PMCID: PMC7872318 DOI: 10.1021/acs.bioconjchem.0c00617] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
Radiolabeled peptides
are a relatively new, very specific radiotracer
group, which is still expanding. This group is very diverse in terms
of peptide size. It contains very small structures containing several
amino acids and whole antibodies. Moreover, radiolabeled peptides
are diverse in terms of the binding aim and therapeutic or diagnostic
applications. The majority of this class of radiotracers is utilized
in oncology, where the same structure can be used in therapy and diagnostic
imaging by varying the radionuclide. In this study, we collected new
reports of radiolabeled peptide applications in diagnosis and therapy
in oncology and other fields of medicine. Radiolabeled peptides are
also increasingly being used in rheumatology, cardiac imaging, or
neurology. The studies collected in this review concern new therapeutic
and diagnostic procedures in humans and new structures tested on animals.
We also performed an analysis of clinical trials, which concerns application
of radiolabeled peptides and antibodies that were reported in the
clinicaltrials.gov database between 2008 and 2018.
Collapse
Affiliation(s)
- Paweł Kręcisz
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland
| | - Kamila Czarnecka
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland
| | - Leszek Królicki
- Department of Nuclear Medicine, Medical University of Warsaw, ul. Banacha 1 a, 02-097, Warsaw, Poland
| | - Elżbieta Mikiciuk-Olasik
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland
| | - Paweł Szymański
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland
| |
Collapse
|
18
|
Hormati A, Ahmadpour S, Afkhami Ardekani M, Khodadust F, Refahi S. Radioprotective effects of montelukast, a selective leukotriene CysLT1 receptor antagonist, against nephrotoxicity induced by gamma radiation in mice. J Biochem Mol Toxicol 2020; 34:e22479. [PMID: 32125029 DOI: 10.1002/jbt.22479] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/24/2019] [Accepted: 02/14/2020] [Indexed: 12/30/2022]
Abstract
PURPOSE In this study, we evaluated the renal protective effects of montelukast (MLK) against ionizing radiation (IR) induced nephrotoxicity in mice. MATERIALS AND METHODS Radioprotective effects of MLK were assessed by biochemical analysis including measurements of kidney malondialdehyde (MDA), reduced glutathione (GSH), and serum creatinine and urea levels. Besides, for further evaluation of protective effects of MLK on renal system, 99m Tc-dimercaptosuccinic acid (DMSA) has been applied. The total antioxidant capacity of MLK was measured by using 1,1-diphenyl-2-picryl hydrazyl radical reagents and compared with butylated hydroxyl toluene standard antioxidant. RESULTS The biochemical evaluation revealed that better results have been achieved for the groups administered with MLK than the only radiation group. Besides only IR-treated mice group, those treated with MLK demonstrated a significant decrease in urea and creatinine levels. Statistically, significant differences of MDA and SHG levels (P < .05) were found between the radiation group and MLK plus IR-treated group. Also, 99m Tc-DMSA kidney uptake value (%ID/g) was observed lower for MLK plus IR-treated mice group than only radiation-treated mice group. CONCLUSIONS According to our findings, MLK has a potential role to be used as a renal protective agent against gamma radiation in radiotherapy.
Collapse
Affiliation(s)
- Ahmad Hormati
- Gastroenterology and Hepatology Disease Research Center, Qom University of Medical Sciences, Qom, Iran.,Gastrointestinal and Liver Disease Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sajjad Ahmadpour
- Gastroenterology and Hepatology Disease Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Mahdieh Afkhami Ardekani
- Department of Radiology, Faculty of Para-Medicine, Hormozgan University of Medical Sciences, Bandare Abbas, Iran
| | - Fatemeh Khodadust
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Soheila Refahi
- Department of Medical Physics, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
19
|
Ahmadpour S, Hosseinimehr SJ. Recent developments in peptide-based SPECT radiopharmaceuticals for breast tumor targeting. Life Sci 2019; 239:116870. [DOI: 10.1016/j.lfs.2019.116870] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 12/31/2022]
|
20
|
Farzipour S, Hosseinimehr SJ. Correlation between in vitro and in vivo Data of Radiolabeled Peptide for Tumor Targeting. Mini Rev Med Chem 2019; 19:950-960. [DOI: 10.2174/1389557519666190304120011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/28/2018] [Accepted: 02/22/2019] [Indexed: 12/12/2022]
Abstract
Tumor-targeting peptides have been generally developed for the overexpression of tumor specific receptors in cancer cells. The use of specific radiolabeled peptide allows tumor visualization by single photon emission computed tomography (SPECT) and positron emission tomography (PET) tools. The high affinity and specific binding of radiolabeled peptide are focusing on tumoral receptors. The character of the peptide itself, in particular, its complex molecular structure and behaviors influence on its specific interaction with receptors which are overexpressed in tumor. This review summarizes various strategies which are applied for the expansion of radiolabeled peptides for tumor targeting based on in vitro and in vivo specific tumor data and then their data were compared to find any correlation between these experiments. With a careful look at previous studies, it can be found that in vitro unblock-block ratio was unable to correlate the tumor to muscle ratio and the success of radiolabeled peptide for in vivo tumor targeting. The introduction of modifiers’ approaches, nature of peptides, and type of chelators and co-ligands have mixed effect on the in vitro and in vivo specificity of radiolabeled peptides.
Collapse
Affiliation(s)
- Soghra Farzipour
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|