1
|
Komedchikova EN, Kolesnikova OA, Obozina AS, Antonova AO, Dukat AM, Fedotova PA, Khardikova DS, Sokol DV, Shimanskaia IO, Svetlakova AV, Shipunova VO. It takes Two: Advancing cancer treatment with two-step nanoparticle delivery. Biochem Biophys Res Commun 2025; 767:151921. [PMID: 40318380 DOI: 10.1016/j.bbrc.2025.151921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/07/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
The rapid advancement of nanobiotechnology has resulted in the development of numerous targeted nanoformulations and sophisticated nanobiorobots for biomedical applications. Despite the potential of nanostructures to improve drug delivery and therapeutic efficacy, their clinical application is still constrained by insufficient accumulation in tumor tissues. Current methodologies result in only an average of 0.6 % of administered nanoparticles reaching tumors, prompting the development of innovative strategies to improve targeting and influence the pharmacokinetics and pharmacodynamics of drugs. One such approach is two-step targeting, which includes either the concept of tumor pre-targeting with specific recognizing elements or the stimuli-sensitive activation of nanostructures. This review critically evaluates advancements in two-step drug delivery systems utilizing nanobiotechnology for targeted cancer therapy. For instance, two-step delivery based on the pre-targeting concept involves an initial injection of targeting molecules that bind to tumor-specific antigens, followed by the administration of drug-loaded nanocarriers modified with complementary adaptors. This approach enhances nanoparticle accumulation in tumors and improves therapeutic outcomes by increasing interaction avidity and overcoming steric hindrances. We critically assess existing adaptor systems for two-step drug delivery and synthesize findings from various studies demonstrating their efficacy in both in vitro and in vivo settings, while addressing challenges in clinical translation. We also explore future directions for developing novel adaptor systems to enhance two-step delivery mechanisms. This review aims to contribute to optimizing nanobiotechnology in oncology for more effective cancer therapies.
Collapse
Affiliation(s)
| | - Olga A Kolesnikova
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592, Moscow, Russia
| | | | - Arina O Antonova
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592, Moscow, Russia
| | - Alexei M Dukat
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592, Moscow, Russia
| | - Polina A Fedotova
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592, Moscow, Russia
| | - Daria S Khardikova
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592, Moscow, Russia
| | - Daniil V Sokol
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592, Moscow, Russia
| | - Iana O Shimanskaia
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592, Moscow, Russia
| | - Anna V Svetlakova
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592, Moscow, Russia
| | | |
Collapse
|
2
|
Zhao J, Li X, Ma T, Chang B, Zhang B, Fang J. Glutathione-triggered prodrugs: Design strategies, potential applications, and perspectives. Med Res Rev 2024; 44:1013-1054. [PMID: 38140851 DOI: 10.1002/med.22007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/20/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023]
Abstract
The burgeoning prodrug strategy offers a promising avenue toward improving the efficacy and specificity of cytotoxic drugs. Elevated intracellular levels of glutathione (GSH) have been regarded as a hallmark of tumor cells and characteristic feature of the tumor microenvironment. Considering the pivotal involvement of elevated GSH in the tumorigenic process, a diverse repertoire of GSH-triggered prodrugs has been developed for cancer therapy, facilitating the attenuation of deleterious side effects associated with conventional chemotherapeutic agents and/or the attainment of more efficacious therapeutic outcomes. These prodrug formulations encompass a spectrum of architectures, spanning from small molecules to polymer-based and organic-inorganic nanomaterial constructs. Although the GSH-triggered prodrugs have been gaining increasing interests, a comprehensive review of the advancements made in the field is still lacking. To fill the existing lacuna, this review undertakes a retrospective analysis of noteworthy research endeavors, based on a categorization of these molecules by their diverse recognition units (i.e., disulfides, diselenides, Michael acceptors, and sulfonamides/sulfonates). This review also focuses on explaining the distinct benefits of employing various chemical architecture strategies in the design of these prodrug agents. Furthermore, we highlight the potential for synergistic functionality by incorporating multiple-targeting conjugates, theranostic entities, and combinational treatment modalities, all of which rely on the GSH-triggering. Overall, an extensive overview of the emerging field is presented in this review, highlighting the obstacles and opportunities that lie ahead. Our overarching goal is to furnish methodological guidance for the development of more efficacious GSH-triggered prodrugs in the future. By assessing the pros and cons of current GSH-triggered prodrugs, we expect that this review will be a handful reference for prodrug design, and would provide a guidance for improving the properties of prodrugs and discovering novel trigger scaffolds for constructing GSH-triggered prodrugs.
Collapse
Affiliation(s)
- Jintao Zhao
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Xinming Li
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
| | - Tao Ma
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Bingbing Chang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Baoxin Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Jianguo Fang
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
3
|
Din SRU, Saeed S, Khan SU, Arbi FM, Xuefang G, Zhong M. Bacteria-driven cancer therapy: Exploring advancements and challenges. Crit Rev Oncol Hematol 2023; 191:104141. [PMID: 37742883 DOI: 10.1016/j.critrevonc.2023.104141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/26/2023] Open
Abstract
Cancer, a serious fatal disease caused by the uncontrolled growth of cells, is the biggest challenge flagging around medicine and health fields. Conventionally, various treatments-based strategies such as radiotherapy, chemotherapy, and alternative cancer therapies possess drugs that cannot reach the cancerous tissues and make them toxic to noncancerous cells. Cancer immunotherapy has made outstanding achievements in reducing the chances of cancer. Our considerable attention towards cancer-directed immune responses and the mechanisms behind which immune cells kill cancer cells have progressively been helpful in the advancement of new therapies. Among them, bacteria-based cancer immunotherapy has achieved much more attention due to smart and robust mechanisms in activating the host anti-tumor response. Moreover, bacterial-based therapy can be utilized as a single monotherapy or in combination with multiple anticancer immunotherapies to accelerate productive clinical results. Herein, we comprehensively reviewed recent advancements, challenges, and future perspectives in developing bacterial-based cancer immunotherapies.
Collapse
Affiliation(s)
- Syed Riaz Ud Din
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Sumbul Saeed
- School of Environment and Science, Griffith University, Nathan, QLD 4111, Australia
| | - Shahid Ullah Khan
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City and Southwest University, College of Agronomy and Biotechnology, Southwest University, Beibei, Chongqing 400715, China; Engineering Research Center of South Upland Agriculture, Ministry of Education, Chongqing 400715, China; Women Medical and Dental College, Khyber Medical University, Peshawar, KPK 22020, Pakistan
| | - Fawad Mueen Arbi
- Quaid-e-Azam Medical College, Bahawalpur, Punjab 63100, Pakistan
| | - Guo Xuefang
- Department of Medical Microbiology, Dalian Medical University, Dalian 116044, China
| | - Mintao Zhong
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
4
|
Tyagi K, Kumari R, Venkatesh V. Alkaline phosphatase (ALP) activatable small molecule-based prodrugs for cancer theranostics. Org Biomol Chem 2023; 21:4455-4464. [PMID: 37191120 DOI: 10.1039/d3ob00510k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Highly water-soluble small molecule-based prodrugs (5-FUPD and SAHAPD) are formulated. They comprise a phosphate group to lock the active drug payload (5-fluorouracil and SAHA) along with a turn-on fluorophore consisting of a glutathione (GSH) depletory feature. Installation of the phosphate group along with purification of final product has been accomplished in an operationally facile manner. Activation of the prodrugs is facilitated by alkaline phosphatase (ALP)-mediated hydrolysis of the phosphate group followed by 1,8-elimination. The prodrugs were found to be highly effective against ALP flared human cervical cancer (HeLa) and liver cancer (HepG2) cell lines. Most notably, they were found to be innocuous to normal liver cells (WRL-68).
Collapse
Affiliation(s)
- Kartikay Tyagi
- Laboratory of Chemical Biology and Medicinal Chemistry, Department of Chemistry, Indian Institute of Technology Roorkee, Uttarakhand-247667, India.
| | - Reena Kumari
- Laboratory of Chemical Biology and Medicinal Chemistry, Department of Chemistry, Indian Institute of Technology Roorkee, Uttarakhand-247667, India.
| | - V Venkatesh
- Laboratory of Chemical Biology and Medicinal Chemistry, Department of Chemistry, Indian Institute of Technology Roorkee, Uttarakhand-247667, India.
| |
Collapse
|
5
|
Queen A, Bhutto HN, Yousuf M, Syed MA, Hassan MI. Carbonic anhydrase IX: A tumor acidification switch in heterogeneity and chemokine regulation. Semin Cancer Biol 2022; 86:899-913. [PMID: 34998944 DOI: 10.1016/j.semcancer.2022.01.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/30/2021] [Accepted: 01/03/2022] [Indexed: 02/07/2023]
Abstract
The primary physiological process of respiration produces carbon dioxide (CO2) that reacts with water molecules which subsequently liberates bicarbonate (HCO-3) and protons. Carbonic anhydrases (CAs) are the primary catalyst involved in this conversion. More than 16 isoforms of human CAs show organ or subcellular specific activity. Dysregulation of each CA is associated with multiple pathologies. Out of these members, the overexpression of membrane-bound carbonic anhydrase IX (CAIX) is associated explicitly with hypoxic tumors or various solid cancers. CAIX helps tumors deal with higher CO2 by sequestering it with bicarbonate ions and helping cancer cells to grow in a comparatively hypoxic or acidic environment, thus acting as a pH adaptation switch. CAIX-mediated adaptations in cancer cells include angiogenesis, metabolic alterations, tumor heterogeneity, drug resistance, and regulation of cancer-specific chemokines. This review comprehensively collects and describe the cancer-specific expression mechanism and role of CAIX in cancer growth, progression, heterogeneity, and its structural insight to develop future combinatorial targeted cancer therapies.
Collapse
Affiliation(s)
- Aarfa Queen
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Humaira Naaz Bhutto
- Department of Biotechnology, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Mohd Yousuf
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Mansoor Ali Syed
- Department of Biotechnology, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India.
| |
Collapse
|
6
|
Soleimani N, Javadi MM. Future prospects of bacteria-mediated cancer therapies: Affliction or opportunity? Microb Pathog 2022; 172:105795. [PMID: 36155065 DOI: 10.1016/j.micpath.2022.105795] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/05/2022] [Accepted: 09/19/2022] [Indexed: 01/10/2023]
Abstract
Cancer, as a disease characterized by uncontrolled growth of cells, is recognized as one of the significant challenges in the field of health and medicine. There are various treatments for cancer like surgery, hormone therapy, chemotherapy, etc., but they have negative effects on the patient's lifestyle. Numerous side effects, and recently the emergence of drug resistance to these methods are weaknesses of these treatments. The utilization of bacteria as a treatment for cancer has attracted scientists' attention in the last decade. There are various methods of using bacteria to treat cancer, including the use of live, attenuated, or genetically engineered microbes, the use of bacterial toxins as an immunotoxin or conjugated to tumor antigens, bacteria-based cancer immunotherapy, bacterial vectors for gene-directed enzyme prodrug, and also the undeniable role of probiotics in treatment, are the cases that today are used for treatment. Bacterial therapy has shown a greater promise in cancer treatment due to its ability to lyse the tumor cells and deliver therapeutic products. However, the potential cytotoxicity of bacteria for healthy tissues, their inability to entirely lyse cancerous cells, and the possibility of mutations in their genomes are among the challenges of bacteriotherapy for cancer. Herein, we summarize the mechanism of bacteria, their potential benefits and harms, and the future of research in this field.
Collapse
Affiliation(s)
- Neda Soleimani
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Mahtab Moshref Javadi
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
7
|
Rommasi F. Bacterial-Based Methods for Cancer Treatment: What We Know and Where We Are. Oncol Ther 2022; 10:23-54. [PMID: 34780046 PMCID: PMC9098760 DOI: 10.1007/s40487-021-00177-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 10/25/2021] [Indexed: 01/10/2023] Open
Abstract
A severe disease, cancer is caused by the exponential and uncontrolled growth of cells, leading to organ dysfunction as well as disorders. This disease has been recognized as one of the significant challenges to health and medicine. Various treatment procedures for cancer are associated with diverse side effects; the most conventional cancer treatments include chemotherapy, surgery, and radiotherapy, among others. Numerous adverse and side effects, low specificity and sensitivity, narrow therapeutic windows, and, recently, the emergence of tumor cells resistant to such treatments have been documented as the shortcomings of conventional treatment strategies. As a group of prokaryotic microorganisms, bacteria have great potential for use in cancer therapy. Currently, utilizing bacteria for cancer treatment has attracted the attention of scientists. The high potential of bacteria to become non-pathogenic by genetic manipulation, their distinguished virulence factors (which can be used as weapons against tumors), their ability to proliferate in tissues, and the contingency to control their population by administrating antibiotics, etc., have made bacteria viable candidates and live micro-medication for cancer therapies. However, the possible cytotoxicity impacts of bacteria, their inability to entirely lyse cancerous cells, as well as the probability of mutations in their genomes are among the significant challenges of bacteria-based methods for cancer treatment. In this article, various available data on bacterial therapeutics, along with their pros and cons, are discussed.
Collapse
Affiliation(s)
- Foad Rommasi
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
8
|
Ahmadi M, Potlitz F, Link A, von Woedtke T, Nasri Z, Wende K. Flucytosine-based prodrug activation by cold physical plasma. Arch Pharm (Weinheim) 2022; 355:e2200061. [PMID: 35621706 DOI: 10.1002/ardp.202200061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/05/2022] [Accepted: 04/26/2022] [Indexed: 11/11/2022]
Abstract
Reactive oxygen species (ROS) are known to trigger drug release from arylboronate-containing ROS-responsive prodrugs. In cancer cells, elevated levels of ROS can be exploited for the selective activation of prodrugs via Baeyer-Villiger type oxidation rearrangement sequences. Here, we report a proof of concept to demonstrate that these cascades can as well be initiated by cold physical plasma (CPP). An analog of a recently reported fluorouracil prodrug based on the less toxic drug 5-fluorocytosine (5-FC) was synthesized with a view to laboratory safety reasons and used as a model compound to prove our hypothesis that CPP is suitable as a trigger for the prodrug activation. Although the envisioned oxidation and rearrangement with successive loss of boronic acid species could be achieved by plasma treatment, the anticipated spontaneous liberation of 5-FC was inefficient in the model case. However, the obtained results suggest that custom-tailored CPP-responsive prodrugs might become an evolving research field.
Collapse
Affiliation(s)
- Mohsen Ahmadi
- Center for Innovation Competence (ZIK) Plasmatis, Leibniz Institute for Plasma Science and Technology (INP Greifswald), Greifswald, Germany
| | - Felix Potlitz
- Institute of Pharmacy, University of Greifswald, Greifswald, Germany
| | - Andreas Link
- Institute of Pharmacy, University of Greifswald, Greifswald, Germany
| | - Thomas von Woedtke
- Center for Innovation Competence (ZIK) Plasmatis, Leibniz Institute for Plasma Science and Technology (INP Greifswald), Greifswald, Germany.,Leibniz Institute for Plasma Science and Technology (INP Greifswald), Greifswald, Germany.,Institute for Hygiene and Environmental Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Zahra Nasri
- Center for Innovation Competence (ZIK) Plasmatis, Leibniz Institute for Plasma Science and Technology (INP Greifswald), Greifswald, Germany
| | - Kristian Wende
- Center for Innovation Competence (ZIK) Plasmatis, Leibniz Institute for Plasma Science and Technology (INP Greifswald), Greifswald, Germany
| |
Collapse
|
9
|
Yaghoubi A, Khazaei M, Ghazvini K, Hasanian SM, Avan A, Soleimanpour S. Bacterial Peptide and Bacteriocins in Treating Gynecological Cancers. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10411-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
10
|
Martin H, Lázaro LR, Gunnlaugsson T, Scanlan EM. Glycosidase activated prodrugs for targeted cancer therapy. Chem Soc Rev 2022; 51:9694-9716. [DOI: 10.1039/d2cs00379a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In this review glycosidase activated prodrugs that target cancer cells are discussed.
Collapse
Affiliation(s)
- Harlei Martin
- School of Chemistry and Trinity Bioscience Institute, The University of Dublin, Trinity College Dublin, Dublin 2, Ireland
| | - Laura Ramírez Lázaro
- School of Chemistry and Trinity Bioscience Institute, The University of Dublin, Trinity College Dublin, Dublin 2, Ireland
- SFI Synthesis and Solid State Pharmaceutical Centre (SSPC), Ireland
| | - Thorfinnur Gunnlaugsson
- School of Chemistry and Trinity Bioscience Institute, The University of Dublin, Trinity College Dublin, Dublin 2, Ireland
- SFI Synthesis and Solid State Pharmaceutical Centre (SSPC), Ireland
| | - Eoin M. Scanlan
- School of Chemistry and Trinity Bioscience Institute, The University of Dublin, Trinity College Dublin, Dublin 2, Ireland
- SFI Synthesis and Solid State Pharmaceutical Centre (SSPC), Ireland
| |
Collapse
|
11
|
Chowdhury M, Kumar Das P. Paclitaxel-Loaded Biotinylated Fe 2+-Doped Carbon Dot: Combination Therapy in Cancer Treatment. ACS APPLIED BIO MATERIALS 2021; 4:5132-5144. [PMID: 35006997 DOI: 10.1021/acsabm.1c00348] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The present research work delineates the design and preparation of covalently tailored biotinylated Fe2+-doped carbon dots (FCDb). The FCDb was successfully used as a pro-drug activator, diagnostic probe, and target-specific delivery vehicle for anticancer drug paclitaxel in pro-drug-free drug combination therapy of cancer treatment. Fe2+-doped carbon dot was synthesized via the hydrothermal method (FCD). The surface of FCD was covalently modified with cancer cell targeting ligand biotin (FCDb). Microscopic and spectroscopic methods were used to characterize aqueous soluble FCD and FCDb. Both FCD and FCDb emit blue fluorescence under UV light irradiation. FCD and FCDb can effectively sense H2O2 by fluorescence quenching as well as activate H2O2 (pro-drug), which oxidatively damage the DNA through the generation of reactive oxygen species (ROS: superoxide (O2•-), hydroxyl radical (•OH), etc). Both FCD and FCDb were utilized as selective cellular markers for cancer cell B16F10 owing to their high H2O2 content, which was more distinct in the case of FCDb due to the overexpression of biotin receptor in cancer cell. Anticancer drug paclitaxel (PTX)-loaded FCDb (FCDb-PTX) was employed for the selective killing of B16F10 cancer cells. This pro-drug-free drug formulation (FCDb-PTX) exhibited ∼2.7- to 3.5-fold higher killing of B16F10 cells mostly via early as well as late apoptotic path in comparison to noncancer NIH3T3 cells through the synergistic action of ROS (generated from H2O2 in the presence of FCDb) and anticancer effect of PTX. Hence, this newly developed FCDb-PTX can act as a potential theranostic agent in the domain of combination therapy of cancer treatment.
Collapse
Affiliation(s)
- Monalisa Chowdhury
- School of Biological Sciences, Indian Association for the Cultivation of Science Jadavpur, Kolkata 700 032, India
| | - Prasanta Kumar Das
- School of Biological Sciences, Indian Association for the Cultivation of Science Jadavpur, Kolkata 700 032, India
| |
Collapse
|
12
|
Besse HC, Chen Y, Scheeren HW, Metselaar JM, Lammers T, Moonen CTW, Hennink WE, Deckers R. A Doxorubicin-Glucuronide Prodrug Released from Nanogels Activated by High-Intensity Focused Ultrasound Liberated β-Glucuronidase. Pharmaceutics 2020; 12:E536. [PMID: 32532061 PMCID: PMC7355552 DOI: 10.3390/pharmaceutics12060536] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 11/16/2022] Open
Abstract
The poor pharmacokinetics and selectivity of low-molecular-weight anticancer drugs contribute to the relatively low effectiveness of chemotherapy treatments. To improve the pharmacokinetics and selectivity of these treatments, the combination of a doxorubicin-glucuronide prodrug (DOX-propGA3) nanogel formulation and the liberation of endogenous β-glucuronidase from cells exposed to high-intensity focused ultrasound (HIFU) were investigated in vitro. First, a DOX-propGA3-polymer was synthesized. Subsequently, DOX-propGA3-nanogels were formed from this polymer dissolved in water using inverse mini-emulsion photopolymerization. In the presence of bovine β-glucuronidase, the DOX-propGA3 in the nanogels was quantitatively converted into the chemotherapeutic drug doxorubicin. Exposure of cells to HIFU efficiently induced liberation of endogenous β-glucuronidase, which in turn converted the prodrug released from the DOX-propGA3-nanogels into doxorubicin. β-glucuronidase liberated from cells exposed to HIFU increased the cytotoxicity of DOX-propGA3-nanogels to a similar extend as bovine β-glucuronidase, whereas in the absence of either bovine β-glucuronidase or β-glucuronidase liberated from cells exposed to HIFU, the DOX-propGA3-nanogels hardly showed cytotoxicity. Overall, DOX-propGA3-nanogels systems might help to further improve the outcome of HIFU-related anticancer therapy.
Collapse
Affiliation(s)
- Helena C. Besse
- Division of Imaging and Oncology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (H.C.B.); (C.T.W.M.)
| | - Yinan Chen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (Y.C.); (T.L.); (W.E.H.)
| | - Hans W. Scheeren
- Cluster for Molecular Chemistry, Radboud University, 6525 XZ Nijmegen, The Netherlands;
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, 52074 Aachen, Germany;
| | - Josbert M. Metselaar
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, 52074 Aachen, Germany;
- Department of Targeted Therapeutics, MIRA Institute for Biomedical Engineering and Technical Medicine, University of Twente, 7500 AE Enschede, The Netherlands
| | - Twan Lammers
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (Y.C.); (T.L.); (W.E.H.)
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, 52074 Aachen, Germany;
- Department of Targeted Therapeutics, MIRA Institute for Biomedical Engineering and Technical Medicine, University of Twente, 7500 AE Enschede, The Netherlands
| | - Chrit T. W. Moonen
- Division of Imaging and Oncology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (H.C.B.); (C.T.W.M.)
| | - Wim E. Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (Y.C.); (T.L.); (W.E.H.)
| | - Roel Deckers
- Division of Imaging and Oncology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (H.C.B.); (C.T.W.M.)
| |
Collapse
|
13
|
Soleimanpour S, Hasanian SM, Avan A, Yaghoubi A, Khazaei M. Bacteriotherapy in gastrointestinal cancer. Life Sci 2020; 254:117754. [PMID: 32389833 DOI: 10.1016/j.lfs.2020.117754] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/27/2020] [Accepted: 05/04/2020] [Indexed: 01/13/2023]
Abstract
The most prevalent gastrointestinal (GI) cancers include colorectal cancer, stomach cancer, and liver cancer, known as the most common causes of cancer-related death in both men and women populations in the world. Traditional therapeutic approaches, including surgery, radiotherapy, and chemotherapy have failed in the effective treatment of cancer. Therefore, there is an urgent need for finding new effective anticancer agents. The available evidence and also the promising results of using bacteria as the anticancer agents on numerous cancer cell lines have attracted the attention of scientists for the therapeutic role of bacteria in the field of cancer therapy. Moreover, several studies on the bacteriotherapy agents have used genetic engineering to overcome the challenges and enhance the efficacy with the least drawbacks. Numerous bacterial species that can specifically target and internalize into the tumor cells are used live, attenuated, or genetically as compared to selectively consider the hypoxic condition of tumor, which results in the tumor suppression. The present study is a comprehensive review of the current literature on the use of bacteria and their substances such as bacteriocins and toxins in the treatment of different types of gastrointestinal cancers.
Collapse
Affiliation(s)
- Saman Soleimanpour
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hasanian
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atieh Yaghoubi
- Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Majid Khazaei
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
14
|
Bacteria and cancer: Different sides of the same coin. Life Sci 2020; 246:117398. [PMID: 32032647 DOI: 10.1016/j.lfs.2020.117398] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/20/2020] [Accepted: 02/01/2020] [Indexed: 12/14/2022]
Abstract
Conventional cancer therapies such as chemotherapy, radiation therapy, and immunotherapy due to the complexity of cancer have been unsuccessful in the complete eradication of tumor cells. Thus, there is a need for new therapeutic strategies toward cancer. Recently, the therapeutic role of bacteria in different fields of medicine and pharmaceutical research has attracted attention in recent decades. Although several bacteria are notorious as cancer-causing agents, recent research revealed intriguing results suggesting the bacterial potential in cancer therapy. Thus, bacterial cancer therapy is an alternative anticancer approach that has promising results on tumor cells in-vivo. Moreover, with the aid of genetic engineering, some natural or genetically modified bacterial strains can directly target hypoxic regions of tumors and secrete therapeutic molecules leading to cancer cell death. Additionally, stimulation of immune cells by bacteria, bacterial cancer DNA vaccine and antitumor bacterial metabolites are other therapeutic applications of bacteria in cancer therapy. The present study is a comprehensive review of different aspects of bacterial cancer therapy alone and in combination with conventional methods, for improving cancer therapy.
Collapse
|
15
|
Areas ES, de Assunção Paiva JL, Ribeiro FV, Pereira TM, Kummerle AE, Silva H, Guedes GP, Cellis do Nascimento AC, da Silva Miranda F, Neves AP. Redox-Activated Drug Delivery Properties and Cytotoxicity of Cobalt Complexes Based on a Fluorescent Coumarin-β-Keto Ester Hybrid. Eur J Inorg Chem 2019. [DOI: 10.1002/ejic.201900734] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Esther Saraiva Areas
- Instituto de Química; Universidade Federal Rural do Rio de Janeiro; BR-465, Km 7 Seropédica CEP 23890-000 Rio de Janeiro Brasil
| | | | - Felipe Vitório Ribeiro
- Instituto de Química; Universidade Federal Rural do Rio de Janeiro; BR-465, Km 7 Seropédica CEP 23890-000 Rio de Janeiro Brasil
| | - Thiago Moreira Pereira
- Instituto de Química; Universidade Federal Rural do Rio de Janeiro; BR-465, Km 7 Seropédica CEP 23890-000 Rio de Janeiro Brasil
| | - Arthur Eugen Kummerle
- Instituto de Química; Universidade Federal Rural do Rio de Janeiro; BR-465, Km 7 Seropédica CEP 23890-000 Rio de Janeiro Brasil
| | - Heveline Silva
- Departamento de Química; Universidade Federal de Minas Gerais; Av. Antônio Carlos, 6627 - Pampulha Belo Horizonte Minas Gerais CEP 31270-901 Brasil
| | - Guilherme Pereira Guedes
- Instituto de Química; Universidade Federal Fluminense; Campus do Valonguinho, Centro, Niterói Rio de Janeiro CEP 24020-150 Brasil
| | | | - Fabio da Silva Miranda
- Instituto de Química; Universidade Federal Fluminense; Campus do Valonguinho, Centro, Niterói Rio de Janeiro CEP 24020-150 Brasil
| | - Amanda Porto Neves
- Instituto de Química; Universidade Federal Rural do Rio de Janeiro; BR-465, Km 7 Seropédica CEP 23890-000 Rio de Janeiro Brasil
| |
Collapse
|
16
|
Di Marco L, Zhang JZ, Doan J, Kim BJ, Yamamoto N, Bryce NS, Hambley TW. Modulating the Cellular Uptake of Fluorescently Tagged Substrates of Prostate-Specific Antigen before and after Enzymatic Activation. Bioconjug Chem 2018; 30:124-133. [DOI: 10.1021/acs.bioconjchem.8b00792] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Lina Di Marco
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Jenny Z. Zhang
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - John Doan
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Byung J. Kim
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Natsuho Yamamoto
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Nicole S. Bryce
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Trevor W. Hambley
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
17
|
Renfrew AK, O'Neill ES, Hambley TW, New EJ. Harnessing the properties of cobalt coordination complexes for biological application. Coord Chem Rev 2018. [DOI: 10.1016/j.ccr.2017.11.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
18
|
Sapre AA, Novitskaya E, Vakharia V, Cota A, Wrasidlo W, Hanrahan SM, Derenzo S, Makale MT, Graeve OA. Optimized Scintillator YAG:Pr Nanoparticles for X-ray Inducible Photodynamic Therapy. MATERIALS LETTERS 2018; 228:49-52. [PMID: 30505045 PMCID: PMC6258075 DOI: 10.1016/j.matlet.2018.05.090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
We describe a sol-gel synthetic method for the production of praseodymium-doped yttrium aluminum garnet (YAG) nanoparticles suitable for X-ray inducible photodynamic therapy (X-PDT). Our sol-gel based approach was optimized by varying temperature and time of calcination, resulting in nanoparticles that were smooth, spherical, and 50-200 nm in crystallite size. The powders were uniformly coated with a thin (10 nm) layer of silica to facilitate surface conjugation with functional moieties. Measurements of photon flux revealed that coated and uncoated powders emitted a similar photon emission spectrum in response to 50 keVp X-rays. We also determined that the presence of silica did not significantly reduce flux and the emission peak had a maximum at approximately 320 nm. Thus, these YAG:Pr powders are suitable candidates for future in vivo X-PDT studies.
Collapse
Affiliation(s)
- Ajay A. Sapre
- University of California, San Diego, Department of Bioengineering, 9500 Gilman Drive – MC 0412, La Jolla, CA 92093-0412, USA
| | - Ekaterina Novitskaya
- University of California, San Diego, Department of Mechanical and Aerospace Engineering, 9500 Gilman Drive – MC 0411, La Jolla, CA 92093-0411, USA
| | - Ved Vakharia
- University of California, San Diego, Department of Mechanical and Aerospace Engineering, 9500 Gilman Drive – MC 0411, La Jolla, CA 92093-0411, USA
| | - Alejandro Cota
- University of California, San Diego, Department of Mechanical and Aerospace Engineering, 9500 Gilman Drive – MC 0411, La Jolla, CA 92093-0411, USA
| | - Wolfgang Wrasidlo
- University of California, San Diego, Department of Neurosciences, 9500 Gilman Drive – MC 0662, La Jolla, CA 92093-0662, USA
| | - Stephen M. Hanrahan
- Lawrence Berkeley National Laboratory, Molecular Biophysics and Integrated Bioimaging Division, Cellular and Tissue Imaging Department, 1 Cyclotron Road, M/S 55-121, Berkeley, CA 94720, USA
| | - Stephen Derenzo
- Lawrence Berkeley National Laboratory, Molecular Biophysics and Integrated Bioimaging Division, Cellular and Tissue Imaging Department, 1 Cyclotron Road, M/S 55-121, Berkeley, CA 94720, USA
| | - Milan T. Makale
- University of California, San Diego, Department of Radiation Medicine and Applied Sciences, 3855 Health Sciences Drive #0819, La Jolla, CA 92093-0819, USA
| | - Olivia A. Graeve
- University of California, San Diego, Department of Mechanical and Aerospace Engineering, 9500 Gilman Drive – MC 0411, La Jolla, CA 92093-0411, USA
| |
Collapse
|
19
|
Rashidi FB, AlQhatani AD, Bashraheel SS, Shaabani S, Groves MR, Dömling A, Goda SK. Isolation and molecular characterization of novel glucarpidases: Enzymes to improve the antibody directed enzyme pro-drug therapy for cancer treatment. PLoS One 2018; 13:e0196254. [PMID: 29698433 PMCID: PMC5919439 DOI: 10.1371/journal.pone.0196254] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 04/09/2018] [Indexed: 11/19/2022] Open
Abstract
Repeated cycles of antibody-directed enzyme pro-drug therapy (ADEPT) and the use of glucarpidase in the detoxification of cytotoxic methotrexate (MTX) are highly desirable during cancer therapy but are hampered by the induced human antibody response to glucarpidase. Novel variants of glucarpidase (formal name: carboxypeptidase G2, CPG2) with epitopes not recognized by the immune system are likely to allow repeated cycles of ADEPT for effective cancer therapy. Towards this aim, over two thousand soil samples were collected and screened for folate hydrolyzing bacteria using folate as the sole carbon source. The work led to the isolation and the characterization of three new glucarpidase producing strains, which were designated as: Pseudomonas lubricans strain SF168, Stenotrophomonas sp SA and Xenophilus azovorans SN213. The CPG2 genes of Xenophilus azovorans SN213 (named Xen CPG2) and Stenotrophomonas sp SA (named Sten CPG2) were cloned and molecularly characterized. Both Xen CPG2 and Sten CPG2 share very close amino acid sequences (99%); we therefore, focused on the study of Xen CPG2. Finally, we demonstrated that a polyclonal antibody raised against our new CPG2, Xen CPG2, does not react with the CPG2 from Pseudomonas sp. strain RS-16 (Ps CPG2) that are currently in clinical use. The two enzymes, therefore could potentially be used consecutively in the ADEPT protocol to minimize the effect of the human antibody response that hampers current treatment with Ps CPG2. The identified novel CPG2 in this study will, therefore, pave the way for safer antibody directed enzyme pro-drug therapy for cancer treatment.
Collapse
Affiliation(s)
| | - Alanod D. AlQhatani
- Anti-doping Lab-Qatar, Research Department, Protein Engineering unit, Doha, Qatar
- Drug Design Group, Department of Pharmacy, University of Groningen, Antonius Deusinglaan, AV Groningen, The Netherlands
| | - Sara S. Bashraheel
- Anti-doping Lab-Qatar, Research Department, Protein Engineering unit, Doha, Qatar
- Drug Design Group, Department of Pharmacy, University of Groningen, Antonius Deusinglaan, AV Groningen, The Netherlands
| | - Shabnam Shaabani
- Drug Design Group, Department of Pharmacy, University of Groningen, Antonius Deusinglaan, AV Groningen, The Netherlands
| | - Matthew R. Groves
- Drug Design Group, Department of Pharmacy, University of Groningen, Antonius Deusinglaan, AV Groningen, The Netherlands
| | - Alexander Dömling
- Drug Design Group, Department of Pharmacy, University of Groningen, Antonius Deusinglaan, AV Groningen, The Netherlands
| | - Sayed K. Goda
- Cairo University, Faculty of Science, Giza, Egypt
- Anti-doping Lab-Qatar, Research Department, Protein Engineering unit, Doha, Qatar
| |
Collapse
|
20
|
Zhang R, Feng L, Dong Z, Wang L, Liang C, Chen J, Ma Q, Zhang R, Chen Q, Wang Y, Liu Z. Glucose & oxygen exhausting liposomes for combined cancer starvation and hypoxia-activated therapy. Biomaterials 2018; 162:123-131. [DOI: 10.1016/j.biomaterials.2018.02.004] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/08/2018] [Accepted: 02/02/2018] [Indexed: 12/24/2022]
|
21
|
Lambruschini C, Villa S, Banfi L, Canepa F, Morana F, Relini A, Riani P, Riva R, Silvetti F. Enzymatically promoted release of organic molecules linked to magnetic nanoparticles. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2018; 9:986-999. [PMID: 29719751 PMCID: PMC5905276 DOI: 10.3762/bjnano.9.92] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/27/2018] [Indexed: 06/08/2023]
Abstract
Magnetite-based magnetic nanoparticles have been successfully coupled to an organic system constituted of a fluorescent molecule, a tripeptide specifier and a spacer. The system is able to selectively release the fluorescent molecule upon targeted enzymatic hydrolysis promoted by a lysine/arginine specific protease.
Collapse
Affiliation(s)
- Chiara Lambruschini
- Department of Chemistry and Industrial Chemistry, Università di Genova, via Dodecaneso, 31 16146 Genova, Italy
| | - Silvia Villa
- Department of Chemistry and Industrial Chemistry, Università di Genova, via Dodecaneso, 31 16146 Genova, Italy
| | - Luca Banfi
- Department of Chemistry and Industrial Chemistry, Università di Genova, via Dodecaneso, 31 16146 Genova, Italy
| | - Fabio Canepa
- Department of Chemistry and Industrial Chemistry, Università di Genova, via Dodecaneso, 31 16146 Genova, Italy
| | - Fabio Morana
- Department of Chemistry and Industrial Chemistry, Università di Genova, via Dodecaneso, 31 16146 Genova, Italy
| | - Annalisa Relini
- Department of Chemistry and Industrial Chemistry, Università di Genova, via Dodecaneso, 31 16146 Genova, Italy
| | - Paola Riani
- Department of Chemistry and Industrial Chemistry, Università di Genova, via Dodecaneso, 31 16146 Genova, Italy
| | - Renata Riva
- Department of Chemistry and Industrial Chemistry, Università di Genova, via Dodecaneso, 31 16146 Genova, Italy
| | - Fulvio Silvetti
- Department of Chemistry and Industrial Chemistry, Università di Genova, via Dodecaneso, 31 16146 Genova, Italy
| |
Collapse
|
22
|
Adiyala PR, Tekumalla V, Sayeed IB, Nayak VL, Nagarajan A, Shareef MA, Nagaraju B, Kamal A. Development of pyrrolo[2,1- c ][1,4]benzodiazepine β-glucoside prodrugs for selective therapy of cancer. Bioorg Chem 2018; 76:288-293. [DOI: 10.1016/j.bioorg.2017.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/17/2017] [Accepted: 12/01/2017] [Indexed: 11/24/2022]
|
23
|
F. Tietze L, Gandamala R, Hoekman S, Kangani M, E. Nidhiry J, Penchalaiah K, Singh Raghuvanshi D. Synthesis and Biology of Dimeric, Trimeric and Tetrameric Analogues of Duocarmycin SA. HETEROCYCLES 2018. [DOI: 10.3987/com-17-s(t)16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
24
|
Zheng N, Wang X, Wang Y, Xu G, Zhang H, Dai W, He B, Zhang Q, Ji J, Wang X. A sensitive liquid chromatography/electrospray tandem mass spectroscopy method for simultaneous quantification of a disulfide bond doxorubicin conjugation prodrug and activated doxorubicin: Application to cellular pharmacokinetic and catabolism studies. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1065-1066:96-103. [PMID: 28957779 DOI: 10.1016/j.jchromb.2017.09.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/11/2017] [Accepted: 09/20/2017] [Indexed: 11/29/2022]
Abstract
In recent years, drug conjugates as a prodrug strategy have been widely studied, especially combined with nanotechnology. Disulfide-linked doxorubicin drug-drug conjugate (DOX-S-S-DOX) nanoparticles, have recently been developed as a doxorubicin prodrug nanoparticles with greater anticancer activity and less toxicity than doxorubicin in vivo, while its intracellular kinetics and metabolism is unclear which may provide us with a deeper understanding of its pharmacological mechanism and antitumor effect. Hence, in this study, a rapid and sensitive ultra high-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method was developed to detect doxorubicin (DOX) activated from DOX-S-S-DOX, as well as the prodrug itself in human breast cancer tumor cells (MCF-7). Sample preparation involved acetonitrile precipitation to extract the analytes simultaneously and bath sonication to remove intercalated DOX from DNA. The calibration range was 3-60ng/mL for DOX and 20-400ng/mL for DOX-S-S-DOX with the correlation coefficients (r2)≥0.99, using daunorubicin as internal standard (IS). The inter- and intra-assay precision (relative standard deviation, RSD%) of quality control samples was in the acceptable range (<15%) and relative error (RE%) for accuracy was between -5.35 and 9.18% for all analytes. Recovery (59.28-69.53% for DOX-S-S-DOX and 99.13-100.10% for DOX) and matrix effect (99.69-111.19%) was consistent, precise, and reproducible at different quality control levels in accordance with FDA guidance. Stability studies showed that DOX-S-S-DOX was unstable both during the bench-top and long-term storage, while the stability during sample preparation and LC-MS runtime was suitable for all the analytes. Hence, the samples should be prepared as soon as possible at the time point to prevent the catabolism of DOX-S-S-DOX. The assay was successfully used in the cellular metabolism and pharmacokinetics study of DOX-S-S-DOX and it may give a clue to explore analytical methods of other prodrug forms of DOX.
Collapse
Affiliation(s)
- Nan Zheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), National Drug Clinical Trial Center, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yaoqi Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Guobing Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), National Drug Clinical Trial Center, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Hua Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Wenbing Dai
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bing He
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jiafu Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), National Drug Clinical Trial Center, Peking University Cancer Hospital & Institute, Beijing 100142, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China.
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
25
|
Qu F, Park S, Martinez K, Gray JL, Thowfeik FS, Lundeen JA, Kuhn AE, Charboneau DJ, Gerlach DL, Lockart MM, Law JA, Jernigan KL, Chambers N, Zeller M, Piro NA, Kassel WS, Schmehl RH, Paul JJ, Merino EJ, Kim Y, Papish ET. Ruthenium Complexes are pH-Activated Metallo Prodrugs (pHAMPs) with Light-Triggered Selective Toxicity Toward Cancer Cells. Inorg Chem 2017. [PMID: 28636344 DOI: 10.1021/acs.inorgchem.7b01065] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Fengrui Qu
- Department
of Chemistry, The University of Alabama, Tuscaloosa, Alabama 35487-0336, United States
| | - Seungjo Park
- Department
of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, Alabama 35487-0203, United States
| | - Kristina Martinez
- Department
of Chemistry, Tulane University, New Orleans, Louisiana 70118, United States
| | - Jessica L. Gray
- Department
of Chemistry, The University of Alabama, Tuscaloosa, Alabama 35487-0336, United States
| | | | - John A. Lundeen
- Department
of Chemistry, The University of Alabama, Tuscaloosa, Alabama 35487-0336, United States
| | - Ashley E. Kuhn
- Department
of Chemistry, Villanova University, Villanova, Pennsylvania 19085, United States
| | - David J. Charboneau
- Department
of Chemistry, The University of Alabama, Tuscaloosa, Alabama 35487-0336, United States
- Department
of Chemistry, Villanova University, Villanova, Pennsylvania 19085, United States
| | - Deidra L. Gerlach
- Department
of Chemistry, The University of Alabama, Tuscaloosa, Alabama 35487-0336, United States
| | - Molly M. Lockart
- Department
of Chemistry, The University of Alabama, Tuscaloosa, Alabama 35487-0336, United States
| | - James A. Law
- Department
of Chemistry, The University of Alabama, Tuscaloosa, Alabama 35487-0336, United States
| | - Katherine L. Jernigan
- Department
of Chemistry, The University of Alabama, Tuscaloosa, Alabama 35487-0336, United States
| | - Nicole Chambers
- Department
of Chemistry, The University of Alabama, Tuscaloosa, Alabama 35487-0336, United States
| | - Matthias Zeller
- Department
of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Nicholas A. Piro
- Department
of Chemistry, Albright College, Reading, Pennsylvania 19612, United States
| | - W. Scott Kassel
- Department
of Chemistry, Villanova University, Villanova, Pennsylvania 19085, United States
| | - Russell H. Schmehl
- Department
of Chemistry, Tulane University, New Orleans, Louisiana 70118, United States
| | - Jared J. Paul
- Department
of Chemistry, Villanova University, Villanova, Pennsylvania 19085, United States
| | - Edward J. Merino
- Department
of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221, United States
| | - Yonghyun Kim
- Department
of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, Alabama 35487-0203, United States
| | - Elizabeth T. Papish
- Department
of Chemistry, The University of Alabama, Tuscaloosa, Alabama 35487-0336, United States
| |
Collapse
|
26
|
Dissanayake S, Denny WA, Gamage S, Sarojini V. Recent developments in anticancer drug delivery using cell penetrating and tumor targeting peptides. J Control Release 2017; 250:62-76. [DOI: 10.1016/j.jconrel.2017.02.006] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 02/02/2017] [Accepted: 02/02/2017] [Indexed: 12/13/2022]
|
27
|
Manoochehri Khoshinani H, Afshar S, Najafi R. Hypoxia: A Double-Edged Sword in Cancer Therapy. Cancer Invest 2016; 34:536-545. [PMID: 27824512 DOI: 10.1080/07357907.2016.1245317] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Hypoxia is a common feature of malignant tumors. There is an interactive connection between hypoxia and chemoresistance, radioresistance, invasiveness, and angiogenesis. Therefore, tumor hypoxia has been considered as a validated target for treating cancer. This review focuses on the role of hypoxia on chemoresistance and radioresistance. In addition, we address several approaches targeting tumor hypoxia, known as hypoxia-targeted therapy.
Collapse
Affiliation(s)
| | - Saeid Afshar
- a Research Center for Molecular Medicine, Hamadan University of Medical Sciences , Hamadan , Iran
| | - Rezvan Najafi
- a Research Center for Molecular Medicine, Hamadan University of Medical Sciences , Hamadan , Iran
| |
Collapse
|
28
|
Conditional Toxin Splicing Using a Split Intein System. Methods Mol Biol 2016. [PMID: 27714618 DOI: 10.1007/978-1-4939-6451-2_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Protein toxin splicing mediated by split inteins can be used as a strategy for conditional cell ablation. The approach requires artificial fragmentation of a potent protein toxin and tethering each toxin fragment to a split intein fragment. The toxin-intein fragments are, in turn, fused to dimerization domains, such that addition of a dimerizing agent reconstitutes the split intein. These chimeric toxin-intein fusions remain nontoxic until the dimerizer is added, resulting in activation of intein splicing and ligation of toxin fragments to form an active toxin. Considerations for the engineering and implementation of conditional toxin splicing (CTS) systems include: choice of toxin split site, split site (extein) chemistry, and temperature sensitivity. The following method outlines design criteria and implementation notes for CTS using a previously engineered system for splicing a toxin called sarcin, as well as for developing alternative CTS systems.
Collapse
|
29
|
Synthesis and antiproliferative activity of 9-benzylamino-6-chloro-2-methoxy-acridine derivatives as potent DNA-binding ligands and topoisomerase II inhibitors. Eur J Med Chem 2016; 116:59-70. [DOI: 10.1016/j.ejmech.2016.03.066] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/11/2016] [Accepted: 03/25/2016] [Indexed: 12/20/2022]
|
30
|
El Aissi R, Miladi I, Chezal JM, Chavignon O, Miot-Noirault E, Moreau E. Melanoma-targeted delivery system (part 2): Synthesis, radioiodination and biological evaluation in B16F0 bearing mice. Eur J Med Chem 2016; 120:304-12. [PMID: 27214141 DOI: 10.1016/j.ejmech.2016.05.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 05/05/2016] [Accepted: 05/06/2016] [Indexed: 10/21/2022]
Abstract
Here we report the synthesis and radiolabelling with iodine-125 of a melanoma-selective prodrug (17a*) and its parent drug IUdR. The in vivo and ex vivo biodistributions of [(125)I](17a*) and [(125)I]IUdR were evaluated in a model of melanoma B16F0-bearing mice. The pharmacokinetic profile of [(125)I](17a*) suggests rapid release of the active drug [(125)I]IUdR after i.v. administration of [(125)I](17a*). Preliminary metabolism studies in dedicated compartments (i.e. blood, urine and tumour) yielded results consistent with this hypothesis.
Collapse
Affiliation(s)
- Radhia El Aissi
- INSERM - Université d'Auvergne, UMR 990, IMTV, BP 184, F-63005 Clermont-Ferrand Cedex, France; Clermont Université, Université d'Auvergne, Imagerie Moléculaire et Thérapie Vectorisée, BP 10448, F-63005 Clermont-Ferrand Cedex, France
| | - Imen Miladi
- INSERM - Université d'Auvergne, UMR 990, IMTV, BP 184, F-63005 Clermont-Ferrand Cedex, France; Clermont Université, Université d'Auvergne, Imagerie Moléculaire et Thérapie Vectorisée, BP 10448, F-63005 Clermont-Ferrand Cedex, France
| | - Jean-Michel Chezal
- INSERM - Université d'Auvergne, UMR 990, IMTV, BP 184, F-63005 Clermont-Ferrand Cedex, France; Clermont Université, Université d'Auvergne, Imagerie Moléculaire et Thérapie Vectorisée, BP 10448, F-63005 Clermont-Ferrand Cedex, France
| | - Olivier Chavignon
- INSERM - Université d'Auvergne, UMR 990, IMTV, BP 184, F-63005 Clermont-Ferrand Cedex, France; Clermont Université, Université d'Auvergne, Imagerie Moléculaire et Thérapie Vectorisée, BP 10448, F-63005 Clermont-Ferrand Cedex, France
| | - Elisabeth Miot-Noirault
- INSERM - Université d'Auvergne, UMR 990, IMTV, BP 184, F-63005 Clermont-Ferrand Cedex, France; Clermont Université, Université d'Auvergne, Imagerie Moléculaire et Thérapie Vectorisée, BP 10448, F-63005 Clermont-Ferrand Cedex, France
| | - Emmanuel Moreau
- INSERM - Université d'Auvergne, UMR 990, IMTV, BP 184, F-63005 Clermont-Ferrand Cedex, France; Clermont Université, Université d'Auvergne, Imagerie Moléculaire et Thérapie Vectorisée, BP 10448, F-63005 Clermont-Ferrand Cedex, France.
| |
Collapse
|
31
|
Abstract
INTRODUCTION Utilizing the prodrug approach as a method to overcome various pharmaceutical and pharmacokinetic barriers to drug delivery is significantly accelerating and achieving successes. In contrast to the older traditional prodrugs which suffer from decreased bioavailability and a high profile of side effects, due to activation at undesired sites, the targeted prodrug approach utilizes delivery systems to improve delivery for a wide range of therapeutics including anti-cancer, anti-bacterial and anti-inflammatory drugs. AREAS COVERED Recent updates in utilization of prodrugs in drug delivery between 2013 and 2015 are discussed. Targeted prodrugs against cancer, solid tumors, microbial infections, inflammation and other diseases using advanced delivery systems such as theranostic approaches, siRNA, DOX immunoconjugate, C 60-ser carrier vector, biotinylated prodrug, human serum albumin (HSA) carrier and others are presented. EXPERT OPINION Recent research efforts have been directed at developing targeted prodrugs to replace the classical prodrugs. The use of this approach has accelerated following the emergence of encouraging results from several studies on targeted prodrugs that have highlighted their higher efficiency and improved safety profiles. Targeted prodrug delivery is now considered more than a chemical modification method. It is an applicable and promising approach and, in the future, better knowledge and wide application of this approach may be attained which may pave the way for more forward-thinking and creative techniques.
Collapse
Affiliation(s)
- Wajd Amly
- a Pharmaceutical Sciences Department, Faculty of Pharmacy , Al-Quds University , Jerusalem , Palestine , Israel
| | - Rafik Karaman
- a Pharmaceutical Sciences Department, Faculty of Pharmacy , Al-Quds University , Jerusalem , Palestine , Israel.,b Department of Sciences , University of Basilicata , Potenza , Italy
| |
Collapse
|
32
|
Nitroreductase gene-directed enzyme prodrug therapy: insights and advances toward clinical utility. Biochem J 2015; 471:131-53. [PMID: 26431849 DOI: 10.1042/bj20150650] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This review examines the vast catalytic and therapeutic potential offered by type I (i.e. oxygen-insensitive) nitroreductase enzymes in partnership with nitroaromatic prodrugs, with particular focus on gene-directed enzyme prodrug therapy (GDEPT; a form of cancer gene therapy). Important first indications of this potential were demonstrated over 20 years ago, for the enzyme-prodrug pairing of Escherichia coli NfsB and CB1954 [5-(aziridin-1-yl)-2,4-dinitrobenzamide]. However, it has become apparent that both the enzyme and the prodrug in this prototypical pairing have limitations that have impeded their clinical progression. Recently, substantial advances have been made in the biodiscovery and engineering of superior nitroreductase variants, in particular development of elegant high-throughput screening capabilities to enable optimization of desirable activities via directed evolution. These advances in enzymology have been paralleled by advances in medicinal chemistry, leading to the development of second- and third-generation nitroaromatic prodrugs that offer substantial advantages over CB1954 for nitroreductase GDEPT, including greater dose-potency and enhanced ability of the activated metabolite(s) to exhibit a local bystander effect. In addition to forging substantial progress towards future clinical trials, this research is supporting other fields, most notably the development and improvement of targeted cellular ablation capabilities in small animal models, such as zebrafish, to enable cell-specific physiology or regeneration studies.
Collapse
|
33
|
Hattinger CM, Serra M. Role of pharmacogenetics of drug-metabolizing enzymes in treating osteosarcoma. Expert Opin Drug Metab Toxicol 2015; 11:1449-63. [PMID: 26095223 DOI: 10.1517/17425255.2015.1060220] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Drug-metabolizing enzymes (DMEs) biotransform several toxins and xenobiotics in both tumor and normal cells, resulting in either their detoxification or their activation. Since DMEs also metabolize several chemotherapeutic drugs, they can significantly influence tumor response to chemotherapy and susceptibility of normal tissues to collateral toxicity of anticancer treatments. AREAS COVERED This review discusses the pharmacogenetics of DMEs involved in the metabolism of drugs which constitute the backbone of osteosarcoma (OS) chemotherapy, highlighting what is presently known for this tumor and their possible impact on the modulation of future treatment approaches. EXPERT OPINION Achieving further insight into pharmacogenetic markers and biological determinants related to treatment response in OS may ultimately lead to individualized treatment regimens, based on a combination of genotype and tumor characteristics of each patient.
Collapse
Affiliation(s)
- Claudia Maria Hattinger
- a Orthopaedic Rizzoli Institute, Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit , Via di Barbiano 1/10, I-40136 Bologna, Italy +390 516 366 762 ; +390 516 366 763 ;
| | | |
Collapse
|
34
|
Tian B, Wong WY, Hegmann E, Gaspar K, Kumar P, Chao H. Production and characterization of a camelid single domain antibody-urease enzyme conjugate for the treatment of cancer. Bioconjug Chem 2015; 26:1144-55. [PMID: 25938892 DOI: 10.1021/acs.bioconjchem.5b00237] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A novel immunoconjugate (L-DOS47) was developed and characterized as a therapeutic agent for tumors expressing CEACAM6. The single domain antibody AFAIKL2, which targets CEACAM6, was expressed in the Escherichia coli BL21 (DE3) pT7-7 system. High purity urease (HPU) was extracted and purified from Jack bean meal. AFAIKL2 was activated using N-succinimidyl [4-iodoacetyl] aminobenzoate (SIAB) as the cross-linker and then conjugated to urease. The activation and conjugation reactions were controlled by altering pH. Under these conditions, the material ratio achieved conjugation ratios of 8-11 antibodies per urease molecule, the residual free urease content was practically negligible (<2%), and high purity (>95%) L-DOS47 conjugate was produced using only ultradiafiltration to remove unreacted antibody and hydrolyzed cross-linker. L-DOS47 was characterized by a panel of analytical techniques including SEC, IEC, Western blot, ELISA, and LC-MS(E) peptide mapping. As the antibody-urease conjugate ratio increased, a higher binding signal was observed. The specificity and cytotoxicity of L-DOS47 was confirmed by screening in four cell lines (BxPC-3, A549, MCF7, and CEACAM6-transfected H23). BxPC-3, a CEACAM6-expressing cell line was found to be most susceptible to L-DOS47. L-DOS47 is being investigated as a potential therapeutic agent in human phase I clinical studies for nonsmall cell lung cancer.
Collapse
Affiliation(s)
- Baomin Tian
- †Helix BioPharma Corporation, 3-305 Industrial Parkway South, Aurora, Ontario L4G 6X7 Canada
| | - Wah Yau Wong
- †Helix BioPharma Corporation, 3-305 Industrial Parkway South, Aurora, Ontario L4G 6X7 Canada
| | - Elda Hegmann
- ‡Liquid Crystal Institute, Kent State University, Kent, Ohio 44242, United States
| | - Kim Gaspar
- †Helix BioPharma Corporation, 3-305 Industrial Parkway South, Aurora, Ontario L4G 6X7 Canada
| | - Praveen Kumar
- †Helix BioPharma Corporation, 3-305 Industrial Parkway South, Aurora, Ontario L4G 6X7 Canada
| | - Heman Chao
- †Helix BioPharma Corporation, 3-305 Industrial Parkway South, Aurora, Ontario L4G 6X7 Canada
| |
Collapse
|
35
|
Cereda V, Formica V, Menghi A, Pellicori S, Roselli M. Kallikrein-related peptidases targeted therapies in prostate cancer: perspectives and challenges. Expert Opin Investig Drugs 2015; 24:929-47. [PMID: 25858813 DOI: 10.1517/13543784.2015.1035708] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Despite the emergence of several new effective treatments for metastatic castration-resistant prostate cancer patients, disease progression inevitably occurs, leading scientific community to carefully look for novel therapeutic targets of prostate cancer. Kallikrein (KLK)-related peptidases have been demonstrated to facilitate prostate tumorigenesis and disease progression through the development of an oncogenic microenvironment for prostate cells. AREAS COVERED This review first summarizes the large amount of preclinical data showing the involvement of KLKs in prostate cancer pathobiology. In the second part, the authors assess the current status and future directions for KLK-targeted therapy and briefly describe the advances and challenges implicated in the design of effective manufactured drugs. The authors then focus on the preclinical data and on Phase I/II studies of the most promising KLK-targeted agents in prostate cancer. The drugs discussed here are divided on the basis of their mechanism of action: KLK-engineered inhibitors; KLK-activated pro-drugs; KLK-targeted microRNAs and small interfering RNAs(-/)small hairpin RNAs; KLK vaccines and antibodies. EXPERT OPINION Targeting KLK expression and/or activity could be a promising direction in prostate cancer treatment. Future human clinical trials will help us to evaluate the real benefits, toxicities and the consequent optimal use of KLK-targeted drugs, as mono-therapy or in combination regimens.
Collapse
Affiliation(s)
- Vittore Cereda
- 1 University of Rome Tor Vergata, Tor Vergata University Clinical Center, Department of Systems Medicine, Medical Oncology , Viale Oxford 81, 00133 Rome , Italy +39 0620908190 ; +39 0620903504 ;
| | | | | | | | | |
Collapse
|
36
|
Mavridis K, Avgeris M, Scorilas A. Targeting kallikrein-related peptidases in prostate cancer. Expert Opin Ther Targets 2014; 18:365-83. [PMID: 24571737 DOI: 10.1517/14728222.2014.880693] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
37
|
Wang ZC, Duan YT, Qiu HY, Huang WY, Wang PF, Yan XQ, Zhang SF, Zhu HL. Novel metronidazole-sulfonamide derivatives as potent and selective carbonic anhydrase inhibitors: design, synthesis and biology analysis. RSC Adv 2014. [DOI: 10.1039/c4ra03819c] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Metronidazole–sulfonamide derivatives, a new class of human carbonic anhydrase inhibitors (hCA), were designed, synthesized, isolated, and evaluated for their ability to inhibit the enzymatic activity of the isozymes hCA II and hCA IX.
Collapse
Affiliation(s)
- Zhong-Chang Wang
- State Key Laboratory of Pharmaceutical Biotechnology
- Nanjing University
- Nanjing 210093, China
| | - Yong-Tao Duan
- State Key Laboratory of Pharmaceutical Biotechnology
- Nanjing University
- Nanjing 210093, China
| | - Han-Yue Qiu
- State Key Laboratory of Pharmaceutical Biotechnology
- Nanjing University
- Nanjing 210093, China
| | - Wan-Yun Huang
- State Key Laboratory of Pharmaceutical Biotechnology
- Nanjing University
- Nanjing 210093, China
- Department of Pharmacology
- Guilin Medical University
| | - Peng-Fei Wang
- State Key Laboratory of Pharmaceutical Biotechnology
- Nanjing University
- Nanjing 210093, China
| | - Xiao-Qiang Yan
- State Key Laboratory of Pharmaceutical Biotechnology
- Nanjing University
- Nanjing 210093, China
| | - Shu-Feng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology
- Nanjing University
- Nanjing 210093, China
- College of Chemistry
- Tianjin Normal University
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology
- Nanjing University
- Nanjing 210093, China
| |
Collapse
|
38
|
Alford SC, O'Sullivan C, Obst J, Christie J, Howard PL. Conditional protein splicing of α-sarcin in live cells. MOLECULAR BIOSYSTEMS 2014; 10:831-7. [DOI: 10.1039/c3mb70387h] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
39
|
Ferrous iron-dependent drug delivery enables controlled and selective release of therapeutic agents in vivo. Proc Natl Acad Sci U S A 2013; 110:18244-9. [PMID: 24145449 DOI: 10.1073/pnas.1312782110] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The precise targeting of cytotoxic agents to specific cell types or cellular compartments is of significant interest in medicine, with particular relevance for infectious diseases and cancer. Here, we describe a method to exploit aberrant levels of mobile ferrous iron (Fe(II)) for selective drug delivery in vivo. This approach makes use of a 1,2,4-trioxolane moiety, which serves as an Fe(II)-sensitive "trigger," making drug release contingent on Fe(II)-promoted trioxolane fragmentation. We demonstrate in vivo validation of this approach with the Plasmodium berghei model of murine malaria. Malaria parasites produce high concentrations of mobile ferrous iron as a consequence of their catabolism of host hemoglobin in the infected erythrocyte. Using activity-based probes, we successfully demonstrate the Fe(II)-dependent and parasite-selective delivery of a potent dipeptidyl aminopeptidase inhibitor. We find that delivery of the compound in its Fe(II)-targeted form leads to more sustained target inhibition with greatly reduced off-target inhibition of mammalian cathepsins. This selective drug delivery translates into improved efficacy and tolerability. These findings demonstrate the utility of a purely chemical means to achieve selective drug targeting in vivo. This approach may find useful application in parasitic infections and more broadly in any disease state characterized by aberrant production of reactive ferrous iron.
Collapse
|
40
|
Wang ZC, Qin YJ, Wang PF, Yang YA, Wen Q, Zhang X, Qiu HY, Duan YT, Wang YT, Sang YL, Zhu HL. Sulfonamides containing coumarin moieties selectively and potently inhibit carbonic anhydrases II and IX: Design, synthesis, inhibitory activity and 3D-QSAR analysis. Eur J Med Chem 2013; 66:1-11. [DOI: 10.1016/j.ejmech.2013.04.035] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 04/13/2013] [Accepted: 04/15/2013] [Indexed: 11/27/2022]
|
41
|
Ibsen S, Su Y, Norton J, Zahavy E, Hayashi T, Adams S, Wrasidlo W, Esener S. Extraction protocol and mass spectrometry method for quantification of doxorubicin released locally from prodrugs in tumor tissue. JOURNAL OF MASS SPECTROMETRY : JMS 2013; 48:768-73. [PMID: 23832932 PMCID: PMC4110111 DOI: 10.1002/jms.3221] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 02/26/2013] [Accepted: 04/10/2013] [Indexed: 05/15/2023]
Abstract
The localized conversion of inactive doxorubicin prodrug chemotherapeutics to pharmacalogically active forms is difficult to quantify in mouse tumor models because it occurs only in small regions of tissue. The tumor tissue extraction protocol and LC-MS/MS analysis method described here were optimized to obtain a detection limit of 7.8 pg for the activated doxorubicin and 0.36 ng for the doxorubicin prodrug. This method can be useful for determining the biodistribution and activation efficiency for many different doxorubicin prodrugs. It can also be used for quantification of doxorubicin from tumor models that have poor vascularization resulting in low tissue accumulation.
Collapse
Affiliation(s)
- Stuart Ibsen
- Department of Bioengineering, Moores Cancer Center, University of California San Diego, 3855 Health Sciences Dr. # 0815, La Jolla, CA 92093-0815, USA.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Green LK, Syddall SP, Carlin KM, Bell GD, Guise CP, Mowday AM, Hay MP, Smaill JB, Patterson AV, Ackerley DF. Pseudomonas aeruginosa NfsB and nitro-CBI-DEI--a promising enzyme/prodrug combination for gene directed enzyme prodrug therapy. Mol Cancer 2013; 12:58. [PMID: 23758947 PMCID: PMC3695803 DOI: 10.1186/1476-4598-12-58] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 06/05/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The nitro-chloromethylbenzindoline prodrug nitro-CBI-DEI appears a promising candidate for the anti-cancer strategy gene-directed enzyme prodrug therapy, based on its ability to be converted to a highly cytotoxic cell-permeable derivative by the nitroreductase NfsB from Escherichia coli. However, relative to some other nitroaromatic prodrugs, nitro-CBI-DEI is a poor substrate for E. coli NfsB. To address this limitation we evaluated other nitroreductase candidates from E. coli and Pseudomonas aeruginosa. FINDINGS Initial screens of candidate genes in the E. coli reporter strain SOS-R2 identified two additional nitroreductases, E. coli NfsA and P. aeruginosa NfsB, as being more effective activators of nitro-CBI-DEI than E. coli NfsB. In monolayer cytotoxicity assays, human colon carcinoma (HCT-116) cells transfected with P. aeruginosa NfsB were >4.5-fold more sensitive to nitro-CBI-DEI than cells expressing either E. coli enzyme, and 23.5-fold more sensitive than untransfected HCT-116. In three dimensional mixed cell cultures, not only were the P. aeruginosa NfsB expressing cells 540-fold more sensitive to nitro-CBI-DEI than pure cultures of untransfected HCT-116, the activated drug that they generated also displayed an unprecedented local bystander effect. CONCLUSION We posit that the discrepancy in the fold-sensitivity to nitro-CBI-DEI between the two and three dimensional cytotoxicity assays stems from loss of activated drug into the media in the monolayer cultures. This emphasises the importance of evaluating high-bystander GDEPT prodrugs in three dimensional models. The high cytotoxicity and bystander effect exhibited by the NfsB_Pa/nitro-CBI-DEI combination suggest that further preclinical development of this GDEPT pairing is warranted.
Collapse
Affiliation(s)
- Laura K Green
- School of Biological Sciences, Victoria University of Wellington, Kelburn Parade, Wellington, New Zealand
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Wirth T, Pestel GF, Ganal V, Kirmeier T, Schuberth I, Rein T, Tietze PLF, Sieber PSA. The Two Faces of Potent Antitumor Duocarmycin-Based Drugs: A Structural Dissection Reveals Disparate Motifs for DNA versus Aldehyde Dehydrogenase 1 Affinity. Angew Chem Int Ed Engl 2013; 52:6921-5. [DOI: 10.1002/anie.201208941] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 03/27/2013] [Indexed: 01/15/2023]
|
44
|
Wirth T, Pestel GF, Ganal V, Kirmeier T, Schuberth I, Rein T, Tietze PLF, Sieber PSA. The Two Faces of Potent Antitumor Duocarmycin-Based Drugs: A Structural Dissection Reveals Disparate Motifs for DNA versus Aldehyde Dehydrogenase 1 Affinity. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201208941] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
45
|
Ibsen S, Zahavy E, Wrasidlo W, Hayashi T, Norton J, Su Y, Adams S, Esener S. Localized in vivo activation of a photoactivatable doxorubicin prodrug in deep tumor tissue. Photochem Photobiol 2013; 89:698-708. [PMID: 23311544 DOI: 10.1111/php.12045] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 01/05/2013] [Indexed: 01/21/2023]
Abstract
Sparing sensitive healthy tissue from chemotherapy exposure is a critical challenge in the treatment of cancer. The work described here demonstrates the localized in vivo photoactivation of a new chemotherapy prodrug of doxorubicin (DOX). The DOX prodrug (DOX-PCB) was 200 times less toxic than DOX and was designed to release pure DOX when exposed to 365 nm light. This wavelength was chosen because it had good tissue penetration through a 1 cm diameter tumor, but had very low skin penetration, due to melanin absorption, preventing uncontrolled activation from outside sources. The light was delivered specifically to the tumor tissue using a specialized fiber-optic LED system. Pharmacokinetic studies showed that DOX-PCB had an α circulation half-life of 10 min which was comparable to that of DOX at 20 min. DOX-PCB demonstrated resistance to metabolic cleavage ensuring that exposure to 365 nm light was the main mode of in vivo activation. Tissue extractions from tumors exposed to 365 nm light in vivo showed the presence of DOX-PCB as well as activated DOX. The exposed tumors had six times more DOX concentration than nearby unexposed control tumors. This in vivo proof of concept demonstrates the first preferential activation of a photocleavable prodrug in deep tumor tissue.
Collapse
Affiliation(s)
- Stuart Ibsen
- Department of Bioengineering, Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Mulet X, Boyd BJ, Drummond CJ. Advances in drug delivery and medical imaging using colloidal lyotropic liquid crystalline dispersions. J Colloid Interface Sci 2013; 393:1-20. [DOI: 10.1016/j.jcis.2012.10.014] [Citation(s) in RCA: 213] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 10/08/2012] [Accepted: 10/08/2012] [Indexed: 10/27/2022]
|
47
|
Tietze LF, Müller M, Duefert SC, Schmuck K, Schuberth I. Photoactivatable Prodrugs of Highly Potent Duocarmycin Analogues for a Selective Cancer Therapy. Chemistry 2012; 19:1726-31. [DOI: 10.1002/chem.201202773] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 10/24/2012] [Indexed: 12/11/2022]
|
48
|
Tietze LF, Behrendt F, Pestel GF, Schuberth I, Mitkovski M. Synthesis, biological evaluation, and live cell imaging of novel fluorescent duocarmycin analogs. Chem Biodivers 2012; 9:2559-70. [PMID: 23161634 DOI: 10.1002/cbdv.201200289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Indexed: 11/07/2022]
Abstract
For a better understanding of the mode of action of duocarmycin and its analogs, the novel fluorescent duocarmycin derivatives 13-15 and 17b-19b were synthesized, and their bioactivity as well as their cellular uptake investigated using confocal laser scanning microscopy (CLSM) in live-cell imaging experiments.
Collapse
Affiliation(s)
- Lutz F Tietze
- Georg-August-Universität Göttingen, Institut für Organische und Biomolekulare Chemie, Tammannstrasse 2, D-37077 Göttingen, Germany.
| | | | | | | | | |
Collapse
|
49
|
Alterio V, Di Fiore A, D'Ambrosio K, Supuran CT, De Simone G. Multiple binding modes of inhibitors to carbonic anhydrases: how to design specific drugs targeting 15 different isoforms? Chem Rev 2012; 112:4421-68. [PMID: 22607219 DOI: 10.1021/cr200176r] [Citation(s) in RCA: 977] [Impact Index Per Article: 75.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Vincenzo Alterio
- Istituto di Biostrutture e Bioimmagini-CNR, via Mezzocannone 16, 80134 Napoli, Italy
| | | | | | | | | |
Collapse
|
50
|
Crielaard BJ, van der Wal S, Lammers T, Le HT, Hennink WE, Schiffelers RM, Storm G, Fens MH. A polymeric colchicinoid prodrug with reduced toxicity and improved efficacy for vascular disruption in cancer therapy. Int J Nanomedicine 2011; 6:2697-703. [PMID: 22114500 PMCID: PMC3218583 DOI: 10.2147/ijn.s24450] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Colchicinoids are very potent tubulin-binding compounds, which interfere with microtubule formation, giving them strong cytotoxic properties, such as cell mitosis inhibition and induction of microcytoskeleton depolymerization. While this makes them promising vascular disrupting agents (VDAs) in cancer therapy, their dose-limiting toxicity has prevented any clinical application for this purpose. Therefore, colchicinoids are considered attractive lead molecules for the development of novel vascular disrupting nanomedicine. In a previous study, a polymeric colchicinoid prodrug that showed favorable hydrolysis characteristics at physiological conditions was developed. In the current study, this polymeric colchicinoid prodrug was evaluated in vitro and in vivo for its toxicity and vascular disrupting potential. Cell viability studies with human umbilical vein endothelial cells, as an in vitro measure for colchicine activity, reflected the degradation kinetics of the prodrug accordingly. Upon intravenous treatment, in vivo, of B16F10 melanoma-bearing mice with colchicine or with the polymeric colchicinoid prodrug, apparent vascular disruption and consequent tumor necrosis was observed for the prodrug but not for free colchicine at an equivalent dose. Moreover, a five-times-higher dose of the prodrug was well tolerated, indicating reduced toxicity. These findings demonstrate that the polymeric colchicinoid prodrug has a substantially improved efficacy/toxicity ratio compared with that of colchicine, making it a promising VDA for cancer therapy.
Collapse
Affiliation(s)
- Bart J Crielaard
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|