1
|
Weijie S. Annexin A2: the feasibility of being a therapeutic target associated with cancer metastasis and drug resistance in cancer microenvironment. Discov Oncol 2024; 15:783. [PMID: 39692932 DOI: 10.1007/s12672-024-01693-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/10/2024] [Indexed: 12/19/2024] Open
Abstract
At present, there is still a lack of effective treatment strategies for cancer metastasis and drug resistance, so finding effective biomarkers is particularly important. AnnexinA2 (ANXA2), a vital membrane protein, critically influences cancer progression, tumor invasion, and tumor microenvironment modulation. To assess the possible application of ANXA2 as a therapeutic target against cancer cell metastasis and drug resistance to chemotherapeutic drugs in the tumor microenvironment, we elucidated the functionality of ANXA2 in stromal cells, angiogenic vascular cells, and infiltrated immune cells that mediate metastasis and drug resistance, as well as its potential as a therapeutic target. ANXA2 shows a high expression level in many tissues, and its expression level is even higher in several tumors and their microenvironments. ANXA2 is a crucial regulator of many factors and may serve as a target against drug-resistant cancers.
Collapse
Affiliation(s)
- Song Weijie
- Laboratory Animal Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin, 300060, China.
| |
Collapse
|
2
|
Mabunda IG, Zinyemba NK, Pillay S, Offor BC, Muller B, Piater LA. The geographical distribution of scorpions, implication of venom toxins, envenomation, and potential therapeutics in Southern and Northern Africa. Toxicol Res (Camb) 2024; 13:tfae118. [PMID: 39100857 PMCID: PMC11298049 DOI: 10.1093/toxres/tfae118] [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: 05/28/2024] [Revised: 07/01/2024] [Accepted: 07/25/2024] [Indexed: 08/06/2024] Open
Abstract
Scorpions are predatory arachnids whose venomous sting primarily affects people in tropical and subtropical regions. Most scorpion stings can only cause localized pain without severe envenomation. Less than one-third of the stings cause systemic envenoming and possibly lead to death. About 350,000 scorpion stings in Northern Africa are recorded yearly, resulting in about 810 deaths. In Eastern/Southern Africa, there are about 79,000 stings recorded yearly, resulting in 245 deaths. Farmers and those living in poverty-stricken areas are among the most vulnerable to getting stung by scorpions. However, compared to adults, children are at greater risk of severe envenomation. Scorpion venom is made up of complex mixtures dominated by peptides and proteins that confer its potency and toxicity. These venom toxins have intra- and interspecies variations associated with the scorpion's habitat, sex, diet, and age. These variations alter the activity of antivenoms used to treat scorpion sting envenomation. Thus, the study of the proteome composition of medically important scorpion venoms needs to be scaled up along their geographical distribution and contributions to envenomation in Southern and Northern Africa. This will help the production of safer, more effective, and broad-spectrum antivenoms within these regions. Here, we review the clinical implications of scorpion sting envenomation in Southern and Northern Africa. We further highlight the compositions of scorpion venoms and tools used in scorpion venomics. We discuss current antivenoms used against scorpion sting envenomation and suggestions for future production of better antivenoms or alternatives. Finally, we discuss the therapeutic properties of scorpion venom.
Collapse
Affiliation(s)
- Isac G Mabunda
- Department of Biochemistry, Corner of Kingsway and University Road, Auckland Park Campus, University of Johannesburg, Auckland Park, 2006, Gauteng, South Africa
| | - Nodji K Zinyemba
- Department of Biochemistry, Corner of Kingsway and University Road, Auckland Park Campus, University of Johannesburg, Auckland Park, 2006, Gauteng, South Africa
| | - Shanelle Pillay
- Department of Biochemistry, Corner of Kingsway and University Road, Auckland Park Campus, University of Johannesburg, Auckland Park, 2006, Gauteng, South Africa
| | - Benedict C Offor
- Department of Biochemistry, Corner of Kingsway and University Road, Auckland Park Campus, University of Johannesburg, Auckland Park, 2006, Gauteng, South Africa
| | - Beric Muller
- South Africa Venom Suppliers cc, 41 Louis, Trichardt 0920, South Africa
| | - Lizelle A Piater
- Department of Biochemistry, Corner of Kingsway and University Road, Auckland Park Campus, University of Johannesburg, Auckland Park, 2006, Gauteng, South Africa
| |
Collapse
|
3
|
Chow CY, King GF. Shining a Light on Venom-Peptide Receptors: Venom Peptides as Targeted Agents for In Vivo Molecular Imaging. Toxins (Basel) 2024; 16:307. [PMID: 39057947 PMCID: PMC11281729 DOI: 10.3390/toxins16070307] [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: 05/13/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Molecular imaging has revolutionised the field of biomedical research by providing a non-invasive means to visualise and understand biochemical processes within living organisms. Optical fluorescent imaging in particular allows researchers to gain valuable insights into the dynamic behaviour of a target of interest in real time. Ion channels play a fundamental role in cellular signalling, and they are implicated in diverse pathological conditions, making them an attractive target in the field of molecular imaging. Many venom peptides exhibit exquisite selectivity and potency towards ion channels, rendering them ideal agents for molecular imaging applications. In this review, we illustrate the use of fluorescently-labelled venom peptides for disease diagnostics and intraoperative imaging of brain tumours and peripheral nerves. Finally, we address challenges for the development and clinical translation of venom peptides as nerve-targeted imaging agents.
Collapse
Affiliation(s)
- Chun Yuen Chow
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
- Australia Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Glenn F. King
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
- Australia Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St. Lucia, QLD 4072, Australia
| |
Collapse
|
4
|
Xia Z, He D, Wu Y, Kwok HF, Cao Z. Scorpion venom peptides: Molecular diversity, structural characteristics, and therapeutic use from channelopathies to viral infections and cancers. Pharmacol Res 2023; 197:106978. [PMID: 37923027 DOI: 10.1016/j.phrs.2023.106978] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/07/2023]
Abstract
Animal venom is an important evolutionary innovation in nature. As one of the most representative animal venoms, scorpion venom contains an extremely diverse set of bioactive peptides. Scorpion venom peptides not only are 'poisons' that immobilize, paralyze, kill, or dissolve preys but also become important candidates for drug development and design. Here, the review focuses on the molecular diversity of scorpion venom peptides, their typical structural characteristics, and their multiple therapeutic or pharmaceutical applications in channelopathies, viral infections and cancers. Especially, the group of scorpion toxin TRPTx targeting transient receptor potential (TRP) channels is systematically summarized and worthy of attention because TRP channels play a crucial role in the regulation of homeostasis and the occurrence of diseases in human. We also further establish the potential relationship between the molecular characteristics and functional applications of scorpion venom peptides to provide a research basis for modern drug development and clinical utilization of scorpion venom resources.
Collapse
Affiliation(s)
- Zhiqiang Xia
- School of Biological and Food Processing Engineering, Huanghuai University, Zhumadian, China
| | - Dangui He
- State Key Laboratory of Virology, College of Life Sciences, Shenzhen Research Institute, Wuhan University, Wuhan, China; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao; Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao
| | - Yingliang Wu
- State Key Laboratory of Virology, College of Life Sciences, Shenzhen Research Institute, Wuhan University, Wuhan, China
| | - Hang Fai Kwok
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao; Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao; MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macao.
| | - Zhijian Cao
- State Key Laboratory of Virology, College of Life Sciences, Shenzhen Research Institute, Wuhan University, Wuhan, China; Bio-drug Research Center, Wuhan University, Wuhan, China.
| |
Collapse
|
5
|
Wang Q, Xin X, Dai Q, Sun M, Chen J, Mostafavi E, Shen Y, Li X. Medulloblastoma targeted therapy: From signaling pathways heterogeneity and current treatment dilemma to the recent advances in development of therapeutic strategies. Pharmacol Ther 2023; 250:108527. [PMID: 37703952 DOI: 10.1016/j.pharmthera.2023.108527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/27/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Abstract
Medulloblastoma (MB) is a major pediatric malignant brain tumor that arises in the cerebellum. MB tumors exhibit highly heterogeneous driven by diverse genetic alterations and could be divided into four major subgroups based on their different biological drivers and molecular features (Wnt, Sonic hedgehog (Shh), group 3, and group 4 MB). Even though the therapeutic strategies for each MB subtype integrate their pathogenesis and were developed to focus on their specific target sites, the unexpected drug non-selective cytotoxicity, low drug accumulation in the brain, and complexed MB tumor microenvironment still be huge obstacles to achieving satisfied MB therapeutic efficiency. This review discussed the current advances in modern MB therapeutic strategy development. Through the recent advances in knowledge of the origin, molecular pathogenesis of MB subtypes and their current therapeutic barriers, we particularly reviewed the current development in advanced MB therapeutic strategy committed to overcome MB treatment obstacles, focusing on novel signaling pathway targeted therapeutic agents and their combination discovery, advanced drug delivery systems design, and MB immunotherapy strategy development.
Collapse
Affiliation(s)
- Qiyue Wang
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing 211816, China
| | - Xiaofei Xin
- Center for Research Development and Evaluation of Pharmaceutical Excipients and Generic Drugs, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Qihao Dai
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing 211816, China
| | - Mengjuan Sun
- Center for Research Development and Evaluation of Pharmaceutical Excipients and Generic Drugs, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Jinhua Chen
- Center for Research Development and Evaluation of Pharmaceutical Excipients and Generic Drugs, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Ebrahim Mostafavi
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Yan Shen
- Center for Research Development and Evaluation of Pharmaceutical Excipients and Generic Drugs, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Xueming Li
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing 211816, China.
| |
Collapse
|
6
|
Farkas S, Cioca D, Murányi J, Hornyák P, Brunyánszki A, Szekér P, Boros E, Horváth P, Hujber Z, Rácz GZ, Nagy N, Tóth R, Nyitray L, Péterfi Z. Chlorotoxin binds to both matrix metalloproteinase 2 and neuropilin 1. J Biol Chem 2023; 299:104998. [PMID: 37394009 PMCID: PMC10477481 DOI: 10.1016/j.jbc.2023.104998] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/03/2023] [Accepted: 06/28/2023] [Indexed: 07/04/2023] Open
Abstract
Chlorotoxin (CTX), a scorpion venom-derived 36-residue miniprotein, binds to and is taken up selectively by glioblastoma cells. Previous studies provided controversial results concerning target protein(s) of CTX. These included CLC3 chloride channel, matrix metalloproteinase 2 (MMP-2), regulators of MMP-2, annexin A2, and neuropilin 1 (NRP1). The present study aimed at clarifying which of the proposed binding partners can really interact with CTX using biochemical methods and recombinant proteins. For this purpose, we established two new binding assays based on anchoring the tested proteins to microbeads and quantifying the binding of CTX by flow cytometry. Screening of His-tagged proteins anchored to cobalt-coated beads indicated strong interaction of CTX with MMP-2 and NRP1, whereas binding to annexin A2 was not confirmed. Similar results were obtained with fluorophore-labeled CTX and CTX-displaying phages. Affinity of CTX to MMP-2 and NRP1 was assessed by the "immunoglobulin-coated bead" test, in which the proteins were anchored to beads by specific antibodies. This assay yielded highly reproducible data using both direct titration and displacement approach. The affinities of labeled and unlabeled CTX appeared to be similar for both MMP-2 and NRP1 with estimated KD values of 0.5 to 0.7 μM. Contrary to previous reports, we found that CTX does not inhibit the activity of MMP-2 and that CTX not only with free carboxyl end but also with carboxamide terminal end binds to NRP1. We conclude that the presented robust assays could also be applied for affinity-improving studies of CTX to its genuine targets using phage display libraries.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Eszter Boros
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Patrik Horváth
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | | | | | | | | | - László Nyitray
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | | |
Collapse
|
7
|
Boltman T, Meyer M, Ekpo O. Diagnostic and Therapeutic Approaches for Glioblastoma and Neuroblastoma Cancers Using Chlorotoxin Nanoparticles. Cancers (Basel) 2023; 15:3388. [PMID: 37444498 DOI: 10.3390/cancers15133388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 07/15/2023] Open
Abstract
Glioblastoma multiforme (GB) and high-risk neuroblastoma (NB) are known to have poor therapeutic outcomes. As for most cancers, chemotherapy and radiotherapy are the current mainstay treatments for GB and NB. However, the known limitations of systemic toxicity, drug resistance, poor targeted delivery, and inability to access the blood-brain barrier (BBB), make these treatments less satisfactory. Other treatment options have been investigated in many studies in the literature, especially nutraceutical and naturopathic products, most of which have also been reported to be poorly effective against these cancer types. This necessitates the development of treatment strategies with the potential to cross the BBB and specifically target cancer cells. Compounds that target the endopeptidase, matrix metalloproteinase 2 (MMP-2), have been reported to offer therapeutic insights for GB and NB since MMP-2 is known to be over-expressed in these cancers and plays significant roles in such physiological processes as angiogenesis, metastasis, and cellular invasion. Chlorotoxin (CTX) is a promising 36-amino acid peptide isolated from the venom of the deathstalker scorpion, Leiurus quinquestriatus, demonstrating high selectivity and binding affinity to a broad-spectrum of cancers, especially GB and NB through specific molecular targets, including MMP-2. The favorable characteristics of nanoparticles (NPs) such as their small sizes, large surface area for active targeting, BBB permeability, etc. make CTX-functionalized NPs (CTX-NPs) promising diagnostic and therapeutic applications for addressing the many challenges associated with these cancers. CTX-NPs may function by improving diffusion through the BBB, enabling increased localization of chemotherapeutic and genotherapeutic drugs to diseased cells specifically, enhancing imaging modalities such as magnetic resonance imaging (MRI), single-photon emission computed tomography (SPECT), optical imaging techniques, image-guided surgery, as well as improving the sensitization of radio-resistant cells to radiotherapy treatment. This review discusses the characteristics of GB and NB cancers, related treatment challenges as well as the potential of CTX and its functionalized NP formulations as targeting systems for diagnostic, therapeutic, and theranostic purposes. It also provides insights into the potential mechanisms through which CTX crosses the BBB to bind cancer cells and provides suggestions for the development and application of novel CTX-based formulations for the diagnosis and treatment of GB and NB in the future.
Collapse
Affiliation(s)
- Taahirah Boltman
- Department of Medical Biosciences, University of the Western Cape, Robert Sobukwe Road, Bellville, Cape Town 7535, South Africa
| | - Mervin Meyer
- Department of Science and Innovation/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, University of the Western Cape, Robert Sobukwe Road, Bellville, Cape Town 7535, South Africa
| | - Okobi Ekpo
- Department of Anatomy and Cellular Biology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| |
Collapse
|
8
|
Bioactive peptides from scorpion venoms: therapeutic scaffolds and pharmacological tools. Chin J Nat Med 2023; 21:19-35. [PMID: 36641229 DOI: 10.1016/s1875-5364(23)60382-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Indexed: 01/14/2023]
Abstract
Evolution and natural selection have endowed animal venoms, including scorpion venoms, with a wide range of pharmacological properties. Consequently, scorpions, their venoms, and/or their body parts have been used since time immemorial in traditional medicines, especially in Africa and Asia. With respect to their pharmacological potential, bioactive peptides from scorpion venoms have become an important source of scientific research. With the rapid increase in the characterization of various components from scorpion venoms, a large number of peptides are identified with an aim of combating a myriad of emerging global health problems. Moreover, some scorpion venom-derived peptides have been established as potential scaffolds helpful for drug development. In this review, we summarize the promising scorpion venoms-derived peptides as drug candidates. Accordingly, we highlight the data and knowledge needed for continuous characterization and development of additional natural peptides from scorpion venoms, as potential drugs that can treat related diseases.
Collapse
|
9
|
Ma H, Jiang K, Hong Y, Lei Y, Fan Y, Jiang W, Zhao L, Liu J, Yao W, Xu J, He M, Wei M. Screening of an annexin-A2-targeted heptapeptide for pancreatic adenocarcinoma localization. Mol Oncol 2022; 17:872-886. [PMID: 36453020 PMCID: PMC10158761 DOI: 10.1002/1878-0261.13352] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/04/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022] Open
Abstract
Annexin A2 (ANXA2) encodes an oncoprotein whose expression has been found to correlate with poorer overall survival (OS) of pancreatic adenocarcinoma (PAAD) patients. Although peptides are available for targeting ANXA2, none of these were initially selected to target this protein specifically. Here, we took ANXA2 as a molecular target for PAAD and employed the phage display technique to screen for a new ANXA2-targeted peptide. The resultant heptapeptide, YW7, was firstly labeled with fluorescein isothiocyanate (FITC) to evaluate its selectivity in cellular uptake, and further with the near-infrared fluorescent (NIRF) dye Cy7 to assess in vivo distribution in a mouse model bearing PANC-1 human pancreatic cancer xenografic tumors. We found that both FITC-YW7 and Cy7-YW7 probes showed significantly higher uptake in PANC-1 cells compared to the HPDE6-C7 pancreatic epithelium cells. Mice intravenously injected with Cy7-YW7 showed higher tumor-to-background ratios (TBRs) (~ 2.7-fold) in tumor tissues compared to those injected with Cy7 alone. Our study suggested that YW7 is a novel peptide targeting ANXA2 and Cy7-YW7 is an NIRF probe potentially useful for the early detection of PAAD.
Collapse
Affiliation(s)
- Heyao Ma
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Kai Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Yuhan Hong
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Yu Lei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Yue Fan
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Wenjian Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Lei Zhao
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of China Medical University, Shenyang, China
| | - Jinyang Liu
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of China Medical University, Shenyang, China
| | - Weifan Yao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Jiao Xu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Miao He
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China.,Liaoning Medical Diagnosis and Treatment Center, Shenyang, China
| |
Collapse
|
10
|
Shrestha A, Lahooti B, Mikelis CM, Mattheolabakis G. Chlorotoxin and Lung Cancer: A Targeting Perspective for Drug Delivery. Pharmaceutics 2022; 14:pharmaceutics14122613. [PMID: 36559106 PMCID: PMC9786857 DOI: 10.3390/pharmaceutics14122613] [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: 10/21/2022] [Revised: 11/11/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
In the generational evolution of nano-based drug delivery carriers, active targeting has been a major milestone for improved and selective drug accumulation in tissues and cell types beyond the existing passive targeting capabilities. Among the various active targeting moieties, chlorotoxin, a peptide extracted from scorpions, demonstrated promising tumor cell accumulation and selection. With lung cancer being among the leading diagnoses of cancer-related deaths in both men and women, novel therapeutic methodologies utilizing nanotechnology for drug delivery emerged. Given chlorotoxin's promising biological activity, we explore its potential against lung cancer and its utilization for active targeting against this cancer's tumor cells. Our analysis indicates that despite the extensive chlorotoxin's research against glioblastoma, lung cancer research with the molecule has been limited, despite some promising early results.
Collapse
Affiliation(s)
- Archana Shrestha
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209, USA
| | - Behnaz Lahooti
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Constantinos M. Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, 26504 Patras, Greece
| | - George Mattheolabakis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209, USA
- Correspondence:
| |
Collapse
|
11
|
Shetty K, Yasaswi S, Dutt S, Yadav KS. Multifunctional nanocarriers for delivering siRNA and miRNA in glioblastoma therapy: advances in nanobiotechnology-based cancer therapy. 3 Biotech 2022; 12:301. [PMID: 36276454 PMCID: PMC9525514 DOI: 10.1007/s13205-022-03365-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 09/17/2022] [Indexed: 11/30/2022] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most lethal cancer due to poor diagnosis and rapid resistance developed towards the drug. Genes associated to cancer-related overexpression of proteins, enzymes, and receptors can be suppressed using an RNA silencing technique. This assists in obtaining tumour targetability, resulting in less harm caused to the surrounding healthy cells. RNA interference (RNAi) has scientific basis for providing potential therapeutic applications in improving GBM treatment. However, the therapeutic application of RNAi is challenging due to its poor permeability across blood-brain barrier (BBB). Nanobiotechnology has evolved the use of nanocarriers such as liposomes, polymeric nanoparticles, gold nanoparticles, dendrimers, quantum dots and other nanostructures in encasing the RNAi entities like siRNA and miRNA. The review highlights the role of these carriers in encasing siRNA and miRNA and promising therapy in delivering them to the glioma cells.
Collapse
Affiliation(s)
- Karishma Shetty
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’S NMIMS (Deemed to be University), Mumbai, India
| | - Soma Yasaswi
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’S NMIMS (Deemed to be University), Mumbai, India
| | - Shilpee Dutt
- Shilpee Dutt Laboratory, Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, 410210 India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085 India
| | - Khushwant S. Yadav
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’S NMIMS (Deemed to be University), Mumbai, India
| |
Collapse
|
12
|
Recombinantly expressed MeICT, a new toxin from Mesobuthus eupeus scorpion, inhibits glioma cell proliferation and downregulates Annexin A2 and FOXM1 genes. Biotechnol Lett 2022; 44:703-712. [PMID: 35524923 DOI: 10.1007/s10529-022-03254-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 04/12/2022] [Indexed: 11/02/2022]
Abstract
Gliomas are highly invasive and lethal malignancy that do not respond to current therapeutic approaches. Novel therapeutic agents are required to target molecular mechanisms involved in glioma progression. MeICT is a new short-chain toxin isolated from Mesobuthus eupeus scorpion venom. This toxin contained 34 amino acid residues and belongs to chloride channels toxins. In this study, the coding sequence of MeICT was cloned into the pET32Rh vector and a high yield of soluble recombinant MeICT was expressed and purified. Recombinant MeICT-His significantly inhibited the proliferation and migration of glioma cells at low concentration. In vivo studies showed that MeICT was not toxic when administrated to mice at high doses. We also determined the effect of MeICT on the mRNA expression of MMP-2, Annexin A2 and FOXM-2 that are key molecules in the progression and invasion of glioma. Expression of Annexin A2 and FOXM1 mRNA was significantly down-regulated following treatment with MeICT. However, no significant decrease in the expression of MMP-2 gene was identified. In this study a short toxin with four disulfide bonds was successfully produced and its anti-cancer effects was detected. Our findings suggest that recombinant MeICT can be considered as a new potent agent for glioma targeting.
Collapse
|
13
|
Kalra RS, Soman GS, Parab PB, Mali AM, Varankar SS, Naik RR, Kamble SC, Dhanjal JK, Bapat SA. A monoclonal antibody against annexin A2 targets stem and progenitor cell fractions in tumors. Transl Oncol 2021; 15:101257. [PMID: 34715620 PMCID: PMC8564672 DOI: 10.1016/j.tranon.2021.101257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/26/2022] Open
Abstract
Development of a novel antibody (termed as mAb150) developed in our lab which targets annexin A2. Although there are earlier reports of another monoclonal antibody with the same target, the epitope recognized by mAb150 is novel. mAb150 is specifically recognized to target the achilles heel of cancer viz. cancer stem cells and progenitors that persist after treatments and potentially give rise to minimal residual disease.
The involvement of cancer stem cells (CSCs) in driving tumor dormancy and drug resistance is well established. Most therapeutic regimens however are ineffective in targeting these regenerative populations. We report the development and evaluation of a monoclonal antibody, mAb150, which targets the metastasis associated antigen, Annexin A2 (AnxA2) through recognition of a N-terminal epitope. Treatment with mAb150 potentiated re-entry of CSCs into the cell cycle that perturbed tumor dormancy and facilitated targeting of CSCs as was validated by in vitro and in vivo assays. Epigenetic potentiation further improved mAb150 efficacy in achieving total tumor regression by targeting regenerative populations to achieve tumor regression, specifically in high-grade serous ovarian adenocarcinoma.
Collapse
Affiliation(s)
- Rajkumar S Kalra
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India; Savitribai Phule Pune University, Ganeshkhind, Pune 411007, India; Immune Signal Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa, 904-0495, Japan
| | - Gaurav S Soman
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India
| | - Pradeep B Parab
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India
| | - Avinash M Mali
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India
| | - Sagar S Varankar
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India; Savitribai Phule Pune University, Ganeshkhind, Pune 411007, India; Wellcome-MRC Cambridge Stem Cell Institute, Puddicombe Way, Cambridge, CB2 0AW
| | - Rutika R Naik
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India; Savitribai Phule Pune University, Ganeshkhind, Pune 411007, India
| | - Swapnil C Kamble
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India; Savitribai Phule Pune University, Ganeshkhind, Pune 411007, India; Department of Technology, Savitribai Phule Pune University, Ganeshkhind, Pune 411007, India
| | - Jaspreet K Dhanjal
- Department of Computational Biology, Indraprastha Institute of Information Technology Delhi, Okhla Industrial Estate, Phase III, New Delhi 110020, India
| | - Sharmila A Bapat
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India; Savitribai Phule Pune University, Ganeshkhind, Pune 411007, India.
| |
Collapse
|
14
|
Sharma G, Braga CB, Chen KE, Jia X, Ramanujam V, Collins BM, Rittner R, Mobli M. Structural basis for the binding of the cancer targeting scorpion toxin, ClTx, to the vascular endothelia growth factor receptor neuropilin-1. Curr Res Struct Biol 2021; 3:179-186. [PMID: 34401749 PMCID: PMC8358460 DOI: 10.1016/j.crstbi.2021.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/23/2021] [Accepted: 07/29/2021] [Indexed: 11/28/2022] Open
Abstract
Chlorotoxin (ClTx) is a 36-residue disulfide-rich peptide isolated from the venom of the scorpion Leiurus quinquestriatus. This peptide has been shown to selectively bind to brain tumours (gliomas), however, with conflicting reports regarding its direct cellular target. Recently, the vascular endothelial growth factor receptor, neuropilin-1 (NRP1) has emerged as a potential target of the peptide. Here, we sought to characterize the details of the binding of ClTx to the b1-domain of NRP1 (NRP1-b1) using solution state nuclear magnetic resonance (NMR) spectroscopy. The 3D structure of the isotope labelled peptide was solved using multidimensional heteronuclear NMR spectroscopy to produce a well-resolved structural ensemble. The structure points to three putative protein-protein interaction interfaces, two basic patches (R14/K15/K23 and R25/K27/R36) and a hydrophobic patch (F6/T7/T8/H10). The NRP1-b1 binding interface of ClTx was elucidated using 15N chemical shift mapping and included the R25/K27/R36 region of the peptide. The thermodynamics of binding was determined using isothermal titration calorimetry (ITC). In both NMR and ITC measurements, the binding was shown to be competitive with a known NRP1-b1 inhibitor. Finally, combining all of this data we generate a model of the ClTx:NRP1-b1 complex. The data shows that the peptide binds to the same region of NRP1 that is used by the SARS-CoV-2 virus for cell entry, however, via a non-canonical binding mode. Our results provide evidence for a non-standard NRP1 binding motif, while also providing a basis for further engineering of ClTx to generate peptides with improved NRP1 binding for future biomedical applications. Structural details of the binding of the scorpion toxin, chlorotoxin (ClTx) to the VEGF receptor neuropilin-1 (NRP1). ClTx was produced in its native fold and isotope labelled using a new, high-yield, bacterial expression system. Multidimensional heteronuclear NMR experiments reveal the high-resolution structure of ClTx and its binding to NRP1. ClTx binds to NRP1 via a non-canonical primary sequence that satisfies the receptor binding motif through its tertiary fold.
Collapse
Affiliation(s)
- Gagan Sharma
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, Australia
| | - Carolyne B. Braga
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, Australia
- Chemistry Institute, University of Campinas, P.O. Box 6154, 13083-970, Campinas, SP, Brazil
| | - Kai-En Chen
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Xinying Jia
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, Australia
| | | | - Brett M. Collins
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Roberto Rittner
- Chemistry Institute, University of Campinas, P.O. Box 6154, 13083-970, Campinas, SP, Brazil
| | - Mehdi Mobli
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, Australia
- Corresponding author.
| |
Collapse
|
15
|
Gerard L, Duvivier L, Gillet JP. Targeting tumor resistance mechanisms. Fac Rev 2021; 10:6. [PMID: 33659924 PMCID: PMC7894262 DOI: 10.12703/r/10-6] [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] [Indexed: 12/24/2022] Open
Abstract
Cancer develops resistance to treatments through many mechanisms. Single-cell analyses reveal the intratumor heterogeneity and dynamic relationships between cancer cell subpopulations. These analyses also highlight that various mechanisms of resistance may coexist in a given tumor. Studies have unraveled how the microenvironment affects tumor response to treatments and how cancer cells may adapt to these treatments. Though challenging, individualized treatment based on the molecular characterization of the tumor should become the new standard of care. In the meantime, the success rate of clinical trials in oncology remains dramatically low. There is a need to do better and improve the predictability of preclinical models. This requires innovative changes in ex vivo models and the culture system currently being used. An innovative ligand design is also urgently needed. The limited arsenal of medicinal chemistry reactions and the biases of scaffold selection favor structurally similar compounds with linear shapes at the expense of disc and spherical shapes, which leave a large chemical shape space untouched. In this regard, venoms have received increasing interest as a wellspring for drug candidates. Overall, the characterization of tumor heterogeneity has contributed to advancing our understanding of the mechanisms that underlie cancer resistance to treatments. Targeting these mechanisms will require setting key milestones to significantly improve the translatability of preclinical studies to the clinic with the hope of increasing the success rate of clinical trials.
Collapse
Affiliation(s)
- Louise Gerard
- Laboratory of Molecular Cancer Biology, Molecular Physiology Research Unit (URPhyM), Namur Research Institute for Life Sciences (NARILIS), Faculty of Medicine, University of Namur, Namur, Belgium
| | - Laurent Duvivier
- Laboratory of Molecular Cancer Biology, Molecular Physiology Research Unit (URPhyM), Namur Research Institute for Life Sciences (NARILIS), Faculty of Medicine, University of Namur, Namur, Belgium
| | - Jean-Pierre Gillet
- Laboratory of Molecular Cancer Biology, Molecular Physiology Research Unit (URPhyM), Namur Research Institute for Life Sciences (NARILIS), Faculty of Medicine, University of Namur, Namur, Belgium
| |
Collapse
|
16
|
Wang K, Kievit FM, Chiarelli PA, Stephen ZR, Lin G, Silber JR, Ellenbogen RG, Zhang M. siRNA nanoparticle suppresses drug-resistant gene and prolongs survival in an orthotopic glioblastoma xenograft mouse model. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2007166. [PMID: 33708035 PMCID: PMC7942690 DOI: 10.1002/adfm.202007166] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Indexed: 05/31/2023]
Abstract
Temozolomide (TMZ) is the standard of care chemotherapy drug for treating glioblastomas (GBMs), the most aggressive cancer that affects people of all ages. However, its therapeutic efficacy is limited by the drug resistance mediated by a DNA repair protein, O6-methylguanine-DNA methyltransferase (MGMT), which eliminates the TMZ-induced DNA lesions. Here we report the development of an iron oxide nanoparticle (NP) system for targeted delivery of siRNAs to suppress the TMZ-resistance gene (MGMT). We show that our NP is able to overcome biological barriers, bind specifically to tumor cells, and reduce MGMT expression in tumors of mice bearing orthotopic GBM serially-passaged patient-derived xenografts. The treatment with sequential administration of this NP and TMZ resulted in increased apoptosis of GBM stem-like cells, reduced tumor growth, and significantly-prolonged survival as compared to mice treated with TMZ alone. This study introduces an approach that holds great promise to improve the outcomes of GBM patients.
Collapse
Affiliation(s)
- Kui Wang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, United States
| | - Forrest M Kievit
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, United States
| | - Peter A Chiarelli
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, United States
| | - Zachary R Stephen
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, United States
| | - Guanyou Lin
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, United States
| | - John R Silber
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, United States
| | - Richard G Ellenbogen
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, United States
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, United States; Department of Neurological Surgery, University of Washington, Seattle, WA 98195, United States
| |
Collapse
|
17
|
Castro J, Ribó M, Vilanova M, Benito A. Strengths and Challenges of Secretory Ribonucleases as AntiTumor Agents. Pharmaceutics 2021; 13:82. [PMID: 33435285 PMCID: PMC7828032 DOI: 10.3390/pharmaceutics13010082] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/25/2022] Open
Abstract
Approaches to develop effective drugs to kill cancer cells are mainly focused either on the improvement of the currently used chemotherapeutics or on the development of targeted therapies aimed at the selective destruction of cancer cells by steering specific molecules and/or enhancing the immune response. The former strategy is limited by its genotoxicity and severe side effects, while the second one is not always effective due to tumor cell heterogeneity and variability of targets in cancer cells. Between these two strategies, several approaches target different types of RNA in tumor cells. RNA degradation alters gene expression at different levels inducing cell death. However, unlike DNA targeting, it is a pleotropic but a non-genotoxic process. Among the ways to destroy RNA, we find the use of ribonucleases with antitumor properties. In the last few years, there has been a significant progress in the understanding of the mechanism by which these enzymes kill cancer cells and in the development of more effective variants. All the approaches seek to maintain the requirements of the ribonucleases to be specifically cytotoxic for tumor cells. These requirements start with the competence of the enzymes to interact with the cell membrane, a process that is critical for their internalization and selectivity for tumor cells and continue with the downstream effects mainly relying on changes in the RNA molecular profile, which are not only due to the ribonucleolytic activity of these enzymes. Although the great improvements achieved in the antitumor activity by designing new ribonuclease variants, some drawbacks still need to be addressed. In the present review, we will focus on the known mechanisms used by ribonucleases to kill cancer cells and on recent strategies to solve the shortcomings that they show as antitumor agents, mainly their pharmacokinetics.
Collapse
Affiliation(s)
- Jessica Castro
- Laboratori d’Enginyeria de Proteïnes, Departament de Biologia, Facultat de Ciències, Universitat de Girona, Campus de Montilivi, Carrer Maria Aurèlia Capmany, 40, 17003 Girona, Spain; (J.C.); (M.R.)
- Institut d’Investigació Biomèdica de Girona Josep Trueta, (IdIBGi), Hospital de Santa Caterina, Carrer del Dr. Castany, s/n, 17190 Salt, Spain
| | - Marc Ribó
- Laboratori d’Enginyeria de Proteïnes, Departament de Biologia, Facultat de Ciències, Universitat de Girona, Campus de Montilivi, Carrer Maria Aurèlia Capmany, 40, 17003 Girona, Spain; (J.C.); (M.R.)
- Institut d’Investigació Biomèdica de Girona Josep Trueta, (IdIBGi), Hospital de Santa Caterina, Carrer del Dr. Castany, s/n, 17190 Salt, Spain
| | - Maria Vilanova
- Laboratori d’Enginyeria de Proteïnes, Departament de Biologia, Facultat de Ciències, Universitat de Girona, Campus de Montilivi, Carrer Maria Aurèlia Capmany, 40, 17003 Girona, Spain; (J.C.); (M.R.)
- Institut d’Investigació Biomèdica de Girona Josep Trueta, (IdIBGi), Hospital de Santa Caterina, Carrer del Dr. Castany, s/n, 17190 Salt, Spain
| | - Antoni Benito
- Laboratori d’Enginyeria de Proteïnes, Departament de Biologia, Facultat de Ciències, Universitat de Girona, Campus de Montilivi, Carrer Maria Aurèlia Capmany, 40, 17003 Girona, Spain; (J.C.); (M.R.)
- Institut d’Investigació Biomèdica de Girona Josep Trueta, (IdIBGi), Hospital de Santa Caterina, Carrer del Dr. Castany, s/n, 17190 Salt, Spain
| |
Collapse
|
18
|
Delinois LJ, Peón H, Villalobos-Santos JC, Ramírez-Paz J, Miller J, Griebenow KH, Tinoco AD. A Cytochrome c-Chlorotoxin Hybrid Protein as a Possible Antiglioma Drug. ChemMedChem 2020; 15:2185-2192. [PMID: 32918396 PMCID: PMC7722079 DOI: 10.1002/cmdc.202000373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 09/02/2020] [Indexed: 11/06/2022]
Abstract
Malignant gliomas are the most lethal form of primary brain tumors. Despite advances in cancer therapy, the prognosis of glioma patients has remained poor. Cytochrome c (Cytc), an endogenous heme-based protein, holds tremendous potential to treat gliomas because of its innate capacity to trigger apoptosis. To this end, a hybrid cytochrome c-chlorotoxin (Cytc-CTX) protein was biosynthesized to enable cellular uptake of the cell impenetrable Cytc using CTX transporters. A nucleotide sequence containing 1 : 1 Cytc and CTX was constructed and separated by a hexahistidine-tag and an enterokinase cleavage site. The sequence was cloned into a pBTR1 plasmid, expressed in Escherichia coli, purified via 2-dimensional chromatography. The identity and size of the protein were determined by Western blot and mass spectrometry. Cytc in this soluble hybrid protein has similar structure and stability as human Cytc and the hybrid protein is endocytosed into a glioma cell line, while displaying potent cytotoxicity and a favorable therapeutic index. Its facile, low-cost, and high yield synthesis, biocompatibility, and robustness suggest that the hybrid protein is a promising candidate for antiglioma drug evaluation.
Collapse
Affiliation(s)
- Louis J Delinois
- Chemistry Department, Faculty of Natural Sciences of the University of Puerto Rico at Río Piedras, San Juan, Puerto Rico, 00925, USA
| | - Harold Peón
- Academia del Perpetuo Socorro High School, San Juan, Puerto Rico, 00907, USA
| | - Juan C Villalobos-Santos
- Biology Department, Faculty of Natural Sciences of the University of Puerto Rico at Río Piedras, San Juan, Puerto Rico, 00925, USA
| | - Josell Ramírez-Paz
- Chemistry Department, Faculty of Natural Sciences of the University of Puerto Rico at Río Piedras, San Juan, Puerto Rico, 00925, USA
| | - Jennifer Miller
- Chemistry Department, Faculty of Natural Sciences of the University of Puerto Rico at Río Piedras, San Juan, Puerto Rico, 00925, USA
| | - Kai H Griebenow
- Chemistry Department, Faculty of Natural Sciences of the University of Puerto Rico at Río Piedras, San Juan, Puerto Rico, 00925, USA
| | - Arthur D Tinoco
- Chemistry Department, Faculty of Natural Sciences of the University of Puerto Rico at Río Piedras, San Juan, Puerto Rico, 00925, USA
| |
Collapse
|
19
|
C-Terminal Amidation of Chlorotoxin Does Not Affect Tumour Cell Proliferation and Has No Effect on Toxin Cytotoxicity. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-020-10117-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
20
|
Sharma MC, Jain D. Important role of annexin A2 (ANXA2) in new blood vessel development in vivo and human triple negative breast cancer (TNBC) growth. Exp Mol Pathol 2020; 116:104523. [PMID: 32866522 DOI: 10.1016/j.yexmp.2020.104523] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/11/2020] [Accepted: 08/26/2020] [Indexed: 11/18/2022]
Abstract
Development of new blood vessels in the tumor microenvironment is an essential component of tumor progression during which newly formed blood vessels nourish tumor cells and play a critical role in rapid tumor growth, invasion and metastasis. Nevertheless, how tumor cells develop new blood vessels in the tumor microenvironment (TME) have been enigmatic. Previously, we have shown specific overexpression of ANX A2 in TNBC cells regulates plasmin generation and suspected a role in neoangiogenesis. In this report, we used Matrigel plug model of in vivo angiogenesis and confirmed its role in new blood vessel development. Next, we tested if blocking of ANX A2 in aggressive human breast TME can inhibit angiogenesis and tumor growth in vivo. We showed that aggressive human breast tumor cells growing in nude mice can induce intense neoangiogenesis in the tumor mass. Blocking of ANXA2 significantly inhibited neoangiogenesis and resulted in inhibition of tumor growth. Interestingly, we identified that blocking of ANXA2 significantly inhibited tyrosine phosphorylation (Tyr-P) of ANXA2 implying its involvement in tyrosine signaling pathway and suggesting it may regulate angiogenesis. Taken together, our experimental evidence suggests that ANX A2 could be a novel strategy for disruption of tyrosine signaling and inhibition of neoangiogenesis in breast tumor.
Collapse
Affiliation(s)
- Mahesh C Sharma
- Research Service, Veterans Affairs Medical Center, Washington, DC 20422, United States of America; Department of Biochemistry and Molecular Medicine, George Washington University, Washington, DC, United States of America.
| | - Diwakar Jain
- Westchester Medical Center, NY 10595, United States of America
| |
Collapse
|
21
|
Noble K, Rohaj A, Abegglen LM, Schiffman JD. Cancer therapeutics inspired by defense mechanisms in the animal kingdom. Evol Appl 2020. [DOI: 10.1111/eva.12963] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Kathleen Noble
- Huntsman Cancer Institute University of Utah Salt Lake City Utah
| | - Aarushi Rohaj
- Huntsman Cancer Institute University of Utah Salt Lake City Utah
| | - Lisa M. Abegglen
- Huntsman Cancer Institute University of Utah Salt Lake City Utah
- Department of Pediatrics University of Utah Salt Lake City Utah
| | - Joshua D. Schiffman
- Huntsman Cancer Institute University of Utah Salt Lake City Utah
- Department of Pediatrics University of Utah Salt Lake City Utah
- PEEL Therapeutics, Inc. Salt Lake City Utah
| |
Collapse
|
22
|
Bordon KDCF, Cologna CT, Fornari-Baldo EC, Pinheiro-Júnior EL, Cerni FA, Amorim FG, Anjolette FAP, Cordeiro FA, Wiezel GA, Cardoso IA, Ferreira IG, de Oliveira IS, Boldrini-França J, Pucca MB, Baldo MA, Arantes EC. From Animal Poisons and Venoms to Medicines: Achievements, Challenges and Perspectives in Drug Discovery. Front Pharmacol 2020; 11:1132. [PMID: 32848750 PMCID: PMC7396678 DOI: 10.3389/fphar.2020.01132] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/13/2020] [Indexed: 12/16/2022] Open
Abstract
Animal poisons and venoms are comprised of different classes of molecules displaying wide-ranging pharmacological activities. This review aims to provide an in-depth view of toxin-based compounds from terrestrial and marine organisms used as diagnostic tools, experimental molecules to validate postulated therapeutic targets, drug libraries, prototypes for the design of drugs, cosmeceuticals, and therapeutic agents. However, making these molecules applicable requires extensive preclinical trials, with some applications also demanding clinical trials, in order to validate their molecular target, mechanism of action, effective dose, potential adverse effects, as well as other fundamental parameters. Here we go through the pitfalls for a toxin-based potential therapeutic drug to become eligible for clinical trials and marketing. The manuscript also presents an overview of the current picture for several molecules from different animal venoms and poisons (such as those from amphibians, cone snails, hymenopterans, scorpions, sea anemones, snakes, spiders, tetraodontiformes, bats, and shrews) that have been used in clinical trials. Advances and perspectives on the therapeutic potential of molecules from other underexploited animals, such as caterpillars and ticks, are also reported. The challenges faced during the lengthy and costly preclinical and clinical studies and how to overcome these hindrances are also discussed for that drug candidates going to the bedside. It covers most of the drugs developed using toxins, the molecules that have failed and those that are currently in clinical trials. The article presents a detailed overview of toxins that have been used as therapeutic agents, including their discovery, formulation, dosage, indications, main adverse effects, and pregnancy and breastfeeding prescription warnings. Toxins in diagnosis, as well as cosmeceuticals and atypical therapies (bee venom and leech therapies) are also reported. The level of cumulative and detailed information provided in this review may help pharmacists, physicians, biotechnologists, pharmacologists, and scientists interested in toxinology, drug discovery, and development of toxin-based products.
Collapse
Affiliation(s)
- Karla de Castro Figueiredo Bordon
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Camila Takeno Cologna
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Ernesto Lopes Pinheiro-Júnior
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Felipe Augusto Cerni
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Fernanda Gobbi Amorim
- Postgraduate Program in Pharmaceutical Sciences, Vila Velha University, Vila Velha, Brazil
| | | | - Francielle Almeida Cordeiro
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Gisele Adriano Wiezel
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Iara Aimê Cardoso
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Isabela Gobbo Ferreira
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Isadora Sousa de Oliveira
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | | | - Mateus Amaral Baldo
- Health and Science Institute, Paulista University, São José do Rio Pardo, Brazil
| | - Eliane Candiani Arantes
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
23
|
Scorpion Toxins and Ion Channels: Potential Applications in Cancer Therapy. Toxins (Basel) 2020; 12:toxins12050326. [PMID: 32429050 PMCID: PMC7290751 DOI: 10.3390/toxins12050326] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 12/24/2022] Open
Abstract
Apoptosis, a genetically directed process of cell death, has been studied for many years, and the biochemical mechanisms that surround it are well known and described. There are at least three pathways by which apoptosis occurs, and each pathway depends on extra or intracellular processes for activation. Apoptosis is a vital process, but disturbances in proliferation and cell death rates can lead to the development of diseases like cancer. Several compounds, isolated from scorpion venoms, exhibit inhibitory effects on different cancer cells. Indeed, some of these compounds can differentiate between healthy and cancer cells within the same tissue. During the carcinogenic process, morphological, biochemical, and biological changes occur that enable these compounds to modulate cancer but not healthy cells. This review highlights cancer cell features that enable modulation by scorpion neurotoxins. The properties of the isolated scorpion neurotoxins in cancer cells and the potential uses of these compounds as alternative treatments for cancer are discussed.
Collapse
|
24
|
Formicola B, Dal Magro R, Montefusco-Pereira CV, Lehr CM, Koch M, Russo L, Grasso G, Deriu MA, Danani A, Bourdoulous S, Re F. The synergistic effect of chlorotoxin-mApoE in boosting drug-loaded liposomes across the BBB. J Nanobiotechnology 2019; 17:115. [PMID: 31711496 PMCID: PMC6844026 DOI: 10.1186/s12951-019-0546-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 10/27/2019] [Indexed: 12/30/2022] Open
Abstract
We designed liposomes dually functionalized with ApoE-derived peptide (mApoE) and chlorotoxin (ClTx) to improve their blood–brain barrier (BBB) crossing. Our results demonstrated the synergistic activity of ClTx-mApoE in boosting doxorubicin-loaded liposomes across the BBB, keeping the anti-tumour activity of the drug loaded: mApoE acts promoting cellular uptake, while ClTx promotes exocytosis of liposomes.
Collapse
Affiliation(s)
- Beatrice Formicola
- School of Medicine and Surgery, University of Milano-Bicocca, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy.
| | - Roberta Dal Magro
- School of Medicine and Surgery, University of Milano-Bicocca, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy
| | - Carlos V Montefusco-Pereira
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland, Saarland University, 66123, Saarbrücken, Germany
| | - Claus-Michael Lehr
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland, Saarland University, 66123, Saarbrücken, Germany
| | - Marcus Koch
- Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
| | - Laura Russo
- Bio Organic Chemistry Laboratory, Department of Biotechnology and Biosciences, University of Milano-Bicocca, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy
| | - Gianvito Grasso
- Istituto Dalle Molle di Studi Sull'Intelligenza Artificiale (IDSIA), Scuola Universitaria Professionale Della Svizzera Italiana (SUPSI), Università Della Svizzera Italiana (USI), Manno, Switzerland
| | - Marco A Deriu
- Department of Mechanical and Aerospace Engineering (DIMEAS), Politecnico di Torino, Corso Duca degli Abruzzi 24, 10128, Turin, Italy
| | - Andrea Danani
- Istituto Dalle Molle di Studi Sull'Intelligenza Artificiale (IDSIA), Scuola Universitaria Professionale Della Svizzera Italiana (SUPSI), Università Della Svizzera Italiana (USI), Manno, Switzerland
| | | | - Francesca Re
- School of Medicine and Surgery, University of Milano-Bicocca, Via Raoul Follereau 3, 20854, Vedano al Lambro, MB, Italy
| |
Collapse
|
25
|
Egloff-Juras C, Bezdetnaya L, Dolivet G, Lassalle HP. NIR fluorescence-guided tumor surgery: new strategies for the use of indocyanine green. Int J Nanomedicine 2019; 14:7823-7838. [PMID: 31576126 PMCID: PMC6768149 DOI: 10.2147/ijn.s207486] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 07/27/2019] [Indexed: 12/15/2022] Open
Abstract
Surgery is the frontline treatment for a large number of cancers. The objective of these excisional surgeries is the complete removal of the primary tumor with sufficient safety margins. Removal of the entire tumor is essential to improve the chances of a full recovery. To help surgeons achieve this objective, near-infrared fluorescence-guided surgical techniques are of great interest. The concomitant use of fluorescence and indocyanine green (ICG) has proved effective in the identification and characterization of tumors. Moreover, ICG is authorized by the Food and Drug Administration and the European Medicines Agency and is therefore the subject of a large number of studies. ICG is one of the most commonly used fluorophores in near-infrared fluorescence-guided techniques. However, it also has some disadvantages, such as limited photostability, a moderate fluorescence quantum yield, a high plasma protein binding rate, and undesired aggregation in aqueous solution. In addition, ICG does not specifically target tumor cells. One way to exploit the capabilities of ICG while offsetting these drawbacks is to develop high-performance near-infrared nanocomplexes formulated with ICG (with high selectivity for tumors, high tumor-to-background ratios, and minimal toxicity). In this review article, we focus on recent developments in ICG complexation strategies to improve near-infrared fluorescence-guided tumor surgery. We describe targeted and nontargeted ICG nanoparticle models and ICG complexation with targeting agents.
Collapse
Affiliation(s)
- Claire Egloff-Juras
- Université de Lorraine, CNRS, CRAN, Nancy F-54000, France.,Université de Lorraine, CHRU-Nancy, Institut de Cancérologie de Lorraine, Nancy F-54000, France
| | - Lina Bezdetnaya
- Université de Lorraine, CNRS, CRAN, Nancy F-54000, France.,Institut de Cancérologie de Lorraine, Nancy F-54000, France
| | - Gilles Dolivet
- Université de Lorraine, CNRS, CRAN, Nancy F-54000, France.,Institut de Cancérologie de Lorraine, Nancy F-54000, France
| | - Henri-Pierre Lassalle
- Université de Lorraine, CNRS, CRAN, Nancy F-54000, France.,Institut de Cancérologie de Lorraine, Nancy F-54000, France
| |
Collapse
|
26
|
Stephen ZR, Chiarelli PA, Revia RA, Wang K, Kievit F, Dayringer C, Jeon M, Ellenbogen R, Zhang M. Time-Resolved MRI Assessment of Convection-Enhanced Delivery by Targeted and Nontargeted Nanoparticles in a Human Glioblastoma Mouse Model. Cancer Res 2019; 79:4776-4786. [PMID: 31331912 PMCID: PMC6744959 DOI: 10.1158/0008-5472.can-18-2998] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 05/08/2019] [Accepted: 07/12/2019] [Indexed: 11/16/2022]
Abstract
Convection-enhanced delivery (CED) provides direct access of infusates to brain tumors; however, clinical translation of this technology has not been realized because of the inability to accurately visualize infusates in real-time and lack of targeting modalities against diffuse cancer cells. In this study, we use time-resolved MRI to reveal the kinetics of CED processes in a glioblastoma (GBM) model using iron oxide nanoparticles (NP) modified with a glioma-targeting ligand, chlorotoxin (CTX). Mice bearing orthotopic human GBM tumors were administered a single dose of targeted CTX-conjugated NP (NPCP-CTX) or nontargeted NP (NPCP) via CED. High-resolution T2-weighted, T2*-weighted, and quantitative T2 MRI were utilized to image NP delivery in real time and determined the volume of distribution (VD) of NPs at multiple time points over the first 48 hours post-CED. GBM-specific targeting was evaluated by flow cytometry and intracellular NP localization by histologic assessment. NPCP-CTX produced a VD of 121 ± 39 mm3 at 24 hours, a significant increase compared with NPCP, while exhibiting GBM specificity and localization to cell nuclei. Notably, CED of NPCP-CTX resulted in a sustained expansion of VD well after infusion, suggesting a possible active transport mechanism, which was further supported by the presence of NPs in endothelial and red blood cells. In summary, we show that time-resolved MRI is a suitable modality to study CED kinetics, and CTX-mediated CED facilitates extensive distribution of infusate and specific targeting of tumor cells. SIGNIFICANCE: MRI is used to monitor convection-enhanced delivery in real time using a nanoparticle-based contrast agent, and glioma-specific targeting significantly improves the volume of distribution in tumors.
Collapse
Affiliation(s)
- Zachary R Stephen
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington
| | - Peter A Chiarelli
- Department of Neurological Surgery, University of Washington, Seattle, Washington
- Department of Neurosurgery, Children's Hospital Los Angeles, Los Angeles, California
| | - Richard A Revia
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington
| | - Kui Wang
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington
| | - Forrest Kievit
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington
- Department of Neurological Surgery, University of Washington, Seattle, Washington
| | - Chris Dayringer
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington
| | - Mike Jeon
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington
| | - Richard Ellenbogen
- Department of Neurological Surgery, University of Washington, Seattle, Washington.
- Department of Radiology, University of Washington, Seattle, Washington
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington.
- Department of Neurological Surgery, University of Washington, Seattle, Washington
| |
Collapse
|
27
|
Dintzis SM, Hansen S, Harrington KM, Tan LC, Miller DM, Ishak L, Parrish-Novak J, Kittle D, Perry J, Gombotz C, Fortney T, Porenta S, Hales L, Calhoun KE, Anderson BO, Javid SH, Byrd DR. Real-time Visualization of Breast Carcinoma in Pathology Specimens From Patients Receiving Fluorescent Tumor-Marking Agent Tozuleristide. Arch Pathol Lab Med 2019; 143:1076-1083. [PMID: 30550350 PMCID: PMC11781288 DOI: 10.5858/arpa.2018-0197-oa] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
CONTEXT.— Resection of breast carcinoma with adequate margins reduces the risk of local recurrence and reoperation. Tozuleristide (BLZ-100) is an investigational peptide-fluorophore agent that may aid in intraoperative tumor detection and margin assessment. In this study, fluorescence imaging was conducted ex vivo on gross breast pathology specimens. OBJECTIVES.— To determine the potential of tozuleristide to detect breast carcinoma in fresh pathology specimens and the feasibility of fluorescence-guided intraoperative pathology assessment of surgical margins. DESIGN.— Twenty-three patients received an intravenous bolus dose of 6 or 12 mg of tozuleristide at least 1 hour before surgery. Fifteen lumpectomy and 12 mastectomy specimens were evaluated for fluorescence by the site's clinical pathology staff using the SIRIS, an investigational near-infrared imaging device. The breast tissue was then processed per usual procedures. Fluorescent patterns were correlated with the corresponding hematoxylin-eosin-stained sections. Clinical pathology reports were used to correlate fluorescent signal to grade, histotype, prognostic marker status, and margin measurements. RESULTS.— Tozuleristide fluorescence was readily observed in invasive and in situ breast carcinoma specimens. Most invasive carcinomas were bright and focal, whereas in situ lesions demonstrated a less intense, more diffuse pattern. Tozuleristide was detected in ductal and lobular carcinomas with a similar fluorescent pattern. Fluorescence was detected in high- and low-grade lesions, and molecular marker/hormone receptor status did not affect signal. Fluorescence could be used to identify the relationship of carcinoma to margins intraoperatively. CONCLUSIONS.— Tumor targeting with tozuleristide allowed visual real-time distinction between pathologically confirmed breast carcinoma and normal tissue.
Collapse
MESH Headings
- Breast Carcinoma In Situ/diagnostic imaging
- Breast Carcinoma In Situ/pathology
- Breast Carcinoma In Situ/surgery
- Breast Neoplasms/diagnostic imaging
- Breast Neoplasms/surgery
- Carcinoma, Ductal, Breast/diagnostic imaging
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/surgery
- Carcinoma, Lobular/diagnostic imaging
- Carcinoma, Lobular/pathology
- Carcinoma, Lobular/surgery
- Female
- Fluorescent Dyes
- Humans
- Indocyanine Green/analogs & derivatives
- Intraoperative Care/methods
- Margins of Excision
- Mastectomy
- Mastectomy, Segmental
- Neoplasm Invasiveness/diagnostic imaging
- Neoplasm Invasiveness/pathology
- Prognosis
- Scorpion Venoms
Collapse
Affiliation(s)
- Suzanne M Dintzis
- From the Departments of Pathology (Dr Dintzis) and Surgery (Ms Hales and Drs Calhoun, Javid, and Byrd), University of Washington Medical Center, Seattle; Breast Surgery Clinic (Dr Harrington), Department of Pathology (Dr Tan), and Clinical Trials (Mses Fortney and Porenta), Overlake Hospital Medical Center, Bellevue, Washington; Development (Dr Miller), Clinical Operations (Mses Ishak and Gombotz), Research (Ms Hansen and Dr Parrish-Novak), and Device Development (Dr Kittle and Mr Perry), Blaze Bioscience, Inc., Seattle, Washington; and the Department of Surgery, University of Washington, Seattle (Dr Anderson)
| | - Stacey Hansen
- From the Departments of Pathology (Dr Dintzis) and Surgery (Ms Hales and Drs Calhoun, Javid, and Byrd), University of Washington Medical Center, Seattle; Breast Surgery Clinic (Dr Harrington), Department of Pathology (Dr Tan), and Clinical Trials (Mses Fortney and Porenta), Overlake Hospital Medical Center, Bellevue, Washington; Development (Dr Miller), Clinical Operations (Mses Ishak and Gombotz), Research (Ms Hansen and Dr Parrish-Novak), and Device Development (Dr Kittle and Mr Perry), Blaze Bioscience, Inc., Seattle, Washington; and the Department of Surgery, University of Washington, Seattle (Dr Anderson)
| | - Kristi M Harrington
- From the Departments of Pathology (Dr Dintzis) and Surgery (Ms Hales and Drs Calhoun, Javid, and Byrd), University of Washington Medical Center, Seattle; Breast Surgery Clinic (Dr Harrington), Department of Pathology (Dr Tan), and Clinical Trials (Mses Fortney and Porenta), Overlake Hospital Medical Center, Bellevue, Washington; Development (Dr Miller), Clinical Operations (Mses Ishak and Gombotz), Research (Ms Hansen and Dr Parrish-Novak), and Device Development (Dr Kittle and Mr Perry), Blaze Bioscience, Inc., Seattle, Washington; and the Department of Surgery, University of Washington, Seattle (Dr Anderson)
| | - Lennart C Tan
- From the Departments of Pathology (Dr Dintzis) and Surgery (Ms Hales and Drs Calhoun, Javid, and Byrd), University of Washington Medical Center, Seattle; Breast Surgery Clinic (Dr Harrington), Department of Pathology (Dr Tan), and Clinical Trials (Mses Fortney and Porenta), Overlake Hospital Medical Center, Bellevue, Washington; Development (Dr Miller), Clinical Operations (Mses Ishak and Gombotz), Research (Ms Hansen and Dr Parrish-Novak), and Device Development (Dr Kittle and Mr Perry), Blaze Bioscience, Inc., Seattle, Washington; and the Department of Surgery, University of Washington, Seattle (Dr Anderson)
| | - Dennis M Miller
- From the Departments of Pathology (Dr Dintzis) and Surgery (Ms Hales and Drs Calhoun, Javid, and Byrd), University of Washington Medical Center, Seattle; Breast Surgery Clinic (Dr Harrington), Department of Pathology (Dr Tan), and Clinical Trials (Mses Fortney and Porenta), Overlake Hospital Medical Center, Bellevue, Washington; Development (Dr Miller), Clinical Operations (Mses Ishak and Gombotz), Research (Ms Hansen and Dr Parrish-Novak), and Device Development (Dr Kittle and Mr Perry), Blaze Bioscience, Inc., Seattle, Washington; and the Department of Surgery, University of Washington, Seattle (Dr Anderson)
| | - Laura Ishak
- From the Departments of Pathology (Dr Dintzis) and Surgery (Ms Hales and Drs Calhoun, Javid, and Byrd), University of Washington Medical Center, Seattle; Breast Surgery Clinic (Dr Harrington), Department of Pathology (Dr Tan), and Clinical Trials (Mses Fortney and Porenta), Overlake Hospital Medical Center, Bellevue, Washington; Development (Dr Miller), Clinical Operations (Mses Ishak and Gombotz), Research (Ms Hansen and Dr Parrish-Novak), and Device Development (Dr Kittle and Mr Perry), Blaze Bioscience, Inc., Seattle, Washington; and the Department of Surgery, University of Washington, Seattle (Dr Anderson)
| | - Julia Parrish-Novak
- From the Departments of Pathology (Dr Dintzis) and Surgery (Ms Hales and Drs Calhoun, Javid, and Byrd), University of Washington Medical Center, Seattle; Breast Surgery Clinic (Dr Harrington), Department of Pathology (Dr Tan), and Clinical Trials (Mses Fortney and Porenta), Overlake Hospital Medical Center, Bellevue, Washington; Development (Dr Miller), Clinical Operations (Mses Ishak and Gombotz), Research (Ms Hansen and Dr Parrish-Novak), and Device Development (Dr Kittle and Mr Perry), Blaze Bioscience, Inc., Seattle, Washington; and the Department of Surgery, University of Washington, Seattle (Dr Anderson)
| | - David Kittle
- From the Departments of Pathology (Dr Dintzis) and Surgery (Ms Hales and Drs Calhoun, Javid, and Byrd), University of Washington Medical Center, Seattle; Breast Surgery Clinic (Dr Harrington), Department of Pathology (Dr Tan), and Clinical Trials (Mses Fortney and Porenta), Overlake Hospital Medical Center, Bellevue, Washington; Development (Dr Miller), Clinical Operations (Mses Ishak and Gombotz), Research (Ms Hansen and Dr Parrish-Novak), and Device Development (Dr Kittle and Mr Perry), Blaze Bioscience, Inc., Seattle, Washington; and the Department of Surgery, University of Washington, Seattle (Dr Anderson)
| | - Jeff Perry
- From the Departments of Pathology (Dr Dintzis) and Surgery (Ms Hales and Drs Calhoun, Javid, and Byrd), University of Washington Medical Center, Seattle; Breast Surgery Clinic (Dr Harrington), Department of Pathology (Dr Tan), and Clinical Trials (Mses Fortney and Porenta), Overlake Hospital Medical Center, Bellevue, Washington; Development (Dr Miller), Clinical Operations (Mses Ishak and Gombotz), Research (Ms Hansen and Dr Parrish-Novak), and Device Development (Dr Kittle and Mr Perry), Blaze Bioscience, Inc., Seattle, Washington; and the Department of Surgery, University of Washington, Seattle (Dr Anderson)
| | - Carolyn Gombotz
- From the Departments of Pathology (Dr Dintzis) and Surgery (Ms Hales and Drs Calhoun, Javid, and Byrd), University of Washington Medical Center, Seattle; Breast Surgery Clinic (Dr Harrington), Department of Pathology (Dr Tan), and Clinical Trials (Mses Fortney and Porenta), Overlake Hospital Medical Center, Bellevue, Washington; Development (Dr Miller), Clinical Operations (Mses Ishak and Gombotz), Research (Ms Hansen and Dr Parrish-Novak), and Device Development (Dr Kittle and Mr Perry), Blaze Bioscience, Inc., Seattle, Washington; and the Department of Surgery, University of Washington, Seattle (Dr Anderson)
| | - Tina Fortney
- From the Departments of Pathology (Dr Dintzis) and Surgery (Ms Hales and Drs Calhoun, Javid, and Byrd), University of Washington Medical Center, Seattle; Breast Surgery Clinic (Dr Harrington), Department of Pathology (Dr Tan), and Clinical Trials (Mses Fortney and Porenta), Overlake Hospital Medical Center, Bellevue, Washington; Development (Dr Miller), Clinical Operations (Mses Ishak and Gombotz), Research (Ms Hansen and Dr Parrish-Novak), and Device Development (Dr Kittle and Mr Perry), Blaze Bioscience, Inc., Seattle, Washington; and the Department of Surgery, University of Washington, Seattle (Dr Anderson)
| | - Stephanie Porenta
- From the Departments of Pathology (Dr Dintzis) and Surgery (Ms Hales and Drs Calhoun, Javid, and Byrd), University of Washington Medical Center, Seattle; Breast Surgery Clinic (Dr Harrington), Department of Pathology (Dr Tan), and Clinical Trials (Mses Fortney and Porenta), Overlake Hospital Medical Center, Bellevue, Washington; Development (Dr Miller), Clinical Operations (Mses Ishak and Gombotz), Research (Ms Hansen and Dr Parrish-Novak), and Device Development (Dr Kittle and Mr Perry), Blaze Bioscience, Inc., Seattle, Washington; and the Department of Surgery, University of Washington, Seattle (Dr Anderson)
| | - Lisa Hales
- From the Departments of Pathology (Dr Dintzis) and Surgery (Ms Hales and Drs Calhoun, Javid, and Byrd), University of Washington Medical Center, Seattle; Breast Surgery Clinic (Dr Harrington), Department of Pathology (Dr Tan), and Clinical Trials (Mses Fortney and Porenta), Overlake Hospital Medical Center, Bellevue, Washington; Development (Dr Miller), Clinical Operations (Mses Ishak and Gombotz), Research (Ms Hansen and Dr Parrish-Novak), and Device Development (Dr Kittle and Mr Perry), Blaze Bioscience, Inc., Seattle, Washington; and the Department of Surgery, University of Washington, Seattle (Dr Anderson)
| | - Kristine E Calhoun
- From the Departments of Pathology (Dr Dintzis) and Surgery (Ms Hales and Drs Calhoun, Javid, and Byrd), University of Washington Medical Center, Seattle; Breast Surgery Clinic (Dr Harrington), Department of Pathology (Dr Tan), and Clinical Trials (Mses Fortney and Porenta), Overlake Hospital Medical Center, Bellevue, Washington; Development (Dr Miller), Clinical Operations (Mses Ishak and Gombotz), Research (Ms Hansen and Dr Parrish-Novak), and Device Development (Dr Kittle and Mr Perry), Blaze Bioscience, Inc., Seattle, Washington; and the Department of Surgery, University of Washington, Seattle (Dr Anderson)
| | - Benjamin O Anderson
- From the Departments of Pathology (Dr Dintzis) and Surgery (Ms Hales and Drs Calhoun, Javid, and Byrd), University of Washington Medical Center, Seattle; Breast Surgery Clinic (Dr Harrington), Department of Pathology (Dr Tan), and Clinical Trials (Mses Fortney and Porenta), Overlake Hospital Medical Center, Bellevue, Washington; Development (Dr Miller), Clinical Operations (Mses Ishak and Gombotz), Research (Ms Hansen and Dr Parrish-Novak), and Device Development (Dr Kittle and Mr Perry), Blaze Bioscience, Inc., Seattle, Washington; and the Department of Surgery, University of Washington, Seattle (Dr Anderson)
| | - Sara H Javid
- From the Departments of Pathology (Dr Dintzis) and Surgery (Ms Hales and Drs Calhoun, Javid, and Byrd), University of Washington Medical Center, Seattle; Breast Surgery Clinic (Dr Harrington), Department of Pathology (Dr Tan), and Clinical Trials (Mses Fortney and Porenta), Overlake Hospital Medical Center, Bellevue, Washington; Development (Dr Miller), Clinical Operations (Mses Ishak and Gombotz), Research (Ms Hansen and Dr Parrish-Novak), and Device Development (Dr Kittle and Mr Perry), Blaze Bioscience, Inc., Seattle, Washington; and the Department of Surgery, University of Washington, Seattle (Dr Anderson)
| | - David R Byrd
- From the Departments of Pathology (Dr Dintzis) and Surgery (Ms Hales and Drs Calhoun, Javid, and Byrd), University of Washington Medical Center, Seattle; Breast Surgery Clinic (Dr Harrington), Department of Pathology (Dr Tan), and Clinical Trials (Mses Fortney and Porenta), Overlake Hospital Medical Center, Bellevue, Washington; Development (Dr Miller), Clinical Operations (Mses Ishak and Gombotz), Research (Ms Hansen and Dr Parrish-Novak), and Device Development (Dr Kittle and Mr Perry), Blaze Bioscience, Inc., Seattle, Washington; and the Department of Surgery, University of Washington, Seattle (Dr Anderson)
| |
Collapse
|
28
|
Neuropilin-1 drives tumor-specific uptake of chlorotoxin. Cell Commun Signal 2019; 17:67. [PMID: 31208428 PMCID: PMC6580635 DOI: 10.1186/s12964-019-0368-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/14/2019] [Indexed: 12/12/2022] Open
Abstract
Background Chlorotoxin (Cltx) isolated from scorpion venom is an established tumor targeting and antiangiogenic peptide. Radiolabeled Cltx therapeutic (131I-TM601) yielded promising results in human glioma clinical studies, and the imaging agent tozuleristide, is under investigation in CNS cancer studies. Several binding targets have previously been proposed for Cltx but none effectively explain its pleiotropic effects; its true target remains ambiguous and is the focus of this study. Methods A peptide-drug conjugate (ER-472) composed of Cltx linked to cryptophycin as warhead was developed as a tool to probe the molecular target and mechanism of action of Cltx, using multiple xenograft models. Results Neuropilin-1 (NRP1), an endocytic receptor on tumor and endothelial cells, was identified as a novel Cltx target, and NRP1 binding by Cltx increased drug uptake into tumor. Metabolism of Cltx to peptide bearing free C-terminal arginine, a prerequisite for NRP1 binding, took place in the tumor microenvironment, while native scorpion Cltx with amidated C-terminal arginine did not bind NRP1, and instead acts as a cryptic peptide. Antitumor activity of ER-472 in xenografts correlated to tumor NRP1 expression. Potency was significantly reduced by treatment with NRP1 blocking antibodies or knockout in tumor cells, confirming a role for NRP1-binding in ER-472 activity. Higher cryptophycin metabolite levels were measured in NRP1-expressing tumors, evidence of NRP1-mediated enhanced drug uptake and presumably responsible for the superior antitumor efficacy. Conclusions NRP1 was identified as a novel Cltx target which enhances tumor drug uptake. This finding should facilitate tumor selection for chlorotoxin-based therapeutics and diagnostics. Electronic supplementary material The online version of this article (10.1186/s12964-019-0368-9) contains supplementary material, which is available to authorized users.
Collapse
|
29
|
Patil CG, Walker DG, Miller DM, Butte P, Morrison B, Kittle DS, Hansen SJ, Nufer KL, Byrnes-Blake KA, Yamada M, Lin LL, Pham K, Perry J, Parrish-Novak J, Ishak L, Prow T, Black K, Mamelak AN. Phase 1 Safety, Pharmacokinetics, and Fluorescence Imaging Study of Tozuleristide (BLZ-100) in Adults With Newly Diagnosed or Recurrent Gliomas. Neurosurgery 2019; 85:E641-E649. [DOI: 10.1093/neuros/nyz125] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 12/20/2018] [Indexed: 11/13/2022] Open
Abstract
AbstractBACKGROUNDFluorescence-guided surgery (FGS) can improve extent of resection in gliomas. Tozuleristide (BLZ-100), a near-infrared imaging agent composed of the peptide chlorotoxin and a near-infrared fluorophore indocyanine green, is a candidate molecule for FGS of glioma and other tumor types.OBJECTIVETo perform a phase 1 dose-escalation study to characterize the safety, pharmacokinetics, and fluorescence imaging of tozuleristide in adults with suspected glioma.METHODSPatients received a single intravenous dose of tozuleristide 3 to 29 h before surgery. Fluorescence images of tumor and cavity in Situ before and after resection and of excised tissue ex Vivo were acquired, along with safety and pharmacokinetic measures.RESULTSA total of 17 subjects received doses between 3 and 30 mg. No dose-limiting toxicity was observed, and no reported adverse events were considered related to tozuleristide. At doses of 9 mg and above, the terminal serum half-life for tozuleristide was approximately 30 min. Fluorescence signal was detected in both high- and low-grade glial tumors, with high-grade tumors generally showing greater fluorescence intensity compared to lower grade tumors. In high-grade tumors, signal intensity increased with increased dose levels of tozuleristide, regardless of the time of dosing relative to surgery.CONCLUSIONThese results support the safety of tozuleristide at doses up to 30 mg and suggest that tozuleristide imaging may be useful for FGS of gliomas.
Collapse
Affiliation(s)
| | - David G Walker
- NEWRO Foundation, Brisbane, Australia
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Brisbane, Australia
| | | | - Pramod Butte
- Cedars-Sinai Medical Center, Los Angeles, California
| | | | | | | | - Kaitlin L Nufer
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Brisbane, Australia
| | | | - Miko Yamada
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Brisbane, Australia
- Future Industries Institute, University of South Australia, Adelaide, Australia
| | - Lynlee L Lin
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Brisbane, Australia
| | - Kim Pham
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Brisbane, Australia
| | - Jeff Perry
- Blaze Bioscience, Inc, Seattle, Washington
| | | | | | - Tarl Prow
- Dermatology Research Centre, The University of Queensland, The University of Queensland Diamantina Institute, Brisbane, Australia
- Future Industries Institute, University of South Australia, Adelaide, Australia
| | - Keith Black
- Cedars-Sinai Medical Center, Los Angeles, California
| | | |
Collapse
|
30
|
Doerflinger A, Quang NN, Gravel E, Ducongé F, Doris E. Aptamer-decorated polydiacetylene micelles with improved targeting of cancer cells. Int J Pharm 2019; 565:59-63. [PMID: 31029658 DOI: 10.1016/j.ijpharm.2019.04.071] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 04/17/2019] [Accepted: 04/24/2019] [Indexed: 10/27/2022]
Abstract
In this study, a "click and hybridization" strategy was developed for the functionalization of polydiacetylene micelles with a targeting aptamer ligand. Decoration of the nanocarriers with an anti-Annexin A2 sequence efficiently triggered enhanced internalization of the functionalized micelles in the MCF-7 cell line, with a marked increase compared to control micelles.
Collapse
Affiliation(s)
- Anaëlle Doerflinger
- Service de Chimie Bioorganique et de Marquage (SCBM), CEA, Université Paris-Saclay, 91191 Gif-sur-Yvette, France
| | - Nam Nguyen Quang
- Molecular Imaging Research Center (MIRCen), CEA, Université Paris-Saclay, UMR9199, 92265 Fontenay-aux-Roses, France
| | - Edmond Gravel
- Service de Chimie Bioorganique et de Marquage (SCBM), CEA, Université Paris-Saclay, 91191 Gif-sur-Yvette, France
| | - Frédéric Ducongé
- Molecular Imaging Research Center (MIRCen), CEA, Université Paris-Saclay, UMR9199, 92265 Fontenay-aux-Roses, France.
| | - Eric Doris
- Service de Chimie Bioorganique et de Marquage (SCBM), CEA, Université Paris-Saclay, 91191 Gif-sur-Yvette, France.
| |
Collapse
|
31
|
Dastpeyman M, Giacomin P, Wilson D, Nolan MJ, Bansal PS, Daly NL. A C-Terminal Fragment of Chlorotoxin Retains Bioactivity and Inhibits Cell Migration. Front Pharmacol 2019; 10:250. [PMID: 30949052 PMCID: PMC6435586 DOI: 10.3389/fphar.2019.00250] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 02/26/2019] [Indexed: 11/13/2022] Open
Abstract
Chlorotoxin was originally isolated from the venom of the Israeli scorpion Leiurus quinquestriatus, and has potential as a tumor imaging agent based on its selective binding to tumor cells. Several targets have been suggested for chlorotoxin including voltage-gated chloride channels, and it has been shown to have anti-angiogenic activity and inhibit cell migration. The structure of chlorotoxin is stabilized by four disulfide bonds and contains β-sheet and helical structure. Interestingly, the reduced form has previously been shown to inhibit cell migration to the same extent as the wild type, but structural analysis indicates that the reduced form of the peptide does not maintain the native secondary structure and appears unstructured in solution. This lack of structure suggests that a short stretch of amino acids might be responsible for the bioactivity. To explore this hypothesis, we have synthesized fragments of chlorotoxin without disulfide bonds. As expected for such small peptides, NMR analysis indicated that the peptides were unstructured in solution. However, the peptide corresponding to the eight C-terminal residues inhibited cell migration, in contrast to the other fragments. Our results suggest that the C-terminal region plays a critical role in the bioactivity of chlorotoxin.
Collapse
Affiliation(s)
- Mohadeseh Dastpeyman
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Paul Giacomin
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - David Wilson
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Matthew J Nolan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Paramjit S Bansal
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Norelle L Daly
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
32
|
Wang Y, Li K, Han S, Tian YH, Hu PC, Xu XL, He YQ, Pan WT, Gao Y, Zhang Z, Zhang JW, Wei L. Chlorotoxin targets ERα/VASP signaling pathway to combat breast cancer. Cancer Med 2019; 8:1679-1693. [PMID: 30806044 PMCID: PMC6488122 DOI: 10.1002/cam4.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/07/2019] [Accepted: 01/19/2019] [Indexed: 12/29/2022] Open
Abstract
Breast cancer is one of the most common malignant tumors among women worldwide. About 70‐75% of primary breast cancers belong to estrogen receptor (ER)‐positive breast cancer. In the development of ER‐positive breast cancer, abnormal activation of the ERα pathway plays an important role and is also a key point leading to the failure of clinical endocrine therapy. In this study, we found that the small molecule peptide chlorotoxin (CTX) can significantly inhibit the proliferation, migration and invasion of breast cancer cells. In in vitro study, CTX inhibits the expression of ERα in breast cancer cells. Further studies showed that CTX can directly bind to ERα and change the protein secondary structure of its LBD domain, thereby inhibiting the ERα signaling pathway. In addition, we also found that vasodilator stimulated phosphoprotein (VASP) is a target gene of ERα signaling pathway, and CTX can inhibit breast cancer cell proliferation, migration, and invasion through ERα/VASP signaling pathway. In in vivo study, CTX significantly inhibits growth of ER overexpressing breast tumor and, more importantly, based on the mechanism of CTX interacting with ERα, we found that CTX can target ER overexpressing breast tumors in vivo. Our study reveals a new mechanism of CTX anti‐ER‐positive breast cancer, which also provides an important reference for the study of CTX anti‐ER‐related tumors.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China.,Department of Oncology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Kai Li
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Song Han
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yi-Hao Tian
- Department of Anatomy, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Peng-Chao Hu
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Xiao-Long Xu
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yan-Qi He
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Wen-Ting Pan
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yang Gao
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Zun Zhang
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Jing-Wei Zhang
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan University, Wuhan, Hubei, China
| | - Lei Wei
- Department of Pathology and Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
33
|
Cohen G, Burks SR, Frank JA. Chlorotoxin-A Multimodal Imaging Platform for Targeting Glioma Tumors. Toxins (Basel) 2018; 10:E496. [PMID: 30486274 PMCID: PMC6316809 DOI: 10.3390/toxins10120496] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/20/2018] [Accepted: 11/23/2018] [Indexed: 12/20/2022] Open
Abstract
Chlorotoxin (CTX) is a 36-amino-acid disulfide-containing peptide derived from the venom of the scorpion Leiurus quinquestriatus. CTX alters physiology in numerous ways. It interacts with voltage gated chloride channels, Annexin-2, and matrix metalloproteinase-2 (MMP-2). CTX-based bioconjugates have been widely subjected to phase I/II clinical trials and have shown substantial promise. Many studies have demonstrated that CTX preferentially binds to neuroectodermal tumors, such as glioblastoma, without cross-reactivity to normal brain cells. With its ability to penetrate the blood-brain-barrier (BBB) and its tyrosine residue allows covalent conjugation with functional moieties, CTX is an attractive platform to explore development of diagnostic and therapeutic agents for gliomas. In this review, we outline CTX structure and its molecular targets, summarize molecular variations of CTX developed for glioma imaging, and discuss future trends and perspectives for CTX conjugates as a theranostic agent.
Collapse
Affiliation(s)
- Gadi Cohen
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Scott R Burks
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Joseph A Frank
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
34
|
Li X, Liu Y, Tian H. Current Developments in Pt(IV) Prodrugs Conjugated with Bioactive Ligands. Bioinorg Chem Appl 2018; 2018:8276139. [PMID: 30402082 PMCID: PMC6191961 DOI: 10.1155/2018/8276139] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/19/2018] [Accepted: 09/06/2018] [Indexed: 12/21/2022] Open
Abstract
To overcome the side effects of and resistance to cisplatin, a variety of Pt(IV) prodrugs were designed and synthesized via different modifications including combination with lipid chains to increase hydrophobicity, conjugation with short peptide chains or nanoparticles to improve drug delivery, or addition of bioactive ligands to the axial positions of Pt(IV) complexes to exert dual-function effects. This review summarizes the recent progress in the development of Pt(IV) prodrugs conjugated with bioactive-targeting ligands, including histone deacetylase inhibitors, p53 agonists, alkylating agents, and nonsteroidal anti-inflammatory agents. Although Pt(IV) complexes that conjugated with bioactive ligands show satisfactory anticancer effects, none has been approved for clinical use. Therefore, we hope that this review will contribute to further study and development of Pt(IV) complexes conjugated with bioactive and other ligands.
Collapse
Affiliation(s)
- Xuejiao Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yahong Liu
- Tianjin Binjiang Pharma, Inc., Tianjin 300192, China
| | - Hongqi Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|
35
|
Karaca U, Kesici S, Özçubukçu S. Synthesis of Chlorotoxin by Native Chemical Ligation. JOURNAL OF THE TURKISH CHEMICAL SOCIETY, SECTION A: CHEMISTRY 2018. [DOI: 10.18596/jotcsa.408517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
36
|
Wang YS, Li H, Li Y, Zhu H, Jin YH. Identification of natural compounds targeting Annexin A2 with an anti-cancer effect. Protein Cell 2018; 9:568-579. [PMID: 29508276 PMCID: PMC5966357 DOI: 10.1007/s13238-018-0513-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/24/2018] [Indexed: 12/24/2022] Open
Abstract
Annexin A2, a multifunctional tumor associated protein, promotes nuclear factor-kappa B (NF-κB) activation by interacting with NF-κB p50 subunit and facilitating its nuclear translocation. Here we demonstrated that two ginsenosides Rg5 (G-Rg5) and Rk1 (G-Rk1), with similar structure, directly bound to Annexin A2 by molecular docking and cellular thermal shift assay. Both Rg5 and Rk1 inhibited the interaction between Annexin A2 and NF-κB p50 subunit, their translocation to nuclear and NF-κB activation. Inhibition of NF-κB by these two ginsenosides decreased the expression of inhibitor of apoptosis proteins (IAPs), leading to caspase activation and apoptosis. Over expression of K302A Annexin A2, a mutant version of Annexin A2, which fails to interact with G-Rg5 and G-Rk1, effectively reduced the NF-κB inhibitory effect and apoptosis induced by G-Rg5 and G-Rk1. In addition, the knockdown of Annexin A2 largely enhanced NF-κB activation and apoptosis induced by the two molecules, indicating that the effects of G-Rg5 and G-Rk1 on NF-κB were mainly mediated by Annexin A2. Taken together, this study for the first time demonstrated that G-Rg5 and G-Rk1 inhibit tumor cell growth by targeting Annexin A2 and NF-κB pathway, and G-Rg5 and G-Rk1 might be promising natural compounds for targeted cancer therapy.
Collapse
Affiliation(s)
- Yu-Shi Wang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University, Changchun, 130012, China
| | - He Li
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University, Changchun, 130012, China
| | - Yang Li
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University, Changchun, 130012, China
| | - Hongyan Zhu
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University, Changchun, 130012, China
| | - Ying-Hua Jin
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Science, Jilin University, Changchun, 130012, China.
| |
Collapse
|
37
|
Pseudogenes of annexin A2, novel prognosis biomarkers for diffuse gliomas. Oncotarget 2017; 8:106962-106975. [PMID: 29291003 PMCID: PMC5739788 DOI: 10.18632/oncotarget.22197] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 09/20/2017] [Indexed: 12/22/2022] Open
Abstract
Diffuse gliomas is a kind of common malignant primary brain tumor. Pseudogenes have multilayered biological function in the progression of human cancers. In this study, Differentially Expressed Pseudogenes (DEPs) between glioblastomas and non-tumor controls were found by bioinformatics analysis, of which the annexin A2 pseudogenes (ANXA2P1, ANXA2P2 and ANXA2P3) were significantly up-regulated, along with the parent gene annexin A2 (ANXA2). Among four glioblastoma subtypes, ANXA2P1 and ANXA2P2 were preferentially expressed in mesenchymal subtype and less expressed in proneural subtype. Meanwhile, Pearson’s correlation analysis revealed that the expression level of ANXA2 was positively correlated with ANXA2 pseudogenes expression. Then, the expression patterns of ANXA2 and its pseudogenes were validated in diffuse glioma specimens (n=99) and non-tumor tissues (n=12) by quantitative real-time PCR (qRT-PCR). Additionally, Kaplan–Meier analysis revealed that highly expressed ANXA2 and annexin A2 pseudogenes were associated with the poor survival outcome of glioma patients. Cox regression analyses suggested that ANXA2, ANXA2P1 and ANXA2P2 were the independent prognosis factors for gliomas. Furthermore, down-regulation of ANXA2 and ANXA2 pseudogenes might contribute to the improvement of patients’ survival who received chemotherapy and radiotherapy. These results demonstrated that ANXA2 pseudogenes and ANXA2 could be used as the novel biomarkers for diagnosis, prognosis and target therapy of gliomas.
Collapse
|
38
|
Ma R, Mahadevappa R, Kwok HF. Venom-based peptide therapy: insights into anti-cancer mechanism. Oncotarget 2017; 8:100908-100930. [PMID: 29246030 PMCID: PMC5725072 DOI: 10.18632/oncotarget.21740] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 09/22/2017] [Indexed: 01/17/2023] Open
Abstract
The 5-year relative survival rate of all types of cancer has increased significantly over the past three decades partly due to the targeted therapy. However, still there are many targeted therapy drugs could play a role only in a portion of cancer patients with specific molecular alternation. It is necessary to continue to develop new biological agents which could be used alone and/or in combination with current FDA approved drugs to treat complex cancer diseases. Venom-based drugs have been used for hundreds of years in human history. Nevertheless, the venom-origin of the anti-cancer drug do rarely appear in the pharmaceutical market; and this is due to the fact that the mechanism of action for a large number of the venom drug such as venom-based peptide is not clearly understood. In this review, we focus on discussing some identified venom-based peptides and their anti-cancer mechanisms including the blockade of cancer cell proliferation, invasion, angiogenesis, and metastasis (hallmarks of cancer) to fulfill the gap which is hindering their use in cancer therapy. Furthermore, it also highlights the importance of immunotherapy based on venom peptide. Overall, this review provides readers for further understanding the mechanism of venom peptide and elaborates on the need to explore peptide-based therapeutic strategies.
Collapse
Affiliation(s)
- Rui Ma
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR
| | - Ravikiran Mahadevappa
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR
| | - Hang Fai Kwok
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR
| |
Collapse
|
39
|
Liu W, Hajjar KA. The annexin A2 system and angiogenesis. Biol Chem 2017; 397:1005-16. [PMID: 27366903 DOI: 10.1515/hsz-2016-0166] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/28/2016] [Indexed: 01/23/2023]
Abstract
The formation of new blood vessels from pre-existing vasculature, the process known as angiogenesis, is highly regulated by pro- and anti-angiogenic signaling molecules including growth factors and proteases. As an endothelial cell-surface co-receptor for plasminogen and tissue plasminogen activator, the annexin A2 (ANXA2) complex accelerates plasmin generation and facilitates fibrinolysis. Plasmin can subsequently activate a downstream proteolytic cascade involving multiple matrix metalloproteinases. Thus, in addition to maintaining blood vessel patency, the ANXA2 complex can also promote angiogenesis via its pro-fibrinolytic activity. The generation of ANXA2-deficient mice allowed us to first observe the pro-angiogenic role of ANXA2 in vivo. Further investigations have provided additional details regarding the mechanism for ANXA2 regulation of retinal and corneal angiogenesis. Other studies have reported that ANXA2 supports angiogenesis in specific tumor-related settings. Here, we summarize results from in vivo studies that illustrate the pro-angiogenic role of ANXA2, and discuss the critical questions that may lead to an advanced understanding of the molecular mechanisms for ANXA2-mediated angiogenesis. Finally, highlights from studies on ANXA2-interacting agents offer potential therapeutic opportunities for the application of ANXA2-centered pharmaceuticals in angiogenesis-related disorders.
Collapse
|
40
|
Lu L, Qi H, Zhu J, Sun WX, Zhang B, Tang CY, Cheng Q. Vascular-homing peptides for cancer therapy. Biomed Pharmacother 2017; 92:187-195. [PMID: 28544932 DOI: 10.1016/j.biopha.2017.05.054] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 12/11/2022] Open
Abstract
In the past 30 years, a variety of phage libraries have been extensively utilized to identify and develop tumor homing peptides (THPs). THPs specifically bind to tumor cells or elements of the tumor microenvironment while no or low affinity to normal cells. In this regard, the efficacy of therapeutic agents in cancer therapy can be enhanced by targeting strategies based on coupling with THPs that recognize receptors expressed by tumor cells or tumor vasculature. Especially, vascular-homing peptides, targeting tumor vasculature, have their receptors expressed on or around the blood vessel including pro-angiogenic factors, metalloproteinase, integrins, fibrin-fibronectin complexes, etc. This review briefly summarizes recent studies on identification and therapeutic applications of vascular-homing peptides targeting common angiogenic markers or with unknown vascular targets in some certain types of cancers. These newly discovered vascular-homing peptides are promising candidates which could provide novel strategies for cancer therapy.
Collapse
Affiliation(s)
- Lan Lu
- Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, PR China; Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, PR China.
| | - Huan Qi
- School of Life Science and Engineering, Southwest University of Science and Technology, PR China
| | - Jie Zhu
- Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, PR China
| | - Wen Xia Sun
- Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, PR China
| | - Bin Zhang
- Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, PR China
| | - Chun Yan Tang
- Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, PR China
| | - Qiang Cheng
- Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, PR China.
| |
Collapse
|
41
|
Parrish-Novak J, Byrnes-Blake K, Lalayeva N, Burleson S, Fidel J, Gilmore R, Gayheart-Walsten P, Bricker GA, Crumb WJ, Tarlo KS, Hansen S, Wiss V, Malta E, Dernell WS, Olson JM, Miller DM. Nonclinical Profile of BLZ-100, a Tumor-Targeting Fluorescent Imaging Agent. Int J Toxicol 2017; 36:104-112. [PMID: 28403743 DOI: 10.1177/1091581817697685] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BLZ-100 is a single intravenous use, fluorescent imaging agent that labels tumor tissue to enable more complete and precise surgical resection. It is composed of a chlorotoxin peptide covalently bound to the near-infrared fluorophore indocyanine green. BLZ-100 is in clinical development for intraoperative visualization of human tumors. The nonclinical safety and pharmacokinetic (PK) profile of BLZ-100 was evaluated in mice, rats, canines, and nonhuman primates (NHP). Single bolus intravenous administration of BLZ-100 was well tolerated, and no adverse changes were observed in cardiovascular safety pharmacology, PK, and toxicology studies in rats and NHP. The single-dose no-observed-adverse-effect-levels (NOAELs) were 7 mg (28 mg/kg) in rats and 60 mg (20 mg/kg) in NHP, corresponding to peak concentration values of 89 400 and 436 000 ng/mL and area-under-the-curve exposure values of 130 000 and 1 240 000 h·ng/mL, respectively. Based on a human imaging dose of 3 mg, dose safety margins are >100 for rat and monkey. BLZ-100 produced hypersensitivity reactions in canine imaging studies (lethargy, pruritus, swollen muzzle, etc). The severity of the reactions was not dose related. In a follow-up study in dogs, plasma histamine concentrations were increased 5 to 60 minutes after BLZ-100 injection; this coincided with signs of hypersensitivity, supporting the conclusion that the reactions were histamine based. Hypersensitivity reactions were not observed in other species or in BLZ-100 human clinical studies conducted to date. The combined imaging, safety pharmacology, PK, and toxicology studies contributed to an extensive initial nonclinical profile for BLZ-100, supporting first-in-human clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Janean Fidel
- 5 Veterinary Teaching Hospital, Washington State University, Pullman, WA, USA
| | | | | | | | | | - K S Tarlo
- 8 Tarlo Toxicology Consulting, LLC, Applegate, MI, USA
| | | | - Valorie Wiss
- 5 Veterinary Teaching Hospital, Washington State University, Pullman, WA, USA
| | - Errol Malta
- 9 Blaze Bioscience Australia Pty Ltd, Melbourne, Australia
| | - William S Dernell
- 5 Veterinary Teaching Hospital, Washington State University, Pullman, WA, USA
| | - James M Olson
- 10 Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Dennis M Miller
- 1 Blaze Bioscience, Inc, Seattle, WA, USA.,9 Blaze Bioscience Australia Pty Ltd, Melbourne, Australia
| |
Collapse
|
42
|
Kerkis I, de Brandão Prieto da Silva AR, Pompeia C, Tytgat J, de Sá Junior PL. Toxin bioportides: exploring toxin biological activity and multifunctionality. Cell Mol Life Sci 2017; 74:647-661. [PMID: 27554773 PMCID: PMC11107510 DOI: 10.1007/s00018-016-2343-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/27/2016] [Accepted: 08/15/2016] [Indexed: 10/21/2022]
Abstract
Toxins have been shown to have many biological functions and to constitute a rich source of drugs and biotechnological tools. We focus on toxins that not only have a specific activity, but also contain residues responsible for transmembrane penetration, which can be considered bioportides-a class of cell-penetrating peptides that are also intrinsically bioactive. Bioportides are potential tools in pharmacology and biotechnology as they help deliver substances and nanoparticles to intracellular targets. Bioportides characterized so far are peptides derived from human proteins, such as cytochrome c (CYCS), calcitonin receptor (camptide), and endothelial nitric oxide synthase (nosangiotide). However, toxins are usually disregarded as potential bioportides. In this review, we discuss the inclusion of some toxins and molecules derived thereof as a new class of bioportides based on structure activity relationship, minimization, and biological activity studies. The comparative analysis of the amino acid residue composition of toxin-derived bioportides and their short molecular variants is an innovative analytical strategy which allows us to understand natural toxin multifunctionality in vivo and plan novel pharmacological and biotechnological products. Furthermore, we discuss how many bioportide toxins have a rigid structure with amphiphilic properties important for both cell penetration and bioactivity.
Collapse
Affiliation(s)
- Irina Kerkis
- Laboratório de Genética, Instituto Butantan, Av. Vital Brasil 1500, São Paulo, SP, 05503-900, Brazil.
| | | | - Celine Pompeia
- Laboratório de Genética, Instituto Butantan, Av. Vital Brasil 1500, São Paulo, SP, 05503-900, Brazil
| | - Jan Tytgat
- Toxicology and Pharmacology, University of Leuven (KU Leuven), Louvain, Belgium
| | - Paulo L de Sá Junior
- Laboratório de Genética, Instituto Butantan, Av. Vital Brasil 1500, São Paulo, SP, 05503-900, Brazil.
| |
Collapse
|
43
|
Abstract
Malignant gliomas are rarely curable malignant tumors in the central nervous system. Chlorotoxin (CTX) is a peptide derived from scorpion venom, which can selectively target malignant gliomas. Onconase (Onc) is a small cytotoxic ribonuclease derived from frogspawn that exhibits cytotoxicity against some tumor cells. In the present study, we found that CTX-conjugated Onc (CTX-Onc) shows better anti-tumor effect than the physical mixture of CTX and Onc (CTX + Onc) on the nude mice carrying subcutaneous glioblastoma cell-derived tumor. However, CTX-Onc does not show dose-dependent anti-tumor effect. In addition, apoptosis in tumor tissue does not show significant difference between the treatment groups. Our results confirmed that CTX-Onc has better anti-tumor effect than CTX + Onc and suggest that it can be potentially used for glioma therapy.
Collapse
Affiliation(s)
- Xiaomin Wang
- Institute of Protein Research, College of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zhanyun Guo
- Institute of Protein Research, College of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
44
|
Glioblastoma multiforme targeted therapy: The Chlorotoxin story. J Clin Neurosci 2016; 33:52-58. [DOI: 10.1016/j.jocn.2016.04.012] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 03/28/2016] [Accepted: 04/02/2016] [Indexed: 12/12/2022]
|
45
|
Baik FM, Hansen S, Knoblaugh SE, Sahetya D, Mitchell RM, Xu C, Olson JM, Parrish-Novak J, Méndez E. Fluorescence Identification of Head and Neck Squamous Cell Carcinoma and High-Risk Oral Dysplasia With BLZ-100, a Chlorotoxin-Indocyanine Green Conjugate. JAMA Otolaryngol Head Neck Surg 2016; 142:330-8. [PMID: 26892902 DOI: 10.1001/jamaoto.2015.3617] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
IMPORTANCE Surgical cure of head and neck squamous cell carcinoma (HNSCC) remains hampered by inadequately resected tumors and poor recognition of lesions with malignant potential. BLZ-100 is a chlorotoxin-based, tumor-targeting agent that has not yet been studied in HNSCC. OBJECTIVE To evaluate BLZ-100 uptake in models of HNSCC and oral dysplasia. DESIGN, SETTING, AND PARTICIPANTS This was an observational study (including sensitivity and specificity analysis) of BLZ-100 uptake in an orthotopic xenograft mouse model of HNSCC and a carcinogen-induced dysplasia model of hamster cheek pouches. INTERVENTIONS Various HNSCC xenografts were established in the tongues of NOD-scid IL2Rgammanull (NSG) mice. BLZ-100 was intravenously injected and fluorescence uptake was measured. To induce dysplasia, the carcinogen 7,12-dimethylbenz(a)anthracene (DMBA) was applied to the cheek pouch of Golden Syrian hamsters for 9 to16 weeks. BLZ-100 was subcutaneously injected, and fluorescence uptake was measured. MAIN OUTCOMES AND MEASURES The signal-to-background ratio (SBR) of BLZ-100 was measured in tumor xenografts. To calculate the sensitivity and specificity of BLZ-100 uptake, a digital grid was placed over tissue sections and correlative histologic sections to discretely measure fluorescence intensity and presence of tumor; a receiver operating characteristic (ROC) curve was then plotted. In the hamster dysplasia model, cheeks were graded according to dysplasia severity. The SBR of BLZ-100 was compared among dysplasia grades. RESULTS In HNSCC xenografts, BLZ-100 demonstrated a mean (SD) SBR of 2.51 (0.47). The ROC curve demonstrated an area under the curve (AUC) of 0.89; an SBR of 2.50 corresponded to 92% sensitivity and 74% specificity. When this analysis was focused on the tumor and nontumor interface, the AUC increased to 0.97; an SBR of 2.50 corresponded to 95% sensitivity and 91% specificity. DMBA treatment of hamster cheek pouches generated lesions representing all grades of dysplasia. The SBR of high-grade dysplasia was significantly greater than that of mild-to-moderate dysplasia (2.31 [0.71] vs 1.51 [0.34], P = .006). CONCLUSIONS AND RELEVANCE BLZ-100 is a sensitive and specific marker of HNSCC and can distinguish high-risk from low-risk dysplasia. BLZ-100 has the potential to serve as an intraoperative guide for tumor margin excision and identification of premalignant lesions.
Collapse
Affiliation(s)
- Fred M Baik
- Department of Otolaryngology-Head and Neck Surgery, University of Washington, Seattle
| | | | - Sue E Knoblaugh
- Department of Veterinary Biosciences, The Ohio State University, Columbus
| | | | - Ryan M Mitchell
- Department of Otolaryngology-Head and Neck Surgery, University of Washington, Seattle
| | - Chang Xu
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - James M Olson
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Eduardo Méndez
- Department of Otolaryngology-Head and Neck Surgery, University of Washington, Seattle4Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
46
|
Al-Asmari AK, Kunnathodi F, Al Saadon K, Idris MM. Elemental analysis of scorpion venoms. JOURNAL OF VENOM RESEARCH 2016; 7:16-20. [PMID: 27826410 PMCID: PMC5074801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 09/01/2016] [Accepted: 09/04/2016] [Indexed: 11/17/2022]
Abstract
Scorpion venom is a rich source of biomolecules, which can perturb physiological activity of the host on envenomation and may also have a therapeutic potential. Scorpion venoms produced by the columnar cells of venom gland are complex mixture of mucopolysaccharides, neurotoxic peptides and other components. This study was aimed at cataloguing the elemental composition of venoms obtained from medically important scorpions found in the Arabian peninsula. The global elemental composition of the crude venom obtained from Androctonus bicolor, Androctonus crassicauda and Leiurus quinquestriatus scorpions were estimated using ICP-MS analyzer. The study catalogued several chemical elements present in the scorpion venom using ICP-MS total quant analysis and quantitation of nine elements exclusively using appropriate standards. Fifteen chemical elements including sodium, potassium and calcium were found abundantly in the scorpion venom at PPM concentrations. Thirty six chemical elements of different mass ranges were detected in the venom at PPB level. Quantitative analysis of the venoms revealed copper to be the most abundant element in Androctonus sp. venom but at lower level in Leiurus quinquestriatus venom; whereas zinc and manganese was found at higher levels in Leiurus sp. venom but at lower level in Androctonus sp. venom. These data and the concentrations of other different elements present in the various venoms are likely to increase our understanding of the mechanisms of venom activity and their pharmacological potentials.
Collapse
|
47
|
Craig SEL, Wright J, Sloan AE, Brady-Kalnay SM. Fluorescent-Guided Surgical Resection of Glioma with Targeted Molecular Imaging Agents: A Literature Review. World Neurosurg 2016; 90:154-163. [PMID: 26915698 PMCID: PMC4915969 DOI: 10.1016/j.wneu.2016.02.060] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/11/2016] [Accepted: 02/12/2016] [Indexed: 12/11/2022]
Abstract
The median life expectancy after a diagnosis of glioblastoma is 15 months. Although chemotherapeutics may someday cure glioblastoma by killing the highly dispersive malignant cells, the most important contribution that clinicians can currently offer to improve survival is by maximizing the extent of resection and providing concurrent chemo-radiation, which has become standard. Strides have been made in this area with the advent and implementation of methods of improved intraoperative tumor visualization. One of these techniques, optical fluorescent imaging with targeted molecular imaging agents, allows the surgeon to view fluorescently labeled tumor tissue during surgery with the use of special microscopy, thereby highlighting where to resect and indicating when tumor-free margins have been obtained. This advantage is especially important at the difficult-to-observe margins where tumor cells infiltrate normal tissue. Targeted fluorescent agents also may be valuable for identifying tumor versus nontumor tissue. In this review, we briefly summarize nontargeted fluorescent tumor imaging agents before discussing several novel targeted fluorescent agents being developed for glioma imaging in the context of fluorescent-guided surgery or live molecular navigation. Many of these agents are currently undergoing preclinical testing. As the agents become available, however, it is necessary to understand the strengths and weaknesses of each.
Collapse
Affiliation(s)
- Sonya E L Craig
- Department of Molecular Biology and Microbiology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - James Wright
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Andrew E Sloan
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA; Brain Tumor and Neuro-Oncology Center, University Hospitals Case Medical Center, Cleveland, Ohio, USA
| | - Susann M Brady-Kalnay
- Department of Molecular Biology and Microbiology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.
| |
Collapse
|
48
|
Cheng Y, Zhu J, Zhao L, Xiong Z, Tang Y, Liu C, Guo L, Qiao W, Shi X, Zhao J. 131I-labeled multifunctional dendrimers modified with BmK CT for targeted SPECT imaging and radiotherapy of gliomas. Nanomedicine (Lond) 2016; 11:1253-66. [PMID: 26940668 DOI: 10.2217/nnm-2016-0001] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Aim: The poly(amidoamine) dendrimers modified with Buthus martensii Karsch chlorotoxin (BmK CT) were developed as a 131I delivery system for glioma-targeted imaging and therapy. Materials & methods: Dendrimers before and after labeling 131I were synthetized and their physicochemical properties were tested. The targeting and therapeutic efficacy of 131I-G5.NHAc-HPAO-(PEG-BmK CT)-(mPEG) dendrimer against glioma was evaluated in vitro and in vivo. Results: All the dendrimers were stable under different conditions. BmK CT modification increased the cellular uptake of dendrimers in C6 glioma cells, but not in the normal RLE-6TN cells. 131I-G5.NHAc-HPAO-(PEG-BmK CT)-(mPEG) dendrimer was radiochemically pure and could be applied in glioma-targeting single-photon emission CT (SPECT) imaging and radiotherapy. Conclusion: 131I-G5.NHAc-HPAO-(PEG-BmK CT)-(mPEG) complex is a promising multifunctional nanoplatform for glioma-specific nuclear imaging and radiotherapy.
Collapse
Affiliation(s)
- Yongjun Cheng
- Department of Nuclear Medicine, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, People's Republic of China
| | - Jingyi Zhu
- State Key Laboratory for Modification of Chemical Fibers & Polymer Materials, College of Materials Science & Engineering, Donghua University, Shanghai 201620, People's Republic of China
| | - Lingzhou Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, People's Republic of China
| | - Zhijuan Xiong
- College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, People's Republic of China
| | - Yueqin Tang
- Experiment Center, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, People's Republic of China
| | - Changcun Liu
- Department of Nuclear Medicine, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, People's Republic of China
| | - Lilei Guo
- Department of Nuclear Medicine, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, People's Republic of China
| | - Wenli Qiao
- Department of Nuclear Medicine, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, People's Republic of China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers & Polymer Materials, College of Materials Science & Engineering, Donghua University, Shanghai 201620, People's Republic of China
- College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, People's Republic of China
| | - Jinhua Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, People's Republic of China
| |
Collapse
|
49
|
Fidel J, Kennedy KC, Dernell WS, Hansen S, Wiss V, Stroud MR, Molho JI, Knoblaugh SE, Meganck J, Olson JM, Rice B, Parrish-Novak J. Preclinical Validation of the Utility of BLZ-100 in Providing Fluorescence Contrast for Imaging Spontaneous Solid Tumors. Cancer Res 2016; 75:4283-91. [PMID: 26471914 DOI: 10.1158/0008-5472.can-15-0471] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
There is a need in surgical oncology for contrast agents that can enable real-time intraoperative visualization of solid tumors that can enable complete resections while sparing normal surrounding tissues. The Tumor Paint agent BLZ-100 is a peptide-fluorophore conjugate that can specifically bind solid tumors and fluoresce in the near-infrared range, minimizing light scatter and signal attenuation. In this study, we provide a preclinical proof of concept for use of this imaging contrast agent as administered before surgery to dogs with a variety of naturally occurring spontaneous tumors. Imaging was performed on excised tissues as well as intraoperatively in a subset of cases. Actionable contrast was achieved between tumor tissue and surrounding normal tissues in adenocarcinomas, squamous cell carcinomas, mast cell tumors, and soft tissue sarcomas. Subcutaneous soft tissue sarcomas were labeled with the highest fluorescence intensity and greatest tumor-to-background signal ratio. Our results establish a foundation that rationalizes clinical studies in humans with soft tissue sarcoma, an indication with a notably high unmet need.
Collapse
Affiliation(s)
- Janean Fidel
- College of Veterinary Medicine, Washington State University, Pullman, Washington
| | - Katie C Kennedy
- College of Veterinary Medicine, Washington State University, Pullman, Washington
| | - William S Dernell
- College of Veterinary Medicine, Washington State University, Pullman, Washington
| | | | | | | | | | | | | | - James M Olson
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Brad Rice
- PerkinElmer, Inc., Waltham, Massachusetts
| | | |
Collapse
|
50
|
Ojeda PG, Wang CK, Craik DJ. Chlorotoxin: Structure, activity, and potential uses in cancer therapy. Biopolymers 2016; 106:25-36. [DOI: 10.1002/bip.22748] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/18/2015] [Accepted: 09/18/2015] [Indexed: 01/09/2023]
Affiliation(s)
- Paola G. Ojeda
- Institute for Molecular Bioscience, the University of Queensland; Brisbane QLD 4072 Australia
| | - Conan K. Wang
- Institute for Molecular Bioscience, the University of Queensland; Brisbane QLD 4072 Australia
| | - David J. Craik
- Institute for Molecular Bioscience, the University of Queensland; Brisbane QLD 4072 Australia
| |
Collapse
|